1
|
Lee HS, Samolyk BL, Pins GD. Extrusion-Based Printing of Myoblast-Loaded Fibrin Microthreads to Induce Myogenesis. J Funct Biomater 2025; 16:21. [PMID: 39852577 PMCID: PMC11765554 DOI: 10.3390/jfb16010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/19/2024] [Accepted: 01/04/2025] [Indexed: 01/26/2025] Open
Abstract
Large skeletal muscle injuries such as volumetric muscle loss (VML) disrupt native tissue structures, including biophysical and biochemical signaling cues that promote the regeneration of functional skeletal muscle. Various biofabrication strategies have been developed to create engineered skeletal muscle constructs that mimic native matrix and cellular microenvironments to enhance muscle regeneration; however, there remains a need to create scalable engineered tissues that provide mechanical stability as well as structural and spatiotemporal signaling cues to promote cell-mediated regeneration of contractile skeletal muscle. We describe a novel strategy for bioprinting multifunctional myoblast-loaded fibrin microthreads (myothreads) that recapitulate the cellular microniches to drive myogenesis and aligned myotube formation. We characterized myoblast alignment, myotube formation, and tensile properties of myothreads as a function of cell-loading density and culture time. We showed that increasing myoblast loading densities enhances myotube formation. Additionally, alignment analyses indicate that the bioprinting process confers myoblast alignment in the constructs. Finally, tensile characterizations suggest that myothreads possess the structural stability to serve as a potential platform for developing scalable muscle scaffolds. We anticipate that our myothread biofabrication approach will enable us to strategically investigate biophysical and biochemical signaling cues and cellular mechanisms that enhance functional skeletal muscle regeneration for the treatment of VML.
Collapse
Affiliation(s)
| | | | - George D. Pins
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA; (H.S.L.); (B.L.S.)
| |
Collapse
|
2
|
Gahlawat S, Oruc D, Paul N, Ragheb M, Patel S, Fasasi O, Sharma P, Shreiber DI, Freeman JW. Tissue Engineered 3D Constructs for Volumetric Muscle Loss. Ann Biomed Eng 2024; 52:2325-2347. [PMID: 39085677 PMCID: PMC11329418 DOI: 10.1007/s10439-024-03541-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/08/2024] [Indexed: 08/02/2024]
Abstract
Severe injuries to skeletal muscles, including cases of volumetric muscle loss (VML), are linked to substantial tissue damage, resulting in functional impairment and lasting disability. While skeletal muscle can regenerate following minor damage, extensive tissue loss in VML disrupts the natural regenerative capacity of the affected muscle tissue. Existing clinical approaches for VML, such as soft-tissue reconstruction and advanced bracing methods, need to be revised to restore tissue function and are associated with limitations in tissue availability and donor-site complications. Advancements in tissue engineering (TE), particularly in scaffold design and the delivery of cells and growth factors, show promising potential for regenerating damaged skeletal muscle tissue and restoring function. This article provides a brief overview of the pathophysiology of VML and critiques the shortcomings of current treatments. The subsequent section focuses on the criteria for designing TE scaffolds, offering insights into various natural and synthetic biomaterials and cell types for effectively regenerating skeletal muscle. We also review multiple TE strategies involving both acellular and cellular scaffolds to encourage the development and maturation of muscle tissue and facilitate integration, vascularization, and innervation. Finally, the article explores technical challenges hindering successful translation into clinical applications.
Collapse
Affiliation(s)
- Sonal Gahlawat
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Doga Oruc
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Nikhil Paul
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Mark Ragheb
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Swati Patel
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Oyinkansola Fasasi
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Peeyush Sharma
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - David I Shreiber
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Joseph W Freeman
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA.
| |
Collapse
|
3
|
Wroblewski OM, Kennedy CS, Vega-Soto EE, Forester CE, Su EY, Nguyen MH, Cederna PS, Larkin LM. Impact of Passaging Primary Skeletal Muscle Cell Isolates on the Engineering of Skeletal Muscle. Tissue Eng Part A 2024. [PMID: 38874526 DOI: 10.1089/ten.tea.2024.0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024] Open
Abstract
Volumetric muscle loss (VML) is a clinical state that results in impaired skeletal muscle function. Engineered skeletal muscle can serve as a treatment for VML. Currently, large biopsies are required to achieve the cells necessary for the fabrication of engineered muscle, leading to donor-site morbidity. Amplification of cell numbers using cell passaging may increase the usefulness of a single muscle biopsy for engineering muscle tissue. In this study, we evaluated the impact of passaging cells obtained from donor muscle tissue by analyzing characteristics of in vitro cellular growth and tissue-engineered skeletal muscle unit (SMU) structure and function. Human skeletal muscle cell isolates from three separate donors (P0-Control) were compared with cells passaged once (P1), twice (P2), or three times (P3) by monitoring SMU force production and determining muscle content and structure using immunohistochemistry. Data indicated that passaging decreased the number of satellite cells and increased the population doubling time. P1 SMUs had slightly greater contractile force and P2 SMUs showed statistically significant greater force production compared with P0 SMUs with no change in SMU muscle content. In conclusion, human skeletal muscle cells can be passaged twice without negatively impacting SMU muscle content or contractile function, providing the opportunity to potentially create larger SMUs from smaller biopsies, thereby producing clinically relevant sized grafts to aid in VML repair.
Collapse
Affiliation(s)
- Olga M Wroblewski
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher S Kennedy
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Emmanuel E Vega-Soto
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Celeste E Forester
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Eileen Y Su
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew H Nguyen
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Paul S Cederna
- Department of Plastic Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Lisa M Larkin
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
4
|
Stellpflug A, Walls J, Hansen C, Joshi A, Wang B. From bone to nanoparticles: development of a novel generation of bone derived nanoparticles for image guided orthopedic regeneration. Biomater Sci 2024; 12:3633-3648. [PMID: 38856671 PMCID: PMC11238765 DOI: 10.1039/d4bm00391h] [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] [Indexed: 06/11/2024]
Abstract
Bone related diseases such as osteoporosis, osteoarthritis, metastatic bone cancer, osteogenesis imperfecta, and Paget's disease, are primarily treated with pharmacologic therapies that often exhibit limited efficacy and substantial side effects. Bone injuries or fractures are primarily repaired with biocompatible materials that produce mixed results in sufficiently regenerating healthy and homogenous bone tissue. Each of these bone conditions, both localized and systemic, use different strategies with the same goal of achieving a healthy and homeostatic bone environment. In this study, we developed a new type of bone-based nanoparticle (BPs) using the entire organic extracellular matrix (ECM) of decellularized porcine bone, additionally encapsulating indocyanine green dye (ICG) for an in vivo monitoring capability. Utilizing the regenerative capability of bone ECM and the functionality of nanoparticles, the ICG encapsulated BPs (ICG/BPs) have been demonstrated to be utilized as a therapeutic option for localized and systemic orthopedic conditions. Additionally, ICG enables an in situ monitoring capability in the Short-Wave Infrared (SWIR) spectrum, capturing the degradation or the biodistribution of the ICG/BPs after both local implantation and intravenous administration, respectively. The efficacy and safety of the ICG/BPs shown within this study lay the foundation for future investigations, which will delve into optimization for clinical translation.
Collapse
Affiliation(s)
- Austin Stellpflug
- Joint Department of Biomedical Engineering, Marquette University and the Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Jacob Walls
- Joint Department of Biomedical Engineering, Marquette University and the Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Christopher Hansen
- Joint Department of Biomedical Engineering, Marquette University and the Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Amit Joshi
- Joint Department of Biomedical Engineering, Marquette University and the Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Bo Wang
- Joint Department of Biomedical Engineering, Marquette University and the Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
5
|
Han J, Rindone AN, Elisseeff JH. Immunoengineering Biomaterials for Musculoskeletal Tissue Repair across Lifespan. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311646. [PMID: 38416061 PMCID: PMC11239302 DOI: 10.1002/adma.202311646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/23/2024] [Indexed: 02/29/2024]
Abstract
Musculoskeletal diseases and injuries are among the leading causes of pain and morbidity worldwide. Broad efforts have focused on developing pro-regenerative biomaterials to treat musculoskeletal conditions; however, these approaches have yet to make a significant clinical impact. Recent studies have demonstrated that the immune system is central in orchestrating tissue repair and that targeting pro-regenerative immune responses can improve biomaterial therapeutic outcomes. However, aging is a critical factor negatively affecting musculoskeletal tissue repair and immune function. Hence, understanding how age affects the response to biomaterials is essential for improving musculoskeletal biomaterial therapies. This review focuses on the intersection of the immune system and aging in response to biomaterials for musculoskeletal tissue repair. The article introduces the general impacts of aging on tissue physiology, the immune system, and the response to biomaterials. Then, it explains how the adaptive immune system guides the response to injury and biomaterial implants in cartilage, muscle, and bone and discusses how aging impacts these processes in each tissue type. The review concludes by highlighting future directions for the development and translation of personalized immunomodulatory biomaterials for musculoskeletal tissue repair.
Collapse
Affiliation(s)
- Jin Han
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
| | - Alexandra N. Rindone
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
| | - Jennifer H. Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine; Baltimore, MD 21231, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
| |
Collapse
|
6
|
Kiratitanaporn W, Guan J, Berry DB, Lao A, Chen S. Multimodal Three-Dimensional Printing for Micro-Modulation of Scaffold Stiffness Through Machine Learning. Tissue Eng Part A 2024; 30:280-292. [PMID: 37747804 DOI: 10.1089/ten.tea.2023.0193] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023] Open
Abstract
The ability to precisely control a scaffold's microstructure and geometry with light-based three-dimensional (3D) printing has been widely demonstrated. However, the modulation of scaffold's mechanical properties through prescribed printing parameters is still underexplored. This study demonstrates a novel 3D-printing workflow to create a complex, elastomeric scaffold with precision-engineered stiffness control by utilizing machine learning. Various printing parameters, including the exposure time, light intensity, printing infill, laser pump current, and printing speed were modulated to print poly (glycerol sebacate) acrylate (PGSA) scaffolds with mechanical properties ranging from 49.3 ± 3.3 kPa to 2.8 ± 0.3 MPa. This enables flexibility in spatial stiffness modulation in addition to high-resolution scaffold fabrication. Then, a neural network-based machine learning model was developed and validated to optimize printing parameters to yield scaffolds with user-defined stiffness modulation for two different vat photopolymerization methods: a digital light processing (DLP)-based 3D printer was utilized to rapidly fabricate stiffness-modulated scaffolds with features on the hundreds of micron scale and a two-photon polymerization (2PP) 3D printer was utilized to print fine structures on the submicron scale. A novel 3D-printing workflow was designed to utilize both DLP-based and 2PP 3D printers to create multiscale scaffolds with precision-tuned stiffness control over both gross and fine geometric features. The described workflow can be used to fabricate scaffolds for a variety of tissue engineering applications, specifically for interfacial tissue engineering for which adjacent tissues possess heterogeneous mechanical properties (e.g., muscle-tendon).
Collapse
Affiliation(s)
- Wisarut Kiratitanaporn
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Jiaao Guan
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, California, USA
| | - David B Berry
- Department of Orthopedic Surgery, University of California San Diego, La Jolla, California, USA
| | - Alison Lao
- Department of NanoEngineering, University of California San Diego, La Jolla, California, USA
| | - Shaochen Chen
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, California, USA
- Department of NanoEngineering, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
7
|
Chen LS, Chen CK, Pang JHS, Lin LP, Yu TY, Tsai WC. Leukocyte-poor platelet-rich plasma and leukocyte-rich platelet-rich plasma promote myoblast proliferation through the upregulation of cyclin A, cdk1, and cdk2. J Orthop Res 2024; 42:32-42. [PMID: 37442643 DOI: 10.1002/jor.25666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/05/2023] [Accepted: 07/11/2023] [Indexed: 07/15/2023]
Abstract
Muscle injuries are common among athletes and often treated with platelet-rich plasma (PRP). However, whether the leukocyte concentration affects the efficacy of PRP in treating muscle injuries remains unclear. This study investigated the effects of leukocyte-poor platelet-rich plasma (LP-PRP) and leukocyte-rich platelet-rich plasma (LR-PRP) on myoblast proliferation and the molecular mechanisms underlying these effects. Myoblasts were treated with 0.5% LP-PRP, 0.5% LR-PRP, 1% LP-PRP, or 1% LR-PRP for 24 h. The gene expression of the LP-PRP- and LR-PRP-treated myoblasts was determined using RNA sequencing analysis. Cell proliferation was evaluated using an bromodeoxyuridine (BrdU) assay, and cell cycle progression was assessed through flow cytometry. The expression of cyclin A, cyclin-dependent kinase 1 (cdk1), and cdk2 was examined using Western blotting. The expression of myoblast determination protein 1 (MyoD1) was examined through Western blotting and immunofluorescence staining. The LP-PRP and LR-PRP both promoted the proliferation of myoblasts and increased differential gene expression of myoblasts. Moreover, the LP-PRP and LR-PRP substantially upregulated the expression of cyclin A, cdk1, and cdk2. MyoD1 expression was induced in the LP-PRP and LR-PRP-treated myoblasts. Our results corroborate the finding that LP-PRP and LR-PRP have similar positive effects on myoblast proliferation and MyoD1 expression.
Collapse
Affiliation(s)
- Li-Siou Chen
- Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chih-Kuang Chen
- Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jong-Hwei Su Pang
- Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Li-Ping Lin
- Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Tung-Yang Yu
- Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Wen-Chung Tsai
- Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
- Center of Comprehensive Sports Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
8
|
Zhang YC, Yang YX, Liu Y, Liu XJ, Dai JH, Gao RS, Hu YY, Fei WY. Combining Porous Se@SiO 2 Nanocomposites and dECM Enhances the Myogenic Differentiation of Adipose-Derived Stem Cells. Int J Nanomedicine 2023; 18:7661-7676. [PMID: 38111844 PMCID: PMC10726970 DOI: 10.2147/ijn.s436081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 12/05/2023] [Indexed: 12/20/2023] Open
Abstract
Background Volumetric Muscle Loss (VML) denotes the traumatic loss of skeletal muscle, a condition that can result in chronic functional impairment and even disability. While the body can naturally repair injured skeletal muscle within a limited scope, patients experiencing local and severe muscle loss due to VML surpass the compensatory capacity of the muscle itself. Currently, clinical treatments for VML are constrained and demonstrate minimal efficacy. Selenium, a recognized antioxidant, plays a crucial role in regulating cell differentiation, anti-inflammatory responses, and various other physiological functions. Methods We engineered a porous Se@SiO2 nanocomposite (SeNPs) with the purpose of releasing selenium continuously and gradually. This nanocomposite was subsequently combined with a decellularized extracellular matrix (dECM) to explore their collaborative protective and stimulatory effects on the myogenic differentiation of adipose-derived mesenchymal stem cells (ADSCs). The influence of dECM and NPs on the myogenic level, reactive oxygen species (ROS) production, and mitochondrial respiratory chain (MRC) activity of ADSCs was evaluated using Western Blot, ELISA, and Immunofluorescence assay. Results Our findings demonstrate that the concurrent application of SeNPs and dECM effectively mitigates the apoptosis and intracellular ROS levels in ADSCs. Furthermore, the combination of dECM with SeNPs significantly upregulated the expression of key myogenic markers, including MYOD, MYOG, Desmin, and myosin heavy chain in ADSCs. Notably, this combination also led to an increase in both the number of mitochondria and the respiratory chain activity in ADSCs. Conclusion The concurrent application of SeNPs and dECM effectively diminishes ROS production, boosts mitochondrial function, and stimulates the myogenic differentiation of ADSCs. This study lays the groundwork for future treatments of VML utilizing the combination of SeNPs and dECM.
Collapse
Affiliation(s)
- Yu-Cheng Zhang
- Clinical Medical College, Dalian Medical University, Dalian, 116044, People’s Republic of China
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People’s Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, People’s Republic of China
| | - Yu-Xia Yang
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People’s Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, People’s Republic of China
- Clinical Medical College, Yangzhou University, Yangzhou, 225001, People’s Republic of China
| | - Yu Liu
- Department of Orthopedics, Wuxi Ninth People’s Hospital Affiliated to Soochow University, Wuxi, 214062, People’s Republic of China
| | - Xi-Jian Liu
- School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, People’s Republic of China
| | - Ji-Hang Dai
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People’s Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, People’s Republic of China
| | - Rang-Shan Gao
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People’s Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, People’s Republic of China
| | - Yang-Yang Hu
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People’s Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, People’s Republic of China
| | - Wen-Yong Fei
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People’s Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, People’s Republic of China
| |
Collapse
|
9
|
Ahmad K, Shaikh S, Chun HJ, Ali S, Lim JH, Ahmad SS, Lee EJ, Choi I. Extracellular matrix: the critical contributor to skeletal muscle regeneration-a comprehensive review. Inflamm Regen 2023; 43:58. [PMID: 38008778 PMCID: PMC10680355 DOI: 10.1186/s41232-023-00308-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/01/2023] [Indexed: 11/28/2023] Open
Abstract
The regenerative ability of skeletal muscle (SM) in response to damage, injury, or disease is a highly intricate process that involves the coordinated activities of multiple cell types and biomolecular factors. Of these, extracellular matrix (ECM) is considered a fundamental component of SM regenerative ability. This review briefly discusses SM myogenesis and regeneration, the roles played by muscle satellite cells (MSCs), other cells, and ECM components, and the effects of their dysregulations on these processes. In addition, we review the various types of ECM scaffolds and biomaterials used for SM regeneration, their applications, recent advances in ECM scaffold research, and their impacts on tissue engineering and SM regeneration, especially in the context of severe muscle injury, which frequently results in substantial muscle loss and impaired regenerative capacity. This review was undertaken to provide a comprehensive overview of SM myogenesis and regeneration, the stem cells used for muscle regeneration, the significance of ECM in SM regeneration, and to enhance understanding of the essential role of the ECM scaffold during SM regeneration.
Collapse
Affiliation(s)
- Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Sibhghatulla Shaikh
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Hee Jin Chun
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Shahid Ali
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Jeong Ho Lim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Syed Sayeed Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea.
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea.
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea.
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea.
| |
Collapse
|
10
|
Vazirzadeh M, Azarpira N, Vosough M, Ghaedi K. Galactosylation of rat natural scaffold for MSC differentiation into hepatocyte-like cells: A comparative analysis of 2D vs. 3D cell culture techniques. Biochem Biophys Rep 2023; 35:101503. [PMID: 37601454 PMCID: PMC10439353 DOI: 10.1016/j.bbrep.2023.101503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/05/2023] [Accepted: 06/21/2023] [Indexed: 08/22/2023] Open
Abstract
The liver plays a crucial role in drug detoxification, and the main source of liver transplants is brain-dead patients. However, the demand for transplants exceeds the available supply, leading to controversies in selecting suitable candidates for acute liver diseases. This research aimed to differentiate mesenchymal stem cells (MSCs) into hepatocyte-like cells using galactosylated rat natural scaffolds and comparing 2-D and 3-D cell culture methods. The study involved isolating and culturing Wharton's jelly cells from the umbilical cord, examining surface markers and adipogenic differentiation potential of MSCs, and culturing mesenchymal cells on galactosylated scaffolds. The growth and proliferation of stem cells on the scaffolds were evaluated using the MTT test, and urea synthesis was measured in different culture environments. Changes in gene expression were analyzed using real-time PCR. Flow cytometry results confirmed the presence of specific surface antigens on MSCs, indicating their identity, while the absence of a specific antigen indicated their differentiation into adipocytes. The MTT test revealed higher cell attachment to galactosylated scaffolds compared to the control groups. Urea secretion was observed in differentiated cells, with the highest levels in cells cultured on galactosylated scaffolds. Gene expression analysis showed differential expression patterns for OCT-4, HNF1, ALB, AFP, and CYP genes under different conditions. The findings indicated that hepatocyte-like cells derived from 3D cultures on galactosylated scaffolds exhibited superior characteristics compared to cells in other culture conditions. These cells demonstrated enhanced proliferation, stability, and urea secretion ability. The study also supported the differentiation potential of MSCs derived from Wharton's jelly umbilical cord into liver-like cells.
Collapse
Affiliation(s)
- Masoud Vazirzadeh
- Department of Cell and Molecular Biology and Microbiology, University of Isfahan, Isfahan, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Kamran Ghaedi
- Department of Cell and Molecular Biology and Microbiology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
11
|
Zhu C, Karvar M, Koh DJ, Sklyar K, Endo Y, Quint J, Samandari M, Tamayol A, Sinha I. Acellular collagen-glycosaminoglycan matrix promotes functional recovery in a rat model of volumetric muscle loss. Regen Med 2023; 18:623-633. [PMID: 37491948 DOI: 10.2217/rme-2023-0060] [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] [Indexed: 07/27/2023] Open
Abstract
Aim: Volumetric muscle loss (VML) is a composite loss of skeletal muscle, which heals with fibrosis, minimal muscle regeneration, and incomplete functional recovery. This study investigated whether collagen-glycosaminoglycan scaffolds (CGS) improve functional recovery following VML. Methods: 15 Sprague-Dawley rats underwent either sham injury or bilateral tibialis anterior (TA) VML injury, with or without CGS implantation. Results: In rats with VML injuries treated with CGS, the TA exhibited greater in vivo tetanic forces and in situ twitch and tetanic dorsiflexion forces compared with those in the non-CGS group at 4- and 6-weeks following injury, respectively. Histologically, the VML with CGS group demonstrated reduced fibrosis and increased muscle regeneration. Conclusion: Taken together, CGS implantation has potential augment muscle recovery following VML.
Collapse
Affiliation(s)
- Christina Zhu
- Division of Plastic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX 79430, USA
| | - Mehran Karvar
- Division of Plastic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel J Koh
- Division of Plastic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Karina Sklyar
- Division of Plastic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yori Endo
- Division of Plastic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jacob Quint
- Department of Biomedical Engineering, University of Connecticut, Farmington, CT 06269, USA
| | - Mohamadmahdi Samandari
- Department of Biomedical Engineering, University of Connecticut, Farmington, CT 06269, USA
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut, Farmington, CT 06269, USA
| | - Indranil Sinha
- Division of Plastic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
12
|
Du Q, Dong T, Liu Y, Zhu X, Li N, Dang L, Cao J, Jin Q, Sun J. Screening criteria of mRNA indicators for wound age estimation. Forensic Sci Res 2023; 7:714-725. [PMID: 36817234 PMCID: PMC9930757 DOI: 10.1080/20961790.2021.1986770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Wound age estimation is a crucial and challenging problem in forensic pathology. Although mRNA is the most commonly used indicator for wound age estimation, screening criteria are lacking. In the present study, the feasibility of screening criteria using mRNA to determine injury time based on the adenylate-uridylate-rich element (ARE) structure and gene ontology (GO) categories were evaluated. A total of 78 Sprague-Dawley male rats were contused and sampled at 4, 8, 12, 16, 20, 24, 28, 32, 36, 40, 44, and 48 h after inflicting injury. The candidate mRNAs were classified based on with or without ARE structure and GO category function. The mRNA expression levels were detected using qRT-PCR. In addition, the standard deviation (STD), mean deviation (MD), relative average deviation (d%), and coefficient of variation (CV) were calculated based on mRNA expression levels. The CV score (CVs) and the CV of CV (CV'CV) were calculated to measure heterogeneity. Finally, based on classic principles, the accuracy of combination of candidate mRNAs was assessed using discriminant analysis to construct a multivariate model for inferring wound age. The results of homogeneity evaluation of each group based on CVs were consistent with the MD, STD, d%, and CV results, indicating the credibility of the evaluation results based on CVs. The candidate mRNAs without ARE structure and classified as cellular component (CC) GO category (ARE-CC) had the highest CVs, showing the mRNAs with these characteristics are the most homogenous mRNAs and best suited for wound age estimation. The highest accuracy was 91.0% when the mRNAs without ARE structure were used to infer the wound age based on the discrimination model. The accuracy of mRNAs classified into CC or multiple function (MF) GO category was higher than mRNAs in the biological process (BP) category. In all subgroups, the accuracy of the composite identification model of mRNA composition without ARE structure and classified as CC was higher than other subgroups. The mRNAs without ARE structure and belonging to the CC GO category were more homogenous, showed higher accuracy for estimating wound age, and were appropriate for rat skeletal muscle wound age estimation. Supplemental data for this article is available online at https://doi.org/10.1080/20961790.2021.1986770 .
Collapse
Affiliation(s)
- Qiuxiang Du
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Tana Dong
- Shandong Public Security Department, The Institute of Criminal Science and Technology, Jinan, China
| | - Yuanxin Liu
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Xiyan Zhu
- Department of Military Traffic Medicine, Army Characteristic Medical Center, Chongqing, China
| | - Na Li
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Lihong Dang
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Jie Cao
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Qianqian Jin
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Junhong Sun
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China,CONTACT Junhong Sun
| |
Collapse
|
13
|
Somers SM, Gilbert-Honick J, Choi IY, K. W. Lo E, Lim H, Dias S, Wagner KR, Mao HQ, Cahan P, Lee G, Grayson WL. Engineering Skeletal Muscle Grafts with PAX7::GFP-Sorted Human Pluripotent Stem Cell-Derived Myogenic Progenitors on Fibrin Microfiber Bundles for Tissue Regeneration. Bioengineering (Basel) 2022; 9:693. [PMID: 36421094 PMCID: PMC9687588 DOI: 10.3390/bioengineering9110693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/01/2022] [Accepted: 11/06/2022] [Indexed: 10/29/2023] Open
Abstract
Tissue engineering strategies that combine human pluripotent stem cell-derived myogenic progenitors (hPDMs) with advanced biomaterials provide promising tools for engineering 3D skeletal muscle grafts to model tissue development in vitro and promote muscle regeneration in vivo. We recently demonstrated (i) the potential for obtaining large numbers of hPDMs using a combination of two small molecules without the overexpression of transgenes and (ii) the application of electrospun fibrin microfiber bundles for functional skeletal muscle restoration following volumetric muscle loss. In this study, we aimed to demonstrate that the biophysical cues provided by the fibrin microfiber bundles induce hPDMs to form engineered human skeletal muscle grafts containing multinucleated myotubes that express desmin and myosin heavy chains and that these grafts could promote regeneration following skeletal muscle injuries. We tested a genetic PAX7 reporter line (PAX7::GFP) to sort for more homogenous populations of hPDMs. RNA sequencing and gene set enrichment analyses confirmed that PAX7::GFP-sorted hPDMs exhibited high expression of myogenic genes. We tested engineered human skeletal muscle grafts derived from PAX7::GFP-sorted hPDMs within in vivo skeletal muscle defects by assessing myogenesis, engraftment and immunogenicity using immunohistochemical staining. The PAX7::GFP-sorted groups had moderately high vascular infiltration and more implanted cell association with embryonic myosin heavy chain (eMHC) regions, suggesting they induced pro-regenerative microenvironments. These findings demonstrated the promise for the use of PAX7::GFP-sorted hPDMs on fibrin microfiber bundles and provided some insights for improving the cell-biomaterial system to stimulate more robust in vivo skeletal muscle regeneration.
Collapse
Affiliation(s)
- Sarah M. Somers
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jordana Gilbert-Honick
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - In Young Choi
- The Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pathology, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Emily K. W. Lo
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - HoTae Lim
- The Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- College of Veterinary Medicine, Chungbuk National University, Chungbuk 28644, Republic of Korea
| | - Shaquielle Dias
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kathryn R. Wagner
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Solomon H. Synder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hai-Quan Mao
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Institute for NanoBioTechnology (INBT), Johns Hopkins University School of Engineering, Baltimore, MD 21218, USA
- Department of Material Sciences & Engineering, Johns Hopkins University School of Engineering, Baltimore, MD 21218, USA
| | - Patrick Cahan
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Gabsang Lee
- The Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Solomon H. Synder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Warren L. Grayson
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Institute for NanoBioTechnology (INBT), Johns Hopkins University School of Engineering, Baltimore, MD 21218, USA
- Department of Material Sciences & Engineering, Johns Hopkins University School of Engineering, Baltimore, MD 21218, USA
- Department of Chemical & Biomolecular, Johns Hopkins University School of Engineering, Baltimore, MD 21218, USA
| |
Collapse
|
14
|
Kiratitanaporn W, Berry DB, Mudla A, Fried T, Lao A, Yu C, Hao N, Ward SR, Chen S. 3D printing a biocompatible elastomer for modeling muscle regeneration after volumetric muscle loss. BIOMATERIALS ADVANCES 2022; 142:213171. [PMID: 36341746 DOI: 10.1016/j.bioadv.2022.213171] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 09/21/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
Abstract
Volumetric muscle loss (VML) injuries due to trauma, tumor ablation, or other degenerative muscle diseases are debilitating and currently have limited options for self-repair. Advancements in 3D printing allow for the rapid fabrication of biocompatible scaffolds with designer patterns. However, the materials chosen are often stiff or brittle, which is not optimal for muscle tissue engineering. This study utilized a photopolymerizable biocompatible elastomer - poly (glycerol sebacate) acrylate (PGSA) - to develop an in vitro model of muscle regeneration and proliferation into an acellular scaffold after VML injury. Mechanical properties of the scaffold were tuned by controlling light intensity during the 3D printing process to match the specific tension of skeletal muscle. The effect of both geometric (channel sizes between 300 and 600 μm) and biologic (decellularized muscle extracellular matrix (dECM)) cues on muscle progenitor cell infiltration, proliferation, organization, and maturation was evaluated in vitro using a near-infrared fluorescent protein (iRFP) transfected cell line to assess cells in the 3D scaffold. Larger channel sizes and dECM coating were found to enhance cell proliferation and maturation, while no discernable effect on cell alignment was observed. In addition, a pilot experiment was carried out to evaluate the regenerative capacity of this scaffold in vivo after a VML injury. Overall, this platform demonstrates a simple model to study muscle progenitor recruitment and differentiation into acellular scaffolds after VML repair.
Collapse
|
15
|
Preclinical Development of Bioengineered Allografts Derived from Decellularized Human Diaphragm. Biomedicines 2022; 10:biomedicines10040739. [PMID: 35453490 PMCID: PMC9031975 DOI: 10.3390/biomedicines10040739] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/01/2022] [Accepted: 03/17/2022] [Indexed: 11/16/2022] Open
Abstract
Volumetric muscle loss (VML) is the traumatic/surgical loss of skeletal muscle, causing aesthetic damage and functional impairment. Suboptimal current surgical treatments are driving research towards the development of optimised regenerative therapies. The grafting of bioengineered scaffolds derived from decellularized skeletal muscle may be a valid option to promote structural and functional healing. In this work, a cellular human diaphragm was considered as a scaffold material for VML treatment. Decellularization occurred through four detergent-enzymatic protocols involving (1) sodium dodecyl sulfate (SDS), (2) SDS + TergitolTM, (3) sodium deoxycholate, and (4) TergitolTM. After decellularization, cells, DNA (≤50 ng/mg of tissue), and muscle fibres were efficiently removed, with the preservation of collagen/elastin and 60%–70% of the glycosaminoglycan component. The detergent-enzymatic treatments did not affect the expression of specific extracellular matrix markers (Collagen I and IV, Laminin), while causing the loss of HLA-DR expression to produce non-immunogenic grafts. Adipose-derived stem cells grown by indirect co-culture with decellularized samples maintained 80%–90% viability, demonstrating the biosafety of the scaffolds. Overall, the tested protocols were quite equivalent, with the patches treated by SDS + TergitolTM showing better collagen preservation. After subcutaneous implant in Balb/c mice, these acellular diaphragmatic grafts did not elicit a severe immune reaction, integrating with the host tissue.
Collapse
|
16
|
Samandari M, Quint J, Rodríguez-delaRosa A, Sinha I, Pourquié O, Tamayol A. Bioinks and Bioprinting Strategies for Skeletal Muscle Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2105883. [PMID: 34773667 PMCID: PMC8957559 DOI: 10.1002/adma.202105883] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/28/2021] [Indexed: 05/16/2023]
Abstract
Skeletal muscles play important roles in critical body functions and their injury or disease can lead to limitation of mobility and loss of independence. Current treatments result in variable functional recovery, while reconstructive surgery, as the gold-standard approach, is limited due to donor shortage, donor-site morbidity, and limited functional recovery. Skeletal muscle tissue engineering (SMTE) has generated enthusiasm as an alternative solution for treatment of injured tissue and serves as a functional disease model. Recently, bioprinting has emerged as a promising tool for recapitulating the complex and highly organized architecture of skeletal muscles at clinically relevant sizes. Here, skeletal muscle physiology, muscle regeneration following injury, and current treatments following muscle loss are discussed, and then bioprinting strategies implemented for SMTE are critically reviewed. Subsequently, recent advancements that have led to improvement of bioprinting strategies to construct large muscle structures, boost myogenesis in vitro and in vivo, and enhance tissue integration are discussed. Bioinks for muscle bioprinting, as an essential part of any bioprinting strategy, are discussed, and their benefits, limitations, and areas to be improved are highlighted. Finally, the directions the field should expand to make bioprinting strategies more translational and overcome the clinical unmet needs are discussed.
Collapse
Affiliation(s)
- Mohamadmahdi Samandari
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Jacob Quint
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | - Indranil Sinha
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, USA
| | - Olivier Pourquié
- Department of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
| |
Collapse
|
17
|
Strategies of the acupuncture treatment for delayed-onset muscle soreness based on pathophysiology. WORLD JOURNAL OF ACUPUNCTURE-MOXIBUSTION 2022. [DOI: 10.1016/j.wjam.2022.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
18
|
Rodriguez BL, Novakova SS, Vega-Soto EE, Nutter GP, Macpherson PCD, Larkin LM. Repairing Volumetric Muscle Loss in the Ovine Peroneus Tertius Following a 6-Month Recovery. Tissue Eng Part A 2021; 28:606-620. [PMID: 34937425 DOI: 10.1089/ten.tea.2021.0187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Tissue-engineered skeletal muscle is a promising novel therapy for the treatment of volumetric muscle loss (VML). Our laboratory has developed tissue-engineered skeletal muscle units (SMUs) and engineered neural conduits (ENCs), and modularly scaled them to clinically relevant sizes for the treatment of VML in a large animal (sheep) model. In a previous study, we evaluated the effects of the SMUs and ENCs in treating a 30% VML injury in the ovine peroneus tertius muscle after a 3-month recovery period. The goal of the current study was to expand on our 3-month study and evaluate the SMUs and ENCs in restoring muscle function after a 6-month recovery period. Six months after implantation, we found that the repair groups with the SMU (VML+SMU and VML+SMU+ENC) restored muscle mass to a level that was statistically indistinguishable from the uninjured contralateral muscle. In contrast, the muscle mass in the VML-Only group was significantly less than groups repaired with an SMU. Following the 6-month recovery from VML, the maximum tetanic force was significantly lower for all VML injured groups compared to the uninjured contralateral muscle. However, we did demonstrate the ability of our ENCs to effectively regenerate nerve between the distal stump of the native nerve and the repair site in 93% of the animals.
Collapse
Affiliation(s)
- Brittany Lynn Rodriguez
- University of Michigan, Biomedical Engineering, BSRB 2328, 109 Zina Pitcher Pl, Ann Arbor, Michigan, United States, 48109;
| | | | | | | | | | - Lisa Marie Larkin
- University of Michian, Physiology, 109 Zina Pitcher Place, 2025 BSRB, Ann Arbor, Michigan, United States, 48109;
| |
Collapse
|
19
|
Pandanaboina SC, RanguMagar AB, Sharma KD, Chhetri BP, Parnell CM, Xie JY, Srivatsan M, Ghosh A. Functionalized Nanocellulose Drives Neural Stem Cells toward Neuronal Differentiation. J Funct Biomater 2021; 12:64. [PMID: 34842752 PMCID: PMC8628960 DOI: 10.3390/jfb12040064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/10/2021] [Accepted: 11/13/2021] [Indexed: 11/23/2022] Open
Abstract
Transplantation of differentiated and fully functional neurons may be a better therapeutic option for the cure of neurodegenerative disorders and brain injuries than direct grafting of neural stem cells (NSCs) that are potentially tumorigenic. However, the differentiation of NSCs into a large population of neurons has been a challenge. Nanomaterials have been widely used as substrates to manipulate cell behavior due to their nano-size, excellent physicochemical properties, ease of synthesis, and versatility in surface functionalization. Nanomaterial-based scaffolds and synthetic polymers have been fabricated with topology resembling the micro-environment of the extracellular matrix. Nanocellulose materials are gaining attention because of their availability, biocompatibility, biodegradability and bioactivity, and affordable cost. We evaluated the role of nanocellulose with different linkage and surface features in promoting neuronal differentiation. Nanocellulose coupled with lysine molecules (CNC-Lys) provided positive charges that helped the cells to attach. Embryonic rat NSCs were differentiated on the CNC-Lys surface for up to three weeks. By the end of the three weeks of in vitro culture, 87% of the cells had attached to the CNC-Lys surface and more than half of the NSCs had differentiated into functional neurons, expressing endogenous glutamate, generating electrical activity and action potentials recorded by the multi-electrode array.
Collapse
Affiliation(s)
- Sahitya Chetan Pandanaboina
- Department of Biological Sciences and Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401, USA; (S.C.P.); (K.D.S.)
| | - Ambar B. RanguMagar
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, AR 72204, USA; (A.B.R.); (B.P.C.); (C.M.P.)
| | - Krishna D. Sharma
- Department of Biological Sciences and Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401, USA; (S.C.P.); (K.D.S.)
| | - Bijay P. Chhetri
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, AR 72204, USA; (A.B.R.); (B.P.C.); (C.M.P.)
| | - Charlette M. Parnell
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, AR 72204, USA; (A.B.R.); (B.P.C.); (C.M.P.)
| | - Jennifer Yanhua Xie
- Department of Basic Sciences, New York Institute of Technology College of Osteopathic Medicine, Arkansas State University, Jonesboro, AR 72401, USA
| | - Malathi Srivatsan
- Department of Biological Sciences and Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401, USA; (S.C.P.); (K.D.S.)
| | - Anindya Ghosh
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, AR 72204, USA; (A.B.R.); (B.P.C.); (C.M.P.)
| |
Collapse
|
20
|
Eugenis I, Wu D, Rando TA. Cells, scaffolds, and bioactive factors: Engineering strategies for improving regeneration following volumetric muscle loss. Biomaterials 2021; 278:121173. [PMID: 34619561 PMCID: PMC8556323 DOI: 10.1016/j.biomaterials.2021.121173] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/01/2021] [Accepted: 08/14/2021] [Indexed: 12/20/2022]
Abstract
Severe traumatic skeletal muscle injuries, such as volumetric muscle loss (VML), result in the obliteration of large amounts of skeletal muscle and lead to permanent functional impairment. Current clinical treatments are limited in their capacity to regenerate damaged muscle and restore tissue function, promoting the need for novel muscle regeneration strategies. Advances in tissue engineering, including cell therapy, scaffold design, and bioactive factor delivery, are promising solutions for VML therapy. Herein, we review tissue engineering strategies for regeneration of skeletal muscle, development of vasculature and nerve within the damaged muscle, and achievements in immunomodulation following VML. In addition, we discuss the limitations of current state of the art technologies and perspectives of tissue-engineered bioconstructs for muscle regeneration and functional recovery following VML.
Collapse
Affiliation(s)
- Ioannis Eugenis
- Department of Bioengineering, Stanford University, Stanford, CA, USA; Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Di Wu
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA; Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
21
|
Wroblewski OM, Nguyen MH, Cederna P, Larkin LM. Impact of Cell-Seeding Density and Cell Confluence on Human Tissue Engineered Skeletal Muscle. Tissue Eng Part A 2021; 28:420-432. [PMID: 34652973 PMCID: PMC9131361 DOI: 10.1089/ten.tea.2021.0132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Tissue engineering methodologies have the potential to treat volumetric muscle loss (VML) via the growth of exogenous skeletal muscle grafts from small autogenous muscle biopsies. A significant obstacle preventing the widespread use of engineered skeletal muscle grafts in a clinical setting is the high number of skeletal muscle stem cells, known as satellite cells, required for fabrication of human-sized skeletal muscle tissue. Additionally, there is a lack of work adapting engineered constructs created for animal models into skeletal muscle engineered from a primary human skeletal muscle cell source. For this study, we used scaffold-free tissue-engineered skeletal muscle units (SMUs) to determine the impact of cell seeding density on the ability to fabricate functional human engineered skeletal muscle. Following established protocols, human skeletal muscle isolates were cultured into SMUs at five different cell-seeding densities: 1,000 cells/cm2, 2,500 cells/cm2, 5,000 cells/cm2, 10,000 cells/cm2, and 25,000 cells/cm2. Following previous human SMU work, SMUs prepared at a cell seeding density of 10,000 cells/cm2 served as controls. Additionally, the impact of cell monolayer confluency on the outcome of human cell-sourced SMU fabrication was investigated at both the 1,000 cells/cm2 and 10,000 cells/cm2 seeding densities. Light microscopy was used to examine myotube formation and hypertrophy in cell monolayers. After the formation of three-dimensional constructs, SMUs underwent maximum tetanic isometric force production measurements and immunohistochemical staining to examine SMU contractile function and muscle-like structure, respectively. Results indicate that the 25,000 cells/cm2 cell-seeding density was detrimental to the contractile function of human cell-sourced SMUs, which had significantly lower maximum tetanic forces compared to SMUs seeded at lower densities. Compared to control, low cell-seeding densities (1,000 cells/cm2 - 5,000 cells/cm2) have no detrimental impact on SMU skeletal muscle growth, maturation, or contractility. Cell cultures seeded at 1,000 cells/cm2 and allowed to proliferate to 90-100% confluency prior to treatment in muscle differentiation media (MDM) resulted in SMUs with greater contractile forces and total muscle-structure compared to cell cultures switched to MDM when underconfluent or overconfluent. In conclusion, initial cell-seeding density for SMU fabrication can be decreased to as low as 1,000 cells/cm2 without negatively impacting SMU muscle-like structure and function.
Collapse
Affiliation(s)
- Olga Maria Wroblewski
- University of Michigan Department of Biomedical Engineering, 505527, Ann Arbor, Michigan, United States;
| | - Matthew Hung Nguyen
- University of Michigan Department of Molecular and Integrative Physiology, 200746, Ann Arbor, Michigan, United States;
| | - Paul Cederna
- University of Michigan Medical School, 12266, Surgery, Ann Arbor, Michigan, United States;
| | - Lisa Marie Larkin
- University of Michigan Department of Molecular and Integrative Physiology, 200746, Ann Arbor, Michigan, United States;
| |
Collapse
|
22
|
Leiva-Cepas F, Benito-Ysamat A, Jimena I, Jimenez-Diaz F, Gil-Belmonte MJ, Ruz-Caracuel I, Villalba R, Peña-Amaro J. Ultrasonographic and Histological Correlation after Experimental Reconstruction of a Volumetric Muscle Loss Injury with Adipose Tissue. Int J Mol Sci 2021; 22:ijms22136689. [PMID: 34206557 PMCID: PMC8268690 DOI: 10.3390/ijms22136689] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/16/2021] [Accepted: 06/19/2021] [Indexed: 02/05/2023] Open
Abstract
Different types of scaffolds are used to reconstruct muscle volume loss injuries. In this experimental study, we correlated ultrasound observations with histological findings in a muscle volume loss injury reconstructed with autologous adipose tissue. The outcome is compared with decellularized and porous matrix implants. Autologous adipose tissue, decellularized matrix, and a porous collagen matrix were implanted in volumetric muscle loss (VML) injuries generated on the anterior tibial muscles of Wistar rats. Sixty days after implantation, ultrasound findings were compared with histological and histomorphometric analysis. The muscles with an autologous adipose tissue implant exhibited an ultrasound pattern that was quite similar to that of the regenerative control muscles. From a histological point of view, the defects had been occupied by newly formed muscle tissue with certain structural abnormalities that would explain the differences between the ultrasound patterns of the normal control muscles and the regenerated ones. While the decellularized muscle matrix implant resulted in fibrosis and an inflammatory response, the porous collagen matrix implant was replaced by regenerative muscle fibers with neurogenic atrophy and fibrosis. In both cases, the ultrasound images reflected echogenic, echotextural, and vascular changes compatible with the histological findings of failed muscle regeneration. The ultrasound analysis confirmed the histological findings observed in the VML injuries reconstructed by autologous adipose tissue implantation. Ultrasound can be a useful tool for evaluating the structure of muscles reconstructed through tissue engineering.
Collapse
Affiliation(s)
- Fernando Leiva-Cepas
- Research Group in Muscle Regeneration, Department of Morphological Sciences, Faculty of Medicine and Nursing, University of Cordoba, 14004 Cordoba, Spain; (F.L.-C.); (A.B.-Y.); (I.J.); (M.J.G.-B.); (I.R.-C.)
- Department of Pathology, Reina Sofia University Hospital, 14004 Cordoba, Spain
- Maimonides Institute for Biomedical Research IMIBIC, Reina Sofia University Hospital, University of Cordoba, 14004 Cordoba, Spain
| | - Alberto Benito-Ysamat
- Research Group in Muscle Regeneration, Department of Morphological Sciences, Faculty of Medicine and Nursing, University of Cordoba, 14004 Cordoba, Spain; (F.L.-C.); (A.B.-Y.); (I.J.); (M.J.G.-B.); (I.R.-C.)
- Maimonides Institute for Biomedical Research IMIBIC, Reina Sofia University Hospital, University of Cordoba, 14004 Cordoba, Spain
- Radiology Department, Musculoskeletal Section, Reina Sofia University Hospital, 14004 Cordoba, Spain
| | - Ignacio Jimena
- Research Group in Muscle Regeneration, Department of Morphological Sciences, Faculty of Medicine and Nursing, University of Cordoba, 14004 Cordoba, Spain; (F.L.-C.); (A.B.-Y.); (I.J.); (M.J.G.-B.); (I.R.-C.)
- Maimonides Institute for Biomedical Research IMIBIC, Reina Sofia University Hospital, University of Cordoba, 14004 Cordoba, Spain
| | - Fernando Jimenez-Diaz
- Sport Sciences Faculty, Castilla La Mancha University, 45071 Toledo, Spain;
- Department of Health Sciences, Faculty of Medicine, Campus de los Jerónimos, San Antonio Catholic University (UCAM), 30107 Murcia, Spain
| | - Maria Jesus Gil-Belmonte
- Research Group in Muscle Regeneration, Department of Morphological Sciences, Faculty of Medicine and Nursing, University of Cordoba, 14004 Cordoba, Spain; (F.L.-C.); (A.B.-Y.); (I.J.); (M.J.G.-B.); (I.R.-C.)
| | - Ignacio Ruz-Caracuel
- Research Group in Muscle Regeneration, Department of Morphological Sciences, Faculty of Medicine and Nursing, University of Cordoba, 14004 Cordoba, Spain; (F.L.-C.); (A.B.-Y.); (I.J.); (M.J.G.-B.); (I.R.-C.)
- Department of Pathology, Ramon y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - Rafael Villalba
- Tissue of Establishment of the Center for Transfusion, Tissues and Cells, 14004 Cordoba, Spain;
| | - Jose Peña-Amaro
- Research Group in Muscle Regeneration, Department of Morphological Sciences, Faculty of Medicine and Nursing, University of Cordoba, 14004 Cordoba, Spain; (F.L.-C.); (A.B.-Y.); (I.J.); (M.J.G.-B.); (I.R.-C.)
- Maimonides Institute for Biomedical Research IMIBIC, Reina Sofia University Hospital, University of Cordoba, 14004 Cordoba, Spain
- Correspondence:
| |
Collapse
|
23
|
Alarcin E, Bal-Öztürk A, Avci H, Ghorbanpoor H, Dogan Guzel F, Akpek A, Yesiltas G, Canak-Ipek T, Avci-Adali M. Current Strategies for the Regeneration of Skeletal Muscle Tissue. Int J Mol Sci 2021; 22:5929. [PMID: 34072959 PMCID: PMC8198586 DOI: 10.3390/ijms22115929] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
Traumatic injuries, tumor resections, and degenerative diseases can damage skeletal muscle and lead to functional impairment and severe disability. Skeletal muscle regeneration is a complex process that depends on various cell types, signaling molecules, architectural cues, and physicochemical properties to be successful. To promote muscle repair and regeneration, various strategies for skeletal muscle tissue engineering have been developed in the last decades. However, there is still a high demand for the development of new methods and materials that promote skeletal muscle repair and functional regeneration to bring approaches closer to therapies in the clinic that structurally and functionally repair muscle. The combination of stem cells, biomaterials, and biomolecules is used to induce skeletal muscle regeneration. In this review, we provide an overview of different cell types used to treat skeletal muscle injury, highlight current strategies in biomaterial-based approaches, the importance of topography for the successful creation of functional striated muscle fibers, and discuss novel methods for muscle regeneration and challenges for their future clinical implementation.
Collapse
Affiliation(s)
- Emine Alarcin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, 34854 Istanbul, Turkey;
| | - Ayca Bal-Öztürk
- Department of Analytical Chemistry, Faculty of Pharmacy, Istinye University, 34010 Istanbul, Turkey;
- Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University, 34010 Istanbul, Turkey
| | - Hüseyin Avci
- Department of Metallurgical and Materials Engineering, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey;
- Cellular Therapy and Stem Cell Research Center, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
- AvciBio Research Group, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey;
- Translational Medicine Research and Clinical Center, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
| | - Hamed Ghorbanpoor
- AvciBio Research Group, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey;
- Department of Biomedical Engineering, Ankara Yildirim Beyazit University, 06010 Ankara, Turkey;
- Department of Biomedical Engineering, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
| | - Fatma Dogan Guzel
- Department of Biomedical Engineering, Ankara Yildirim Beyazit University, 06010 Ankara, Turkey;
| | - Ali Akpek
- Department of Bioengineering, Gebze Technical University, 41400 Gebze, Turkey; (A.A.); (G.Y.)
| | - Gözde Yesiltas
- Department of Bioengineering, Gebze Technical University, 41400 Gebze, Turkey; (A.A.); (G.Y.)
| | - Tuba Canak-Ipek
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany;
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany;
| |
Collapse
|
24
|
Wroblewski OM, Vega-Soto EE, Nguyen MH, Cederna PS, Larkin LM. Impact of Human Epidermal Growth Factor on Tissue-Engineered Skeletal Muscle Structure and Function. Tissue Eng Part A 2021; 27:1151-1159. [PMID: 33203338 DOI: 10.1089/ten.tea.2020.0255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Skeletal muscle tissue engineering technologies have the potential to treat volumetric muscle loss (VML) by growing exogenous muscle tissue. However, there has been limited success in engineering human cell-sourced skeletal muscle with structure and function comparable to native adult human muscle. The use of growth factors at optimal concentrations and delivery times is critical in enhancing the in vitro myogenesis of satellite cells used in engineered skeletal muscle. The mitogenic protein human epidermal growth factor (hEGF) is of particular interest because it enhances satellite cell proliferation and sarcomeric structure formation in myogenic cell cultures. In this study, we used our scaffold-free tissue-engineered skeletal muscle units (SMUs) to examine the effects of hEGF on the structure and function of human cell-sourced engineered skeletal muscle. During our established SMU fabrication process, human muscle cell isolates were exposed to media treated with 7.5 nM hEGF at three different time spans during the 21-day cell culture period: 0 to 6 days postseeding (hEGF-treated Muscle Growth Media [MGM] Only), 7 to 21 days postseeding (hEGF-treated Muscle Differentiation Media (MDM) Only), and 0 to 21 days postseeding (hEGF-treated MGM+MDM). Control cell cultures were fed standard MGM and MDM (no hEGF treatment). During the fabrication process, light microscopy was used to examine proliferation and differentiation of myogenic cells in the monolayer. After SMU formation, the three-dimensional constructs underwent tetanic force production measurements to evaluate contractile function and immunohistochemical staining to examine SMU structure. Results indicated that hEGF administration impacted myogenesis, by increasing myotube diameter in hEGF-treated MGM only and hEGF-treated MDM-only cell cultures, and by increasing myotube density in hEGF-treated MGM+MDM cultures. The exposure of myogenic cells to hEGF during any time period of the fabrication process led to a significant increase in SMU myosin heavy-chain content. SMUs exposed to hEGF-treated MDM and hEGF-treated MGM+MDM exhibited greater cross-sectional areas and more organized sarcomeric structure. Furthermore, hEGF-treated MGM+MDM SMUs displayed significantly enhanced contractile function compared with controls, indicating advanced functional maturation. In conclusion, hEGF supplementation in human primary myogenic cell cultures advances tissue-engineered skeletal muscle structural and functional characteristics. Impact statement Our research suggests that human epidermal growth factor (hEGF) serves as a critical growth factor in enhancing in vitro skeletal muscle cell proliferation and differentiation during myogenesis and advances human skeletal muscle engineered tissues toward a more native adult skeletal muscle phenotype. Understanding the impact of hEGF on engineered skeletal muscle function and structure is valuable in determining the optimal culture conditions for the development of tissue engineering-based therapies for volumetric muscle loss.
Collapse
Affiliation(s)
- Olga M Wroblewski
- Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Emmanuel E Vega-Soto
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew H Nguyen
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Paul S Cederna
- Plastic Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Lisa M Larkin
- Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA.,Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
25
|
Wang YH, Wang DR, Guo YC, Liu JY, Pan J. The application of bone marrow mesenchymal stem cells and biomaterials in skeletal muscle regeneration. Regen Ther 2020; 15:285-294. [PMID: 33426231 PMCID: PMC7770413 DOI: 10.1016/j.reth.2020.11.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/07/2020] [Accepted: 11/16/2020] [Indexed: 02/08/2023] Open
Abstract
Skeletal muscle injuries have bothered doctors and caused great burdens to the public medical insurance system for a long time. Once injured, skeletal muscles usually go through the processes of inflammation, repairing and remodeling. If repairing and remodeling stages are out of balance, scars will be formed to replace injured skeletal muscles. At present, clinicians usually use conventional methods to restore the injured skeletal muscles, such as flap transplantation. However, flap transplantation sometimes needs to sacrifice healthy autologous tissues and will bring extra harm to patients. In recent years, stem cells-based tissue engineering provides us new treatment ideas for skeletal muscle injuries. Stem cells are cells with multiple differentiation potential and have ability to differentiate into adult cells under special condition. Skeletal muscle tissues also have stem cells, called satellite cells, but they are in small amount and new muscle fibers that derived from them may not be enough to replace injured fibers. Bone marrow mesenchymal stem cells (BM-MSCs) could promote musculoskeletal tissue regeneration and activate the myogenic differentiation of satellite cells. Biomaterial is another important factor to promote tissue regeneration and greatly enhance physiological activities of stem cells in vivo. The combined use of stem cells and biomaterials will gradually become a mainstream to restore injured skeletal muscles in the future. This review article mainly focuses on the review of research about the application of BM-MSCs and several major biomaterials in skeletal muscle regeneration over the past decades.
Collapse
Key Words
- 3D-ECM, three dimensional extracellular matrix
- ASCs, adipose stem cells
- BDNF, brain derived neurotrophic factor
- BM-MSCs
- BM-MSCs, bone marrow mesenchymal stem cells
- Biomaterial
- CREB, cAMP- response element binding protein
- DPSCs, dental pulp stem cells
- Differentiation
- ECM, extracellular matrix
- ECs, endothelial cells
- EGF, epidermal growth factor
- FGF, fibroblast growth factor
- FGF-2, fibroblast growth factor-2
- GCSF, granulocyte colony-stimulating factor
- GDNF, glial derived neurotrophic factor
- GPT, gelatin-poly(ethylene glycol)- tyramine
- HGF, hepatocyte growth factor
- IGF-1, insulin-like growth factor-1
- IL, interleukin
- LIF, leukemia inhibitory factor
- MRF, myogenic muscle factor
- NSAIDs, non-steroidal drugs
- PDGF-BB, platelet derived growth factor-BB
- PGE2, prostaglandin E2
- PRP, platelet rich plasma
- S1P, sphingosine 1-phosphate
- SDF-1, stromal cell derived factor-1
- Skeletal muscle injury
- TGF-β, transforming growth factor-β
- Tissue regeneration
- TrkB, tyrosine kinaseB
- VEGF, vascular endothelial growth factor
- VML, volumetric muscle loss
Collapse
Affiliation(s)
- Yu-Hao Wang
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China.,National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, 610041, PR China
| | - Dian-Ri Wang
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China.,National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, 610041, PR China
| | - Yu-Chen Guo
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China.,National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Ji-Yuan Liu
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China.,National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Jian Pan
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China.,National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, 610041, PR China
| |
Collapse
|
26
|
Skeletal Muscle Tissue Engineering: Biomaterials-Based Strategies for the Treatment of Volumetric Muscle Loss. Bioengineering (Basel) 2020; 7:bioengineering7030085. [PMID: 32751847 PMCID: PMC7552659 DOI: 10.3390/bioengineering7030085] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/17/2020] [Accepted: 07/28/2020] [Indexed: 12/13/2022] Open
Abstract
Millions of Americans suffer from skeletal muscle injuries annually that can result in volumetric muscle loss (VML), where extensive musculoskeletal damage and tissue loss result in permanent functional deficits. In the case of small-scale injury skeletal muscle is capable of endogenous regeneration through activation of resident satellite cells (SCs). However, this is greatly reduced in VML injuries, which remove native biophysical and biochemical signaling cues and hinder the damaged tissue's ability to direct regeneration. The current clinical treatment for VML is autologous tissue transfer, but graft failure and scar tissue formation leave patients with limited functional recovery. Tissue engineering of instructive biomaterial scaffolds offers a promising approach for treating VML injuries. Herein, we review the strategic engineering of biophysical and biochemical cues in current scaffold designs that aid in restoring function to these preclinical VML injuries. We also discuss the successes and limitations of the three main biomaterial-based strategies to treat VML injuries: acellular scaffolds, cell-delivery scaffolds, and in vitro tissue engineered constructs. Finally, we examine several innovative approaches to enhancing the design of the next generation of engineered scaffolds to improve the functional regeneration of skeletal muscle following VML injuries.
Collapse
|
27
|
Miranda CMFDC, Therrien J, Leonel LCPC, Smith OE, Miglino MA, Smith LC. Decellularization of Extracellular Matrix from Equine Skeletal Muscle. J Equine Vet Sci 2020; 90:102962. [PMID: 32534761 DOI: 10.1016/j.jevs.2020.102962] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 07/17/2019] [Accepted: 02/06/2020] [Indexed: 01/29/2023]
Abstract
Equine represents an attractive animal model for musculoskeletal tissue diseases, exhibiting much similarity to the injuries that occur in humans. Cell therapy and tissue bioengineering have been widely used as a therapeutic alternative by regenerative medicine in musculoskeletal diseases. Thus, the aim of this study was to produce an acellular biomaterial of equine skeletal muscle and to evaluate its effectiveness in supporting the in vitro culture of equine induced pluripotency stem cells (iPSCs). Biceps femoris samples were frozen at -20°C for 4 days and incubated in 1% sodium dodecyl sulfate (SDS), 5 mM EDTA + 50 mM Tris and 1% Triton X-100; the effectiveness of the decellularization was evaluated by the absence of remnant nuclei (histological and 4',6-diamidino-2-phenylindole [DAPI] analysis), preservation of extracellular matrix (ECM) proteins (immunofluorescence and immunohistochemistry) and organization of ECM ultrastructure (scanning electron microscopy). Decellularized samples were recellularized with iPSCs at the concentration of 50,000 cells/cm2 and cultured in vitro for 9 days, and the presence of the cells in the biomaterial was evaluated by histological analysis and presence of nuclei. Decellularized biomaterial showed absence of remnant nuclei and muscle fibers, as well as the preservation of ECM architecture, vascular network and proteins, laminin, fibronectin, elastin, collagen III and IV. After cellularization, iPSC nuclei were present at 9 days after incubation, indicating the decellularized biomaterial-supported iPSC survival. It is concluded that the ECM biomaterial produced from the decellularized equine skeletal muscle has potential for iPSC adhesion, representing a promising biomaterial for regenerative medicine in the therapy of musculoskeletal diseases.
Collapse
Affiliation(s)
- Carla Maria Figueiredo de Carvalho Miranda
- Centre de recherche en reproduction et fertilité, Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada; Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil.
| | - Jacinthe Therrien
- Centre de recherche en reproduction et fertilité, Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | | | - Olivia Eilers Smith
- Centre de recherche en reproduction et fertilité, Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Maria Angélica Miglino
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Lawrence Charles Smith
- Centre de recherche en reproduction et fertilité, Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| |
Collapse
|
28
|
Novakova SS, Rodriguez BL, Vega-Soto EE, Nutter GP, Armstrong RE, Macpherson PCD, Larkin LM. Repairing Volumetric Muscle Loss in the Ovine Peroneus Tertius Following a 3-Month Recovery. Tissue Eng Part A 2020; 26:837-851. [PMID: 32013753 DOI: 10.1089/ten.tea.2019.0288] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Much effort has been made to fabricate engineered tissues on a scale that is clinically relevant to humans; however, scale-up remains one of the most significant technological challenges of tissue engineering to date. To address this limitation, our laboratory has developed tissue-engineered skeletal muscle units (SMUs) and engineered neural conduits (ENCs), and modularly scaled them to clinically relevant sizes for the treatment of volumetric muscle loss (VML). The goal of this study was to evaluate the SMUs and ENCs in vitro, and to test the efficacy of our SMUs and ENCs in restoring muscle function in a clinically relevant large animal (sheep) model. The animals received a 30% VML injury to the peroneus tertius muscle and were allowed to recover for 3 months. The animals were divided into three experimental groups: VML injury without a repair (VML only), repair with an SMU (VML+SMU), or repair with an SMU and ENC (VML+SMU+ENC). We evaluated the SMUs before implantation and found that our single scaled-up SMUs were characterized by the presence of contracting myotubes, linearly aligned extracellular matrix proteins, and Pax7+ satellite cells. Three months after implantation, we found that the repair groups (VML+SMU and VML+SMU+ENC) had restored muscle mass and tetanic force production to a level that was statistically indistinguishable from the uninjured contralateral muscle after 3 months in vivo. Furthermore, we demonstrated the ability of our ENCs to effectively bridge the gap between native nerve and the repair site by eliciting a muscle contraction through direct electrical stimulation of the re-routed nerve. Impact statement The fabrication of tissues of clinically relevant sizes is one of the largest obstacles preventing engineered tissues from achieving widespread use in the clinic. This study aimed to combat this limitation by developing a fabrication method to scale-up tissue-engineered skeletal muscle for the treatment of volumetric muscle loss in a large animal (sheep) model and evaluating the efficacy of the tissue-engineered constructs after a 3-month recovery.
Collapse
Affiliation(s)
- Stoyna S Novakova
- Department of Molecular and Integrative Physiology and University of Michigan, Ann Arbor, Michigan, USA
| | - Brittany L Rodriguez
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Emmanuel E Vega-Soto
- Department of Molecular and Integrative Physiology and University of Michigan, Ann Arbor, Michigan, USA
| | - Genevieve P Nutter
- Department of Molecular and Integrative Physiology and University of Michigan, Ann Arbor, Michigan, USA
| | - Rachel E Armstrong
- Department of Molecular and Integrative Physiology and University of Michigan, Ann Arbor, Michigan, USA
| | - Peter C D Macpherson
- Department of Molecular and Integrative Physiology and University of Michigan, Ann Arbor, Michigan, USA
| | - Lisa M Larkin
- Department of Molecular and Integrative Physiology and University of Michigan, Ann Arbor, Michigan, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
29
|
Mintz EL, Passipieri JA, Franklin IR, Toscano VM, Afferton EC, Sharma PR, Christ GJ. Long-Term Evaluation of Functional Outcomes Following Rat Volumetric Muscle Loss Injury and Repair. Tissue Eng Part A 2020; 26:140-156. [PMID: 31578935 DOI: 10.1089/ten.tea.2019.0126] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Volumetric muscle loss (VML) injuries, by definition, exceed the endogenous repair capacity of skeletal muscle resulting in permanent structural and functional deficits. VML injuries present a significant burden for both civilian and military medicine. Despite progress, there is still considerable room for therapeutic improvement. In this regard, tissue-engineered constructs show promise for VML repair, as they provide an opportunity to introduce both scaffolding and cellular components. We have pioneered the development of a tissue-engineered muscle repair (TEMR) technology created by seeding muscle progenitor cells onto a porcine-derived bladder acellular matrix followed by cyclic stretch preconditioning before implantation. Our work to date has demonstrated significant functional repair (60-90% functional recovery) in progressively larger rodent models of VML injury following TEMR implantation. Notwithstanding this success, TEMR implantation in cylindrically shaped VML injuries in the tibialis anterior (TA) muscle was associated with more variable functional outcomes than has been observed in sheet-like muscles such as the latissimus dorsi. In fact, previous observations documented a dichotomy of responses following TEMR implantation in a rodent TA VML injury model; with an ≈61% functional improvement observed in fewer than half (46%) of TEMR-implanted animals at 12 weeks postinjury. This current study builds directly from those observations as we modified the geometry of both the VML injury and the TEMR construct to determine if improved matching of the implanted TEMR construct to the surgically created VML injury resulted in increased functional recovery posttreatment. Following these modifications, we observed a comparable degree of functional improvement in a larger proportion of animals (≈67%) that was durable up to 24 weeks post-TEMR implantation. Moreover, in ≈25% of all TEMR-implanted animals, functional recovery was virtually complete (TEMR max responders), and furthermore, the functional recovery in all 67% of responding animals was accompanied by the presence of native-like muscle properties within the repaired TA muscle, including fiber cross-sectional area, fiber type, vascularization, and innervation. This study emphasizes the importance of tuning the application of tissue engineering technology platforms to the specific requirements of diverse VML injuries to improve functional outcomes. Impact Statement This report confirms and extends previous observations with our implantable tissue-engineered technology platform for repair of volumetric muscle loss (VML) injuries. Based on our prior work, we addressed factors hypothesized to be responsible for significant outcome variability following treatment of VML injuries in a rat tibialis anterior model. Through customization of the muscle repair technology to a specific VML injury, we were able to significantly increase the frequency at which functional recovery occurred, and furthermore, demonstrate durability out to 6 months. In addition, the enhanced biomimetic qualities of repaired muscle tissue were associated with the most robust functional outcomes.
Collapse
Affiliation(s)
- Ellen L Mintz
- Department of Pathology, University of Virginia, Charlottesville, Virginia
| | - Juliana A Passipieri
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | | | - Victoria M Toscano
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - Emma C Afferton
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - Poonam R Sharma
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - George J Christ
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia.,Department of Orthopaedics, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
30
|
Gilbert-Honick J, Grayson W. Vascularized and Innervated Skeletal Muscle Tissue Engineering. Adv Healthc Mater 2020; 9:e1900626. [PMID: 31622051 PMCID: PMC6986325 DOI: 10.1002/adhm.201900626] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/27/2019] [Indexed: 12/12/2022]
Abstract
Volumetric muscle loss (VML) is a devastating loss of muscle tissue that overwhelms the native regenerative properties of skeletal muscle and results in lifelong functional deficits. There are currently no treatments for VML that fully recover the lost muscle tissue and function. Tissue engineering presents a promising solution for VML treatment and significant research has been performed using tissue engineered muscle constructs in preclinical models of VML with a broad range of defect locations and sizes, tissue engineered construct characteristics, and outcome measures. Due to the complex vascular and neural anatomy within skeletal muscle, regeneration of functional vasculature and nerves is vital for muscle recovery following VML injuries. This review aims to summarize the current state of the field of skeletal muscle tissue engineering using 3D constructs for VML treatment with a focus on studies that have promoted vascular and neural regeneration within the muscle tissue post-VML.
Collapse
Affiliation(s)
- Jordana Gilbert-Honick
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Warren Grayson
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Material Sciences & Engineering, Johns Hopkins University, School of Engineering, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology (INBT), Johns Hopkins University School of Engineering, Baltimore, MD 21218, USA
| |
Collapse
|
31
|
Panayi AC, Smit L, Hays N, Udeh K, Endo Y, Li B, Sakthivel D, Tamayol A, Neppl RL, Orgill DP, Nuutila K, Sinha I. A porous collagen-GAG scaffold promotes muscle regeneration following volumetric muscle loss injury. Wound Repair Regen 2020; 28:61-74. [PMID: 31603580 DOI: 10.1111/wrr.12768] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 09/03/2019] [Accepted: 09/20/2019] [Indexed: 12/13/2022]
Abstract
Volumetric muscle loss (VML) is a segmental loss of skeletal muscle which commonly heals with fibrosis, minimal muscle regeneration, and loss of muscle strength. Treatment options for these wounds which promote functional recovery are currently lacking. This study was designed to investigate whether the collagen-GAG scaffold (CGS) promotes functional muscle recovery following VML. A total of 66 C57/Bl6 mice were used in a three-stage experiment. First, 24 animals were split into three groups which underwent sham injury or unilateral quadriceps VML injury with or without CGS implantation. Two weeks post-surgery, muscle was harvested for histological and gene expression analysis. In the second stage, 18 mice underwent bilateral quadriceps VML injury, followed by weekly functional testing using a treadmill. In the third stage, 24 mice underwent sham or bilateral quadriceps VML injury with or without CGS implantation, with tissue harvested six weeks post-surgery for histological and gene expression analysis. VML mice treated with CGS demonstrated increased remnant fiber hypertrophy versus both the VML with no CGS and uninjured groups. Both VML groups showed greater muscle fiber hypertrophy than non-injured muscle. This phenomenon was still evident in the longer-term experiment. The gene array indicated that the CGS promoted upregulation of factors involved in promoting wound healing and regeneration. In terms of functional improvement, the VML mice treated with CGS ran at higher maximum speeds than VML without CGS. A CGS was shown to enhance muscle hypertrophy in response to VML injury with a resultant improvement in functional performance. A gene array highlighted increased gene expression of multiple growth factors following CGS implantation. This suggests that implantation of a CGS could be a promising treatment for VML wounds.
Collapse
Affiliation(s)
- Adriana C Panayi
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Lucindi Smit
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Nicole Hays
- University of Maryland School of Medicine, Baltimore, Maryland
| | - Kodi Udeh
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Yori Endo
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Bin Li
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Dharaniya Sakthivel
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Ali Tamayol
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Nebraska
| | - Ronald L Neppl
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Dennis P Orgill
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Kristo Nuutila
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Indranil Sinha
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
32
|
Can Dynamic Contrast-Enhanced CT Quantify Perfusion in a Stimulated Muscle of Limited Size? A Rat Model. Clin Orthop Relat Res 2020; 478:179-188. [PMID: 31794491 PMCID: PMC7000042 DOI: 10.1097/corr.0000000000001045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Muscle injury may result in damage to the vasculature, rendering it unable to meet the metabolic demands of muscle regeneration and healing. Therefore, therapies frequently aim to maintain, restore, or improve blood supply to the injured muscle. Although there are several options to assess the vascular outcomes of these therapies, few are capable of spatially assessing perfusion in large volumes of tissue. QUESTIONS/PURPOSES Can dynamic contrast-enhanced CT (DCE-CT) imaging acquired with a clinical CT scanner be used in a rat model to quantify perfusion in the anterior tibialis muscle at spatially relevant volumes, as assessed by (1) the blood flow rate and tissue blood volume in the muscle after three levels of muscle stimulation (low, medium, and maximum) relative to baseline as determined by the non-stimulated contralateral leg; and (2) how do these measurements compare with those obtained by the more standard approach of microsphere perfusion? METHODS The right anterior tibialis muscles of adult male Sprague Dawley rats were randomized to low- (n = 10), medium- (n = 6), or maximum- (n = 3) level (duty cycles of 2.5%, 5.0%, and 20%, respectively) nerve electrode coupled muscle stimulation directly followed by DCE-CT imaging. Tissue blood flow and blood volume maps were created using commercial software and volumetrically measured using NIH software. Although differences in blood flow were detectable across the studied levels of muscle stimulation, a review of the evidence suggested the absolute blood flow quantified was underestimated. Therefore, at a later date, a separate set of adult male Sprague Dawley rats were randomized for microsphere perfusion (n = 7) to define blood flow in the animal model with an accepted standard. With this technique, intra-arterial particles sized to freely flow in blood but large enough to lodge in tissue capillaries were injected. Simultaneously, blood sampling at a fixed flow rate was simultaneously performed to provide a fixed blood flow rate sample. The tissues of interest were then explanted and assessed for the total number of particles per tissue volume. Tissue blood flow rate was then calculated based on the particle count ratio within the reference sample. Note that a tissue's blood volume cannot be calculated with this method. Comparison analysis to the non-stimulated baseline leg was performed using two-tailed paired student t-test. An ANOVA was used to compare difference between stimulation groups. RESULTS DCE-CT measured (mean ± SD) increasing tissue blood flow differences in stimulated anterior tibialis muscle at 2.5% duty cycle (32 ± 5 cc/100 cc/min), 5.0% duty cycle (46 ± 13 cc/100 cc/min), and 20% duty cycle (73 ± 3 cc/100 cc/min) compared with the paired contralateral non-stimulated anterior tibialis muscle (10 ± 2 cc/100 cc/min, mean difference 21 cc/100 cc/min [95% CI 17.08 to 25.69]; 9 ± 1 cc/100 cc/min, mean difference 37 cc/100 cc/min [95% CI 23.06 to 50.11]; and 11 ± 2 cc/100 cc/min, mean difference 62 cc/100 cc/min [95% CI 53.67 to 70.03]; all p < 0.001). Similarly, DCE-CT showed increasing differences in tissue blood volumes within the stimulated anterior tibialis muscle at 2.5% duty cycle (23.2 ± 4.2 cc/100 cc), 5.0% duty cycle (39.2 ± 7.2 cc/100 cc), and 20% duty cycle (52.5 ± 13.1 cc/100 cc) compared with the paired contralateral non-stimulated anterior tibialis muscle (3.4 ± 0.7 cc/100 cc, mean difference 19.8 cc/100 cc [95% CI 16.46 to 23.20]; p < 0.001; 3.5 ± 0.4 cc/100 cc, mean difference 35.7 cc/100 cc [95% CI 28.44 to 43.00]; p < 0.001; and 4.2 ± 1.3 cc/100 cc, mean difference 48.3 cc/100 cc [95% CI 17.86 to 78.77]; p = 0.010). Microsphere perfusion measurements also showed an increasing difference in tissue blood flow in the stimulated anterior tibialis muscle at 2.5% duty cycle (62 ± 43 cc/100 cc/min), 5.0% duty cycle (89 ± 52 cc/100 cc/min), and 20% duty cycle (313 ± 269 cc/100 cc/min) compared with the paired contralateral non-stimulated anterior tibialis muscle (8 ± 4 cc/100 cc/min, mean difference 55 cc/100 cc/min [95% CI 15.49 to 94.24]; p = 0.007; 9 ± 9 cc/100 cc/min, mean difference 79 cc/100 cc/min [95% CI 33.83 to 125.09]; p = 0.003; and 18 ± 18 cc/100 cc/min, mean difference 295 cc/100 cc/min [95% CI 8.45 to 580.87]; p = 0.023). Qualitative comparison between the methods suggests that DCE-CT values underestimate tissue blood flow with a post-hoc ANOVA showing DCE-CT blood flow values within the 2.5% duty cycle group (32 ± 5 cc/100 cc/min) to be less than the microsphere perfusion value (62 ± 43 cc/100 cc/min) with a mean difference of 31 cc/100 cc/min (95% CI 2.46 to 60.23; p = 0.035). CONCLUSIONS DCE-CT using a clinical scanner is a feasible modality to measure incremental changes of blood flow and tissue blood volume within a spatially challenged small animal model. Care should be taken in studies where true blood flow values are needed, as this particular small-volume muscle model suggests true blood flow is underestimated using the specific adaptions of DCE-CT acquisition and image processing chosen. CLINICAL RELEVANCE CT perfusion is a clinically available modality allowing for translation of science from bench to bedside. Adapting the modality to fit small animal models that are relevant to muscle healing may hasten time to clinical utility.
Collapse
|
33
|
Greising SM, Corona BT, McGann C, Frankum JK, Warren GL. Therapeutic Approaches for Volumetric Muscle Loss Injury: A Systematic Review and Meta-Analysis. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:510-525. [PMID: 31578930 DOI: 10.1089/ten.teb.2019.0207] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [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
|
34
|
Rodriguez BL, Nguyen MH, Armstrong RE, Vega-Soto EE, Polkowski PM, Larkin LM. A Comparison of Ovine Facial and Limb Muscle as a Primary Cell Source for Engineered Skeletal Muscle. Tissue Eng Part A 2019; 26:167-177. [PMID: 31469044 DOI: 10.1089/ten.tea.2019.0087] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Volumetric muscle loss (VML) contributes to the number of soft tissue injuries that necessitate reconstructive surgery, but treatment options are often limited by tissue availability and donor site morbidity. To combat these issues, our laboratory has developed scaffold-free tissue-engineered skeletal muscle units (SMUs) as a novel treatment for VML injuries. Recently, we have begun experiments addressing VML in facial muscle, and the optimal starting cell population for engineered skeletal muscle tissue for this application may not be cells derived from hindlimb muscles due to reported heterogeneity of cell populations. Thus, the purpose of this study was to compare SMUs fabricated from both craniofacial and hindlimb sources to determine which cell source is best suited for the engineering of skeletal muscle. Herein, we assessed the development, structure, and function of SMUs derived from four muscle sources, including two hindlimb muscles (i.e., soleus and semimembranosus [SM]) and two craniofacial muscles (i.e., zygomaticus major and masseter). Overall, the zygomaticus major exhibited the least efficient digestion, and SMUs fabricated from this muscle exhibited the least aligned myosin heavy chain staining and consequently, the lowest average force production. Conversely, the SM muscle exhibited the most efficient digestion and the highest number of myotubes/mm2; however, the SM, masseter, and soleus groups were roughly equivalent in terms of force production and histological structure. Impact Statement An empirical comparison of the development, structure, and function of engineered skeletal muscle tissue fabricated from different muscles, including both craniofacial and hindlimb sources, will not only provide insight into innate regenerative mechanisms of skeletal muscle but also will give our team and other researchers the information necessary to determine which cell sources are best suited for the skeletal muscle tissue engineering.
Collapse
Affiliation(s)
| | - Matthew H Nguyen
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Rachel E Armstrong
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Emmanuel E Vega-Soto
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Phillip M Polkowski
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Lisa M Larkin
- Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
35
|
Ilanlou S, Khakbiz M, Amoabediny G, Mohammadi J. Preclinical studies of acellular extracellular matrices as small-caliber vascular grafts. Tissue Cell 2019; 60:25-32. [DOI: 10.1016/j.tice.2019.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 07/28/2019] [Accepted: 07/30/2019] [Indexed: 01/09/2023]
|
36
|
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 2019; 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] [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
|
37
|
Stephens CJ, Spector JA, Butcher JT. Biofabrication of thick vascularized neo-pedicle flaps for reconstructive surgery. Transl Res 2019; 211:84-122. [PMID: 31170376 PMCID: PMC6702068 DOI: 10.1016/j.trsl.2019.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 05/06/2019] [Accepted: 05/14/2019] [Indexed: 01/01/2023]
Abstract
Wound chronicity due to intrinsic and extrinsic factors perturbs adequate lesion closure and reestablishment of the protective skin barrier. Immediate and proper care of chronic wounds is necessary for a swift recovery and a reduction of patient vulnerability to infection. Advanced therapies supplemented with standard wound care procedures have been clinically implemented to restore aberrant tissue; however, these treatments are ineffective if local vasculature is too compromised to support minimally-invasive strategies. Autologous "flaps", which are tissues equipped with their own hierarchical vascular supply, can be harvested from one region of the patient and transplanted to the wound where it is reperfused upon microsurgical anastomosis to appropriate recipient vessels. Despite the success of autologous flap transfer, these procedures are extremely invasive, incur obligatory donor-site morbidity, and require sufficient donor-tissue availability, microsurgical expertise, and specialized equipment. 3D-bioprinting modalities, such as extrusion-based bioprinting, can be used to address the clinical constraints of autologous flap transfer, primarily addressing donor-site morbidity and tissue availability. This advancement in regenerative medicine allows the biofabrication of heterogeneous tissue structures with high shape fidelity and spatial resolution to generate biomimetic constructs with the anatomically-precise geometries of native tissue to ensure tissue-specific function. Yet, meaningful progress toward this clinical application has been limited by the lack of vascularization required to meet the nutrient and oxygen demands of clinically relevant tissue volumes. Thus, various criteria for the fabrication of functional tissues with hierarchical, patent vasculature must be considered when implementing 3D-bioprinting technologies for deep, chronic wounds.
Collapse
Affiliation(s)
- Chelsea J Stephens
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Jason A Spector
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York; Division of Plastic Surgery, Weill Cornell Medical College, New York, New York
| | - Jonathan T Butcher
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York.
| |
Collapse
|
38
|
Passipieri JA, Hu X, Mintz E, Dienes J, Baker HB, Wallace CH, Blemker SS, Christ GJ. In Silico and In Vivo Studies Detect Functional Repair Mechanisms in a Volumetric Muscle Loss Injury. Tissue Eng Part A 2019; 25:1272-1288. [PMID: 30882277 PMCID: PMC6760186 DOI: 10.1089/ten.tea.2018.0280] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/10/2018] [Indexed: 11/12/2022] Open
Abstract
IMPACT STATEMENT Despite medical advances, volumetric muscle loss (VML) injuries to craniofacial muscles represent an unmet clinical need. We report an implantable tissue-engineered construct that leads to substantial tissue regeneration and functional recovery in a preclinical model of VML injury that is dimensionally relevant to unilateral cleft lip repair, and a series of corresponding computational models that provide biomechanical insight into mechanism(s) responsible for the VML-induced functional deficits and recovery following tissue-engineered muscle repair implantation. This unique combined approach represents a critical first step toward establishing a crucial biomechanical basis for the development of efficacious regenerative technologies, considering the spectrum of VML injuries.
Collapse
Affiliation(s)
- Juliana A. Passipieri
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - Xiao Hu
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - Ellen Mintz
- Department of Pathology, University of Virginia, Charlottesville, Virginia
| | - Jack Dienes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - Hannah B. Baker
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - C. Hunter Wallace
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - Silvia S. Blemker
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
- Department of Mechanical and Aerospace Engineering, University of Virginia, Charlottesville, Virginia
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia
| | - George J. Christ
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
- Department of Orthopaedics, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
39
|
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
|
40
|
Trevisan C, Fallas MEA, Maghin E, Franzin C, Pavan P, Caccin P, Chiavegato A, Carraro E, Boso D, Boldrin F, Caicci F, Bertin E, Urbani L, Milan A, Biz C, Lazzari L, De Coppi P, Pozzobon M, Piccoli M. Generation of a Functioning and Self-Renewing Diaphragmatic Muscle Construct. Stem Cells Transl Med 2019; 8:858-869. [PMID: 30972959 PMCID: PMC6646700 DOI: 10.1002/sctm.18-0206] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 03/04/2019] [Indexed: 12/19/2022] Open
Abstract
Surgical repair of large muscular defects requires the use of autologous graft transfer or prosthetic material. Naturally derived matrices are biocompatible materials obtained by tissue decellularization and are commonly used in clinical practice. Despite promising applications described in the literature, the use of acellular matrices to repair large defects has been only partially successful, highlighting the need for more efficient constructs. Scaffold recellularization by means of tissue engineering may improve not only the structure of the matrix, but also its ability to functionally interact with the host. The development of such a complex construct is challenging, due to the complexity of the native organ architecture and the difficulties in recreating the cellular niche with both proliferative and differentiating potential during growth or after damage. In this study, we tested a mouse decellularized diaphragmatic extracellular matrix (ECM) previously described by our group, for the generation of a cellular skeletal muscle construct with functional features. The decellularized matrix was stored using different conditions to mimic the off‐the‐shelf clinical need. Pediatric human muscle precursors were seeded into the decellularized scaffold, demonstrating proliferation and differentiation capability, giving rise to a functioning three‐dimensional skeletal muscle structure. Furthermore, we exposed the engineered construct to cardiotoxin injury and demonstrated its ability to activate a regenerative response in vitro promoting cell self‐renewal and a positive ECM remodeling. Functional reconstruction of an engineered skeletal muscle with maintenance of a stem cell pool makes this a promising tool toward future clinical applications in diaphragmatic regeneration. stem cells translational medicine2019;8:858&869
Collapse
Affiliation(s)
- Caterina Trevisan
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy.,Department of Women and Children Health, University of Padova, Padova, Italy
| | - Mario Enrique Alvrez Fallas
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy.,Department of Women and Children Health, University of Padova, Padova, Italy
| | - Edoardo Maghin
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy.,Department of Women and Children Health, University of Padova, Padova, Italy
| | - Chiara Franzin
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Piero Pavan
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy.,Department of Industrial Engineering, University of Padova, Padova, Italy.,Centre for Mechanics of Biological Materials, University of Padova, Padova, Italy
| | - Paola Caccin
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Angela Chiavegato
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,CNR Institute for Neuroscience, Padova, Italy
| | - Eugenia Carraro
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Daniele Boso
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | | | | | - Enrica Bertin
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Luca Urbani
- Stem Cells & Regenerative Medicine Section, Developmental Biology & Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,Institute of Hepatology, The Foundation for Liver Research, London, United Kingdom.,Faculty of Life Sciences & Medicine, King's College, London, United Kingdom
| | - Anna Milan
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy.,Department of Women and Children Health, University of Padova, Padova, Italy
| | - Carlo Biz
- Department of Surgery, Oncology, and Gastroenterology DiSCOG, Orthopaedic Clinic, University of Padova, Padua, Italy
| | - Lorenza Lazzari
- Laboratory of Regenerative Medicine - Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Paolo De Coppi
- Stem Cells & Regenerative Medicine Section, Developmental Biology & Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,Specialist Neonatal and Paediatric Surgery, Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Michela Pozzobon
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy.,Department of Women and Children Health, University of Padova, Padova, Italy
| | - Martina Piccoli
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy.,Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
41
|
Patel KH, Dunn AJ, Talovic M, Haas GJ, Marcinczyk M, Elmashhady H, Kalaf EG, Sell SA, Garg K. Aligned nanofibers of decellularized muscle ECM support myogenic activity in primary satellite cells in vitro. ACTA ACUST UNITED AC 2019; 14:035010. [PMID: 30812025 DOI: 10.1088/1748-605x/ab0b06] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Volumetric muscle loss (VML) is a loss of over ∼10% of muscle mass that results in functional impairment. Although skeletal muscle possesses the ability to repair and regenerate itself following minor injuries, VML injuries are irrecoverable. Currently, there are no successful clinical therapies for the treatment of VML. Previous studies have treated VML defects with decellularized extracellular matrix (D-ECM) scaffolds derived from either pig urinary bladder or small intestinal submucosa. These therapies were unsuccessful due to the poor mechanical stability of D-ECM leading to quick degradation in vivo. To circumvent these issues, in this manuscript aligned nanofibers of D-ECM were created using electrospinning that mimicked native muscle architecture and provided topographical cues to primary satellite cells. Additionally, combining D-ECM with polycaprolactone (PCL) improved the tensile mechanical properties of the electrospun scaffold. In vitro testing shows that the electrospun scaffold with aligned nanofibers of PCL and D-ECM supports satellite cell growth, myogenic protein expression, and myokine production.
Collapse
|
42
|
Al Halabi F, Gryshkov O, Kuhn AI, Kapralova VM, Glasmacher B. Force induced piezoelectric effect of polyvinylidene fluoride and polyvinylidene fluoride-co-trifluoroethylene nanofibrous scaffolds. Int J Artif Organs 2018; 41:811-822. [DOI: 10.1177/0391398818785049] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Polyvinylidene fluoride and its co-polymer with trifluoroethylene are promising biomaterials for supporting nerve regeneration processes because of their proven biocompatibility and piezoelectric properties that could stimulate cell ingrowth due to electrical activity upon mechanical deformation. This study reports the piezoelectric effect of electrospun polyvinylidene fluoride scaffolds in response to mechanical loading. An impact test machine was used to evaluate the generation of electrical voltage upon application of an impact load. Scaffolds were produced via electrospinning from polyvinylidene fluoride and polyvinylidene fluoride-co-trifluoroethylene with concentrations of 10–20 wt% dissolved in N,N-dimethylformamide (DMF) and acetone (6:4). The structural and thermal properties of scaffolds were analyzed using Fourier Transform Infrared Spectroscopy and Differential Scanning Calorimetry, respectively. The piezoelectric response of the scaffolds was induced using a custom-made manual impact press machine. Impact forces between 0.4 and 14 N were applied. Fourier Transform Infrared Spectroscopy and Differential Scanning Calorimetry results demonstrated the piezoelectric effect of the electrospun polyvinylidene fluoride and polyvinylidene fluoride-co-trifluoroethylene scaffolds. All the scaffolds exhibited a piezoelectric polar beta-phase formation. Their thermal enthalpies were higher than the value of the initial materials and exhibited a better tendency of crystallization. The electrospun scaffolds exhibited piezoelectric responses in form of voltage by applying impact load. Polyvinylidene fluoride-co-trifluoroethylene scaffolds showed higher values in the range of 6–30 V as compared to pure polyvinylidene fluoride. Here, the mechanically induced electrical impulses measured were between 2.5 and 8 V. Increasing the impact forces did not increase the piezoelectric effect. The results demonstrate the possibility of producing electrospun polyvinylidene fluoride and polyvinylidene fluoride-co-trifluoroethylene scaffolds as nerve guidance with piezoelectric response. Further experiments must be carried out to analyze the piezoelectricity at dynamic conditions.
Collapse
Affiliation(s)
- Fedaa Al Halabi
- Institute for Multiphase Processes, Leibniz Universität Hannover, Hannover, Germany
| | - Oleksandr Gryshkov
- Institute for Multiphase Processes, Leibniz Universität Hannover, Hannover, Germany
| | - Antonia I Kuhn
- Institute for Multiphase Processes, Leibniz Universität Hannover, Hannover, Germany
| | - Viktoria M Kapralova
- Higher School of Applied Physics and Space Technologies, Peter the Great St. Petersburg Polytechnic University, Saint Petersburg, Russia
| | - Birgit Glasmacher
- Institute for Multiphase Processes, Leibniz Universität Hannover, Hannover, Germany
| |
Collapse
|
43
|
Hotfiel T, Seil R, Bily W, Bloch W, Gokeler A, Krifter RM, Mayer F, Ueblacker P, Weisskopf L, Engelhardt M. Nonoperative treatment of muscle injuries - recommendations from the GOTS expert meeting. J Exp Orthop 2018; 5:24. [PMID: 29931565 PMCID: PMC6013414 DOI: 10.1186/s40634-018-0139-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 06/11/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Muscle injuries are some of the most common injuries in sports; they have a high recurrence rate and can result in the loss of ability to participate in training or competition. In clinical practice, a wide variety of treatment strategies are commonly applied. However, a limited amount of evidence-based data exists, and most therapeutic approaches are solely based on "best practice". Thus, there is a need for consensus to provide strategies and recommendations for the treatment of muscle injuries. METHODS The 2016 GOTS Expert Meeting, initiated by the German-Austrian-Swiss Society for Orthopaedic Traumatologic Sports Medicine (GOTS), focused on the topic of muscle and tendon injuries and was held in Spreewald/Berlin, Germany. The committee was composed of twenty-two medical specialists. Nine of them were delegated to a subcommittee focusing on the nonoperative treatment of muscle injuries. The recommendations and statements that were developed were reviewed by the entire consensus committee and voted on by the members. RESULTS The committee reached a consensus on the utility and effectiveness of the management of muscle injuries. MAIN RESULTS the "PRICE" principle to target the first inflammatory response is one of the most relevant steps in the treatment of muscle injuries. Haematoma aspiration may be considered in the early stages after injury. There is presently no clear evidence that intramuscular injections are of use in the treatment of muscle injuries. The ingestion of non-steroidal anti-inflammatory drugs (NSAIDs) should be regarded critically because there is currently no hard evidence to support their use, although they are appropriate in exceptional cases. CONCLUSIONS The present work provides a structured overview of the various nonoperative treatment strategies of muscle injuries and evaluates their effectiveness with respect to the existing scientific evidence and clinical expertise in the context of basic science on the healing process of muscle injuries. The committee agreed that there is a compelling need for further studies, including high-quality randomized investigations to completely evaluate the effectiveness of the existing therapeutic approaches. The given recommendations may be updated and adjusted as further evidence will be generated.
Collapse
Affiliation(s)
- T Hotfiel
- Department of Orthopaedic Surgery, Friedrich-Alexander-University Erlangen-Nuremberg, Rathsbergerstraße 57, D-91054, Erlangen, Germany.
| | - R Seil
- Department of Orthopaedic Surgery, Clinique d'Eich - Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
- Sports Medicine Research Laboratory, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - W Bily
- Department of Physical Medicine and Rehabilitation, Wilhelminenspital, Vienna, Austria
| | - W Bloch
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - A Gokeler
- Luxembourg Institute of Research in Orthopedics, Sports Medicine and Science, Luxembourg City, Luxembourg
- Exercise Science and Neuroscience, Department Exercise & Health Faculty of Science, Paderborn University, Paderborn, Germany
| | - R M Krifter
- ORTHOFOCUS-Orthopedic Competence Center, Graz-Salzburg, Austria
| | - F Mayer
- Outpatient Clinic Potsdam, Sports Medicine & Sports Orthopaedics, University of Potsdam, Potsdam, Germany
| | - P Ueblacker
- MW Center of Orthopedics and Sports Medicine, Munich, Germany
| | - L Weisskopf
- Altius Swiss Sportmed Center, Rheinfelden, Switzerland
| | - M Engelhardt
- Department of Trauma and Orthopedic Surgery, Klinikum Osnabrück, Osnabrück, Germany
| |
Collapse
|
44
|
Regeneration of diaphragm with bio-3D cellular patch. Biomaterials 2018; 167:1-14. [DOI: 10.1016/j.biomaterials.2018.03.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 12/22/2022]
|
45
|
Corona BT, Flanagan KE, Brininger CM, Goldman SM, Call JA, Greising SM. Impact of volumetric muscle loss injury on persistent motoneuron axotomy. Muscle Nerve 2018; 57:799-807. [PMID: 29144551 DOI: 10.1002/mus.26016] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 11/12/2017] [Accepted: 11/14/2017] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Volumetric muscle loss (VML) occurs following significant traumatic injury or surgical removal of skeletal muscle, resulting in nonrecoverable loss of tissue and long-term dysfunction. Perhaps less recognized is that VML injuries inherently disrupt the neuromuscular unit, resulting in fiber denervation and presumptive motor unit rearrangement, expansion, and/or loss. To characterize neural dysfunction we quantified motoneuron axotomy, in efforts to understand how this relates to the temporal coordination of neuromuscular and morphological alterations due to injury. METHODS In an established rat tibialis anterior (TA) VML injury model, we examined the motoneuron, skeletal muscle, and maximal isometric torque at 3, 7, 14, and 21 days postinjury. RESULTS Significant axotomy of 57-79% of all TA muscle motoneurons was observed through 21 days postinjury, which was coupled with a 45-90% TA maximal torque deficit. DISCUSSION A ∼20% partial ablation of the TA muscle causes disproportionate damage across the motor unit acutely postinjury. Muscle Nerve 57: 799-807, 2018.
Collapse
Affiliation(s)
- Benjamin T Corona
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research, 3698 Chambers Pass, BHT1, Fort Sam Houston, Texas, 78234, USA
| | - Kate E Flanagan
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research, 3698 Chambers Pass, BHT1, Fort Sam Houston, Texas, 78234, USA
| | - Christian M Brininger
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research, 3698 Chambers Pass, BHT1, Fort Sam Houston, Texas, 78234, USA
| | - Stephen M Goldman
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research, 3698 Chambers Pass, BHT1, Fort Sam Houston, Texas, 78234, USA
| | - Jarrod A Call
- Department of Kinesiology, University of Georgia, Athens, Georgia, USA.,Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
| | - Sarah M Greising
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research, 3698 Chambers Pass, BHT1, Fort Sam Houston, Texas, 78234, USA
| |
Collapse
|
46
|
Stromal Cell-Derived Factor 1 Plasmid Regenerates Both Smooth and Skeletal Muscle After Anal Sphincter Injury in the Long Term. Dis Colon Rectum 2017; 60:1320-1328. [PMID: 29112569 DOI: 10.1097/dcr.0000000000000940] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Regenerating muscle at a time remote from injury requires re-expression of cytokines to attract stem cells to start and sustain the process of repair. OBJECTIVE We aimed to evaluate the sustainability of muscle regeneration after treatment with a nonviral plasmid expressing stromal cell-derived factor 1. DESIGN This was a randomized study. SETTINGS The study was conducted with animals in a single research facility. INTERVENTIONS Fifty-six female age-/weight-matched Sprague-Dawley rats underwent excision of the ventral half of the anal sphincter complex. Three weeks later, rats were randomly allocated (n = 8) to one of the following groups: no treatment, 100 μg of plasmid encoding stromal cell-derived factor 1 injected locally, local injection of plasmid and 8 × 10 bone marrow-derived mesenchymal stem cells, and plasmid encoding stromal cell-derived factor 1 injected locally with injection of a gelatin scaffold mixed with bone marrow-derived mesenchymal stem cells. MAIN OUTCOME MEASURES Anal manometry, histology, immunohistochemistrym and morphometry were performed 8 weeks after treatment. Protein expression of cytokines CXCR4 and Myf5 was investigated 1 week after treatment (n = 6 per group). ANOVA was used, with p < 0.0083 indicating significant differences for anal manometry and p < 0.05 for all other statistical analysis. RESULTS Eight weeks after treatment, all of the groups receiving the plasmid had significantly higher anal pressures than controls and more organized muscle architecture in the region of the defect. Animals receiving plasmid alone had significantly greater muscle in the defect (p = 0.03) than either animals with injury alone (p = 0.02) or those receiving the plasmid, cells, and scaffold (p = 0.03). Both smooth and skeletal muscles were regenerated significantly more after plasmid treatment. There were no significant differences in the protein levels of CXCR4 or Myf5. LIMITATIONS The study was limited by its small sample size and because stromal cell-derived factor 1 was not blocked. CONCLUSIONS A plasmid expressing stromal cell-derived factor 1 may be sufficient to repair an injured anal sphincter even long after the injury and in the absence of mesenchymal stem cell or scaffold treatments. See Video Abstract at http://links.lww.com/DCR/A451.
Collapse
|
47
|
Hejbøl EK, Sellathurai J, Nair PD, Schrøder HD. Injectable scaffold materials differ in their cell instructive effects on primary human myoblasts. J Tissue Eng 2017; 8:2041731417717677. [PMID: 28717506 PMCID: PMC5502935 DOI: 10.1177/2041731417717677] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/06/2017] [Indexed: 12/15/2022] Open
Abstract
Scaffolds are materials used for delivery of cells for regeneration of tissues. They support three-dimensional organization and improve cell survival. For the repair of small skeletal muscles, injections of small volumes of cells are attractive, and injectable scaffolds for delivery of cells offer a minimally invasive technique. In this study, we examined in vitro the cell instructive effects of three types of injectable scaffolds, fibrin, alginate, and poly(lactic-co-glycolic acid)-based microparticles on primary human myoblasts. The myoblast morphology and progression in the myogenic program differed, depending on the type of scaffold material. In alginate gel, the cells obtained a round morphology, they ceased to proliferate, and entered quiescence. In the fibrin gels, differentiation was promoted, and myotubes were observed within a few days in culture, while poly(lactic-co-glycolic acid)-based microparticles supported prolonged proliferation. Myoblasts released from the alginate and fibrin gels were studied, and cells released from these scaffolds had retained the ability to proliferate and differentiate. Thus, the study shows that human myogenic cells combined with injectable scaffold materials are guided into different states depending on the choice of scaffold. This opens for in vivo experiments, including testing of the significance of the cell state on regeneration potential of primary human myoblasts.
Collapse
Affiliation(s)
- Eva Kildall Hejbøl
- Institute of Clinical Research, SDU Muscle Research Cluster, University of Southern Denmark, Odense, Denmark
| | - Jeeva Sellathurai
- Institute of Clinical Research, SDU Muscle Research Cluster, University of Southern Denmark, Odense, Denmark
| | - Prabha Damodaran Nair
- Division of Tissue Engineering and Regeneration Technologies, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Henrik Daa Schrøder
- Institute of Clinical Research, SDU Muscle Research Cluster, University of Southern Denmark, Odense, Denmark.,Department of Pathology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
48
|
Badylak SF, Dziki JL, Sicari BM, Ambrosio F, Boninger ML. Mechanisms by which acellular biologic scaffolds promote functional skeletal muscle restoration. Biomaterials 2016; 103:128-136. [PMID: 27376561 DOI: 10.1016/j.biomaterials.2016.06.047] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 06/15/2016] [Accepted: 06/20/2016] [Indexed: 12/31/2022]
|
49
|
Ostrovidov S, Shi X, Sadeghian RB, Salehi S, Fujie T, Bae H, Ramalingam M, Khademhosseini A. Stem Cell Differentiation Toward the Myogenic Lineage for Muscle Tissue Regeneration: A Focus on Muscular Dystrophy. Stem Cell Rev Rep 2016; 11:866-84. [PMID: 26323256 DOI: 10.1007/s12015-015-9618-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Skeletal muscle tissue engineering is one of the important ways for regenerating functionally defective muscles. Among the myopathies, the Duchenne muscular dystrophy (DMD) is a progressive disease due to mutations of the dystrophin gene leading to progressive myofiber degeneration with severe symptoms. Although current therapies in muscular dystrophy are still very challenging, important progress has been made in materials science and in cellular technologies with the use of stem cells. It is therefore useful to review these advances and the results obtained in a clinical point of view. This article focuses on the differentiation of stem cells into myoblasts, and their application in muscular dystrophy. After an overview of the different stem cells that can be induced to differentiate into the myogenic lineage, we introduce scaffolding materials used for muscular tissue engineering. We then described some widely used methods to differentiate different types of stem cell into myoblasts. We highlight recent insights obtained in therapies for muscular dystrophy. Finally, we conclude with a discussion on stem cell technology. We discussed in parallel the benefits brought by the evolution of the materials and by the expansion of cell sources which can differentiate into myoblasts. We also discussed on future challenges for clinical applications and how to accelerate the translation from the research to the clinic in the frame of DMD.
Collapse
Affiliation(s)
- Serge Ostrovidov
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai, 980-8577, Japan
| | - Xuetao Shi
- National Engineering Research Center for Tissue Restoration and Reconstruction & School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, People's Republic of China
| | - Ramin Banan Sadeghian
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai, 980-8577, Japan
| | - Sahar Salehi
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai, 980-8577, Japan
| | - Toshinori Fujie
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, 162-8480, Japan
| | - Hojae Bae
- College of Animal Bioscience and Technology, Department of Bioindustrial Technologies, Konkuk University, Hwayang-dong, Kwangjin-gu, Seoul, 143-701, Republic of Korea
| | - Murugan Ramalingam
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai, 980-8577, Japan
- Christian Medical College Bagayam Campus, Centre for Stem Cell Research, Vellore, 632002, India
| | - Ali Khademhosseini
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai, 980-8577, Japan.
- College of Animal Bioscience and Technology, Department of Bioindustrial Technologies, Konkuk University, Hwayang-dong, Kwangjin-gu, Seoul, 143-701, Republic of Korea.
- Division of Biomedical Engineering, Department of Medicine, Harvard Medical School, Biomaterials Innovation Research Center, Brigham and Women's Hospital, Boston, MA, 02139, USA.
- Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.
- Department of Physics, King Abdulaziz University, Jeddah, 21569, Saudi Arabia.
| |
Collapse
|
50
|
Dziki J, Badylak S, Yabroudi M, Sicari B, Ambrosio F, Stearns K, Turner N, Wyse A, Boninger ML, Brown EHP, Rubin JP. An acellular biologic scaffold treatment for volumetric muscle loss: results of a 13-patient cohort study. NPJ Regen Med 2016; 1:16008. [PMID: 29302336 PMCID: PMC5744714 DOI: 10.1038/npjregenmed.2016.8] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 02/08/2016] [Accepted: 03/22/2016] [Indexed: 12/13/2022] Open
Abstract
Volumetric muscle loss (VML) is a severe and debilitating clinical problem. Current standard of care includes physical therapy or orthotics, which do not correct underlying strength deficits, and surgical tendon transfers or muscle transfers, which involve donor site morbidity and fall short of restoring function. The results of a 13-patient cohort study are described herein and involve a regenerative medicine approach for VML treatment. Acellular bioscaffolds composed of mammalian extracellular matrix (ECM) were implanted and combined with aggressive and early physical therapy following treatment. Immunolabeling of ultrasound-guided biopsies, and magnetic resonance imaging and computed tomography imaging were performed to analyse the presence of stem/progenitor cells and formation of new skeletal muscle. Force production, range-of-motion and functional task performance were analysed by physical therapists. Electrodiagnostic evaluation was used to analyse presence of innervated skeletal muscle. This study is registered with ClinicalTrials.gov, numbers NCT01292876. In vivo remodelling of ECM bioscaffolds was associated with mobilisation of perivascular stem cells; formation of new, vascularised, innervated islands of skeletal muscle within the implantation site; increased force production; and improved functional task performance when compared with pre-operative performance. Compared with pre-operative performance, by 6 months after ECM implantation, patients showed an average improvement of 37.3% (P<0.05) in strength and 27.1% improvement in range-of-motion tasks (P<0.05). Implantation of acellular bioscaffolds derived from ECM can improve strength and function, and promotes site-appropriate remodelling of VML defects. These findings provide early evidence of bioscaffolding as a viable treatment of VML.
Collapse
Affiliation(s)
- Jenna Dziki
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mohammad Yabroudi
- Department of Physical Therapy, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Rehabilitation Sciences, Jordan University of Science and Technology, Al Ramtha, Irbid, Jordan
| | - Brian Sicari
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Fabrisia Ambrosio
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Physical Therapy, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kristen Stearns
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA
| | - Neill Turner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aaron Wyse
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael L Boninger
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA
| | - Elke H P Brown
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA
| | - J Peter Rubin
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|