1
|
Sonaye SY, Sikder P. Bioengineered Constructs as a Tissue Engineering-Based Therapy for Volumetric Muscle Loss. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40265282 DOI: 10.1089/ten.teb.2025.0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Severe skeletal muscle injuries involving substantial tissue loss can significantly impair muscle strength and functionality, reducing the quality of life for affected individuals. Such injuries, termed volumetric muscle loss, require extensive clinical intervention, as the body's innate healing mechanisms are insufficient to regenerate functional muscle. The current standard of care primarily involves autologous muscle tissue transfer, with some consideration of acellular synthetic constructs. However, both approaches have limited therapeutic efficacy, presenting challenges such as donor-site morbidity, infection risks, and suboptimal functional recovery. Over the past decade, skeletal muscle tissue engineering (SMTE) has emerged as a promising strategy for regenerating functional muscle through bioengineered constructs. Advanced biofabrication techniques, including bioprinting, have further enabled the development of synthetic constructs that closely mimic native muscle architecture. Given these advancements, a critical review of recent therapeutic strategies, their achievements, and limitations is necessary. This review examines the spectrum of bioengineered constructs developed from various biomaterials and evaluates their therapeutic potential. Special emphasis is placed on 3D bioprinting strategies and their role in creating physiologically relevant constructs for functional muscle restoration. In addition, the integration of machine learning in optimizing construct design, predicting cellular behavior, and enhancing tissue integration is discussed. The review indicates that despite significant progress in SMTE, key challenges remain, including replicating the complex structural organization of muscle tissue, minimizing fibrosis, and achieving vascularization and innervation to regenerate functional, strengthened muscle. Future research should address these barriers while prioritizing the development of translational, clinically relevant regenerative constructs. In addition, efforts should focus on advancing scalable, construct-based regenerative treatments that are readily available at the point of care and easily managed in surgical settings.
Collapse
Affiliation(s)
| | - Prabaha Sikder
- Department of Mechanical Engineering, Cleveland State University, Cleveland, Ohio, USA
| |
Collapse
|
2
|
Sorensen JR, Hoffman DB, Raymond-Pope CJ, Lillquist TJ, Russell AM, Corona BT, Greising SM. Inhibition of ErbB2 mitigates secondary denervation after traumatic muscle injury. J Physiol 2025. [PMID: 40033740 DOI: 10.1113/jp287435] [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: 08/06/2024] [Accepted: 02/13/2025] [Indexed: 03/05/2025] Open
Abstract
Secondary denervation has recently been described as part of the sequela of volumetric muscle loss (VML) injury, occurring along with a significantly elevated neurotrophic response, specifically neuregulin-1 (NRG1). This may contribute to chronic functional impairments associated with the injury, representing an overlooked treatment target. Thus, though paradoxical, the goal of this study was to pharmacologically reduce neurotrophic signalling after VML using a monoclonal antibody (Herceptin) that inhibits ErbB2 receptors. We also assessed whether ErbB2 inhibition combined with a myogenic treatment (i.e. minced muscle graft) would have a synergistically beneficial effect on function. Adult male Lewis rats underwent surgical induction of tibialis anterior muscle VML injury and were randomized into one of four groups: VML untreated, VML Herceptin, VML muscle graft and VML muscle graft + Herceptin, with comparisons to the contralateral (uninjured) control muscle. Rats receiving Herceptin were administered the drug (8 mg/kg i.p.) at the time of surgery and thrice per week for the duration of the study (48 days). Terminally individual NMJs were quantitatively evaluated, and maximal in vivo torque was tested. ErbB2 inhibition fully restored the normal rates of NMJ innervation and morphology after VML injury, and improved innervation of de novo myofibres after a muscle-graft treatment. However ErbB2 inhibition did not improve skeletal muscle function alone or in combination with a muscle-graft treatment. We conclude that ErbB2 inhibition is a promising therapeutic option for treating VML injury, yet more work is needed to optimize the translation of improved NMJ characteristics to recover function. KEY POINTS: In cases of complex traumatic musculoskeletal injury, such as volumetric muscle loss (VML), the endogenous ability of skeletal muscle to regenerate and recover function is lost. Innervation, or the connection of a motor axon to each individual myofibre, is a necessary component of myofibre survival and contractile function, which is disrupted after VML. Paradoxically a monocolonal antibody inhibitor of neurotrophic signalling (receptor tyrosine kinase ErbB2; Herceptin) has been shown to improve regeneration in rodent models of nerve injury. Here we show that pharmaceutical ErbB2 inhibition following a rat model of VML improves muscle innervation; however it did not correspondingly recover muscle function. Although ErbB2 inhibition alone is an ineffective treatment for VML injury, its ability to improve innervation is noteworthy and should be considered as an adjunctive or combinatorial therapy option.
Collapse
Affiliation(s)
- Jacob R Sorensen
- School of Kinesiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Daniel B Hoffman
- School of Kinesiology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Thomas J Lillquist
- School of Kinesiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Amanda M Russell
- School of Kinesiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Benjamin T Corona
- School of Medicine, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Sarah M Greising
- School of Kinesiology, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
3
|
Hu C, Chiang G, Chan AHP, Alcazar C, Nakayama KH, Quarta M, Rando TA, Huang NF. A mouse model of volumetric muscle loss and therapeutic scaffold implantation. Nat Protoc 2025; 20:608-619. [PMID: 39424992 PMCID: PMC11896750 DOI: 10.1038/s41596-024-01059-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 05/24/2024] [Indexed: 10/21/2024]
Abstract
Skeletal myofibers naturally regenerate after damage; however, impaired muscle function can result in cases when a prominent portion of skeletal muscle mass is lost, for example, following traumatic muscle injury. Volumetric muscle loss can be modeled in mice using a surgical model of muscle ablation to study the pathology of volumetric muscle loss and to test experimental treatments, such as the implantation of acellular scaffolds, which promote de novo myogenesis and angiogenesis. Here we provide step-by-step instructions to perform full-thickness surgical ablation, using biopsy punches, and to remove a large volume of the tibialis anterior muscle of the lower limb in mice. This procedure results in a reduction in muscle mass and limited regeneration capacity; the approach is easy to reproduce and can also be applied to larger animal models. For therapeutic applications, we further explain how to implant bioscaffolds into the ablated muscle site. With adequate training and practice, the surgical procedure can be performed within 30 min.
Collapse
Affiliation(s)
- Caroline Hu
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Gladys Chiang
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Alex H-P Chan
- The Stanford Cardiovascular Institute, Stanford University, Palo Alto, CA, USA
- Department of Cardiothoracic Surgery, Stanford University, Palo Alto, CA, USA
| | - Cynthia Alcazar
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
| | - Karina H Nakayama
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
| | - Marco Quarta
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Thomas A Rando
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Ngan F Huang
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
- The Stanford Cardiovascular Institute, Stanford University, Palo Alto, CA, USA.
- Department of Cardiothoracic Surgery, Stanford University, Palo Alto, CA, USA.
- Department of Chemical Engineering, Stanford University, Palo Alto, CA, USA.
| |
Collapse
|
4
|
Cai CW, Grey JA, Hubmacher D, Han WM. Biomaterial-Based Regenerative Strategies for Volumetric Muscle Loss: Challenges and Solutions. Adv Wound Care (New Rochelle) 2025; 14:159-175. [PMID: 38775429 PMCID: PMC11971559 DOI: 10.1089/wound.2024.0079] [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: 03/30/2024] [Accepted: 05/15/2024] [Indexed: 07/11/2024] Open
Abstract
Significance: Volumetric muscle loss (VML) is caused by the loss of significant amounts of skeletal muscle tissue. VML cannot be repaired by intrinsic regenerative processes, resulting in permanent loss of muscle function and disability. Current rehabilitative-focused treatment strategies lack efficacy and do not restore muscle function, indicating the need for the development of effective regenerative strategies. Recent Advances: Recent developments implicate biomaterial-based approaches for promoting muscle repair and functional restoration post-VML. Specifically, bioscaffolds transplanted in the injury site have been utilized to mimic endogenous cues of the ablated tissue to promote myogenic pathways, increase neo-myofiber synthesis, and ultimately restore contractile function to the injured unit. Critical Issues: Despite the development and preclinical testing of various biomaterial-based regenerative strategies, effective therapies for patients are not available. The unique challenges posed for biomaterial-based treatments of VML injuries, including its scalability and clinical applicability beyond small-animal models, impede progress. Furthermore, production of tissue-engineered constructs is technically demanding, with reproducibility issues at scale and complexities in achieving vascularization and innervation of large constructs. Future Directions: Biomaterial-based regenerative strategies designed to comprehensively address the pathophysiology of VML are needed. Considerations for clinical translation, including scalability and regulatory compliance, should also be considered when developing such strategies. In addition, an integrated approach that combines regenerative and rehabilitative strategies is essential for ensuring functional improvement.
Collapse
Affiliation(s)
- Charlene W. Cai
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Biology, The College of New Jersey, Ewing, New Jersey, USA
| | - Josh A. Grey
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute of Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Dirk Hubmacher
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Woojin M. Han
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute of Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
5
|
Lee MC, Jodat YA, Endo Y, Rodríguez-delaRosa A, Zhang T, Karvar M, Tanoury ZA, Quint J, Kamperman T, Kiaee K, Ochoa SL, Shi K, Huang Y, Rosales MP, Lee H, Kim J, Ceron EL, Reyes IG, Panayi AC, Wang X, Kim KT, Moon JI, Park SG, Lee K, Calabrese MA, Lee J, Tamayol A, Lee L, Pourquié O, Kim WJ, Sinha I, Shin SR. Engineering large-scale hiPSC-derived vessel-integrated muscle-like lattices for enhanced volumetric muscle regeneration. Trends Biotechnol 2024; 42:1715-1744. [PMID: 39306493 PMCID: PMC11625013 DOI: 10.1016/j.tibtech.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 12/08/2024]
Abstract
Engineering biomimetic tissue implants with human induced pluripotent stem cells (hiPSCs) holds promise for repairing volumetric tissue loss. However, these implants face challenges in regenerative capability, survival, and geometric scalability at large-scale injury sites. Here, we present scalable vessel-integrated muscle-like lattices (VMLs), containing dense and aligned hiPSC-derived myofibers alongside passively perfusable vessel-like microchannels inside an endomysium-like supporting matrix using an embedded multimaterial bioprinting technology. The contractile and millimeter-long myofibers are created in mechanically tailored and nanofibrous extracellular matrix-based hydrogels. Incorporating vessel-like lattice enhances myofiber maturation in vitro and guides host vessel invasion in vivo, improving implant integration. Consequently, we demonstrate successful de novo muscle formation and muscle function restoration through a combinatorial effect between improved graft-host integration and its increased release of paracrine factors within volumetric muscle loss injury models. The proposed modular bioprinting technology enables scaling up to centimeter-sized prevascularized hiPSC-derived muscle tissues with custom geometries for next-generation muscle regenerative therapies.
Collapse
Affiliation(s)
- Myung Chul Lee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Medicinal Materials Research Center, Korea Institute of Science and Technology, Seoul, 02792 Republic of Korea
| | - Yasamin A. Jodat
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Yori Endo
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Alejandra Rodríguez-delaRosa
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Ting Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
| | - Mehran Karvar
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ziad Al Tanoury
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Jacob Quint
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE, 68588, USA
| | - Tom Kamperman
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Kiavash Kiaee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Sofia Lara Ochoa
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Kun Shi
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Yike Huang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Montserrat Pineda Rosales
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Hyeseon Lee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Jiseong Kim
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Eder Luna Ceron
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Isaac Garcia Reyes
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Adriana C. Panayi
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xichi Wang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Ki-Tae Kim
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Dental Multi-omics Center, Seoul National University, Seoul, 08826, Republic of Korea
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jae-I Moon
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Dental Multi-omics Center, Seoul National University, Seoul, 08826, Republic of Korea
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Gwa Park
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Dental Multi-omics Center, Seoul National University, Seoul, 08826, Republic of Korea
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kangju Lee
- Department of Healthcare and Medical Engineering, Chonnam National University, Yeosu 59626, South Korea
| | - Michelle A. Calabrese
- Chemical Engineering and Materials Science Department, University of Minnesota, Minneapolis, MN 55455, USA
| | - Junmin Lee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE, 68588, USA
| | - Luke Lee
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Korea
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Korea
| | - Olivier Pourquié
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Woo-Jin Kim
- Correspondence: (I.S.), (W.J.K.), (S.R.S.), Twitter: Yasamin A. Jodat: @YasaminJodat
| | - Indranil Sinha
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Lead contact
| |
Collapse
|
6
|
Tanner GI, Schiltz L, Narra N, Figueiredo ML, Qazi TH. Granular Hydrogels Improve Myogenic Invasion and Repair after Volumetric Muscle Loss. Adv Healthc Mater 2024; 13:e2303576. [PMID: 38329892 DOI: 10.1002/adhm.202303576] [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: 10/17/2023] [Revised: 12/26/2023] [Indexed: 02/10/2024]
Abstract
Skeletal muscle injuries including volumetric muscle loss (VML) lead to excessive tissue scarring and permanent functional disability. Despite its high prevalence, there is currently no effective treatment for VML. Bioengineering interventions such as biomaterials that fill the VML defect to support cell and tissue growth are a promising therapeutic strategy. However, traditional biomaterials developed for this purpose lack the pore features needed to support cell infiltration. The present study investigates for the first time, the impact of granular hydrogels on muscle repair - hypothesizing that their flowability will permit conformable filling of the defect site and their inherent porosity will support the invasion of native myogenic cells, leading to effective muscle repair. Small and large microparticle fragments are prepared from photocurable hyaluronic acid polymer via extrusion fragmentation and facile size sorting. In assembled granular hydrogels, particle size and degree of packing significantly influence pore features, rheological behavior, and injectability. Using a mouse model of VML, it is demonstrated that, in contrast to bulk hydrogels, granular hydrogels support early-stage (satellite cell invasion) and late-stage (myofiber regeneration) muscle repair processes. Together, these results highlight the promising potential of injectable and porous granular hydrogels in supporting endogenous repair after severe muscle injury.
Collapse
Affiliation(s)
- Gabrielle I Tanner
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Leia Schiltz
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Niharika Narra
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Marxa L Figueiredo
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Taimoor H Qazi
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| |
Collapse
|
7
|
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
|
8
|
Clark AR, Kulwatno J, Kanovka SS, Klarmann GJ, Hernandez CE, Natoli RM, McKinley TO, Potter BK, Dearth CL, Goldman SM. A Two-Stage Approach Integrating Provisional Biomaterial-Mediated Stabilization Followed by a Definitive Treatment for Managing Volumetric Muscle Loss Injuries. J Funct Biomater 2024; 15:160. [PMID: 38921533 PMCID: PMC11204500 DOI: 10.3390/jfb15060160] [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: 05/14/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/27/2024] Open
Abstract
Treatment of volumetric muscle loss (VML) faces challenges due to its unique pathobiology and lower priority in severe musculoskeletal injury management. Consequently, a need exists for multi-stage VML treatment strategies to accommodate delayed interventions owing to comorbidity management or prolonged casualty care in combat settings. To this end, polyvinyl alcohol (PVA) was used at concentrations of 5%, 7.5%, and 10% to generate provisional muscle void fillers (MVFs) of varying stiffness values (1.125 kPa, 3.700 kPa, and 7.699 kPa) to stabilize VML injuries as part of a two-stage approach. These were implanted into a rat model for a duration of 4 weeks, then explanted and either left untreated (control) or treated through minced muscle grafting (MMG). Additional benchmarks included acute MMG and unrepaired groups. At the MVF explant, the 7.5% PVA group exhibited superior neuromuscular function compared to the 5% and 10% PVA groups, the least fibrosis, and the largest median myofiber size among all groups at the 12-week endpoint. Despite the 7.5% PVA's superiority amongst the two-stage treatment groups, neuromuscular function was neither improved nor impaired relative to acute treatment benchmarks. This suggests that the future success of a two-stage VML treatment strategy will necessitate a more effective definitive intervention.
Collapse
Affiliation(s)
- Andrew R. Clark
- Extremity Trauma and Amputation Center of Excellence, Defense Health Agency, Falls Church, VA 22042, USA
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Jonathan Kulwatno
- Extremity Trauma and Amputation Center of Excellence, Defense Health Agency, Falls Church, VA 22042, USA
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Sergey S. Kanovka
- Extremity Trauma and Amputation Center of Excellence, Defense Health Agency, Falls Church, VA 22042, USA
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - George J. Klarmann
- 4D Bio3 Center for Biotechnology and Department of Radiology and Radiological Sciences, Uniformed Services University of the Health Sciences, Bethesda, MD 20817, USA
- The Geneva Foundation, Tacoma, WA 98402, USA
| | - Claudia E. Hernandez
- Extremity Trauma and Amputation Center of Excellence, Defense Health Agency, Falls Church, VA 22042, USA
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- C2 Alaska, LLC, San Antonio, TX 78249, USA
| | - Roman M. Natoli
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Todd O. McKinley
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Benjamin K. Potter
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Department of Orthopaedic Surgery, Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
| | - Christopher L. Dearth
- Extremity Trauma and Amputation Center of Excellence, Defense Health Agency, Falls Church, VA 22042, USA
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Stephen M. Goldman
- Extremity Trauma and Amputation Center of Excellence, Defense Health Agency, Falls Church, VA 22042, USA
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
9
|
Zhao N, Huang Y, Cheng X, Xie L, Xiao W, Shi B, Li J. A critical size volumetric muscle loss model in mouse masseter with impaired mastication on nutrition. Cell Prolif 2024; 57:e13610. [PMID: 38356342 DOI: 10.1111/cpr.13610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/13/2024] [Accepted: 01/30/2024] [Indexed: 02/16/2024] Open
Abstract
Orofacial muscle defect due to congenital anomalies, tumour ablation or traumatic accident that exceeds endogenous regeneration capacity may lead to sustained deficits in masticatory function and nutrition intake. Functional recovery has always been the goal of muscle tissue repair, but currently, there is no suitable model for quantitative analyses of either functional consequences or treatment efficacy of orofacial muscle defect. This study proposed a critical size volumetric muscle loss (VML) model in mouse masseter with impaired mastication on nutrition. Full-thickness VML defects in diameter of 1.0, 1.5, 2.0 and 3.0 mm were generated in the centre of the mouse masseter using a biopsy punch to determine the critical size for functional impairment. In the VML region, myogenesis was dampened but fibrogenesis was activated, as long with a reduction in the density of the neuromuscular junction and an increase in vascular density. Accordingly, persistent fibrosis was observed in the centre region of VML in all diameters. The 2.0 mm diameter was the critical threshold to masticatory function impairment after VML in the masseter. VML of 3.0 mm diameter led to a significant impact on nutrition intake and body weight gain. Autologous muscle graft effectively relieved the fibrosis and functional deficit after VML injury in the masseter. This model serves as a reliable tool in studying functional recovery strategies for orofacial muscle defects.
Collapse
Affiliation(s)
- Ning Zhao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yixuan Huang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xu Cheng
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| | - Li Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| | - Wenlin Xiao
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bing Shi
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jingtao Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Motherwell JM, Dolan CP, Kanovka SS, Edwards JB, Franco SR, Janakiram NB, Valerio MS, Goldman SM, Dearth CL. Effects of Adjunct Antifibrotic Treatment within a Regenerative Rehabilitation Paradigm for Volumetric Muscle Loss. Int J Mol Sci 2023; 24:3564. [PMID: 36834976 PMCID: PMC9964131 DOI: 10.3390/ijms24043564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
The use of a rehabilitation approach that promotes regeneration has the potential to improve the efficacy of pro-regenerative therapies and maximize functional outcomes in the treatment of volumetric muscle loss (VML). An adjunct antifibrotic treatment could further enhance functional gains by reducing fibrotic scarring. This study aimed to evaluate the potential synergistic effects of losartan, an antifibrotic pharmaceutical, paired with a voluntary wheel running rehabilitation strategy to enhance a minced muscle graft (MMG) pro-regenerative therapy in a rodent model of VML. The animals were randomly assigned into four groups: (1) antifibrotic with rehabilitation, (2) antifibrotic without rehabilitation, (3) vehicle treatment with rehabilitation, and (4) vehicle treatment without rehabilitation. At 56 days, the neuromuscular function was assessed, and muscles were collected for histological and molecular analysis. Surprisingly, we found that the losartan treatment decreased muscle function in MMG-treated VML injuries by 56 days, while the voluntary wheel running elicited no effect. Histologic and molecular analysis revealed that losartan treatment did not reduce fibrosis. These findings suggest that losartan treatment as an adjunct therapy to a regenerative rehabilitation strategy negatively impacts muscular function and fails to promote myogenesis following VML injury. There still remains a clinical need to develop a regenerative rehabilitation treatment strategy for traumatic skeletal muscle injuries. Future studies should consider optimizing the timing and duration of adjunct antifibrotic treatments to maximize functional outcomes in VML injuries.
Collapse
Affiliation(s)
- Jessica M. Motherwell
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Montgomery, MD 20815, USA
- Department of Surgery, Walter Reed National Military Medical Center, Uniformed Services University of the Health Sciences, Montgomery, MD 20815, USA
| | - Connor P. Dolan
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Montgomery, MD 20815, USA
- Department of Surgery, Walter Reed National Military Medical Center, Uniformed Services University of the Health Sciences, Montgomery, MD 20815, USA
| | - Sergey S. Kanovka
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Montgomery, MD 20815, USA
- Department of Surgery, Walter Reed National Military Medical Center, Uniformed Services University of the Health Sciences, Montgomery, MD 20815, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Montgomery, MD 20817, USA
| | - Jorge B. Edwards
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Montgomery, MD 20815, USA
- Department of Surgery, Walter Reed National Military Medical Center, Uniformed Services University of the Health Sciences, Montgomery, MD 20815, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Montgomery, MD 20817, USA
| | - Sarah R. Franco
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Montgomery, MD 20815, USA
- Department of Surgery, Walter Reed National Military Medical Center, Uniformed Services University of the Health Sciences, Montgomery, MD 20815, USA
| | - Naveena B. Janakiram
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Montgomery, MD 20815, USA
- Department of Surgery, Walter Reed National Military Medical Center, Uniformed Services University of the Health Sciences, Montgomery, MD 20815, USA
| | - Michael S. Valerio
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Montgomery, MD 20815, USA
- Department of Surgery, Walter Reed National Military Medical Center, Uniformed Services University of the Health Sciences, Montgomery, MD 20815, USA
| | - Stephen M. Goldman
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Montgomery, MD 20815, USA
- Department of Surgery, Walter Reed National Military Medical Center, Uniformed Services University of the Health Sciences, Montgomery, MD 20815, USA
| | - Christopher L. Dearth
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Montgomery, MD 20815, USA
- Department of Surgery, Walter Reed National Military Medical Center, Uniformed Services University of the Health Sciences, Montgomery, MD 20815, USA
| |
Collapse
|
11
|
Dolan CP, Clark AR, Motherwell JM, Janakiram NB, Valerio MS, Dearth CL, Goldman SM. The impact of bilateral injuries on the pathophysiology and functional outcomes of volumetric muscle loss. NPJ Regen Med 2022; 7:59. [PMID: 36243737 PMCID: PMC9569363 DOI: 10.1038/s41536-022-00255-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/05/2022] [Indexed: 11/09/2022] Open
Abstract
Volumetric muscle loss (VML)-defined as the irrecoverable loss of skeletal muscle tissue with associated persistent functional deficits-is among the most common and highly debilitating combat-related extremity injuries. This is particularly true in cases of severe polytrauma wherein multiple extremities may be involved as a result of high energy wounding mechanisms. As such, significant investment and effort has been made toward developing a clinically viable intervention capable of restoring the form and function of the affected musculature. While these investigations conducted to date have varied with respect to the species, breed, and sex of the chosen pre-clinical in-vivo model system, the majority of these studies have been performed in unilateral injury models, an aspect which may not fully exemplify the clinical representation of the multiply injured patient. Furthermore, while various components of the basal pathophysiology of VML (e.g., fibrosis and inflammation) have been investigated, relatively little effort has focused on how the pathophysiology and efficacy of pro-regenerative technologies is altered when there are multiple VML injuries. Thus, the purpose of this study was two-fold: (1) to investigate if/how the pathophysiology of unilateral VML injuries differs from bilateral VML injuries and (2) to interrogate the effect of bilateral VML injuries on the efficacy of a well-characterized regenerative therapy, minced muscle autograft (MMG). In contrast to our hypothesis, we show that bilateral VML injuries exhibit a similar systemic inflammatory response and improved muscle functional recovery, compared to unilateral injured animals. Furthermore, MMG treatment was found to only be effective at promoting an increase in functional outcomes in unilateral VML injuries. The findings presented herein add to the growing knowledge base of the pathophysiology of VML, and, importantly, reiterate the importance of comprehensively characterizing preclinical models which are utilized for early-stage screening of putative therapies as they can directly influence the translational research pipeline.
Collapse
Affiliation(s)
- Connor P Dolan
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, MD, USA.,Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Andrew R Clark
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, MD, USA.,Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Jessica M Motherwell
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, MD, USA.,Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Naveena B Janakiram
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, MD, USA.,Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Michael S Valerio
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, MD, USA.,Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Christopher L Dearth
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, MD, USA.,Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Stephen M Goldman
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, MD, USA. .,Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, USA.
| |
Collapse
|
12
|
Greising SM, Weiner JI, Garry DJ, Sachs DH, Garry MG. Human muscle in gene edited pigs for treatment of volumetric muscle loss. Front Genet 2022; 13:948496. [PMID: 35957684 PMCID: PMC9358139 DOI: 10.3389/fgene.2022.948496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/07/2022] [Indexed: 11/23/2022] Open
Abstract
Focusing on complex extremity trauma and volumetric muscle loss (VML) injuries, this review highlights: 1) the current pathophysiologic limitations of the injury sequela; 2) the gene editing strategy of the pig as a model that provides a novel treatment approach; 3) the notion that human skeletal muscle derived from gene edited, humanized pigs provides a groundbreaking treatment option; and 4) the impact of this technologic platform and how it will advance to far more multifaceted applications. This review seeks to shed insights on a novel treatment option using gene edited pigs as a platform which is necessary to overcome the clinical challenges and limitations in the field.
Collapse
Affiliation(s)
- Sarah M. Greising
- School of Kinesiology, University of Minnesota, Minneapolis, MN, United States
- *Correspondence: Sarah M. Greising, ; Mary G. Garry,
| | - Joshua I. Weiner
- Departments of Surgery, Columbia Center for Translations Immunology, College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Daniel J. Garry
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States
- NorthStar Genomics, Eagan, MN, United States
| | - David H. Sachs
- Departments of Surgery, Columbia Center for Translations Immunology, College of Physicians and Surgeons, Columbia University, New York, NY, United States
- Department of Surgery, Massachusetts General Hospital, Boston, MA, United States
| | - Mary G. Garry
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States
- NorthStar Genomics, Eagan, MN, United States
- *Correspondence: Sarah M. Greising, ; Mary G. Garry,
| |
Collapse
|
13
|
Hoffman DB, Raymond-Pope CJ, Sorensen JR, Corona BT, Greising SM. Temporal changes in the muscle extracellular matrix due to volumetric muscle loss injury. Connect Tissue Res 2022; 63:124-137. [PMID: 33535825 PMCID: PMC8364566 DOI: 10.1080/03008207.2021.1886285] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE/AIM Volumetric muscle loss (VML) is a devastating orthopedic injury resulting in chronic persistent functional deficits, loss of joint range of motion, pathologic fibrotic deposition and lifelong disability. However, there is only limited mechanistic understanding of VML-induced fibrosis. Herein we examined the temporal changes in the fibrotic deposition at 3, 7, 14, 28, and 48 days post-VML injury. MATERIALS AND METHODS Adult male Lewis rats (n = 39) underwent a full thickness ~20% (~85 mg) VML injury to the tibialis anterior (TA) muscle unilaterally, the contralateral TA muscle served as the control group. All TA muscles were harvested for biochemical and histologic evaluation. RESULTS The ratio of collagen I/III was decreased at 3, 7, and 14 days post-VML, but significantly increased at 48 days. Decorin content followed an opposite trend, significantly increasing by day 3 before dropping to below control levels by 48 days. Histological evaluation of the defect area indicates a shift from loosely packed collagen at early time points post-VML, to a densely packed fibrotic scar by 48 days. CONCLUSIONS The shift from early wound healing efforts to a fibrotic scar with densely packed collagen within the skeletal muscle occurs around 21 days after VML injury through dogmatic synchronous reduction of collagen III and increase in collagen I. Thus, there appears to be an early window for therapeutic intervention to prevent pathologic fibrous tissue formation, potentially by targeting CCN2/CTGF or using decorin as a therapeutic.
Collapse
Affiliation(s)
- Daniel B. Hoffman
- School of Kinesiology, University of Minnesota, Minneapolis MN 55455
| | | | - Jacob R. Sorensen
- School of Kinesiology, University of Minnesota, Minneapolis MN 55455
| | | | - Sarah M. Greising
- School of Kinesiology, University of Minnesota, Minneapolis MN 55455;,For reprints contact: Sarah M. Greising, Ph.D., 1900 University Ave SE, 220A Cooke Hall, Minneapolis MN, 55455, , Phone: 612-626-7890, Fax: 612-626-7700
| |
Collapse
|
14
|
Quint JP, Samandari M, Abbasi L, Mollocana E, Rinoldi C, Mostafavi A, Tamayol A. Nanoengineered myogenic scaffolds for skeletal muscle tissue engineering. NANOSCALE 2022; 14:797-814. [PMID: 34951427 PMCID: PMC8900679 DOI: 10.1039/d1nr06143g] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Extreme loss of skeletal muscle overwhelms the natural regenerative capability of the body, results in permanent disability and substantial economic burden. Current surgical techniques result in poor healing, secondary injury to the autograft donor site, and incomplete recuperation of muscle function. Most current tissue engineering and regenerative strategies fail to create an adequate mechanical and biological environment that enables cell infiltration, proliferation, and myogenic differentiation. In this study, we present a nanoengineered skeletal muscle scaffold based on functionalized gelatin methacrylate (GelMA) hydrogel, optimized for muscle progenitors' proliferation and differentiation. The scaffold was capable of controlling the release of insulin-like growth factor 1 (IGF-1), an important myogenic growth factor, by utilizing the electrostatic interactions with LAPONITE® nanoclays (NCs). Physiologically relevant levels of IGF-1 were maintained during a controlled release over two weeks. The NC was able to retain 50% of the released IGF-1 within the hydrogel niche, significantly improving cellular proliferation and differentiation compared to control hydrogels. IGF-1 supplemented medium controls required 44% more IGF-1 than the comparable NC hydrogel composites. The nanofunctionalized scaffold is a viable option for the treatment of extreme muscle injuries and offers scalable benefits for translational interventions and the growing field of clean meat production.
Collapse
Affiliation(s)
- Jacob P Quint
- Department of Biomedical Engineering, University of Connecticut, Farmington, CT 06030, USA.
| | - Mohamadmahdi Samandari
- Department of Biomedical Engineering, University of Connecticut, Farmington, CT 06030, USA.
| | - Laleh Abbasi
- Department of Molecular, Cellular & Biomedical Sciences, The City College of New York, New York, NY, 10031, USA
| | - Evelyn Mollocana
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE, 68588, USA
| | - Chiara Rinoldi
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Azadeh Mostafavi
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE, 68588, USA
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut, Farmington, CT 06030, USA.
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
15
|
Hadipour A, Bayati V, Rashno M, Orazizadeh M. Aligned Poly(ε-caprolactone) Nanofibers Superimposed on Decellularized Human Amniotic Membrane Promoted Myogenic Differentiation of Adipose Derived Stem Cells. CELL JOURNAL 2021; 23:603-611. [PMID: 34939752 PMCID: PMC8665975 DOI: 10.22074/cellj.2021.7261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 06/10/2020] [Indexed: 11/29/2022]
Abstract
Objective This study was designed to fabricate a suitable permanent scaffold for the normal aligned myotube formation
and improve the process of myogenic differentiation of selected stem cells.
Materials and Methods In this experimental study, an engineered scaffold composed of decellularized human amniotic
membrane (DHAM) and electrospun fibers of poly(ε-caprolactone) (PCL) was fabricated and characterized. PCL
nanofibers were superimposed on DHAM (PCL-DHAM) in two different patterns, including randomized fibers (Random)
and aligned fibers (Aligned). Adipose derived stem cells (ADSCs) were isolated from adult Wistar rats and cultured on
designed scaffold and induced to myotube differentiation. Using an MTT assay, the vitality of cells was determined.
Then, myogenic cell differentiation was assessed using scan electron microscopy (SEM), immunofluorescence assay,
and reverse transcription-polymerase chain reaction (RT-PCR).
Results The mechanical properties of engineered PCL-DHAM composite improved significantly compared to DHAM
as a control. The engineered PCL-DHAM promoted cell growth and high expression of myosin, Mhc2 and myogenin
and thus enhanced the myotube formation.
Conclusion These findings revealed that bio-composite membrane prepared from PCL nanofibers and DHAM, may
represent a promising biomaterial as a desirable scaffold for applying in the bioengineered muscle repair.
Collapse
Affiliation(s)
- Azam Hadipour
- Cellular and Molecular Research Center (CMRC), Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Vahid Bayati
- Cellular and Molecular Research Center (CMRC), Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Rashno
- Cellular and Molecular Research Center (CMRC), Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahmoud Orazizadeh
- Cellular and Molecular Research Center (CMRC), Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
16
|
Westman AM, Peirce SM, Christ GJ, Blemker SS. Agent-based model provides insight into the mechanisms behind failed regeneration following volumetric muscle loss injury. PLoS Comput Biol 2021; 17:e1008937. [PMID: 33970905 PMCID: PMC8110270 DOI: 10.1371/journal.pcbi.1008937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/01/2021] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscle possesses a remarkable capacity for repair and regeneration following a variety of injuries. When successful, this highly orchestrated regenerative process requires the contribution of several muscle resident cell populations including satellite stem cells (SSCs), fibroblasts, macrophages and vascular cells. However, volumetric muscle loss injuries (VML) involve simultaneous destruction of multiple tissue components (e.g., as a result of battlefield injuries or vehicular accidents) and are so extensive that they exceed the intrinsic capability for scarless wound healing and result in permanent cosmetic and functional deficits. In this scenario, the regenerative process fails and is dominated by an unproductive inflammatory response and accompanying fibrosis. The failure of current regenerative therapeutics to completely restore functional muscle tissue is not surprising considering the incomplete understanding of the cellular mechanisms that drive the regeneration response in the setting of VML injury. To begin to address this profound knowledge gap, we developed an agent-based model to predict the tissue remodeling response following surgical creation of a VML injury. Once the model was able to recapitulate key aspects of the tissue remodeling response in the absence of repair, we validated the model by simulating the tissue remodeling response to VML injury following implantation of either a decellularized extracellular matrix scaffold or a minced muscle graft. The model suggested that the SSC microenvironment and absence of pro-differentiation SSC signals were the most important aspects of failed muscle regeneration in VML injuries. The major implication of this work is that agent-based models may provide a much-needed predictive tool to optimize the design of new therapies, and thereby, accelerate the clinical translation of regenerative therapeutics for VML injuries.
Collapse
Affiliation(s)
- Amanda M. Westman
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Shayn M. Peirce
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Ophthalmology, University of Virginia, Charlottesville, Virginia, United States of America
| | - George J. Christ
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail: (GJC); (SSB)
| | - Silvia S. Blemker
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Ophthalmology, University of Virginia, Charlottesville, Virginia, United States of America
- Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
- Mechanical and Aerospace Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail: (GJC); (SSB)
| |
Collapse
|
17
|
Quint JP, Mostafavi A, Endo Y, Panayi A, Russell CS, Nourmahnad A, Wiseman C, Abbasi L, Samandari M, Sheikhi A, Nuutila K, Sinha I, Tamayol A. In Vivo Printing of Nanoenabled Scaffolds for the Treatment of Skeletal Muscle Injuries. Adv Healthc Mater 2021; 10:e2002152. [PMID: 33644996 PMCID: PMC8137605 DOI: 10.1002/adhm.202002152] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Indexed: 01/24/2023]
Abstract
Extremity skeletal muscle injuries result in substantial disability. Current treatments fail to recoup muscle function, but properly designed and implemented tissue engineering and regenerative medicine techniques can overcome this challenge. In this study, a nanoengineered, growth factor-eluting bioink that utilizes Laponite nanoclay for the controlled release of vascular endothelial growth factor (VEGF) and a GelMA hydrogel for a supportive and adhesive scaffold that can be crosslinked in vivo is presented. The bioink is delivered with a partially automated handheld printer for the in vivo formation of an adhesive and 3D scaffold. The effect of the controlled delivery of VEGF alone or paired with adhesive, supportive, and fibrilar architecture has not been studied in volumetric muscle loss (VML) injuries. Upon direct in vivo printing, the constructs are adherent to skeletal muscle and sustained release of VEGF. The in vivo printing of muscle ink in a murine model of VML injury promotes functional muscle recovery, reduced fibrosis, and increased anabolic response compared to untreated mice. The in vivo construction of a therapeutic-eluting 3D scaffold paves the way for the immediate treatment of a variety of soft tissue traumas.
Collapse
Affiliation(s)
- Jacob P. Quint
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE, 68588, USA
- Department of Biomedical Engineering, University of Connecticut, Farmington, CT 06030, USA
| | - Azadeh Mostafavi
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE, 68588, USA
| | - Yori Endo
- Division of Plastic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Adriana Panayi
- Division of Plastic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Carina S. Russell
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE, 68588, USA
| | - Atousa Nourmahnad
- Division of Plastic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Chris Wiseman
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE, 68588, USA
| | - Laleh Abbasi
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE, 68588, USA
| | - Mohamadmahdi Samandari
- Department of Biomedical Engineering, University of Connecticut, Farmington, CT 06030, USA
| | - Amir Sheikhi
- Department of Chemical Engineering, Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Kristo Nuutila
- Division of Plastic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Indranil Sinha
- Division of Plastic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Ali Tamayol
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE, 68588, USA
- Department of Biomedical Engineering, University of Connecticut, Farmington, CT 06030, USA
| |
Collapse
|
18
|
Sorensen JR, Hoffman DB, Corona BT, Greising SM. Secondary denervation is a chronic pathophysiologic sequela of volumetric muscle loss. J Appl Physiol (1985) 2021; 130:1614-1625. [PMID: 33830817 DOI: 10.1152/japplphysiol.00049.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Volumetric muscle loss (VML) is the traumatic loss of muscle tissue that results in long-term functional impairments. Despite the loss of myofibers, there remains an unexplained significant decline in muscle function. VML injury likely extends beyond the defect area, causing negative secondary outcomes to the neuromuscular system, including the neuromuscular junctions (NMJs), yet the extent to which VML induces denervation is unclear. This study systematically examined NMJs surrounding the VML injury, hypothesizing that the sequela of VML includes denervation. The VML injury removed ∼20% of the tibialis anterior (TA) muscle in adult male inbred Lewis rats (n = 43), the noninjured leg served as an intra-animal control. Muscles were harvested up to 48 days post-VML. Synaptic terminals were identified immunohistochemically, and quantitative confocal microscopy evaluated 2,613 individual NMJ. Significant denervation was apparent by 21 and 48 days post-VML. Initially, denervation increased ∼10% within 3 days of injury; with time, denervation further increased to ∼22% and 32% by 21 and 48 days post-VML, respectively, suggesting significant secondary denervation. The appearance of terminal axon sprouting and polyinnervation were observed as early as 7 days post-VML, increasing in number and complexity throughout 48 days. There was no evidence of VML-induced NMJ size alteration, which may be beneficial for interventions aimed at restoring muscle function. This work recognizes VML-induced secondary denervation and poor remodeling of the NMJ as part of the sequela of VML injury; moreover, secondary denervation is a possible contributing factor to the chronic functional impairments and potentially an overlooked treatment target.NEW & NOTEWORTHY This work advances our understanding of the pathophysiologic complexity of volumetric muscle loss injury. Specifically, we identified secondary denervation in the muscle remaining after volumetric muscle loss injuries as a novel aspect of the injury sequela. Denervation increased chronically, in parallel with the appearance of irregular morphological characteristics and destabilization of the neuromuscular junction, which is expected to further confound chronic functional impairments.
Collapse
Affiliation(s)
- Jacob R Sorensen
- School of Kinesiology, University of Minnesota, Minneapolis, Minnesota
| | - Daniel B Hoffman
- School of Kinesiology, University of Minnesota, Minneapolis, Minnesota
| | - Benjamin T Corona
- School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Sarah M Greising
- School of Kinesiology, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
19
|
Khodabukus A. Tissue-Engineered Skeletal Muscle Models to Study Muscle Function, Plasticity, and Disease. Front Physiol 2021; 12:619710. [PMID: 33716768 PMCID: PMC7952620 DOI: 10.3389/fphys.2021.619710] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/25/2021] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle possesses remarkable plasticity that permits functional adaptations to a wide range of signals such as motor input, exercise, and disease. Small animal models have been pivotal in elucidating the molecular mechanisms regulating skeletal muscle adaptation and plasticity. However, these small animal models fail to accurately model human muscle disease resulting in poor clinical success of therapies. Here, we review the potential of in vitro three-dimensional tissue-engineered skeletal muscle models to study muscle function, plasticity, and disease. First, we discuss the generation and function of in vitro skeletal muscle models. We then discuss the genetic, neural, and hormonal factors regulating skeletal muscle fiber-type in vivo and the ability of current in vitro models to study muscle fiber-type regulation. We also evaluate the potential of these systems to be utilized in a patient-specific manner to accurately model and gain novel insights into diseases such as Duchenne muscular dystrophy (DMD) and volumetric muscle loss. We conclude with a discussion on future developments required for tissue-engineered skeletal muscle models to become more mature, biomimetic, and widely utilized for studying muscle physiology, disease, and clinical use.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
20
|
McClure MJ, Olson LC, Cohen DJ, Huang YC, Zhang S, Nguyen T, Boyan BD, Schwartz Z. RNU ( Foxn1
RNU-Nude) Rats Demonstrate an Improved Ability to Regenerate Muscle in a Volumetric Muscle Injury Compared to Sprague Dawley Rats. Bioengineering (Basel) 2021; 8:bioengineering8010012. [PMID: 33467489 PMCID: PMC7830033 DOI: 10.3390/bioengineering8010012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 01/13/2023] Open
Abstract
Products developed for skeletal muscle regeneration frequently incorporate allogeneic and xenogeneic materials to elicit a regenerative response to heal skeletal muscle wounds. To avoid graft rejection in preclinical studies, immunodeficient rodents are used. Whether the immunodeficiency alters the host response to the material in skeletal muscle has not been studied. In this study, we hypothesized that an allogeneic acellular skeletal muscle grafts implanted in an immunodeficient rat (RNU, Foxn1-deficient) would exhibit better new muscle fiber formation compared to grafts implanted in immunocompetent Sprague Dawley (SD) rats. Decellularized SD skeletal muscle matrix (DMM) was implanted in the gastrocnemius (N = 8 rats/group). 56 days after surgery, animal gait was examined and animals were euthanized. Muscle force was assessed and fiber number as well as immune cell infiltrate was measured by histomorphometry and immunohistochemistry. Animal gait and percent recovery of muscle force were unchanged in both groups, but newly regenerated muscle fibers increased in RNU rats. Macrophage staining for CD68 was higher in RNU rats than in SD rats. These data show differences in muscle regeneration between animal models using the same biomaterial treatment, but these differences could not be ascribed to the immune response. Overall, our data provide awareness that more studies are needed to understand how host responses to biomaterials differ based on the animal model used.
Collapse
Affiliation(s)
- Michael J. McClure
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (L.C.O.); (D.J.C.); (S.Z.); (T.N.); (B.D.B.); (Z.S.)
- Correspondence: ; Tel.: +1-804-828-8337
| | - Lucas C. Olson
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (L.C.O.); (D.J.C.); (S.Z.); (T.N.); (B.D.B.); (Z.S.)
| | - David J. Cohen
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (L.C.O.); (D.J.C.); (S.Z.); (T.N.); (B.D.B.); (Z.S.)
| | | | - Shirley Zhang
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (L.C.O.); (D.J.C.); (S.Z.); (T.N.); (B.D.B.); (Z.S.)
| | - Tri Nguyen
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (L.C.O.); (D.J.C.); (S.Z.); (T.N.); (B.D.B.); (Z.S.)
| | - Barbara D. Boyan
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (L.C.O.); (D.J.C.); (S.Z.); (T.N.); (B.D.B.); (Z.S.)
- Wallace H. Coulter Department of Biomedical Engineering, College of Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (L.C.O.); (D.J.C.); (S.Z.); (T.N.); (B.D.B.); (Z.S.)
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
21
|
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
|
22
|
Goldman SM, Feng JP, Corona BT. Volumetric muscle loss disrupts length-dependent architectural and functional characteristics of skeletal muscle. Connect Tissue Res 2021; 62:72-82. [PMID: 32660287 DOI: 10.1080/03008207.2020.1789608] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose/Aim: Skeletal muscle architecture is a primary determinant of function. Volumetric muscle loss (VML) injury is destructive; however, the impact on muscle architecture is uncharacterized. Methods: Architectural and functional effects of VML were assessed in rat tibialis anterior (TA) muscle model 4 weeks post-injury. Results: VML caused a 31% and 33% reduction in muscle weight (p < 0.001) and fiber length (p = 0.002), respectively, culminating a 34% reduction of fiber to muscle length ratio (FL:ML; p < 0.001). Fiber pennation angle (+14%; p = 0.150) and physiological cross-sectional area (PCSA; -12%; p = 0.220) were unchanged. VML injury reduced peak isometric force (Po) by 36% (p < 0.001), specific force (sPo = Po/PCSA) by 41% (vs. Po, p > 0.999), and force per gram muscle weight (Po/mw) by 18% (vs. Po, p < 0.001). VML injury increased the length at which Po was produced (Lo) by 8% (p = 0.009), and reduced functional excursion by 35% (p = 0.035). Conclusion: The architectural changes after VML injury preserved PCSA, and therefore preserved "potential" maximal force-producing capacity. At most, only half the Po deficit was due directly to the cumulative effect of horizontal and longitudinal tissue loss. Highlighting the impact of longitudinal muscle loss, VML injury reduced fiber length, and FL:ML and grossly disrupted length-dependent functional properties. These findings raise the importance of augmenting length-dependent muscle properties to optimize functional recovery after VML injury.
Collapse
Affiliation(s)
- Stephen M Goldman
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research , Fort Sam Houston, TX, USA.,DoD-VA Extremity Trauma and Amputation Center of Excellence , Fort Sam Houston, TX, USA.,Department of Surgery, Uniformed Services University of the Health Sciences , Bethesda, MD, USA.,Department of Surgery, Walter Reed National Military Medical Center , Bethesda, MD, USA
| | - Jonathan P Feng
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research , Fort Sam Houston, TX, USA
| | - Benjamin T Corona
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research , Fort Sam Houston, TX, USA
| |
Collapse
|
23
|
Sicherer ST, Venkatarama RS, Grasman JM. Recent Trends in Injury Models to Study Skeletal Muscle Regeneration and Repair. Bioengineering (Basel) 2020; 7:bioengineering7030076. [PMID: 32698352 PMCID: PMC7552705 DOI: 10.3390/bioengineering7030076] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/14/2020] [Accepted: 07/18/2020] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscle injuries that occur from traumatic incidents, such as those caused by car accidents or surgical resections, or from injuries sustained on the battlefield, result in the loss of functionality of the injured muscle. To understand skeletal muscle regeneration and to better treat these large scale injuries, termed volumetric muscle loss (VML), in vivo injury models exploring the innate mechanisms of muscle injury and repair are essential for the creation of clinically applicable treatments. While the end result of a muscle injury is often the destruction of muscle tissue, the manner in which these injuries are induced as well as the response from the innate repair mechanisms found in muscle in each animal models can vary. This targeted review describes injury models that assess both skeletal muscle regeneration (i.e., the response of muscle to myotoxin or ischemic injury) and skeletal muscle repair (i.e., VML injury). We aimed to summarize the injury models used in the field of skeletal muscle tissue engineering, paying particular attention to strategies to induce muscle damage and how to standardize injury conditions for future experiments.
Collapse
|
24
|
Martin RA, Buckley KH, Mankowski DC, Riley BM, Sidwell AN, Douglas SL, Worth RG, Pizza FX. Myogenic Cell Expression of Intercellular Adhesion Molecule-1 Contributes to Muscle Regeneration after Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:2039-2055. [PMID: 32650005 DOI: 10.1016/j.ajpath.2020.06.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 06/10/2020] [Accepted: 06/15/2020] [Indexed: 10/23/2022]
Abstract
This study investigated intercellular adhesion molecule-1 (ICAM-1), a membrane protein that mediates cell-to-cell adhesion and communication, as a mechanism through which the inflammatory response facilitates muscle regeneration after injury. Toxin-induced muscle injury to tibialis anterior muscles of wild-type mice caused ICAM-1 to be expressed by a population of satellite cells/myoblasts and myofibers. Myogenic cell expression of ICAM-1 contributed to the restoration of muscle structure after injury, as regenerating myofibers were more abundant and myofiber size was larger for wild-type compared with Icam1-/- mice during 28 days of recovery. Contrastingly, restoration of muscle function after injury was similar between the genotypes. ICAM-1 facilitated the restoration of muscle structure after injury through mechanisms involving the regulation of myofiber branching, protein synthesis, and the organization of nuclei within myofibers after myogenic cell fusion. These findings provide support for a paradigm in which ICAM-1 expressed by myogenic cells after muscle injury augments their adhesive and fusogenic properties, which, in turn, facilitates regenerative and hypertrophic processes that restore structure to injured muscle.
Collapse
Affiliation(s)
- Ryan A Martin
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Kole H Buckley
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Drew C Mankowski
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Benjamin M Riley
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Alena N Sidwell
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Stephanie L Douglas
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Randall G Worth
- Department of Medical Microbiology and Immunology, The University of Toledo, Toledo, Ohio
| | - Francis X Pizza
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio.
| |
Collapse
|
25
|
Leiva-Cepas F, Jimena I, Ruz-Caracuel I, Luque E, Villalba R, Peña-Amaro J. Histology of skeletal muscle reconstructed by means of the implantation of autologous adipose tissue: an experimental study. Histol Histopathol 2020; 35:457-474. [PMID: 31523800 DOI: 10.14670/hh-18-163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The purpose of this study was to determine the histological characteristics of a skeletal muscle reconstructed by means of the implantation of autologous adipose tissue following an experimentally-induced volumetric muscle loss. A cylindrical piece in the belly of the rat anterior tibial muscle was removed. In the hole, inguinal subcutaneous adipose tissue of the same rat was grafted. Animals were sacrificed 7, 14, 21, 28 and 60 days posttransplantation. Histological, histochemical, immunohistochemical and morphometric techniques were used. At all times analyzed, the regenerative muscle fibers formed from the edges of the muscle tissue showed histological, histochemical and immunohistochemical differences in comparison with the control group. These differences are related to delays in the maturation process and are related to problems in reinnervation and disorientation of muscle fibers. The stains for MyoD and desmin showed that some myoblasts and myotubes seem to derive from the transplanted adipose tissue. After 60 days, the transplant area was 20% occupied by fibrosis and by 80% skeletal muscle. However, the neo-muscle was chaotically organized showing muscle fiber disorientation and centronucleated fibers with irregular shape and size. Our results support the hypothesis that, at least from a morphological point of view, autologous adipose tissue transplantation favors reconstruction following a volumetric loss of skeletal muscle by combining the inherent regenerative response of the organ itself and the myogenic differentiation of the stem cells present in the adipose tissue. However, in our study, the formed neo-muscle exhibited histological differences in comparison with the normal skeletal muscle.
Collapse
Affiliation(s)
- Fernando Leiva-Cepas
- Department of Morphological Sciences, Section of Histology, Faculty of Medicine and Nursing, University of Cordoba, Córdoba, Spain
- Research Group in Muscle Regeneration, University of Cordoba, Córdoba, Spain
- Maimonides Institute for Biomedical Research IMIBIC, Reina Sofia University Hospital, University of Cordoba, Spain
- Present address: Department of Pathology, Reina Sofia University Hospital, Córdoba, Spain
| | - Ignacio Jimena
- Department of Morphological Sciences, Section of Histology, Faculty of Medicine and Nursing, University of Cordoba, Córdoba, Spain
- Research Group in Muscle Regeneration, University of Cordoba, Córdoba, Spain
- Maimonides Institute for Biomedical Research IMIBIC, Reina Sofia University Hospital, University of Cordoba, Córdoba, Spain
| | - Ignacio Ruz-Caracuel
- Department of Morphological Sciences, Section of Histology, Faculty of Medicine and Nursing, University of Cordoba, Córdoba, Spain
- Research Group in Muscle Regeneration, University of Cordoba, Córdoba, Spain
- Present address: Department of Pathology, Ramón y Cajal University Hospital, Madrid, Spain
| | - Evelio Luque
- Department of Morphological Sciences, Section of Histology, Faculty of Medicine and Nursing, University of Cordoba, Córdoba, Spain
- Maimonides Institute for Biomedical Research IMIBIC, Reina Sofia University Hospital, University of Cordoba, Córdoba, Spain
| | - Rafael Villalba
- Tissue of Establishment of the Center for Transfusion, Tissues and Cells, Córdoba, Spain
| | - Jose Peña-Amaro
- Research Group in Muscle Regeneration, University of Cordoba, Córdoba, Spain
- Maimonides Institute for Biomedical Research IMIBIC, Reina Sofia University Hospital, University of Cordoba, Córdoba, Spain
- Department of Morphological Sciences, Section of Histology, Faculty of Medicine and Nursing, University of Cordoba, Córdoba, Spain.
| |
Collapse
|
26
|
Kim J, Kasukonis B, Roberts K, Dunlap G, Brown L, Washington T, Wolchok J. Graft alignment impacts the regenerative response of skeletal muscle after volumetric muscle loss in a rat model. Acta Biomater 2020; 105:191-202. [PMID: 31978621 DOI: 10.1016/j.actbio.2020.01.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 01/01/2023]
Abstract
A key event in the etiology of volumetric muscle loss (VML) injury is the bulk loss of structural cues provided by the underlying extracellular matrix (ECM). To re-establish the lost cues, there is broad consensus within the literature supporting the utilization of implantable scaffolding. However, while scaffold based regenerative medicine strategies have shown potential, there remains a significant amount of outcome variability observed across the field. We suggest that an overlooked source of outcome variability is differences in scaffolding architecture. The goal of this study was to test the hypothesis that implant alignment has a significant impact on genotypic and phenotypic outcomes following the repair of VML injuries. Using a rat VML model, outcomes across three autograft implant treatment groups (aligned implants, 45° misaligned, and 90° misaligned) and two recovery time points (2 weeks and 12 weeks) were examined (n = 6-8/group). At 2 weeks post-repair there were no significant differences in muscle mass and torque recovery between the treatment groups, however we did observe a significant upregulation of MyoD (2.5 fold increase) and Pax7 (2 fold increase) gene expression as well as the presence of immature myofibers at the implant site for those animals repaired with aligned autografts. By 12 weeks post-repair, functional and structural differences between the treatment groups could be detected. Aligned autografts had significantly greater mass and torque recovery (77 ± 10% of normal) when compared to 45° and 90° misaligned autografts (64 ± 10% and 61 ± 11%, respectively). Examination of tissue structure revealed extensive fibrosis and a significant increase in non-contractile tissue area fraction for only those animals treated using misaligned autografts. When taken together, the results suggest that implant graft orientation has a significant impact on in-vivo outcomes and indicate that the effect of graft alignment on muscle phenotype may be mediated through genotypic changes to myogenesis and fibrosis at the site of injury and repair. STATEMENT OF SIGNIFICANCE: A key event in the etiology of volumetric muscle loss injury is the bulk loss of architectural cues provided by the underlying extracellular matrix. To re-establish the lost cues, there is broad consensus within the literature supporting the utilization of implantable scaffolding. Yet, although native muscle is a highly organized tissue with network and cellular alignment in the direction of contraction, there is little evidence within the field concerning the importance of re-establishing native architectural alignment. The results of this study suggest that critical interactions exist between implant and native muscle alignment cues during healing, which influence the balance between myogenesis and fibrosis. Specifically, it appears that alignment of implant architectural cues with native muscle cues is necessary to create a pro-myogenic environment and contractile force recovery. The results also suggest that misaligned cues may be pathological, leading to fibrosis and poor contractile force recovery.
Collapse
Affiliation(s)
- John Kim
- Department of Biomedical Engineering, College of Engineering, University of Arkansas, Fayetteville, AR, United States
| | - Ben Kasukonis
- Department of Biomedical Engineering, College of Engineering, University of Arkansas, Fayetteville, AR, United States
| | - Kevin Roberts
- Department of Biomedical Engineering, College of Engineering, University of Arkansas, Fayetteville, AR, United States; Department of Health, Human Performance, and Recreation, College of Education and Health Professions, University of Arkansas, Fayetteville, AR, United States
| | - Grady Dunlap
- Department of Biomedical Engineering, College of Engineering, University of Arkansas, Fayetteville, AR, United States
| | - Lemuel Brown
- Department of Health, Human Performance, and Recreation, College of Education and Health Professions, University of Arkansas, Fayetteville, AR, United States
| | - Tyrone Washington
- Department of Health, Human Performance, and Recreation, College of Education and Health Professions, University of Arkansas, Fayetteville, AR, United States
| | - Jeffrey Wolchok
- Department of Biomedical Engineering, College of Engineering, University of Arkansas, Fayetteville, AR, United States.
| |
Collapse
|
27
|
Jia W, Hu H, Li A, Deng H, Hogue CL, Mauro JC, Zhang C, Fu Q. Glass-activated regeneration of volumetric muscle loss. Acta Biomater 2020; 103:306-317. [PMID: 31830584 DOI: 10.1016/j.actbio.2019.12.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/02/2019] [Accepted: 12/05/2019] [Indexed: 12/15/2022]
Abstract
Volumetric muscle loss (VML) resulting from injuries to skeletal muscles has profound consequences in healthcare. Current VML treatment based on the use of soft materials including biopolymers and decellularized extracellular matrix (dECM) is challenging due to their incapability of stimulating the formation of satellite cells (SCs), muscle stem cells, which are required for muscle regeneration. Additional stem cells and/or growth factors have to be incorporated in these constructs for improved efficacy. Here we report an approach by using bioactive glasses capable of regenerating VML without growth factors or stem cells. One silicate and two borate compositions with different degradation rates (2.4% for silicate 45S5; 5.3% and 30.4% for borate 8A3B and 13-93B3, respectively, in simulated body fluid (SBF) at 37 °C for 30 days) were used for this study. Our in vitro models demonstrate the ability of ions released from bioactive glasses in promoting angiogenesis and stimulating cells to secrete critical muscle-related growth factors. We further show the activation of SCs and the regeneration of skeletal muscles in a rat VML model. Considering these promising results, this work reveals a potentially simple and safe approach to regenerating skeletal muscle defects. STATEMENT OF SIGNIFICANCE: (1) This is the first report on an inorganic material used in skeletal muscle regeneration through in vitro and in vivo models. (2) Bioactive glass is found to activate the production of satellite cells (SCs), muscle stem cells, without the incorporation of extra stem cells or growth factors. (3) The work represents a simple, safe, low-cost yet efficient means for healing muscle defects.
Collapse
|
28
|
Kim JT, Kasukonis B, Dunlap G, Perry R, Washington T, Wolchok JC. Regenerative Repair of Volumetric Muscle Loss Injury is Sensitive to Age. Tissue Eng Part A 2020; 26:3-14. [PMID: 31064280 PMCID: PMC6983754 DOI: 10.1089/ten.tea.2019.0034] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/25/2019] [Indexed: 12/20/2022] Open
Abstract
In this study, the influence of age on effectiveness of regenerative repair for the treatment of volumetric muscle loss (VML) injury was explored. Tibialis anterior (TA) VML injuries were repaired in both 3- and 18-month-old animal models (Fischer 344 rat) using allogeneic decellularized skeletal muscle (DSM) scaffolds supplemented with autologous minced muscle (MM) paste. Within the 3-month animal group, TA peak contractile force was significantly improved (79% of normal) in response to DSM+MM repair. However, within the 18-month animal group, muscle force following repair (57% of normal) was not significantly different from unrepaired VML controls (59% of normal). Within the 3-month animal group, repair with DSM+MM generally reduced scarring at the site of VML repair, whereas scarring and a loss of contractile tissue was notable at the site of repair within the 18-month group. Within 3-month animals, expression of myogenic genes (MyoD, MyoG), extracellular matrix genes (Col I, Col III, TGF-β), and key wound healing genes (TNF-α and IL-1β) were increased. Alternatively, expression was unchanged across all genes examined within the 18-month animal group. The findings suggest that a decline in regenerative capacity and increased fibrosis with age may present an obstacle to regenerative medicine strategies targeting VML injury. Impact Statement This study compared the recovery following volumetric muscle loss (VML) injury repair using a combination of minced muscle paste and decellularized muscle extracellular matrix carrier in both a younger (3 months) and older (18 months) rat population. Currently, VML repair research is being conducted with the young patient population in mind, but our group is the first to look at the effects of age on the efficacy of VML repair. Our findings highlight the importance of considering age-related changes in response to VML when developing repair strategies targeting an elderly patient population.
Collapse
Affiliation(s)
- John T. Kim
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Benjamin Kasukonis
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Grady Dunlap
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Richard Perry
- Department of Health, Human Performance, and Recreation, College of Education and Health Professions, University of Arkansas, Fayetteville, Arkansas
| | - Tyrone Washington
- Department of Health, Human Performance, and Recreation, College of Education and Health Professions, University of Arkansas, Fayetteville, Arkansas
| | - Jeffrey C. Wolchok
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| |
Collapse
|
29
|
Greising SM, Corona BT, McGann C, Frankum JK, Warren GL. Therapeutic Approaches for Volumetric Muscle Loss Injury: A Systematic Review and Meta-Analysis. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:510-525. [PMID: 31578930 DOI: 10.1089/ten.teb.2019.0207] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Our goal was to understand the impact of regenerative therapies on the functional capacity of skeletal muscle following volumetric muscle loss (VML) injury. An extensive database search (e.g., PubMed, Cochrane Library, and ClinicalTrials.gov) was conducted up through January 2019 to evaluate the following: "In humans or animals with VML injury, is treatment better than no treatment at recovering functional capacity?" Study eligibility criteria required studies to have both an untreated and at least one treated VML injury group. From 2312 study reports, 44 studies met the inclusion criteria. Quantitative functional capacity data (absolute and/or normalized strength) or proportional measures (histological analysis quantifying viable muscle tissue, mitochondrial function, and/or exhaustive treadmill running) were extracted for use. While both human and animal studies were included in the searches, only animal studies met the eligibility criteria. Using a random-effects model, Hedges' g was used as the effect size (ES) and calculated such that a positive ES indicated treatment efficacy. The overall ES was 0.75 (95% confidence interval: 0.53-0.96; p < 0.0000001), indicating that the treatments, on average, resulted in a significant improvement in functional capacity. From network meta-analyses, it was determined that an acellular biomaterial combined with stem and/or progenitor cells had the greatest treatment effectiveness. The findings indicate that various treatments in animal models of VML improve the functional capacity of muscle compared to leaving the injury untreated; however, the ∼16% beneficial effect is small. Our results suggest that current regenerative therapy paradigms require further maturation to achieve clinically meaningful improvements in the functional capacity of the muscle. Impact Statement Our most salient findings are that (1) various treatment approaches used in animal models of volumetric muscle loss (VML) injury improve functional capacity compared to leaving the injury untreated and (2) an acellular biomaterial in combination with cellular components was the most effective treatment to improve functional capacity following VML injury to date. The nature of our findings has substantial implications for regenerative medicine, biomedical engineering, and rehabilitative techniques currently being evaluated and developed for VML injury repair, and are pivotal to the progression of the regenerative medicine effort aimed at restoring maximal function to traumatized and disabled limbs.
Collapse
Affiliation(s)
- Sarah M Greising
- School of Kinesiology, University of Minnesota, Minneapolis, Minnesota
| | - Benjamin T Corona
- School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Christopher McGann
- Department of Physical Therapy, Georgia State University, Atlanta, Georgia
| | - Jeremy K Frankum
- Department of Physical Therapy, Georgia State University, Atlanta, Georgia
| | - Gordon L Warren
- Department of Physical Therapy, Georgia State University, Atlanta, Georgia
| |
Collapse
|
30
|
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
|
31
|
Dienes JA, Hu X, Janson KD, Slater C, Dooley EA, Christ GJ, Russell SD. Analysis and Modeling of Rat Gait Biomechanical Deficits in Response to Volumetric Muscle Loss Injury. Front Bioeng Biotechnol 2019; 7:146. [PMID: 31275932 PMCID: PMC6593045 DOI: 10.3389/fbioe.2019.00146] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/31/2019] [Indexed: 12/22/2022] Open
Abstract
There is currently a substantial volume of research underway to develop more effective approaches for the regeneration of functional muscle tissue as treatment for volumetric muscle loss (VML) injury, but few studies have evaluated the relationship between injury and the biomechanics required for normal function. To address this knowledge gap, the goal of this study was to develop a novel method to quantify the changes in gait of rats with tibialis anterior (TA) VML injuries. This method should be sensitive enough to identify biomechanical and kinematic changes in response to injury as well as during recovery. Control rats and rats with surgically-created VML injuries were affixed with motion capture markers on the bony landmarks of the back and hindlimb and were recorded walking on a treadmill both prior to and post-surgery. Data collected from the motion capture system was exported for post-hoc analysis in OpenSim and Matlab. In vivo force testing indicated that the VML injury was associated with a significant deficit in force generation ability. Analysis of joint kinematics showed significant differences at all three post-surgical timepoints and gait cycle phase shifting, indicating augmented gait biomechanics in response to VML injury. In conclusion, this method identifies and quantifies key differences in the gait biomechanics and joint kinematics of rats with VML injuries and allows for analysis of the response to injury and recovery. The comprehensive nature of this method opens the door for future studies into dynamics and musculoskeletal control of injured gait that can inform the development of regenerative technologies focused on the functional metrics that are most relevant to recovery from VML injury.
Collapse
Affiliation(s)
- Jack A Dienes
- Biomedical Engineering Department, University of Virginia, Charlottesville, VA, United States
| | - Xiao Hu
- Biomedical Engineering Department, University of Virginia, Charlottesville, VA, United States
| | - Kevin D Janson
- Biomedical Engineering Department, University of Virginia, Charlottesville, VA, United States
| | - Conrad Slater
- Biomedical Engineering Department, University of Virginia, Charlottesville, VA, United States
| | - Emily A Dooley
- Mechanical and Aerospace Engineering Department, University of Virginia, Charlottesville, VA, United States
| | - George J Christ
- Biomedical Engineering Department, University of Virginia, Charlottesville, VA, United States.,Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA, United States
| | - Shawn D Russell
- Biomedical Engineering Department, University of Virginia, Charlottesville, VA, United States.,Mechanical and Aerospace Engineering Department, University of Virginia, Charlottesville, VA, United States.,Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
32
|
Nakayama KH, Quarta M, Paine P, Alcazar C, Karakikes I, Garcia V, Abilez OJ, Calvo NS, Simmons CS, Rando TA, Huang NF. Treatment of volumetric muscle loss in mice using nanofibrillar scaffolds enhances vascular organization and integration. Commun Biol 2019; 2:170. [PMID: 31098403 PMCID: PMC6505043 DOI: 10.1038/s42003-019-0416-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/05/2019] [Indexed: 12/15/2022] Open
Abstract
Traumatic skeletal muscle injuries cause irreversible tissue damage and impaired revascularization. Engineered muscle is promising for enhancing tissue revascularization and regeneration in injured muscle. Here we fabricated engineered skeletal muscle composed of myotubes interspersed with vascular endothelial cells using spatially patterned scaffolds that induce aligned cellular organization, and then assessed their therapeutic benefit for treatment of murine volumetric muscle loss. Murine skeletal myoblasts co-cultured with endothelial cells in aligned nanofibrillar scaffolds form endothelialized and aligned muscle with longer myotubes, more synchronized contractility, and more abundant secretion of angiogenic cytokines, compared to endothelialized engineered muscle formed from randomly-oriented scaffolds. Treatment of traumatically injured muscle with endothelialized and aligned skeletal muscle promotes the formation of highly organized myofibers and microvasculature, along with greater vascular perfusion, compared to treatment of muscle derived from randomly-oriented scaffolds. This work demonstrates the potential of endothelialized and aligned engineered skeletal muscle to promote vascular regeneration following transplantation.
Collapse
Affiliation(s)
- Karina H. Nakayama
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305 USA
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305 USA
| | - Marco Quarta
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94304 USA
| | - Patrick Paine
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94304 USA
| | - Cynthia Alcazar
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA
| | - Ioannis Karakikes
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305 USA
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305 USA
| | - Victor Garcia
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA
| | - Oscar J. Abilez
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305 USA
| | - Nicholas S. Calvo
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainsville, FL 32611 USA
| | - Chelsey S. Simmons
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainsville, FL 32611 USA
| | - Thomas A. Rando
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94304 USA
| | - Ngan F. Huang
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305 USA
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305 USA
| |
Collapse
|
33
|
Aguilar CA, Greising SM, Watts A, Goldman SM, Peragallo C, Zook C, Larouche J, Corona BT. Multiscale analysis of a regenerative therapy for treatment of volumetric muscle loss injury. Cell Death Discov 2018; 4:33. [PMID: 29531830 PMCID: PMC5841404 DOI: 10.1038/s41420-018-0027-8] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 12/22/2017] [Indexed: 01/01/2023] Open
Abstract
Skeletal muscle possesses a remarkable capacity to regenerate when injured, but when confronted with major traumatic injury resulting in volumetric muscle loss (VML), the regenerative process consistently fails. The loss of muscle tissue and function from VML injury has prompted development of a suite of therapeutic approaches but these strategies have proceeded without a comprehensive understanding of the molecular landscape that drives the injury response. Herein, we administered a VML injury in an established rodent model and monitored the evolution of the healing phenomenology over multiple time points using muscle function testing, histology, and expression profiling by RNA sequencing. The injury response was then compared to a regenerative medicine treatment using orthotopic transplantation of autologous minced muscle grafts (~1 mm3 tissue fragments). A chronic inflammatory and fibrotic response was observed at all time points following VML. These results suggest that the pathological response to VML injury during the acute stage of the healing response overwhelms endogenous and therapeutic regenerative processes. Overall, the data presented delineate key molecular characteristics of the pathobiological response to VML injury that are critical effectors of effective regenerative treatment paradigms.
Collapse
Affiliation(s)
- Carlos A. Aguilar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Sarah M. Greising
- Extremity Trauma and Regenerative Medicine, United States Army Institute of Surgical Research, Fort Sam Houston, San Antonio, TX USA
| | - Alain Watts
- Massachusetts Institute of Technology - Lincoln Laboratory, Lexington, MA USA
| | - Stephen M. Goldman
- Extremity Trauma and Regenerative Medicine, United States Army Institute of Surgical Research, Fort Sam Houston, San Antonio, TX USA
| | - Chelsea Peragallo
- Massachusetts Institute of Technology - Lincoln Laboratory, Lexington, MA USA
| | - Christina Zook
- Massachusetts Institute of Technology - Lincoln Laboratory, Lexington, MA USA
| | - Jacqueline Larouche
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Benjamin T. Corona
- Extremity Trauma and Regenerative Medicine, United States Army Institute of Surgical Research, Fort Sam Houston, San Antonio, TX USA
| |
Collapse
|
34
|
Goldman SM, Henderson BEP, Walters TJ, Corona BT. Co-delivery of a laminin-111 supplemented hyaluronic acid based hydrogel with minced muscle graft in the treatment of volumetric muscle loss injury. PLoS One 2018; 13:e0191245. [PMID: 29329332 PMCID: PMC5766229 DOI: 10.1371/journal.pone.0191245] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 01/02/2018] [Indexed: 12/21/2022] Open
Abstract
Minced muscle autografting mediates de novo myofiber regeneration and promotes partial recovery of neuromuscular strength after volumetric muscle loss injury (VML). A major limitation of this approach is the availability of sufficient donor tissue for the treatment of relatively large VMLs without inducing donor site morbidity. This study evaluated a laminin-111 supplemented hyaluronic acid based hydrogel (HA+LMN) as a putative myoconductive scaffolding to be co-delivered with minced muscle grafts. In a rat tibialis anterior muscle VML model, delivery of a reduced dose of minced muscle graft (50% of VML defect) within HA+LMN resulted in a 42% improvement of peak tetanic torque production over unrepaired VML affected limbs. However, the improvement in strength was not improved compared to a 50% minced graft-only control group. Moreover, histological analysis revealed that the improvement in in vivo functional capacity mediated by minced grafts in HA+LMN was not accompanied by a particularly robust graft mediated regenerative response as determined through donor cell tracking of the GFP+ grafting material. Characterization of the spatial distribution and density of macrophage and satellite cell populations indicated that the combination therapy damps the heightened macrophage response while re-establishing satellite content 14 days after VML to a level consistent with an endogenously healing ischemia-reperfusion induced muscle injury. Moreover, regional analysis revealed that the combination therapy increased satellite cell density mostly in the remaining musculature, as opposed to the defect area. Based on the results, the following salient conclusions were drawn: 1) functional recovery mediated by the combination therapy is likely due to a superposition of de novo muscle fiber regeneration and augmented repair of muscle fibers within the remaining musculature, and 2) The capacity for VML therapies to augment regeneration and repair within the remaining musculature may have significant clinical impact and warrants further exploration.
Collapse
Affiliation(s)
- Stephen M. Goldman
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, Texas, United States of America
| | - Beth E. P. Henderson
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, Texas, United States of America
| | - Thomas J. Walters
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, Texas, United States of America
| | - Benjamin T. Corona
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, Texas, United States of America
| |
Collapse
|
35
|
Corona BT, Rivera JC, Wenke JC, Greising SM. Tacrolimus as an adjunct to autologous minced muscle grafts for the repair of a volumetric muscle loss injury. J Exp Orthop 2017; 4:36. [PMID: 29127611 PMCID: PMC5681453 DOI: 10.1186/s40634-017-0112-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/20/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Volumetric muscle loss (VML) following extremity orthopaedic trauma or surgery results in chronic functional deficits and disability. A current translational approach to address the devastating functional limitations due to VML injury is the use of an autologous minced muscle graft (~1 mm3 pieces of muscle tissue) replacement into the injured defect area, although limitations related to donor site morbidity are still unaddressed. This study was designed to explore adjunct pharmacological immunomodulation to enhance graft efficacy and promote muscle function following VML injury, and thereby reduce the amount of donor tissue required. FINDINGS Using a validated VML porcine injury model in which 20% of the muscle volume was surgically removed, this study examined muscle function over 3 months post-VML injury. In vivo isometric torque of the peroneus teritus (PT) muscle was not different before surgery among sham, non-repaired, non-repaired with tacrolimus, graft-repaired, and graft-repaired with tacrolimus VML groups. Bi-weekly torque analysis of the VML injured musculature presented a significant strength deficit of ~26% compared to pre-injury in the non-repaired, non-repaired with tacrolimus, and graft-repaired groups. Comparatively, the strength deficit in the graft-repair with systemic tacrolimus was marginally improved (~19%; p = 0.056). Both of the minced graft repaired groups presented a greater proportion of muscle tissue in full-thickness histology specimen. CONCLUSIONS We demonstrate that adjunctive use of tacrolimus with an ~50% minced muscle graft replacement resulted in modest improvements in muscle function 3 months after injury and repair, but the magnitude of improvement is not expected to elicit clinically meaningful functional improvements.
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, TX, 78234, USA
| | - Jessica C Rivera
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research, 3698 Chambers Pass, BHT1, Fort Sam Houston, TX, 78234, USA
| | - Joseph C Wenke
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research, 3698 Chambers Pass, BHT1, Fort Sam Houston, TX, 78234, 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, TX, 78234, USA.
| |
Collapse
|
36
|
Goldman SM, Corona BT. Co-delivery of micronized urinary bladder matrix damps regenerative capacity of minced muscle grafts in the treatment of volumetric muscle loss injuries. PLoS One 2017; 12:e0186593. [PMID: 29040321 PMCID: PMC5645132 DOI: 10.1371/journal.pone.0186593] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 10/04/2017] [Indexed: 12/31/2022] Open
Abstract
Minced muscle grafts (MG) promote de novo muscle fiber regeneration and neuromuscular strength recovery in small and large animal models of volumetric muscle loss. The most noteworthy limitation of this approach is its reliance on a finite supply of donor tissue. To address this shortcoming, this study sought to evaluate micronized acellular urinary bladder matrix (UBM) as a scaffolding to promote in vivo expansion of this MG therapy in a rat model. Rats received volumetric muscle loss injuries to the tibialis anterior muscle of their left hind limb which were either left untreated or repaired with minced muscle graft at dosages of 50% and 100% of the defect mass, urinary bladder matrix in isolation, or a with an expansion product consisting of a combination of the two putative therapies in which the minced graft is delivered at a dosage of 50% of the defect mass. Rats survived to 2 and 8 weeks post injury before functional (in vivo neuromuscular strength), histological, morphological, and biochemical analyses were performed. Rats treated with the expansion product exhibited improved neuromuscular function relative to untreated VML after an 8 week time period following injury. This improvement in functional capacity, however, was accompanied with a concomitant reduction in graft mediated regeneration, as evidenced cell lineage tracing enable by a transgenic GFP expressing donor, and a mixed histological outcome indicating coincident fibrous matrix deposition with interspersed islands of nascent muscle fibers. Furthermore, quantitative immunofluorescence and transcriptional analysis following the 2 week time point suggests an exacerbated immune response to the UBM as a possible nidus for the observed suboptimal regenerative outcome. Moving forward, efforts related to the development of a MG expansion product should carefully consider the effects of the host immune response to candidate biomaterials in order to avoid undesirable dysregulation of pro-regenerative cross talk between the immune system and myogenic processes.
Collapse
Affiliation(s)
- Stephen M. Goldman
- United States Army Institute of Surgical Research, Fort Sam Houston, TX, United States of America
| | - Benjamin T. Corona
- United States Army Institute of Surgical Research, Fort Sam Houston, TX, United States of America
| |
Collapse
|
37
|
Hurtgen BJ, Ward CL, Leopold Wager CM, Garg K, Goldman SM, Henderson BEP, McKinley TO, Greising SM, Wenke JC, Corona BT. Autologous minced muscle grafts improve endogenous fracture healing and muscle strength after musculoskeletal trauma. Physiol Rep 2017; 5:e13362. [PMID: 28747511 PMCID: PMC5532491 DOI: 10.14814/phy2.13362] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 06/26/2017] [Accepted: 06/27/2017] [Indexed: 12/18/2022] Open
Abstract
The deleterious impact of concomitant muscle injury on fracture healing and limb function is commonly considered part of the natural sequela of orthopedic trauma. Recent reports suggest that heightened inflammation in the surrounding traumatized musculature is a primary determinant of fracture healing. Relatedly, there are emerging potential therapeutic approaches for severe muscle trauma (e.g., volumetric muscle loss [VML] injury), such as autologous minced muscle grafts (1 mm3 pieces of muscle; GRAFT), that can partially prevent chronic functional deficits and appear to have an immunomodulatory effect within VML injured muscle. The primary goal of this study was to determine if repair of VML injury with GRAFT rescues impaired fracture healing and improves the strength of the traumatized muscle in a male Lewis rat model of tibia open fracture. The most salient findings of the study were: (1) tibialis anterior (TA) muscle repair with GRAFT improved endogenous healing of fractured tibia and improved the functional outcome of muscle regeneration; (2) GRAFT repair attenuated the monocyte/macrophage (CD45+CDllb+) and T lymphocyte (CD3+) response to VML injury; (3) TA muscle protein concentrations of MCP1, IL-10, and IGF-1 were augmented in a proregenerative manner by GRAFT repair; (4) VML injury concomitant with osteotomy induced a heightened systemic presence of alarmins (e.g., soluble RAGE) and leukocytes (e.g., monocytes), and depressed IGF-1 concentration, which GRAFT repair ameliorated. Collectively, these data indicate that repair of VML injury with a regenerative therapy can modulate the inflammatory and regenerative phenotype of the treated muscle and in association improve musculoskeletal healing.
Collapse
Affiliation(s)
- Brady J Hurtgen
- Extremity Trauma and Regenerative Medicine Task Area, US Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Catherine L Ward
- Extremity Trauma and Regenerative Medicine Task Area, US Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Chrissy M Leopold Wager
- Extremity Trauma and Regenerative Medicine Task Area, US Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Koyal Garg
- Extremity Trauma and Regenerative Medicine Task Area, US Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Stephen M Goldman
- Extremity Trauma and Regenerative Medicine Task Area, US Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Beth E P Henderson
- Extremity Trauma and Regenerative Medicine Task Area, US Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Todd O McKinley
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Sarah M Greising
- Extremity Trauma and Regenerative Medicine Task Area, US Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Joseph C Wenke
- Extremity Trauma and Regenerative Medicine Task Area, US Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Benjamin T Corona
- Extremity Trauma and Regenerative Medicine Task Area, US Army Institute of Surgical Research, Fort Sam Houston, Texas
| |
Collapse
|
38
|
Corona BT, Henderson BEP, Ward CL, Greising SM. Contribution of minced muscle graft progenitor cells to muscle fiber formation after volumetric muscle loss injury in wild-type and immune deficient mice. Physiol Rep 2017; 5:e13249. [PMID: 28400501 PMCID: PMC5392532 DOI: 10.14814/phy2.13249] [Citation(s) in RCA: 36] [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: 02/17/2017] [Revised: 03/10/2017] [Accepted: 03/16/2017] [Indexed: 12/17/2022] Open
Abstract
Volumetric muscle injury (VML) causes an irrecoverable loss of muscle fibers, persistent strength deficits, and chronic disability. A crucial challenge to VML injury and possible regeneration is the removal of all of the in situ native elements necessary for skeletal muscle regeneration. Our first goal was to establish a reliable VML model in the mouse tibialis anterior (TA) muscle. In adult male wild-type and nude mice, a non-repaired ≈20% VML injury to the TA muscle resulted in an ≈59% loss in nerve evoked muscle strength, ≈33% loss in muscle mass, and ≈29% loss of muscle fibers at 28 day post-injury. Our second goal was to investigate if minced muscle grafts (≈1 mm3 tissue fragments) promote recovery of muscle fibers after VML injury and to understand if the graft-derived progenitor cells directly contribute to fiber regeneration. To assess donor cell contribution, donor muscle tissue was derived from UBC-GFP mice in a subset of experiments. Minced grafts restored ≈34% of the lost fibers 28 days post-injury. The number of GFP+ fibers and the estimated number of regenerated fibers were similar, regardless of host mouse strain. The muscle tissue regeneration promoted by minced grafts did not improve TA muscle strength at this time post-injury. These findings demonstrate the direct contribution of minced muscle graft-derived myogenic stem/progenitor cells to recovery of muscle fibers after VML injury and signify the utility of autologous myogenic stem cell therapies for this indication.
Collapse
Affiliation(s)
- Benjamin T Corona
- Extremity Trauma and Regenerative Medicine Task Area, US Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Beth E P Henderson
- Extremity Trauma and Regenerative Medicine Task Area, US Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Catherine L Ward
- Extremity Trauma and Regenerative Medicine Task Area, US Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Sarah M Greising
- Extremity Trauma and Regenerative Medicine Task Area, US Army Institute of Surgical Research, Fort Sam Houston, Texas
| |
Collapse
|