1
|
Yousefi F, Foster LA, Selim OA, Zhao C. Integrating Physical and Biochemical Cues for Muscle Engineering: Scaffolds and Graft Durability. Bioengineering (Basel) 2024; 11:1245. [PMID: 39768063 PMCID: PMC11673930 DOI: 10.3390/bioengineering11121245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Muscle stem cells (MuSCs) are essential for skeletal muscle regeneration, influenced by a complex interplay of mechanical, biochemical, and molecular cues. Properties of the extracellular matrix (ECM) such as stiffness and alignment guide stem cell fate through mechanosensitive pathways, where forces like shear stress translate into biochemical signals, affecting cell behavior. Aging introduces senescence which disrupts the MuSC niche, leading to reduced regenerative capacity via epigenetic alterations and metabolic shifts. Transplantation further challenges MuSC viability, often resulting in fibrosis driven by dysregulated fibro-adipogenic progenitors (FAPs). Addressing these issues, scaffold designs integrated with pharmacotherapy emulate ECM environments, providing cues that enhance graft functionality and endurance. These scaffolds facilitate the synergy between mechanotransduction and intracellular signaling, optimizing MuSC proliferation and differentiation. Innovations utilizing human pluripotent stem cell-derived myogenic progenitors and exosome-mediated delivery exploit bioactive properties for targeted repair. Additionally, 3D-printed and electrospun scaffolds with adjustable biomechanical traits tackle scalability in treating volumetric muscle loss. Advanced techniques like single-cell RNA sequencing and high-resolution imaging unravel muscle repair mechanisms, offering precise mapping of cellular interactions. Collectively, this interdisciplinary approach fortifies tissue graft durability and MuSC maintenance, propelling therapeutic strategies for muscle injuries and degenerative diseases.
Collapse
Affiliation(s)
- Farbod Yousefi
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| | - Lauren Ann Foster
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
- Atlanta Veterans Affairs Medical Center, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Omar A. Selim
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| | - Chunfeng Zhao
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| |
Collapse
|
2
|
Gahlawat S, Oruc D, Paul N, Ragheb M, Patel S, Fasasi O, Sharma P, Shreiber DI, Freeman JW. Tissue Engineered 3D Constructs for Volumetric Muscle Loss. Ann Biomed Eng 2024; 52:2325-2347. [PMID: 39085677 PMCID: PMC11329418 DOI: 10.1007/s10439-024-03541-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/08/2024] [Indexed: 08/02/2024]
Abstract
Severe injuries to skeletal muscles, including cases of volumetric muscle loss (VML), are linked to substantial tissue damage, resulting in functional impairment and lasting disability. While skeletal muscle can regenerate following minor damage, extensive tissue loss in VML disrupts the natural regenerative capacity of the affected muscle tissue. Existing clinical approaches for VML, such as soft-tissue reconstruction and advanced bracing methods, need to be revised to restore tissue function and are associated with limitations in tissue availability and donor-site complications. Advancements in tissue engineering (TE), particularly in scaffold design and the delivery of cells and growth factors, show promising potential for regenerating damaged skeletal muscle tissue and restoring function. This article provides a brief overview of the pathophysiology of VML and critiques the shortcomings of current treatments. The subsequent section focuses on the criteria for designing TE scaffolds, offering insights into various natural and synthetic biomaterials and cell types for effectively regenerating skeletal muscle. We also review multiple TE strategies involving both acellular and cellular scaffolds to encourage the development and maturation of muscle tissue and facilitate integration, vascularization, and innervation. Finally, the article explores technical challenges hindering successful translation into clinical applications.
Collapse
Affiliation(s)
- Sonal Gahlawat
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Doga Oruc
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Nikhil Paul
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Mark Ragheb
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Swati Patel
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Oyinkansola Fasasi
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Peeyush Sharma
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - David I Shreiber
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Joseph W Freeman
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA.
| |
Collapse
|
3
|
Ege D, Lu HH, Boccaccini AR. Bioactive Glass and Silica Particles for Skeletal and Cardiac Muscle Tissue Regeneration. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:448-461. [PMID: 38126329 DOI: 10.1089/ten.teb.2023.0277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
When skeletal and cardiac tissues are damaged, surgical approaches are not always successful and tissue regeneration approaches are investigated. Reports in the literature indicate that silica nanoparticles and bioactive glasses (BGs), including silicate bioactive glasses (e.g., 45S5 BG), phosphate glass fibers, boron-doped mesoporous BGs, borosilicate glasses, and aluminoborates, are promising for repairing skeletal muscle tissue. Silica nanoparticles and BGs have been combined with polymers to obtain aligned nanofibers and to maintain controlled delivery of nanoparticles for skeletal muscle repair. The literature indicates that cardiac muscle regeneration can be also triggered by the ionic products of BGs. This was observed to be due to the release of vascular endothelial growth factor and other growth factors from cardiomyocytes, which regulate endothelial cells to form capillary structures (angiogenesis). Specific studies, including both in vitro and in vivo approaches, are reviewed in this article. The analysis of the literature indicates that although the research field is still very limited, BGs are showing great promise for muscle tissue engineering and further research in the field should be carried out to expand our basic knowledge on the application of BGs in muscle (skeletal and cardiac) tissue regeneration. Impact statement This review highlights the potential of silica particles and bioactive glasses (BGs) for skeletal and cardiac tissue regeneration. These biomaterials create scaffolds triggering muscle cell differentiation. Ionic products from BGs stimulate growth factors, supporting angiogenesis in cardiac tissue repair. Further research is required to expand our know-how on silica particles and BGs in muscle tissue engineering.
Collapse
Affiliation(s)
- Duygu Ege
- Institute of Biomedical Engineering, Bogazici University, Istanbul, Turkey
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Hsuan-Heng Lu
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Aldo R Boccaccini
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
4
|
Snow F, O'Connell C, Yang P, Kita M, Pirogova E, Williams RJ, Kapsa RMI, Quigley A. Engineering interfacial tissues: The myotendinous junction. APL Bioeng 2024; 8:021505. [PMID: 38841690 PMCID: PMC11151436 DOI: 10.1063/5.0189221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 05/06/2024] [Indexed: 06/07/2024] Open
Abstract
The myotendinous junction (MTJ) is the interface connecting skeletal muscle and tendon tissues. This specialized region represents the bridge that facilitates the transmission of contractile forces from muscle to tendon, and ultimately the skeletal system for the creation of movement. MTJs are, therefore, subject to high stress concentrations, rendering them susceptible to severe, life-altering injuries. Despite the scarcity of knowledge obtained from MTJ formation during embryogenesis, several attempts have been made to engineer this complex interfacial tissue. These attempts, however, fail to achieve the level of maturity and mechanical complexity required for in vivo transplantation. This review summarizes the strategies taken to engineer the MTJ, with an emphasis on how transitioning from static to mechanically inducive dynamic cultures may assist in achieving myotendinous maturity.
Collapse
|
5
|
Sun Y, Sheng R, Cao Z, Liu C, Li J, Zhang P, Du Y, Mo Q, Yao Q, Chen J, Zhang W. Bioactive fiber-reinforced hydrogel to tailor cell microenvironment for structural and functional regeneration of myotendinous junction. SCIENCE ADVANCES 2024; 10:eadm7164. [PMID: 38657071 PMCID: PMC11042749 DOI: 10.1126/sciadv.adm7164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 03/18/2024] [Indexed: 04/26/2024]
Abstract
Myotendinous junction (MTJ) injuries are prevalent in clinical practice, yet the treatment approaches are limited to surgical suturing and conservative therapy, exhibiting a high recurrence rate. Current research on MTJ tissue engineering is scarce and lacks in vivo evaluation of repair efficacy. Here, we developed a three-dimensional-printed bioactive fiber-reinforced hydrogel containing mesenchymal stem cells (MSCs) and Klotho for structural and functional MTJ regeneration. In a rat MTJ defect model, the bioactive fiber-reinforced hydrogel promoted the structural restoration of muscle, tendon, and muscle-tendon interface and enhanced the functional recovery of injured MTJ. In vivo proteomics and in vitro cell cultures elucidated the regenerative mechanisms of the bioactive fiber-reinforced hydrogel by modulating oxidative stress and inflammation, thus engineering an optimized microenvironment to support the survival and differentiation of transplanted MSCs and maintain the functional phenotype of resident cells within MTJ tissues, including tendon/muscle cells and macrophages. This strategy provides a promising treatment for MTJ injuries.
Collapse
Affiliation(s)
- Yuzhi Sun
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, China
| | - Renwang Sheng
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Zhicheng Cao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, China
| | - Chuanquan Liu
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Jiaxiang Li
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, China
| | - Po Zhang
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, China
| | - Yan Du
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Qingyun Mo
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310000 Hangzhou, China
| | - Jialin Chen
- School of Medicine, Southeast University, 210009 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310000 Hangzhou, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China
| | - Wei Zhang
- School of Medicine, Southeast University, 210009 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310000 Hangzhou, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China
| |
Collapse
|
6
|
Tong S, Sun Y, Kuang B, Wang M, Chen Z, Zhang W, Chen J. A Comprehensive Review of Muscle-Tendon Junction: Structure, Function, Injury and Repair. Biomedicines 2024; 12:423. [PMID: 38398025 PMCID: PMC10886980 DOI: 10.3390/biomedicines12020423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/31/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
The muscle-tendon junction (MTJ) is a highly specific tissue interface where the muscle's fascia intersects with the extracellular matrix of the tendon. The MTJ functions as the particular structure facilitating the transmission of force from contractive muscle fibers to the skeletal system, enabling movement. Considering that the MTJ is continuously exposed to constant mechanical forces during physical activity, it is susceptible to injuries. Ruptures at the MTJ often accompany damage to both tendon and muscle tissues. In this review, we attempt to provide a precise definition of the MTJ, describe its subtle structure in detail, and introduce therapeutic approaches related to MTJ tissue engineering. We hope that our detailed illustration of the MTJ and summary of the representative research achievements will help researchers gain a deeper understanding of the MTJ and inspire fresh insights and breakthroughs for future research.
Collapse
Affiliation(s)
- Siqi Tong
- School of Medicine, Southeast University, Nanjing 210009, China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing 210009, China
| | - Yuzhi Sun
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing 210009, China
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Baian Kuang
- School of Medicine, Southeast University, Nanjing 210009, China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing 210009, China
| | - Mingyue Wang
- School of Medicine, Southeast University, Nanjing 210009, China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing 210009, China
| | - Zhixuan Chen
- School of Medicine, Southeast University, Nanjing 210009, China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing 210009, China
| | - Wei Zhang
- School of Medicine, Southeast University, Nanjing 210009, China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing 210009, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing 210096, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou 310058, China
| | - Jialin Chen
- School of Medicine, Southeast University, Nanjing 210009, China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing 210009, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing 210096, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou 310058, China
| |
Collapse
|
7
|
Kang MS, Yu Y, Park R, Heo HJ, Lee SH, Hong SW, Kim YH, Han DW. Highly Aligned Ternary Nanofiber Matrices Loaded with MXene Expedite Regeneration of Volumetric Muscle Loss. NANO-MICRO LETTERS 2024; 16:73. [PMID: 38175358 PMCID: PMC10767178 DOI: 10.1007/s40820-023-01293-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/16/2023] [Indexed: 01/05/2024]
Abstract
Current therapeutic approaches for volumetric muscle loss (VML) face challenges due to limited graft availability and insufficient bioactivities. To overcome these limitations, tissue-engineered scaffolds have emerged as a promising alternative. In this study, we developed aligned ternary nanofibrous matrices comprised of poly(lactide-co-ε-caprolactone) integrated with collagen and Ti3C2Tx MXene nanoparticles (NPs) (PCM matrices), and explored their myogenic potential for skeletal muscle tissue regeneration. The PCM matrices demonstrated favorable physicochemical properties, including structural uniformity, alignment, microporosity, and hydrophilicity. In vitro assays revealed that the PCM matrices promoted cellular behaviors and myogenic differentiation of C2C12 myoblasts. Moreover, in vivo experiments demonstrated enhanced muscle remodeling and recovery in mice treated with PCM matrices following VML injury. Mechanistic insights from next-generation sequencing revealed that MXene NPs facilitated protein and ion availability within PCM matrices, leading to elevated intracellular Ca2+ levels in myoblasts through the activation of inducible nitric oxide synthase (iNOS) and serum/glucocorticoid regulated kinase 1 (SGK1), ultimately promoting myogenic differentiation via the mTOR-AKT pathway. Additionally, upregulated iNOS and increased NO- contributed to myoblast proliferation and fiber fusion, thereby facilitating overall myoblast maturation. These findings underscore the potential of MXene NPs loaded within highly aligned matrices as therapeutic agents to promote skeletal muscle tissue recovery.
Collapse
Affiliation(s)
- Moon Sung Kang
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea
| | - Yeuni Yu
- Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Rowoon Park
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea
| | - Hye Jin Heo
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Seok Hyun Lee
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea
- Osstem Implant Inc., Seoul, 07789, Republic of Korea
| | - Suck Won Hong
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea.
- Engineering Research Center for Color‑Modulated Extra‑Sensory Perception Technology, Pusan National University, Busan, 46241, Republic of Korea.
| | - Yun Hak Kim
- Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea.
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea.
- Periodontal Disease Signaling Network Research Center and Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea.
| | - Dong-Wook Han
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea.
- BIO-IT Fusion Technology Research Institute, Pusan National University, Busan, 46241, Republic of Korea.
| |
Collapse
|
8
|
Sato H, Kohyama K, Uchibori T, Takanari K, Huard J, Badylak SF, D'Amore A, Wagner WR. Creating and Transferring an Innervated, Vascularized Muscle Flap Made from an Elastic, Cellularized Tissue Construct Developed In Situ. Adv Healthc Mater 2023; 12:e2301335. [PMID: 37499214 DOI: 10.1002/adhm.202301335] [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: 04/26/2023] [Revised: 07/21/2023] [Indexed: 07/29/2023]
Abstract
Reanimating facial structures following paralysis and muscle loss is a surgical objective that would benefit from improved options for harvesting appropriately sized muscle flaps. The objective of this study is to apply electrohydrodynamic processing to generate a cellularized, elastic, biocomposite scaffold that could develop and mature as muscle in a prepared donor site in vivo, and then be transferred as a thin muscle flap with a vascular and neural pedicle. First, an effective extracellular matrix (ECM) gel type is selected for the biocomposite scaffold from three types of ECM combined with poly(ester urethane)urea microfibers and evaluated in rat abdominal wall defects. Next, two types of precursor cells (muscle-derived and adipose-derived) are compared in constructs placed in rat hind limb defects for muscle regeneration capacity. Finally, with a construct made from dermal ECM and muscle-derived stem cells, protoflaps are implanted in one hindlimb for development and then microsurgically transferred as a free flap to the contralateral limb where stimulated muscle function is confirmed. This construct generation and in vivo incubation procedure may allow the generation of small-scale muscle flaps appropriate for transfer to the face, offering a new strategy for facial reanimation.
Collapse
Affiliation(s)
- Hideyoshi Sato
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Dr., Pittsburgh, PA, 15219, USA
| | - Keishi Kohyama
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Dr., Pittsburgh, PA, 15219, USA
| | - Takafumi Uchibori
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Dr., Pittsburgh, PA, 15219, USA
| | - Keisuke Takanari
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Dr., Pittsburgh, PA, 15219, USA
| | - Johnny Huard
- Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, 181 West Meadow Dr., Vail, CO, 81657, USA
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Dr., Pittsburgh, PA, 15219, USA
- Department of Surgery, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA, 15213, USA
- Department of Bioengineering, University of Pittsburgh, 3700 O'Hara Street, Benedum Hall of Engineering, Pittsburgh, PA, 15261, USA
| | - Antonio D'Amore
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Dr., Pittsburgh, PA, 15219, USA
- Department of Surgery, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA, 15213, USA
- Department of Bioengineering, University of Pittsburgh, 3700 O'Hara Street, Benedum Hall of Engineering, Pittsburgh, PA, 15261, USA
- Fondazione Ri.MED, Palermo, 90133, Italy
| | - William R Wagner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Dr., Pittsburgh, PA, 15219, USA
- Department of Surgery, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA, 15213, USA
- Department of Bioengineering, University of Pittsburgh, 3700 O'Hara Street, Benedum Hall of Engineering, Pittsburgh, PA, 15261, USA
- Department of Chemical Engineering, University of Pittsburgh, 3700 O'Hara Street, Benedum Hall of Engineering, Pittsburgh, PA, 15261, USA
| |
Collapse
|
9
|
Martinello T, Akyürek EE, Ventriglia G, Patruno M, Sacchetto R. The dorsal portion of the bovine diaphragm as a useful tissue for producing a 3D muscle scaffold. J Anat 2023; 243:878-885. [PMID: 37322832 PMCID: PMC10557388 DOI: 10.1111/joa.13915] [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: 02/07/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 06/17/2023] Open
Abstract
Three-dimensional (3D) organoids are an innovative approach to obtain an in vitro model for ex vivo studies to overcome the limitations of monolayer cell culture and reduce the use of animal models. An organoid of skeletal muscle requires the presence of the extracellular matrix to represent a functional muscle in vitro, which is why decellularized tissue is an optimal choice. Various muscles have been considered to produce a muscle organoid, most from rodents or small animals, and only recently some studies have been reported on the muscles of large animals. This work presents a muscular organoid produced from the bovine diaphragm, which has a peculiar multilayered structure with different fibre orientations depending on the considered area. This paper analyses the anatomical structure of the bovine diaphragm, selects the most appropriate portion, and presents a decellularization protocol for a multilayered muscle. In addition, a preliminary test of recellularization with primary bovine myocytes was presented with the future aim of obtaining a 3D muscle allogenic organoid, completely bovine-derived. The results demonstrate that the dorsal portion of bovine diaphragm presents a regular alternation of muscular and fibrous layers and that the complete decellularization does not affect the biocompatibility. These results provide a strong foundation for the potential application of this portion of tissue as a scaffold for in vitro studies of muscle organoids.
Collapse
Affiliation(s)
| | - Eylem Emek Akyürek
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy
| | | | - Marco Patruno
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy
| | - Roberta Sacchetto
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy
| |
Collapse
|
10
|
Song Z, Yang D, Hu Q, Wang Y, Zhang H, Dong W, Yang J, Gu Y. Reconstruction of Abdominal Wall Defect with Composite Scaffold of 3D Printed ADM/PLA in a Rat Model. Macromol Biosci 2023; 23:e2200521. [PMID: 36746773 DOI: 10.1002/mabi.202200521] [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: 11/29/2022] [Revised: 01/15/2023] [Indexed: 02/08/2023]
Abstract
Abdominal wall defects are a frequently occurring condition in surgical practice. The most important are material structure and biocompatibility. In this study, PLA mesh composited with a 3D printing of acellular dermal matrix (ADM) material is used to repair abdominal wall defects. The results show that the adhesion score of ADM/PLA composite scaffolds is smaller than PLA meshes. Immunohistochemical assessment reveals that the ADM/PLA composite scaffold can effectively reduce the inflammatory response at the contact surface between the meshes and the abdominal organs. And the ADM/PLA composite scaffold can effectively reduce the expression levels of the inflammation-related factors IL-6 and IL-10. In addition, the ADM/PLA composite scaffold repair is rich in the expression levels of tissue regeneration-related factors vascular endothelial growth factor and transforming growth factor β. Thus, ADM/PLA composite scaffolds can effectively reduce surrounding inflammation to effectively promote the repair of abdominal wall defects.
Collapse
Affiliation(s)
- Zhicheng Song
- General Surgery, Huadong Hospital Affiliated to Fudan University, 221 Yan 'an West Road, Jing 'an District, Shanghai, Shanghai, 200040, China
| | - Dongchao Yang
- General Surgery, Huadong Hospital Affiliated to Fudan University, 221 Yan 'an West Road, Jing 'an District, Shanghai, Shanghai, 200040, China
| | - Qingxi Hu
- Rapid Manufacturing Engineering Center, Shanghai University, 99 Shangda Road, Baoshan District, Shanghai, Shanghai, 200444, China
| | - Yiming Wang
- Administrative office, Tenth People's Hospital of Tongji University, 301 Yanchang Zhong Lu, Jing 'an District, Shanghai, Shanghai, 200072, China
| | - Haiguang Zhang
- Rapid Manufacturing Engineering Center, Shanghai University, 99 Shangda Road, Baoshan District, Shanghai, Shanghai, 200444, China
| | - Wenpei Dong
- General Surgery, Huadong Hospital Affiliated to Fudan University, 221 Yan 'an West Road, Jing 'an District, Shanghai, Shanghai, 200040, China
| | - Jianjun Yang
- General Surgery, Huadong Hospital Affiliated to Fudan University, 221 Yan 'an West Road, Jing 'an District, Shanghai, Shanghai, 200040, China
| | - Yan Gu
- General Surgery, Huadong Hospital Affiliated to Fudan University, 221 Yan 'an West Road, Jing 'an District, Shanghai, Shanghai, 200040, China
| |
Collapse
|
11
|
Behre A, Tashman JW, Dikyol C, Shiwarski DJ, Crum RJ, Johnson SA, Kommeri R, Hussey GS, Badylak SF, Feinberg AW. 3D Bioprinted Patient-Specific Extracellular Matrix Scaffolds for Soft Tissue Defects. Adv Healthc Mater 2022; 11:e2200866. [PMID: 36063047 PMCID: PMC9780169 DOI: 10.1002/adhm.202200866] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 08/02/2022] [Indexed: 01/28/2023]
Abstract
Soft tissue injuries such as volumetric muscle loss (VML) are often too large to heal normally on their own, resulting in scar formation and functional deficits. Decellularized extracellular matrix (dECM) scaffolds placed into these wounds have shown the ability to modulate the immune response and drive constructive healing. This provides a potential solution for functional tissue regeneration, however, these acellular dECM scaffolds are challenging to fabricate into complex geometries. 3D bioprinting is uniquely positioned to address this, being able to create patient-specific scaffolds based on clinical 3D imaging data. Here, a process to use freeform reversible embedding of suspended hydrogels (FRESH) 3D bioprinting and computed tomography (CT) imaging to build large volume, patient-specific dECM patches (≈12 × 8 × 2 cm) for implantation into canine VML wound models is developed. Quantitative analysis shows that these dECM patches are dimensionally accurate and conformally adapt to the surface of complex wounds. Finally, this approach is extended to a human VML injury to demonstrate the fabrication of clinically relevant dECM scaffolds with precise control over fiber alignment and micro-architecture. Together these advancements represent a step towards an improved, clinically translatable, patient-specific treatment for soft tissue defects from trauma, tumor resection, and other surgical procedures.
Collapse
Affiliation(s)
- Anne Behre
- Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPA15213USA
| | - Joshua W. Tashman
- Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPA15213USA
| | - Caner Dikyol
- Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPA15213USA
| | - Daniel J. Shiwarski
- Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPA15213USA
| | - Raphael J. Crum
- McGowan Institute for Regenerative MedicineUniversity of PittsburghPittsburghPA15219USA
| | - Scott A. Johnson
- McGowan Institute for Regenerative MedicineUniversity of PittsburghPittsburghPA15219USA
| | - Remya Kommeri
- McGowan Institute for Regenerative MedicineUniversity of PittsburghPittsburghPA15219USA
| | - George S. Hussey
- McGowan Institute for Regenerative MedicineUniversity of PittsburghPittsburghPA15219USA
| | - Stephen F. Badylak
- McGowan Institute for Regenerative MedicineUniversity of PittsburghPittsburghPA15219USA
| | - Adam W. Feinberg
- Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPA15213USA
- McGowan Institute for Regenerative MedicineUniversity of PittsburghPittsburghPA15219USA
- Department of Materials Science & EngineeringCarnegie Mellon UniversityPittsburghPA15213USA
| |
Collapse
|
12
|
McInnes AD, Moser MAJ, Chen X. Preparation and Use of Decellularized Extracellular Matrix for Tissue Engineering. J Funct Biomater 2022; 13:jfb13040240. [PMID: 36412881 PMCID: PMC9680265 DOI: 10.3390/jfb13040240] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/22/2022] [Accepted: 11/05/2022] [Indexed: 11/16/2022] Open
Abstract
The multidisciplinary fields of tissue engineering and regenerative medicine have the potential to revolutionize the practise of medicine through the abilities to repair, regenerate, or replace tissues and organs with functional engineered constructs. To this end, tissue engineering combines scaffolding materials with cells and biologically active molecules into constructs with the appropriate structures and properties for tissue/organ regeneration, where scaffolding materials and biomolecules are the keys to mimic the native extracellular matrix (ECM). For this, one emerging way is to decellularize the native ECM into the materials suitable for, directly or in combination with other materials, creating functional constructs. Over the past decade, decellularized ECM (or dECM) has greatly facilitated the advance of tissue engineering and regenerative medicine, while being challenged in many ways. This article reviews the recent development of dECM for tissue engineering and regenerative medicine, with a focus on the preparation of dECM along with its influence on cell culture, the modification of dECM for use as a scaffolding material, and the novel techniques and emerging trends in processing dECM into functional constructs. We highlight the success of dECM and constructs in the in vitro, in vivo, and clinical applications and further identify the key issues and challenges involved, along with a discussion of future research directions.
Collapse
Affiliation(s)
- Adam D. McInnes
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
- Correspondence: ; Tel.: +1-306-966-5435
| | - Michael A. J. Moser
- Department of Surgery, Health Sciences Building, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| |
Collapse
|
13
|
Philips C, Terrie L, Thorrez L. Decellularized skeletal muscle: A versatile biomaterial in tissue engineering and regenerative medicine. Biomaterials 2022; 283:121436. [DOI: 10.1016/j.biomaterials.2022.121436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/27/2022] [Accepted: 02/17/2022] [Indexed: 12/31/2022]
|
14
|
Romagnoli C, Iantomasi T, Brandi ML. Available In Vitro Models for Human Satellite Cells from Skeletal Muscle. Int J Mol Sci 2021; 22:ijms222413221. [PMID: 34948017 PMCID: PMC8706222 DOI: 10.3390/ijms222413221] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle accounts for almost 40% of the total adult human body mass. This tissue is essential for structural and mechanical functions such as posture, locomotion, and breathing, and it is endowed with an extraordinary ability to adapt to physiological changes associated with growth and physical exercise, as well as tissue damage. Moreover, skeletal muscle is the most age-sensitive tissue in mammals. Due to aging, but also to several diseases, muscle wasting occurs with a loss of muscle mass and functionality, resulting from disuse atrophy and defective muscle regeneration, associated with dysfunction of satellite cells, which are the cells responsible for maintaining and repairing adult muscle. The most established cell lines commonly used to study muscle homeostasis come from rodents, but there is a need to study skeletal muscle using human models, which, due to ethical implications, consist primarily of in vitro culture, which is the only alternative way to vertebrate model organisms. This review will survey in vitro 2D/3D models of human satellite cells to assess skeletal muscle biology for pre-clinical investigations and future directions.
Collapse
Affiliation(s)
- Cecilia Romagnoli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (C.R.); (T.I.)
| | - Teresa Iantomasi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (C.R.); (T.I.)
| | - Maria Luisa Brandi
- F.I.R.M.O. Italian Foundation for the Research on Bone Diseases, Via Reginaldo Giuliani 195/A, 50141 Florence, Italy
- Correspondence:
| |
Collapse
|
15
|
Sheng H, Guo Y, Zhang L, Zhang J, Miao M, Tan H, Hu D, Li X, Ding X, Li G, Guo H. Proteomic Studies on the Mechanism of Myostatin Regulating Cattle Skeletal Muscle Development. Front Genet 2021; 12:752129. [PMID: 34868225 PMCID: PMC8635237 DOI: 10.3389/fgene.2021.752129] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/28/2021] [Indexed: 11/25/2022] Open
Abstract
Myostatin (MSTN) is an important negative regulator of muscle growth and development. In this study, we performed comparatively the proteomics analyses of gluteus tissues from MSTN+/− Mongolian cattle (MG.MSTN+/−) and wild type Mongolian cattle (MG.WT) using a shotgun-based tandem mass tag (TMT) 6-plex labeling method to investigate the regulation mechanism of MSTN on the growth and development of bovine skeletal muscle. A total of 1,950 proteins were identified in MG.MSTN+/− and MG.WT. Compared with MG.WT cattle, a total of 320 differentially expressed proteins were identified in MG.MSTN cattle, including 245 up-regulated differentially expressed proteins and 75 down-regulated differentially expressed proteins. Bioinformatics analysis showed that knockdown of the MSTN gene increased the expression of extracellular matrix and ribosome-related proteins, induced activation of focal adhesion, PI3K-AKT, and Ribosomal pathways. The results of proteomic analysis were verified by muscle tissue Western blot test and in vitro MSTN gene knockdown test, and it was found that knockdown MSTN gene expression could promote the proliferation and myogenic differentiation of bovine skeletal muscle satellite cells (BSMSCs). At the same time, Co-Immunoprecipitation (CO-IP) assay showed that MSTN gene interacted with extracellular matrix related protein type I collagen α 1 (COL1A1), and knocking down the expression of COL1A1 could inhibit the activity of adhesion, PI3K-AKT and ribosome pathway, thus inhibit BSMSCs proliferation. These results suggest that the MSTN gene regulates focal adhesion, PI3K-AKT, and Ribosomal pathway through the COL1A1 gene. In general, this study provides new insights into the regulatory mechanism of MSTN involved in muscle growth and development.
Collapse
Affiliation(s)
- Hui Sheng
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Yiwen Guo
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Linlin Zhang
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Junxing Zhang
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Manning Miao
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Haoyun Tan
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Debao Hu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Xin Li
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Xiangbin Ding
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Guangpeng Li
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
| | - Hong Guo
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| |
Collapse
|
16
|
Eugenis I, Wu D, Rando TA. Cells, scaffolds, and bioactive factors: Engineering strategies for improving regeneration following volumetric muscle loss. Biomaterials 2021; 278:121173. [PMID: 34619561 PMCID: PMC8556323 DOI: 10.1016/j.biomaterials.2021.121173] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/01/2021] [Accepted: 08/14/2021] [Indexed: 12/20/2022]
Abstract
Severe traumatic skeletal muscle injuries, such as volumetric muscle loss (VML), result in the obliteration of large amounts of skeletal muscle and lead to permanent functional impairment. Current clinical treatments are limited in their capacity to regenerate damaged muscle and restore tissue function, promoting the need for novel muscle regeneration strategies. Advances in tissue engineering, including cell therapy, scaffold design, and bioactive factor delivery, are promising solutions for VML therapy. Herein, we review tissue engineering strategies for regeneration of skeletal muscle, development of vasculature and nerve within the damaged muscle, and achievements in immunomodulation following VML. In addition, we discuss the limitations of current state of the art technologies and perspectives of tissue-engineered bioconstructs for muscle regeneration and functional recovery following VML.
Collapse
Affiliation(s)
- Ioannis Eugenis
- Department of Bioengineering, Stanford University, Stanford, CA, USA; Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Di Wu
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA; Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
17
|
Carleton MM, Sefton MV. Promoting endogenous repair of skeletal muscle using regenerative biomaterials. J Biomed Mater Res A 2021; 109:2720-2739. [PMID: 34041836 DOI: 10.1002/jbm.a.37239] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/14/2021] [Accepted: 05/18/2021] [Indexed: 02/06/2023]
Abstract
Skeletal muscles normally have a remarkable ability to repair themselves; however, large muscle injuries and several myopathies diminish this ability leading to permanent loss of function. No clinical therapy yet exists that reliably restores muscle integrity and function following severe injury. Consequently, numerous tissue engineering techniques, both acellular and with cells, are being investigated to enhance muscle regeneration. Biomaterials are an essential part of these techniques as they can present physical and biochemical signals that augment the repair process. Successful tissue engineering strategies require regenerative biomaterials that either actively promote endogenous muscle repair or create an environment supportive of regeneration. This review will discuss several acellular biomaterial strategies for skeletal muscle regeneration with a focus on those under investigation in vivo. This includes materials that release bioactive molecules, biomimetic materials and immunomodulatory materials.
Collapse
Affiliation(s)
- Miranda M Carleton
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Michael V Sefton
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
18
|
Dessauge F, Schleder C, Perruchot MH, Rouger K. 3D in vitro models of skeletal muscle: myopshere, myobundle and bioprinted muscle construct. Vet Res 2021; 52:72. [PMID: 34011392 PMCID: PMC8136231 DOI: 10.1186/s13567-021-00942-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/15/2021] [Indexed: 12/25/2022] Open
Abstract
Typical two-dimensional (2D) culture models of skeletal muscle-derived cells cannot fully recapitulate the organization and function of living muscle tissues, restricting their usefulness in in-depth physiological studies. The development of functional 3D culture models offers a major opportunity to mimic the living tissues and to model muscle diseases. In this respect, this new type of in vitro model significantly increases our understanding of the involvement of the different cell types present in the formation of skeletal muscle and their interactions, as well as the modalities of response of a pathological muscle to new therapies. This second point could lead to the identification of effective treatments. Here, we report the significant progresses that have been made the last years to engineer muscle tissue-like structures, providing useful tools to investigate the behavior of resident cells. Specifically, we interest in the development of myopshere- and myobundle-based systems as well as the bioprinting constructs. The electrical/mechanical stimulation protocols and the co-culture systems developed to improve tissue maturation process and functionalities are presented. The formation of these biomimetic engineered muscle tissues represents a new platform to study skeletal muscle function and spatial organization in large number of physiological and pathological contexts.
Collapse
|
19
|
Mihaly E, Altamirano DE, Tuffaha S, Grayson W. Engineering skeletal muscle: Building complexity to achieve functionality. Semin Cell Dev Biol 2021; 119:61-69. [PMID: 33994095 DOI: 10.1016/j.semcdb.2021.04.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022]
Abstract
Volumetric muscle loss (VML) VML is defined as the loss of a critical mass of skeletal muscle that overwhelms the muscle's natural healing mechanisms, leaving patients with permanent functional deficits and deformity. The treatment of these defects is complex, as skeletal muscle is a composite structure that relies closely on the action of supporting tissues such as tendons, vasculature, nerves, and bone. The gold standard of treatment for VML injuries, an autologous muscle flap transfer, suffers from many shortcomings but nevertheless remains the best clinically available avenue to restore function. This review will consider the use of composite tissue engineered constructs, with multiple components that act together to replicate the function of an intact muscle, as an alternative to autologous muscle flaps. We will discuss recent advances in the field of tissue engineering that enable skeletal muscle constructs to more closely reproduce the functionality of an autologous muscle flap by incorporating vasculature, promoting innervation, and reconstructing the muscle-tendon boundary. Additionally, our understanding of the cellular composition of skeletal muscle has evolved to recognize the importance of a diverse variety of cell types in muscle regeneration, including fibro/adipogenic progenitors and immune cells like macrophages and regulatory T cells. We will address recent advances in our understanding of how these cell types interact with, and can be incorporated into, implanted tissue engineered constructs.
Collapse
Affiliation(s)
- Eszter Mihaly
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Dallas E Altamirano
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Sami Tuffaha
- Department of Plastic and Reconstructive Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; Curtis National Hand Center, MedStar Union Memorial Hospital, Baltimore, MD 21218, USA
| | - Warren Grayson
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Materials Science & Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Chemical & Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for NanoBioTechnology (INBT), Johns Hopkins University School of Engineering, Baltimore, MD 21218, USA.
| |
Collapse
|
20
|
Promoting musculoskeletal system soft tissue regeneration by biomaterial-mediated modulation of macrophage polarization. Bioact Mater 2021; 6:4096-4109. [PMID: 33997496 PMCID: PMC8091177 DOI: 10.1016/j.bioactmat.2021.04.017] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/27/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023] Open
Abstract
Musculoskeletal disorders are common in clinical practice. Repairing critical-sized defects in musculoskeletal systems remains a challenge for researchers and surgeons, requiring the application of tissue engineering biomaterials. Successful application depends on the response of the host tissue to the biomaterial and specific healing process of each anatomical structure. The commonly-held view is that biomaterials should be biocompatible to minimize local host immune response. However, a growing number of studies have shown that active modulation of the immune cells, particularly macrophages, via biomaterials is an effective way to control immune response and promote tissue regeneration as well as biomaterial integration. Therefore, we critically review the role of macrophages in the repair of injured musculoskeletal system soft tissues, which have relatively poor regenerative capacities, as well as discuss further enhancement of target tissue regeneration via modulation of macrophage polarization by biomaterial-mediated immunomodulation (biomaterial properties and delivery systems). This active regulation approach rather than passive-evade strategy maximizes the potential of biomaterials to promote musculoskeletal system soft tissue regeneration and provides alternative therapeutic options for repairing critical-sized defects. Different phenotypes of macrophages play a crucial role in musculoskeletal system soft tissue regeneration. Biomaterials and biomaterial-based delivery systems can be utilized to modulate macrophage polarization. This review summarizes immunomodulatory biomaterials to spur musculoskeletal system soft tissue regeneration.
Collapse
|
21
|
Wang D, Zhang X, Huang S, Liu Y, Fu BSC, Mak KKL, Blocki AM, Yung PSH, Tuan RS, Ker DFE. Engineering multi-tissue units for regenerative Medicine: Bone-tendon-muscle units of the rotator cuff. Biomaterials 2021; 272:120789. [PMID: 33845368 DOI: 10.1016/j.biomaterials.2021.120789] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 12/13/2022]
Abstract
Our body systems are comprised of numerous multi-tissue units. For the musculoskeletal system, one of the predominant functional units is comprised of bone, tendon/ligament, and muscle tissues working in tandem to facilitate locomotion. To successfully treat musculoskeletal injuries and diseases, critical consideration and thoughtful integration of clinical, biological, and engineering aspects are necessary to achieve translational bench-to-bedside research. In particular, identifying ideal biomaterial design specifications, understanding prior and recent tissue engineering advances, and judicious application of biomaterial and fabrication technologies will be crucial for addressing current clinical challenges in engineering multi-tissue units. Using rotator cuff tears as an example, insights relevant for engineering a bone-tendon-muscle multi-tissue unit are presented. This review highlights the tissue engineering strategies for musculoskeletal repair and regeneration with implications for other bone-tendon-muscle units, their derivatives, and analogous non-musculoskeletal tissue structures.
Collapse
Affiliation(s)
- Dan Wang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Xu Zhang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR
| | - Shuting Huang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR
| | - Yang Liu
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Bruma Sai-Chuen Fu
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | | | - Anna Maria Blocki
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Patrick Shu-Hang Yung
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Rocky S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR
| | - Dai Fei Elmer Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR.
| |
Collapse
|
22
|
Dienes J, Browne S, Farjun B, Amaral Passipieri J, Mintz EL, Killian G, Healy KE, Christ GJ. Semisynthetic Hyaluronic Acid-Based Hydrogel Promotes Recovery of the Injured Tibialis Anterior Skeletal Muscle Form and Function. ACS Biomater Sci Eng 2021; 7:1587-1599. [PMID: 33660968 DOI: 10.1021/acsbiomaterials.0c01751] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Volumetric muscle loss (VML) injuries are characterized by a degree of tissue loss that exceeds the endogenous regenerative capacity of muscle, resulting in permanent structural and functional deficits. Such injuries are a consequence of trauma, as well as a host of congenital and acquired diseases and disorders. Despite significant preclinical research with diverse biomaterials, as well as early clinical studies with implantation of decellularized extracellular matrices, there are still significant barriers to more complete restoration of muscle form and function following repair of VML injuries. In fact, identification of novel biomaterials with more advantageous regenerative profiles is a critical limitation to the development of improved therapeutics. As a first step in this direction, we evaluated a novel semisynthetic hyaluronic acid-based (HyA) hydrogel that embodies material features more favorable for robust muscle regeneration. This HyA-based hydrogel is composed of an acrylate-modified HyA (AcHyA) macromer, an AcHyA macromer conjugated with the bsp-RGD(15) peptide sequence to enhance cell adhesion, a high-molecular-weight heparin to sequester growth factors, and a matrix metalloproteinase-cleavable cross-linker to allow for cell-dependent remodeling. In a well-established, clinically relevant rat tibialis anterior VML injury model, we report observations of robust functional recovery, accompanied by volume reconstitution, muscle regeneration, and native-like vascularization following implantation of the HyA-based hydrogel at the site of injury. These findings have important implications for the development and clinical application of the improved biomaterials that will be required for stable and complete functional recovery from diverse VML injuries.
Collapse
Affiliation(s)
- Jack Dienes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Shane Browne
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, United States.,Department of Material Science and Engineering, University of California, Berkeley, Berkeley 94720, United States
| | - Bruna Farjun
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Juliana Amaral Passipieri
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Ellen L Mintz
- Pathology Department, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Grant Killian
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Kevin E Healy
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, United States.,Department of Material Science and Engineering, University of California, Berkeley, Berkeley 94720, United States
| | - George J Christ
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, United States.,Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia 22908, United States
| |
Collapse
|
23
|
Awad K, Ahuja N, Fiedler M, Peper S, Wang Z, Aswath P, Brotto M, Varanasi V. Ionic Silicon Protects Oxidative Damage and Promotes Skeletal Muscle Cell Regeneration. Int J Mol Sci 2021; 22:E497. [PMID: 33419056 PMCID: PMC7825403 DOI: 10.3390/ijms22020497] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 12/20/2022] Open
Abstract
Volumetric muscle loss injuries overwhelm the endogenous regenerative capacity of skeletal muscle, and the associated oxidative damage can delay regeneration and prolong recovery. This study aimed to investigate the effect of silicon-ions on C2C12 skeletal muscle cells under normal and excessive oxidative stress conditions to gain insights into its role on myogenesis during the early stages of muscle regeneration. In vitro studies indicated that 0.1 mM Si-ions into cell culture media significantly increased cell viability, proliferation, migration, and myotube formation compared to control. Additionally, MyoG, MyoD, Neurturin, and GABA expression were significantly increased with addition of 0.1, 0.5, and 1.0 mM of Si-ion for 1 and 5 days of C2C12 myoblast differentiation. Furthermore, 0.1-2.0 mM Si-ions attenuated the toxic effects of H2O2 within 24 h resulting in increased cell viability and differentiation. Addition of 1.0 mM of Si-ions significantly aid cell recovery and protected from the toxic effect of 0.4 mM H2O2 on cell migration. These results suggest that ionic silicon may have a potential effect in unfavorable situations where reactive oxygen species is predominant affecting cell viability, proliferation, migration, and differentiation. Furthermore, this study provides a guide for designing Si-containing biomaterials with desirable Si-ion release for skeletal muscle regeneration.
Collapse
Affiliation(s)
- Kamal Awad
- Department of Materials Science and Engineering, College of Engineering, University of Texas at Arlington, Arlington, TX 76019, USA; (K.A.); (P.A.)
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA; (N.A.); (M.F.); (S.P.); (Z.W.)
| | - Neelam Ahuja
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA; (N.A.); (M.F.); (S.P.); (Z.W.)
| | - Matthew Fiedler
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA; (N.A.); (M.F.); (S.P.); (Z.W.)
| | - Sara Peper
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA; (N.A.); (M.F.); (S.P.); (Z.W.)
- Department of Bioengineering, College of Engineering, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Zhiying Wang
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA; (N.A.); (M.F.); (S.P.); (Z.W.)
| | - Pranesh Aswath
- Department of Materials Science and Engineering, College of Engineering, University of Texas at Arlington, Arlington, TX 76019, USA; (K.A.); (P.A.)
| | - Marco Brotto
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA; (N.A.); (M.F.); (S.P.); (Z.W.)
| | - Venu Varanasi
- Department of Materials Science and Engineering, College of Engineering, University of Texas at Arlington, Arlington, TX 76019, USA; (K.A.); (P.A.)
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA; (N.A.); (M.F.); (S.P.); (Z.W.)
| |
Collapse
|
24
|
Allogeneic Decellularized Muscle Scaffold Is Less Fibrogenic and Inflammatory than Acellular Dermal Matrices in a Rat Model of Skeletal Muscle Regeneration. Plast Reconstr Surg 2020; 146:43e-53e. [PMID: 32590650 DOI: 10.1097/prs.0000000000006922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND Skeletal muscle trauma can produce grave functional deficits, but therapeutic options remain limited. The authors studied whether a decellularized skeletal muscle scaffold would provide benefits in inducing skeletal muscle regeneration over acellular dermal matrices. METHODS Eighty-two rat muscle defects were surgically created and assigned to no intervention or implantation of AlloDerm, Strattice, decellularized rat muscle, or decellularized rat dermis to 30 or 60 days. Decellularized rat muscle and dermis were prepared using a negative pressure-assisted protocol. Assessment for cellularity, neovascularization, myogenesis, inflammation and fibrosis were done histologically and by polymerase chain reaction. RESULTS Histology showed relative hypercellularity of AlloDerm (p < 0.003); Strattice appeared encapsulated. Immunofluorescence for CD31 and myosin heavy chain in decellularized rat muscle revealed dense microvasculature and peripheral islands of myogenesis. MyoD expression in muscle scaffolds was 23-fold higher than in controls (p < 0.01). Decellularized rat muscle showed no up-regulation of COX-2 (p < 0.05), with less expression than decellularized rat dermis and Strattice (p < 0.002). Decellularized rat muscle scaffolds expressed tumor necrosis factor-α less than Strattice, AlloDerm, and decellularized rat dermis (p < 0.01); collagen-1a less than decellularized rat dermis and Strattice (p < 0.04); α-smooth muscle actin 7-fold less than AlloDerm (p = 0.04); and connective tissue growth factor less than Strattice, AlloDerm, and decellularized rat dermis (p < 0.02). CONCLUSION Decellularized muscle matrix appears to reduce inflammation and fibrosis in an animal muscle defect as compared with dermal matrices and promotes greater expression of myocyte differentiation-inducing genes.
Collapse
|
25
|
Skeletal Muscle Tissue Engineering: Biomaterials-Based Strategies for the Treatment of Volumetric Muscle Loss. Bioengineering (Basel) 2020; 7:bioengineering7030085. [PMID: 32751847 PMCID: PMC7552659 DOI: 10.3390/bioengineering7030085] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/17/2020] [Accepted: 07/28/2020] [Indexed: 12/13/2022] Open
Abstract
Millions of Americans suffer from skeletal muscle injuries annually that can result in volumetric muscle loss (VML), where extensive musculoskeletal damage and tissue loss result in permanent functional deficits. In the case of small-scale injury skeletal muscle is capable of endogenous regeneration through activation of resident satellite cells (SCs). However, this is greatly reduced in VML injuries, which remove native biophysical and biochemical signaling cues and hinder the damaged tissue's ability to direct regeneration. The current clinical treatment for VML is autologous tissue transfer, but graft failure and scar tissue formation leave patients with limited functional recovery. Tissue engineering of instructive biomaterial scaffolds offers a promising approach for treating VML injuries. Herein, we review the strategic engineering of biophysical and biochemical cues in current scaffold designs that aid in restoring function to these preclinical VML injuries. We also discuss the successes and limitations of the three main biomaterial-based strategies to treat VML injuries: acellular scaffolds, cell-delivery scaffolds, and in vitro tissue engineered constructs. Finally, we examine several innovative approaches to enhancing the design of the next generation of engineered scaffolds to improve the functional regeneration of skeletal muscle following VML injuries.
Collapse
|
26
|
Judson RN, Rossi FMV. Towards stem cell therapies for skeletal muscle repair. NPJ Regen Med 2020; 5:10. [PMID: 32411395 PMCID: PMC7214464 DOI: 10.1038/s41536-020-0094-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 03/31/2020] [Indexed: 01/23/2023] Open
Abstract
Skeletal muscle is an ideal target for cell therapy. The use of its potent stem cell population in the form of autologous intramuscular transplantation represents a tantalizing strategy to slow the progression of congenital muscle diseases (such as Duchenne Muscular Dystrophy) or regenerate injured tissue following trauma. The syncytial nature of skeletal muscle uniquely permits the engraftment of stem/progenitor cells to contribute to new myonuclei and restore the expression of genes mutated in myopathies. Historically however, the implementation of this approach has been significantly limited by the inability to expand undifferentiated muscle stem cells (MuSCs) in culture whilst maintaining transplantation potential. This is crucial, as MuSC expansion and/or genetic manipulation is likely necessary for therapeutic applications. In this article, we review recent studies that have provided a number of important breakthroughs to tackle this problem. Progress towards this goal has been achieved by exploiting biochemical, biophysical and developmental paradigms to construct innovative in vitro strategies that are guiding stem cell therapies for muscle repair towards the clinic.
Collapse
Affiliation(s)
- Robert N Judson
- 1STEMCELL Technologies Inc, Vancouver, BC Canada.,2Biomedical Research Centre, Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC Canada
| | - Fabio M V Rossi
- 2Biomedical Research Centre, Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC Canada
| |
Collapse
|
27
|
Bour RK, Sharma PR, Turner JS, Hess WE, Mintz EL, Latvis CR, Shepherd BR, Presnell SC, McConnell MJ, Highley C, Peirce SM, Christ GJ. Bioprinting on sheet-based scaffolds applied to the creation of implantable tissue-engineered constructs with potentially diverse clinical applications: Tissue-Engineered Muscle Repair (TEMR) as a representative testbed. Connect Tissue Res 2020; 61:216-228. [PMID: 31899969 DOI: 10.1080/03008207.2019.1679800] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose: This report explores the overlooked potential of bioprinting to automate biomanufacturing of simple tissue structures, such as the uniform deposition of (mono)layers of progenitor cells on sheetlike decellularized extracellular matrices (dECM). In this scenario, dECM serves as a biodegradable celldelivery matrix to provide enhanced regenerative microenvironments for tissue repair. The Tissue-Engineered Muscle Repair (TEMR) technology-where muscle progenitor cells are seeded onto a porcine bladder acellular matrix (BAM), serves as a representative testbed for bioprinting applications. Previous work demonstrated that TEMR implantation improved functional outcomes following VML injury in biologically relevant rodent models.Materials and Methods: In the described bioprinting system, a cell-laden hydrogel bioink is used to deposit high cell densities (1.4 × 105-3.5 × 105 cells/cm2), onto both sides of the bladder acellular matrix as proof-of-concept.Results: These bioprinting methods achieve a reproducible and homogeneous distribution of cells, on both sides of the BAM scaffold, after just 24hrs, with cell viability as high as 98%. These preliminary results suggest bioprinting allows for improved dual-sided cell coverage compared to manual-seeding.Conclusions: Bioprinting can enable automated fabrication of TEMR constructs with high fidelity and scalability, while reducing biomanufacturing costs and timelines. Such bioprinting applications are underappreciated, yet critical, to expand the overall biomanufacturing paradigm for tissue engineered medical products. In addition, biofabrication of sheet-like implantable constructs, with cells deposited on both sides, is a process that is both scaffold and cell-type agnostic, and furthermore, is amenable to many geometries, and thus, additional tissue engineering applications beyond skeletal muscle.
Collapse
Affiliation(s)
- R K Bour
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - P R Sharma
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - J S Turner
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - W E Hess
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - E L Mintz
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - C R Latvis
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | | | | | - M J McConnell
- Departments of Biochemistry and Molecular Genetics, and Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - C Highley
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA.,Department of Chemical Engineering, University of Virginia, Charlottesville, VA, USA
| | - S M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA.,Department of Plastic Surgery, University of Virginia, Charlottesville, VA, USA
| | - G J Christ
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA.,Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
28
|
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: 101] [Impact Index Per Article: 20.2] [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
|
29
|
Panayi AC, Smit L, Hays N, Udeh K, Endo Y, Li B, Sakthivel D, Tamayol A, Neppl RL, Orgill DP, Nuutila K, Sinha I. A porous collagen-GAG scaffold promotes muscle regeneration following volumetric muscle loss injury. Wound Repair Regen 2020; 28:61-74. [PMID: 31603580 DOI: 10.1111/wrr.12768] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 09/03/2019] [Accepted: 09/20/2019] [Indexed: 12/13/2022]
Abstract
Volumetric muscle loss (VML) is a segmental loss of skeletal muscle which commonly heals with fibrosis, minimal muscle regeneration, and loss of muscle strength. Treatment options for these wounds which promote functional recovery are currently lacking. This study was designed to investigate whether the collagen-GAG scaffold (CGS) promotes functional muscle recovery following VML. A total of 66 C57/Bl6 mice were used in a three-stage experiment. First, 24 animals were split into three groups which underwent sham injury or unilateral quadriceps VML injury with or without CGS implantation. Two weeks post-surgery, muscle was harvested for histological and gene expression analysis. In the second stage, 18 mice underwent bilateral quadriceps VML injury, followed by weekly functional testing using a treadmill. In the third stage, 24 mice underwent sham or bilateral quadriceps VML injury with or without CGS implantation, with tissue harvested six weeks post-surgery for histological and gene expression analysis. VML mice treated with CGS demonstrated increased remnant fiber hypertrophy versus both the VML with no CGS and uninjured groups. Both VML groups showed greater muscle fiber hypertrophy than non-injured muscle. This phenomenon was still evident in the longer-term experiment. The gene array indicated that the CGS promoted upregulation of factors involved in promoting wound healing and regeneration. In terms of functional improvement, the VML mice treated with CGS ran at higher maximum speeds than VML without CGS. A CGS was shown to enhance muscle hypertrophy in response to VML injury with a resultant improvement in functional performance. A gene array highlighted increased gene expression of multiple growth factors following CGS implantation. This suggests that implantation of a CGS could be a promising treatment for VML wounds.
Collapse
Affiliation(s)
- Adriana C Panayi
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Lucindi Smit
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Nicole Hays
- University of Maryland School of Medicine, Baltimore, Maryland
| | - Kodi Udeh
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Yori Endo
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Bin Li
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Dharaniya Sakthivel
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Ali Tamayol
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Nebraska
| | - Ronald L Neppl
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Dennis P Orgill
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Kristo Nuutila
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Indranil Sinha
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
30
|
Murdock MH, David S, Swinehart IT, Reing JE, Tran K, Gassei K, Orwig KE, Badylak SF. Human Testis Extracellular Matrix Enhances Human Spermatogonial Stem Cell Survival In Vitro. Tissue Eng Part A 2019; 25:663-676. [PMID: 30311859 DOI: 10.1089/ten.tea.2018.0147] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
IMPACT STATEMENT This study developed and characterized human testis extracellular matrix (htECM) and porcine testis ECM (ptECM) for testing in human spermatogonial stem cell (hSSC) culture. Results confirmed the hypothesis that ECM from the homologous species (human) and homologous tissue (testis) is optimal for maintaining hSSCs. We describe a simplified feeder-free, serum-free condition for future iterative testing to achieve the long-term goal of stable hSSC cultures. To facilitate analysis and understand the fate of hSSCs in culture, we describe a multiparameter, high-throughput, quantitative flow cytometry approach to rapidly count undifferentiated spermatogonia, differentiated spermatogonia, apoptotic spermatogonia, and proliferative spermatogonia in hSSC cultures.
Collapse
Affiliation(s)
- Mark H Murdock
- 1 McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sherin David
- 2 Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Women's Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ilea T Swinehart
- 1 McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Janet E Reing
- 1 McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kien Tran
- 2 Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Women's Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kathrin Gassei
- 2 Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Women's Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kyle E Orwig
- 2 Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Women's Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Stephen F Badylak
- 1 McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- 3 Department of Surgery, and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- 4 Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
31
|
Marcinczyk M, Dunn A, Haas G, Madsen J, Scheidt R, Patel K, Talovic M, Garg K. The Effect of Laminin-111 Hydrogels on Muscle Regeneration in a Murine Model of Injury. Tissue Eng Part A 2019; 25:1001-1012. [PMID: 30426851 PMCID: PMC9839345 DOI: 10.1089/ten.tea.2018.0200] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
IMPACT STATEMENT Extremity injuries make up the most common survivable injuries in vehicular accidents and modern military conflicts. A majority of these injuries involve volumetric muscle loss (VML). The potential for donor site morbidity may limit the clinical use of autologous muscle grafts for VML injuries. Treatments that can improve the regeneration of functional muscle tissue are critically needed to improve limb salvage and reduce the rate of delayed amputations. The development of a laminin-111-enriched fibrin hydrogel will offer a potentially transformative and "off-the-shelf" clinically relevant therapy for functional skeletal muscle regeneration.
Collapse
Affiliation(s)
- Madison Marcinczyk
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Andrew Dunn
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Gabriel Haas
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Josh Madsen
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Robert Scheidt
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Krishna Patel
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Muhamed Talovic
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Koyal Garg
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri.,Address correspondence to: Koyal Garg, PhD, Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, 3507 Lindell Boulevard, St. Louis, MO 63103
| |
Collapse
|
32
|
Dunn A, Talovic M, Patel K, Patel A, Marcinczyk M, Garg K. Biomaterial and stem cell-based strategies for skeletal muscle regeneration. J Orthop Res 2019; 37:1246-1262. [PMID: 30604468 DOI: 10.1002/jor.24212] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 12/13/2018] [Indexed: 02/04/2023]
Abstract
Adult skeletal muscle can regenerate effectively after mild physical or chemical insult. Muscle trauma or disease can overwhelm this innate capacity for regeneration and result in heightened inflammation and fibrotic tissue deposition resulting in loss of structure and function. Recent studies have focused on biomaterial and stem cell-based therapies to promote skeletal muscle regeneration following injury and disease. Many stem cell populations besides satellite cells are implicated in muscle regeneration. These stem cells include but are not limited to mesenchymal stem cells, adipose-derived stem cells, hematopoietic stem cells, pericytes, fibroadipogenic progenitors, side population cells, and CD133+ stem cells. However, several challenges associated with their isolation, availability, delivery, survival, engraftment, and differentiation have been reported in recent studies. While acellular scaffolds offer a relatively safe and potentially off-the-shelf solution to cell-based therapies, they are often unable to stimulate host cell migration and activity to a level that would result in clinically meaningful regeneration of traumatized muscle. Combining stem cells and biomaterials may offer a viable therapeutic strategy that may overcome the limitations associated with these therapies when they are used in isolation. In this article, we review the stem cell populations that can stimulate muscle regeneration in vitro and in vivo. We also discuss the regenerative potential of combination therapies that utilize both stem cell and biomaterials for the treatment of skeletal muscle injury and disease. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1246-1262, 2019.
Collapse
Affiliation(s)
- Andrew Dunn
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Muhamed Talovic
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Krishna Patel
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Anjali Patel
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Madison Marcinczyk
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Koyal Garg
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| |
Collapse
|
33
|
Trevisan C, Fallas MEA, Maghin E, Franzin C, Pavan P, Caccin P, Chiavegato A, Carraro E, Boso D, Boldrin F, Caicci F, Bertin E, Urbani L, Milan A, Biz C, Lazzari L, De Coppi P, Pozzobon M, Piccoli M. Generation of a Functioning and Self-Renewing Diaphragmatic Muscle Construct. Stem Cells Transl Med 2019; 8:858-869. [PMID: 30972959 PMCID: PMC6646700 DOI: 10.1002/sctm.18-0206] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 03/04/2019] [Indexed: 12/19/2022] Open
Abstract
Surgical repair of large muscular defects requires the use of autologous graft transfer or prosthetic material. Naturally derived matrices are biocompatible materials obtained by tissue decellularization and are commonly used in clinical practice. Despite promising applications described in the literature, the use of acellular matrices to repair large defects has been only partially successful, highlighting the need for more efficient constructs. Scaffold recellularization by means of tissue engineering may improve not only the structure of the matrix, but also its ability to functionally interact with the host. The development of such a complex construct is challenging, due to the complexity of the native organ architecture and the difficulties in recreating the cellular niche with both proliferative and differentiating potential during growth or after damage. In this study, we tested a mouse decellularized diaphragmatic extracellular matrix (ECM) previously described by our group, for the generation of a cellular skeletal muscle construct with functional features. The decellularized matrix was stored using different conditions to mimic the off‐the‐shelf clinical need. Pediatric human muscle precursors were seeded into the decellularized scaffold, demonstrating proliferation and differentiation capability, giving rise to a functioning three‐dimensional skeletal muscle structure. Furthermore, we exposed the engineered construct to cardiotoxin injury and demonstrated its ability to activate a regenerative response in vitro promoting cell self‐renewal and a positive ECM remodeling. Functional reconstruction of an engineered skeletal muscle with maintenance of a stem cell pool makes this a promising tool toward future clinical applications in diaphragmatic regeneration. stem cells translational medicine2019;8:858&869
Collapse
Affiliation(s)
- Caterina Trevisan
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy.,Department of Women and Children Health, University of Padova, Padova, Italy
| | - Mario Enrique Alvrez Fallas
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy.,Department of Women and Children Health, University of Padova, Padova, Italy
| | - Edoardo Maghin
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy.,Department of Women and Children Health, University of Padova, Padova, Italy
| | - Chiara Franzin
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Piero Pavan
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy.,Department of Industrial Engineering, University of Padova, Padova, Italy.,Centre for Mechanics of Biological Materials, University of Padova, Padova, Italy
| | - Paola Caccin
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Angela Chiavegato
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,CNR Institute for Neuroscience, Padova, Italy
| | - Eugenia Carraro
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Daniele Boso
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | | | | | - Enrica Bertin
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Luca Urbani
- Stem Cells & Regenerative Medicine Section, Developmental Biology & Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,Institute of Hepatology, The Foundation for Liver Research, London, United Kingdom.,Faculty of Life Sciences & Medicine, King's College, London, United Kingdom
| | - Anna Milan
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy.,Department of Women and Children Health, University of Padova, Padova, Italy
| | - Carlo Biz
- Department of Surgery, Oncology, and Gastroenterology DiSCOG, Orthopaedic Clinic, University of Padova, Padua, Italy
| | - Lorenza Lazzari
- Laboratory of Regenerative Medicine - Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Paolo De Coppi
- Stem Cells & Regenerative Medicine Section, Developmental Biology & Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,Specialist Neonatal and Paediatric Surgery, Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Michela Pozzobon
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy.,Department of Women and Children Health, University of Padova, Padova, Italy
| | - Martina Piccoli
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy.,Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
34
|
Tang X, Daneshmandi L, Awale G, Nair LS, Laurencin CT. Skeletal Muscle Regenerative Engineering. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2019; 5:233-251. [PMID: 33778155 DOI: 10.1007/s40883-019-00102-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Skeletal muscles have the intrinsic ability to regenerate after minor injury, but under certain circumstances such as severe trauma from accidents, chronic diseases or battlefield injuries the regeneration process is limited. Skeletal muscle regenerative engineering has emerged as a promising approach to address this clinical issue. The regenerative engineering approach involves the convergence of advanced materials science, stem cell science, physical forces, insights from developmental biology, and clinical translation. This article reviews recent studies showing the potential of the convergences of technologies involving biomaterials, stem cells and bioactive factors in concert with clinical translation, in promoting skeletal muscle regeneration. Several types of biomaterials such as electrospun nanofibers, hydrogels, patterned scaffolds, decellularized tissues, and conductive matrices are being investigated. Detailed discussions are given on how these biomaterials can interact with cells and modulate their behavior through physical, chemical and mechanical cues. In addition, the application of physical forces such as mechanical and electrical stimulation are reviewed as strategies that can further enhance muscle contractility and functionality. The review also discusses established animal models to evaluate regeneration in two clinically relevant muscle injuries; volumetric muscle loss (VML) and muscle atrophy upon rotator cuff injury. Regenerative engineering approaches using advanced biomaterials, cells, and physical forces, developmental cues along with insights from immunology, genetics and other aspects of clinical translation hold significant potential to develop promising strategies to support skeletal muscle regeneration.
Collapse
Affiliation(s)
- Xiaoyan Tang
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA.,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA.,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA.,Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Leila Daneshmandi
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA.,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA.,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA.,Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Guleid Awale
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA.,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA.,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA.,Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Lakshmi S Nair
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA.,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA.,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA.,Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA.,Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Cato T Laurencin
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA.,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA.,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA.,Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA.,Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA.,Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
35
|
Nakayama KH, Shayan M, Huang NF. Engineering Biomimetic Materials for Skeletal Muscle Repair and Regeneration. Adv Healthc Mater 2019; 8:e1801168. [PMID: 30725530 PMCID: PMC6589032 DOI: 10.1002/adhm.201801168] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/21/2018] [Indexed: 11/12/2022]
Abstract
Although skeletal muscle is highly regenerative following injury or disease, endogenous self-regeneration is severely impaired in conditions of volume traumatic muscle loss. Consequently, tissue engineering approaches are a promising means to regenerate skeletal muscle. Biological scaffolds serve as not only structural support for the promotion of cellular ingrowth but also impart potent modulatory signaling cues that may be beneficial for tissue regeneration. In this work, the progress of tissue engineering approaches for skeletal muscle engineering and regeneration is overviewed, with a focus on the techniques to create biomimetic engineered tissue using extracellular cues. These factors include mechanical and electrical stimulation, geometric patterning, and delivery of growth factors or other bioactive molecules. The progress of evaluating the therapeutic efficacy of these approaches in preclinical models of muscle injury is further discussed.
Collapse
Affiliation(s)
- Karina H Nakayama
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Veterans Affairs Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| | - Mahdis Shayan
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Veterans Affairs Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| | - Ngan F Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Veterans Affairs Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
36
|
Wang Z, Li Z, Li Z, Wu B, Liu Y, Wu W. Cartilaginous extracellular matrix derived from decellularized chondrocyte sheets for the reconstruction of osteochondral defects in rabbits. Acta Biomater 2018; 81:129-145. [PMID: 30300711 DOI: 10.1016/j.actbio.2018.10.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/20/2018] [Accepted: 10/04/2018] [Indexed: 12/28/2022]
Abstract
Cartilaginous extracellular matrix (ECM) materials derived from decellularized native articular cartilage are widely used in cartilage regeneration. However, it is difficult for endogenous cells to migrate into ECM derived from native cartilage owing to its nonporous structure and dense nature. Moreover, current decellularization approaches frequently lead to architectural breakdown and potential loss of surface composition of ECM. To solve this problem, we aimed to establish a novel biological ECM scaffold from chondrocyte sheets for cartilage regeneration. We cultured chondrocytes harvested from the auricular cartilage of 4-week-old New Zealand rabbits and enabled them to form cell sheets. These sheets were decellularized using sodium dodecyl sulfate (SDS) with three different concentrations, namely, 1%, 5%, and 10%, followed by 1% Triton X-100 and deoxyribonuclease enzyme solution. In vitro microstructural examination and mechanical tests demonstrated that 1% SDS not only removed chondrocytes completely but also maintained the native architecture and composition of ECM, thus avoiding the use of high-concentration SDS. Application of decellularized chondrocyte sheets for osteochondral defects in rabbits resulted in substantial host remodeling and variant regeneration of osteochondral tissues. One percent SDS-treated decellularized chondrocyte sheets contributed to the superior reconstruction of osteochondral defects as compared with 5% and 10% SDS groups, which includes vascularized subchondral bone, articular cartilage with adequate thickness, and integration with host tissues. Furthermore, ECM from 1% SDS significantly increased the migrating potential of bone marrow mesenchymal stem cells (BMSCs) in vitro. RT-PCR and western blot also revealed that ECM increased the expression of SOX-9 in BMSCs, whereas it decreased COL-X expression. In conclusion, our results suggested that the chondrocyte sheets decellularized with 1% SDS preserved the integrity and bioactivity, which favored cell recruitment and enabled osteochondral regeneration in the knee joints of rabbits, thus offering a promising approach for articular cartilage reconstruction without cell transplantation. STATEMENT OF SIGNIFICANCE: Although biological extracellular matrix (ECM) derived from decellularized native cartilage has been widely used in cartilage regeneration, it is difficult for endogenous cells to migrate into ECM owing to its dense nature. Moreover, current decellularization approaches lead to architectural breakdown of ECM. This study established a novel biological ECM from decellularized chondrocyte sheets for cartilage regeneration. Our results suggested that cartilaginous ECM favored cell recruitment and enabled osteochondral regeneration in rabbits, thus offering a promising approach for articular cartilage reconstruction without cell transplantation. SDS 1% adequately decellularized the chondrocytes in cell sheets, whereas it maintained the native architecture and composition of ECM, thereby avoiding the use of high-concentration SDS and providing a new way to acquire cartilaginous ECM.
Collapse
|
37
|
Edgar L, Altamimi A, García Sánchez M, Tamburrinia R, Asthana A, Gazia C, Orlando G. Utility of extracellular matrix powders in tissue engineering. Organogenesis 2018; 14:172-186. [PMID: 30183489 PMCID: PMC6300104 DOI: 10.1080/15476278.2018.1503771] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Extracellular matrix (ECM) materials have had remarkable success as scaffolds in tissue engineering (TE) and as therapies for tissue injury whereby the ECM microenvironment promotes constructive remodeling and tissue regeneration. ECM powder and solubilized derivatives thereof have novel applications in TE and RM afforded by the capacity of these constructs to be dynamically modulated. The powder form allows for effective incorporation and penetration of reagents; hence, ECM powder is an efficacious platform for 3D cell culture and vehicle for small molecule delivery. ECM powder offers minimally invasive therapy for tissue injury and successfully treatment for wounds refractory to first-line therapies. Comminution of ECM and fabrication of powder-derived constructs, however, may compromise the biological integrity of the ECM. The current lack of optimized fabrication protocols prevents a more extensive and effective clinical application of ECM powders. Further study on methods of ECM powder fabrication and modification is needed.
Collapse
Affiliation(s)
- Lauren Edgar
- Wake Forest School of Medicine, Department of Surgery, Winston-Salem, NC, USA,Correspondence to: Lauren Elizabeth Edgar, E-mail:
| | - Afnan Altamimi
- Wake Forest Institute for Regenerative Medicine, Department of Surgery, Winston-Salem, NC, USA,King Khalid University Hospital, Department of Surgery, Riyadh, Saudi Arabia
| | | | - Riccardo Tamburrinia
- Wake Forest School of Medicine, Department of Surgery, Winston-Salem, NC, USA,Wake Forest Institute for Regenerative Medicine, Department of Surgery, Winston-Salem, NC, USA,School of Experimental Medicine, University of Pavia, Pavia, Italy
| | - Amish Asthana
- Wake Forest Institute for Regenerative Medicine, Department of Surgery, Winston-Salem, NC, USA
| | - Carlo Gazia
- Wake Forest Institute for Regenerative Medicine, Department of Surgery, Winston-Salem, NC, USA
| | - Giuseppe Orlando
- Wake Forest School of Medicine, Department of Surgery, Winston-Salem, NC, USA,Wake Forest Institute for Regenerative Medicine, Department of Surgery, Winston-Salem, NC, USA
| |
Collapse
|
38
|
Zhang CH, Jiang YL, Ning LJ, Li Q, Fu WL, Zhang YJ, Zhang YJ, Xia CC, Li J, Qin TW. Evaluation of Decellularized Bovine Tendon Sheets for Achilles Tendon Defect Reconstruction in a Rabbit Model. Am J Sports Med 2018; 46:2687-2699. [PMID: 30074843 DOI: 10.1177/0363546518787515] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Achilles tendon (AT) defects frequently occur in trauma and chronic injuries. Currently, no method can satisfactorily reconstruct the AT with completely restored function. PURPOSE To evaluate the postoperative outcomes of AT defect reconstruction with decellularized bovine tendon sheets (DBTSs) in a rabbit model. STUDY DESIGN Controlled laboratory study. METHODS DBTSs were prepared from bovine tendons after compression, decellularization, antigen extraction, freeze drying, and sterilization. Platelet-rich plasma (PRP) was obtained by differential centrifugation. Sixty-three rabbits were used in this study, and the AT defect model was created bilaterally. All rabbits were divided into 3 groups (n = 21). In the DBTS group and the DBTS + PRP group, 2-cm-long AT was excised and reconstructed by DBTSs or PRP-treated DBTSs. In the control group, the rabbits underwent AT transection, and stumps were sutured. After surgery, all rabbits were assessed by ultrasonography and magnetic resonance imaging and then sacrificed for histological examination and biomechanical testing at 4, 8, or 12 weeks. RESULTS Gross observations demonstrated the absence of immunologic incompatibility and rejection. Histological examination showed that DBTSs promoted host cell infiltration and new fibrous tissue integration as compared with the control group. In each group, there was an AT-like structure formation and aligned collagen fiber deposition at 12 weeks. Mechanical properties of the reconstructed AT were not significantly different among the 3 groups at 4, 8, and 12 weeks after surgery ( P > .05). Ultrasonography and magnetic resonance imaging results illustrated that the reconstructed AT from each group maintained remodeling, and there was no significant difference in the echogenicity scoring ( P > .05) and percentages of good and excellent ( P > .05) among the 3 groups. CONCLUSION DBTSs, which retain the native tendon structure and bioactive factors, had the ability to remodel and integrate into the rabbit AT and improve the healing process. CLINICAL RELEVANCE DBTSs could serve as an effective bioscaffold to reconstruct AT defects.
Collapse
Affiliation(s)
- Cheng-Hao Zhang
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yan-Lin Jiang
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Liang-Ju Ning
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qi Li
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Wei-Li Fu
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ya-Jing Zhang
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yan-Jing Zhang
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chun-Chao Xia
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Jian Li
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ting-Wu Qin
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Current Methods for Skeletal Muscle Tissue Repair and Regeneration. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1984879. [PMID: 29850487 PMCID: PMC5926523 DOI: 10.1155/2018/1984879] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 02/28/2018] [Accepted: 03/11/2018] [Indexed: 12/11/2022]
Abstract
Skeletal muscle has the capacity of regeneration after injury. However, for large volumes of muscle loss, this regeneration needs interventional support. Consequently, muscle injury provides an ongoing reconstructive and regenerative challenge in clinical work. To promote muscle repair and regeneration, different strategies have been developed within the last century and especially during the last few decades, including surgical techniques, physical therapy, biomaterials, and muscular tissue engineering as well as cell therapy. Still, there is a great need to develop new methods and materials, which promote skeletal muscle repair and functional regeneration. In this review, we give a comprehensive overview over the epidemiology of muscle tissue loss, highlight current strategies in clinical treatment, and discuss novel methods for muscle regeneration and challenges for their future clinical translation.
Collapse
|
40
|
Lin J, Zhou W, Han S, Bunpetch V, Zhao K, Liu C, Yin Z, Ouyang H. Cell-material interactions in tendon tissue engineering. Acta Biomater 2018; 70:1-11. [PMID: 29355716 DOI: 10.1016/j.actbio.2018.01.012] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 12/11/2017] [Accepted: 01/10/2018] [Indexed: 12/19/2022]
Abstract
The interplay between cells and materials is a fundamental topic in biomaterial-based tissue regeneration. One of the principles for biomaterial development in tendon regeneration is to stimulate tenogenic differentiation of stem cells. To this end, efforts have been made to optimize the physicochemical and bio-mechanical properties of biomaterials for tendon tissue engineering. However, recent progress indicated that innate immune cells, especially macrophages, can also respond to the material cues and undergo phenotypical changes, which will either facilitate or hinder tissue regeneration. This process has been, to some extent, neglected by traditional strategies and may partially explain the unsatisfactory outcomes of previous studies; thus, more researchers have turned their focus on developing and designing immunoregenerative biomaterials to enhance tendon regeneration. In this review, we will first summarize the effects of material cues on tenogenic differentiation and paracrine secretion of stem cells. A brief introduction will also be made on how material cues can be manipulated for the regeneration of tendon-to-bone interface. Then, we will discuss the characteristics and influences of macrophages on the repair process of tendon healing and how they respond to different materials cues. These principles may benefit the development of novel biomaterials provided with combinative bioactive cues to activate tenogenic differentiation of stem cells and pro-resolving macrophage phenotype. STATEMENT OF SIGNIFICANCE The progress achieved with the rapid development of biomaterial-based strategies for tendon regeneration has not yielded broad benefits to clinical patients. In addition to the interplay between stem cells and biomaterials, the innate immune response to biomaterials also plays a determinant role in tissue regeneration. Here, we propose that fine-tuning of stem cell behaviors and alternative activation of macrophages through material cues may lead to effective tendon/ligament regeneration. We first review the characteristics of key material cues that have been manipulated to promote tenogenic differentiation and paracrine secretion of stem cells in tendon regeneration. Then, we discuss the potentiality of corresponding material cues in activating macrophages toward a pro-resolving phenotype to promote tissue repair.
Collapse
Affiliation(s)
- Junxin Lin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Zhejiang University, China
| | - Wenyan Zhou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Zhejiang University, China
| | - Shan Han
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Zhejiang University, China
| | - Varitsara Bunpetch
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Zhejiang University, China
| | - Kun Zhao
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Zhejiang University, China; Department of Sports Medicine, School of Medicine, Zhejiang University, China
| | - Chaozhong Liu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Zhejiang University, China
| | - Zi Yin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Zhejiang University, China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Zhejiang University, China; Department of Sports Medicine, School of Medicine, Zhejiang University, China; China Orthopedic Regenerative Medicine Group (CORMed), China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, China.
| |
Collapse
|
41
|
Sarrafian TL, Bodine SC, Murphy B, Grayson JK, Stover SM. Extracellular matrix scaffolds for treatment of large volume muscle injuries: A review. Vet Surg 2018; 47:524-535. [PMID: 29603757 DOI: 10.1111/vsu.12787] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 10/11/2017] [Accepted: 11/03/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Large muscular or musculotendinous defects present a dilemma because of the inadequacies of current treatment strategies. Extracellular matrices (ECM) are potential clinically applicable regenerative biomaterials. This review summarizes information from the preclinical literature evaluating the use of ECM for muscle regeneration in animal models of volumetric muscle loss (VML). STUDY DESIGN Literature review. SAMPLE POPULATION Animal models of VML in which surgical repair was performed with an ECM product, with or without added cell populations. METHODS PubMed, Google Scholar, CAB abstracts, and Scopus were searched for preclinical studies using ECM in animal models of VML. The search terms "extracellular matrix," "VML," "muscle regeneration," "cell seeded," and "scaffold" identified 40 articles that met inclusion criteria of an animal model of VML in which surgical repair was performed with an ECM product, with or without added cell populations. Key skeletal muscle repair mechanisms and experimental findings on scaffold type, VML location, and experimental animal species were summarized. CONCLUSIONS Satellite cells and basal lamina are key endogenous contributors to skeletal muscle regeneration. ECM as a dynamic tissue component may provide structural integrity, signaling molecules, and a 3-dimensional topography conducive to muscle regeneration. Preclinical models of muscle repair most commonly used mice and rats (88%). Most experimental lesions were created in abdominal wall (33%), anterior tibialis (33%), latissimus dorsi (10%), or quadriceps (10%) muscles. Matrices varied markedly in source and preparation. Experimental outcomes of ECM and cell-seeded ECM implantation for muscle regeneration in VML were highly variable and dependent on matrix tissue source, preparation method, and anatomic site of injury. Scar tissue formation likely contributes to load transfer. Nonappendicular lesions had better regenerative results compared with appendicular VML. CLINICAL SIGNIFICANCE The preponderance of current evidence supports the use of ECM for muscle defect repair only in specific instances, such as nonappendicular and/or partial-thickness defects. Consequently, clinical use of ECM in veterinary patients requires careful consideration of the specific ECM product, lesion size and location, and loading circumstances.
Collapse
Affiliation(s)
- Tiffany L Sarrafian
- J. D. Wheat Veterinary Orthopedic Research Laboratory, University of California, Davis, Davis, California.,Clinical Investigation Facility, David Grant US Air Force Medical Center, Travis Air Force Base, Fairfield, California
| | - Sue C Bodine
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, California
| | - Brian Murphy
- J. D. Wheat Veterinary Orthopedic Research Laboratory, University of California, Davis, Davis, California
| | - J Kevin Grayson
- Clinical Investigation Facility, David Grant US Air Force Medical Center, Travis Air Force Base, Fairfield, California
| | - Susan M Stover
- J. D. Wheat Veterinary Orthopedic Research Laboratory, University of California, Davis, Davis, California
| |
Collapse
|
42
|
Correia CR, Gaifem J, Oliveira MB, Silvestre R, Mano JF. The influence of surface modified poly(l-lactic acid) films on the differentiation of human monocytes into macrophages. Biomater Sci 2018; 5:551-560. [PMID: 28128374 DOI: 10.1039/c6bm00920d] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Macrophages play a crucial role in the biological performance of biomaterials, as key factors in defining the optimal inflammation-healing balance towards tissue regeneration and implant integration. Here, we investigate how different surface modifications performed on poly(l-lactic acid) (PLLA) films would influence the differentiation of human monocytes into macrophages. We tested PLLA films without modification, surface-modified by plasma treatment (pPLLA) or by combining plasma treatment with different coating materials, namely poly(l-lysine) and a series of proteins from the extracellular matrix: collagen I, fibronectin, vitronectin, laminin and albumin. While all the tested films are non-cytotoxic, differences in cell adhesion and morphology are observed. Monocyte-derived macrophages (MDM) present a more rounded shape in non-modified films, while a more elongated phenotype is observed containing filopodia-like and podosome-like structures in all modified films. No major differences are found for the expression of HLA-DR+/CD80+ and CD206+/CD163+ surface markers, as well as for the ability of MDM to phagocytize. Interestingly, MDM differentiated on pPLLA present the highest expression of MMP9. Upon differentiation, MDM in all surface modified films present lower amounts of IL-6 and IL-10 compared to non-modified films. After stimulating MDM with the potent pro-inflammatory agent LPS, pPLLA and poly(l-lysine) and fibronectin-modified films reveal a significant reduction in IL-6 secretion, while the opposite effect is observed with IL-10. Of note, in comparison to non-modified films, all surface modified films induce a significant reduction of the IL-6/IL-10 ratio, a valuable prognosticator of the pro- versus anti-inflammatory balance. These findings provide important insights into MDM-biomaterial interactions, while strengthening the need for designing immune-informed biomaterials.
Collapse
Affiliation(s)
- Clara R Correia
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Joana Gaifem
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal. and Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
| | - Mariana B Oliveira
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Ricardo Silvestre
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal. and Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
| | - João F Mano
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
43
|
Prest TA, Yeager E, LoPresti ST, Zygelyte E, Martin MJ, Dong L, Gibson A, Olutoye OO, Brown BN, Cheetham J. Nerve-specific, xenogeneic extracellular matrix hydrogel promotes recovery following peripheral nerve injury. J Biomed Mater Res A 2018; 106:450-459. [PMID: 28891122 PMCID: PMC5745279 DOI: 10.1002/jbm.a.36235] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 08/16/2017] [Accepted: 09/08/2017] [Indexed: 01/07/2023]
Abstract
Peripheral nerve possesses the inherent ability to regrow and recover following injury. However, nerve regeneration is often slow and incomplete due to limitations associated with the local microenvironment during the repair process. Manipulation of the local microenvironment at the site of nerve repair, therefore, represents a significant opportunity for improvement in downstream outcomes. Macrophages and Schwann cells play a key role in the orchestration of early events after peripheral nerve injury. We describe the production, characterization, and use of an injectable, peripheral nerve-specific extracellular matrix-based hydrogel (PNSECM) for promoting modulation of the local macrophage and Schwann cell responses at the site of nerve repair in a rodent model of sciatic nerve injury. We show that PNSECM hydrogels largely maintain the matrix structure associated with normal native peripheral nerve tissue. PNSECM hydrogels were also found to promote increased macrophage invasion, higher percentages of M2 macrophages and enhanced Schwann cell migration when used as a lumen filler in a rodent model of nerve gap repair using an inert nerve guidance conduit. These results suggest that an injectable PNSECM hydrogel can provide a supportive, bioactive scaffold which promotes repair of peripheral nerve in vivo. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 450-459, 2018.
Collapse
Affiliation(s)
- Travis A. Prest
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA
| | - Eric Yeager
- Department of Clinical Sciences, Cornell University, Ithaca, NY
| | - Samuel T. LoPresti
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA
| | | | | | - Longying Dong
- Department of Clinical Sciences, Cornell University, Ithaca, NY
| | - Alexis Gibson
- Department of Clinical Sciences, Cornell University, Ithaca, NY
| | - Oluyinka O. Olutoye
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA
| | - Bryan N. Brown
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA
- Department of Clinical Sciences, Cornell University, Ithaca, NY
| | - Jonathan Cheetham
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA
- Department of Clinical Sciences, Cornell University, Ithaca, NY
| |
Collapse
|
44
|
Corona BT, Flanagan KE, Brininger CM, Goldman SM, Call JA, Greising SM. Impact of volumetric muscle loss injury on persistent motoneuron axotomy. Muscle Nerve 2018; 57:799-807. [PMID: 29144551 DOI: 10.1002/mus.26016] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 11/12/2017] [Accepted: 11/14/2017] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Volumetric muscle loss (VML) occurs following significant traumatic injury or surgical removal of skeletal muscle, resulting in nonrecoverable loss of tissue and long-term dysfunction. Perhaps less recognized is that VML injuries inherently disrupt the neuromuscular unit, resulting in fiber denervation and presumptive motor unit rearrangement, expansion, and/or loss. To characterize neural dysfunction we quantified motoneuron axotomy, in efforts to understand how this relates to the temporal coordination of neuromuscular and morphological alterations due to injury. METHODS In an established rat tibialis anterior (TA) VML injury model, we examined the motoneuron, skeletal muscle, and maximal isometric torque at 3, 7, 14, and 21 days postinjury. RESULTS Significant axotomy of 57-79% of all TA muscle motoneurons was observed through 21 days postinjury, which was coupled with a 45-90% TA maximal torque deficit. DISCUSSION A ∼20% partial ablation of the TA muscle causes disproportionate damage across the motor unit acutely postinjury. Muscle Nerve 57: 799-807, 2018.
Collapse
Affiliation(s)
- Benjamin T Corona
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research, 3698 Chambers Pass, BHT1, Fort Sam Houston, Texas, 78234, USA
| | - Kate E Flanagan
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research, 3698 Chambers Pass, BHT1, Fort Sam Houston, Texas, 78234, USA
| | - Christian M Brininger
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research, 3698 Chambers Pass, BHT1, Fort Sam Houston, Texas, 78234, USA
| | - Stephen M Goldman
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research, 3698 Chambers Pass, BHT1, Fort Sam Houston, Texas, 78234, USA
| | - Jarrod A Call
- Department of Kinesiology, University of Georgia, Athens, Georgia, USA.,Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
| | - Sarah M Greising
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research, 3698 Chambers Pass, BHT1, Fort Sam Houston, Texas, 78234, USA
| |
Collapse
|
45
|
Dziki JL, Badylak SF. Extracellular Matrix for Myocardial Repair. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1098:151-171. [PMID: 30238370 DOI: 10.1007/978-3-319-97421-7_8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Multiple strategies have been investigated to restore functional myocardium following injury or disease including the local administration of cytokines or chemokines, stem/progenitor cell therapy, mechanical circulatory support, pharmacologic use, and the use of inductive biomaterials. The use of xenogeneic biologic scaffolds composed of extracellular matrix (ECM) has been shown to facilitate functional restoration of several tissues and organs including the esophagus, skeletal muscle, skin, and myocardium, among others. The present chapter describes the current understanding of specific components of biologic scaffolds composed of ECM, the mechanisms by which ECM bioscaffolds promote constructive cardiac remodeling after injury, determinants of remodeling outcome, and the versatility of ECM as a potential cardiac therapeutic.
Collapse
Affiliation(s)
- Jenna L Dziki
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA. .,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. .,Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
46
|
Ghetti M, Papa V, Deluca G, Purpura V, Ruscelli P, Melandri D, Capirossi D, Nigrisoli E, Minghetti P, Bondioli E, Cenacchi G. Histological and ultrastructural evaluation of human decellularized matrix as a hernia repair device. Ultrastruct Pathol 2017; 42:32-38. [PMID: 29192810 DOI: 10.1080/01913123.2017.1365788] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Recently, interest has been increasing for human decellularized matrices, due to their ability to reduce numerous side effects related to hernia repair. To date, only animal studies investigated the biological interaction post-implant of human decellularized matrices for soft tissue repair. Therefore, the aim of this study was to evaluate the morphological response one year post implant of human decellularized matrix, through morphological analysis of human biopsies. The histological and ultrastructural results revealed a perfect cellular repopulation and neoangiogenesis, with minimal inflammatory response and a well-organized collagen matrix. The results have indicated that this scaffold can be an effective treatment for hernia.
Collapse
Affiliation(s)
- Martina Ghetti
- a Biomedical and Neuromotor Sciences Department , University of Bologna , Bologna , Italy.,b Burn Center and Emilia Romagna Regional Skin Bank , M. Bufalini Hospital, AUSL Romagna , Cesena , Italy
| | - Valentina Papa
- a Biomedical and Neuromotor Sciences Department , University of Bologna , Bologna , Italy
| | - Giovanni Deluca
- c Pathology Unit , M. Bufalini Hospital, AUSL Romagna , Cesena , Italy
| | - Valeria Purpura
- b Burn Center and Emilia Romagna Regional Skin Bank , M. Bufalini Hospital, AUSL Romagna , Cesena , Italy
| | - Paolo Ruscelli
- d Emergency Surgery Unit, Torrette Hospital, Faculty of Medicine and Surgery , Polytechnic University of Marche , Ancona , Italy
| | - Davide Melandri
- b Burn Center and Emilia Romagna Regional Skin Bank , M. Bufalini Hospital, AUSL Romagna , Cesena , Italy
| | - Daniela Capirossi
- c Pathology Unit , M. Bufalini Hospital, AUSL Romagna , Cesena , Italy
| | - Evandro Nigrisoli
- c Pathology Unit , M. Bufalini Hospital, AUSL Romagna , Cesena , Italy
| | - Paola Minghetti
- b Burn Center and Emilia Romagna Regional Skin Bank , M. Bufalini Hospital, AUSL Romagna , Cesena , Italy
| | - Elena Bondioli
- b Burn Center and Emilia Romagna Regional Skin Bank , M. Bufalini Hospital, AUSL Romagna , Cesena , Italy
| | - Giovanna Cenacchi
- a Biomedical and Neuromotor Sciences Department , University of Bologna , Bologna , Italy
| |
Collapse
|
47
|
Sharifzadeh G, Hosseinkhani H. Biomolecule-Responsive Hydrogels in Medicine. Adv Healthc Mater 2017; 6. [PMID: 29057617 DOI: 10.1002/adhm.201700801] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/17/2017] [Indexed: 12/19/2022]
Abstract
Recent advances and applications of biomolecule-responsive hydrogels, namely, glucose-responsive hydrogels, protein-responsive hydrogels, and nucleic-acid-responsive hydrogels are highlighted. However, achieving the ultimate purpose of using biomolecule-responsive hydrogels in preclinical and clinical areas is still at the very early stage and calls for more novel designing concepts and advance ideas. On the way toward the real/clinical application of biomolecule-responsive hydrogels, plenty of factors should be extensively studied and examined under both in vitro and in vivo conditions. For example, biocompatibility, biointegration, and toxicity of biomolecule-responsive hydrogels should be carefully evaluated. From the living body's point of view, biocompatibility is seriously depended on the interactions at the tissue/polymer interface. These interactions are influenced by physical nature, chemical structure, surface properties, and degradation of the materials. In addition, the developments of advanced hydrogels with tunable biological and mechanical properties which cause no/low side effects are of great importance.
Collapse
Affiliation(s)
- Ghorbanali Sharifzadeh
- Department of Polymer Engineering; Faculty of Chemical Engineering; Universiti Teknologi Malaysia; 81310 Johor Malaysia
| | | |
Collapse
|
48
|
Abstract
Volumetric muscle loss (VML) resulting from extremity trauma presents chronic and persistent functional deficits which ultimately manifest disability. Acellular biological scaffolds, or decellularized extracellular matrices (ECMs), embody an ideal treatment platform due to their current clinical use for soft tissue repair, off-the-shelf availability, and zero autogenous donor tissue burden. ECMs have been reported to promote functional skeletal muscle tissue remodeling in small and large animal models of VML injury, and this conclusion was reached in a recent clinical trial that enrolled 13 patients. However, numerous other pre-clinical reports have not observed ECM-mediated skeletal muscle regeneration. The current study was designed to reconcile these discrepancies. The capacity of ECMs to orchestrate functional muscle tissue remodeling was interrogated in a porcine VML injury model using unbiased assessments of muscle tissue regeneration and functional recovery. Here, we show that VML injury incites an overwhelming inflammatory and fibrotic response that leads to expansive fibrous tissue deposition and chronic functional deficits, which ECM repair does not augment.
Collapse
|
49
|
Bioinductive Scaffolds—Powerhouses of Skeletal Muscle Tissue Engineering. CURRENT PATHOBIOLOGY REPORTS 2017. [DOI: 10.1007/s40139-017-0151-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
50
|
Costa A, Naranjo JD, Londono R, Badylak SF. Biologic Scaffolds. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a025676. [PMID: 28320826 DOI: 10.1101/cshperspect.a025676] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Biologic scaffold materials composed of allogeneic or xenogeneic extracellular matrix are commonly used for the repair and functional reconstruction of injured and missing tissues. These naturally occurring bioscaffolds are manufactured by the removal of the cellular content from source tissues while preserving the structural and functional molecular units of the remaining extracellular matrix (ECM). The mechanisms by which these bioscaffolds facilitate constructive remodeling and favorable clinical outcomes include release or creation of effector molecules that recruit endogenous stem/progenitor cells to the site of scaffold placement and modulation of the innate immune response, specifically the activation of an anti-inflammatory macrophage phenotype. The methods by which ECM biologic scaffolds are prepared, the current understanding of in vivo scaffold remodeling, and the associated clinical outcomes are discussed in this article.
Collapse
Affiliation(s)
- Alessandra Costa
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania 15219
| | - Juan Diego Naranjo
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania 15219
| | - Ricardo Londono
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania 15219.,School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania 15219.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15219.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| |
Collapse
|