1
|
Nie Y, Mu Q, Sun Y, Ferdous Z, Wang L, Chen C, Nakajima T, Gong JP, Tanaka S, Tsuda M. Mechanochemistry-Induced Universal Hydrogel Surface Modification for Orientation and Enhanced Differentiation of Skeletal Muscle Myoblasts. ACS APPLIED BIO MATERIALS 2025. [PMID: 40106521 DOI: 10.1021/acsabm.4c01991] [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: 03/22/2025]
Abstract
Micropatterned surface substrates containing topographic cues offer the possibility of programming tissue organization as a cell template by guiding cell alignment, adhesion, and function. In this study, we developed and used a force stamp method to grow aligned micropatterns with tunable chemical properties and elasticity on the surface of hydrogels based on a force-triggered polymerization mechanism of double-network hydrogels to elucidate the underlying mechanisms by which cells sense and respond to their mechanical and chemical microenvironments. In this work, we describe the impact of aligned micropatterns on the combined effects of microstructural chemistry and mechanics on the selective adhesion, directed migration, and differentiation of myoblasts. Our investigations revealed that topographically engineered substrates with hydrophobic and elevated surface roughness significantly enhanced myoblast adhesion kinetics. Concurrently, spatially ordered architectures facilitated cytoskeletal reorganization in myocytes, establishing biomechanically favorable niches for syncytial myotube development through extracellular matrix (ECM) physical guidance. Reverse transcription PCR analysis and immunofluorescence revealed that the expression of differentiation-specific genes, myosin heavy chain, and myogenic regulatory factors Myf5 and MyoD was upregulated in muscle cells on the aligned patterned scaffolds. These results suggest that the aligned micropatterns can promote muscle cell differentiation, making them potential scaffolds for enhancing skeletal differentiation.
Collapse
Affiliation(s)
- Yuheng Nie
- Graduate School of Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Japan
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
| | - Qifeng Mu
- Graduate School of Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Japan
| | - Yanpeng Sun
- Graduate School of Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Japan
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
| | - Zannatul Ferdous
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, N21W10, Kita-ku, Sapporo 001-0021, Japan
| | - Lei Wang
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, N21W10, Kita-ku, Sapporo 001-0021, Japan
| | - Cewen Chen
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
| | - Tasuku Nakajima
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, N21W10, Kita-ku, Sapporo 001-0021, Japan
- Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Japan
| | - Jian Ping Gong
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, N21W10, Kita-ku, Sapporo 001-0021, Japan
- Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, N21W10, Kita-ku, Sapporo 001-0021, Japan
- Department of Surgical Pathology, Hokkaido University Hospital, N14W5, Kita-ku, Sapporo 060-8648, Japan
| | - Masumi Tsuda
- Graduate School of Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Japan
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, N21W10, Kita-ku, Sapporo 001-0021, Japan
| |
Collapse
|
2
|
Zhang X, Chen Y, Zhou S, Liu Y, Zhu S, Jia X, Lu Z, Zhang Y, Zhang W, Ye Z, Cai B, Kong L, Liu F. RNA Coating Promotes Peri-Implant Osseointegration. ACS Biomater Sci Eng 2024; 10:7030-7042. [PMID: 38943625 PMCID: PMC11558559 DOI: 10.1021/acsbiomaterials.4c00133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 07/01/2024]
Abstract
In addition to transmitting and carrying genetic information, RNA plays an important abiotic role in the world of nanomaterials. RNA is a natural polyanionic biomacromolecule, and its ability to promote osteogenesis by binding with other inorganic materials as an osteogenic induction agent was discovered only recently. However, whether it can promote osseointegration on implants has not been reported. Here, we investigated the effect of the RNA-containing coating materials on peri-implant osseointegration. Total RNA extracted from rat muscle tissue was used as an osteogenic induction agent, and hyaluronic acid (HA) was used to maintain its negative charge. In simulated body fluids (SBF), in vitro studies demonstrated that the resulting material encouraged calcium salt deposition. Cytological experiments showed that the RNA-containing coating induced greater cell adhesion and osteogenic differentiation in comparison to the control. The results of animal experiments showed that the RNA-containing coating had osteoinductive and bone conduction activities, which are beneficial for bone formation and osseointegration. Therefore, the RNA-containing coatings are useful for the surface modification of titanium implants to promote osseointegration.
Collapse
Affiliation(s)
- Xiao Zhang
- College
of Life Sciences, Northwest University, Xi’an 710069, China
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Yicheng Chen
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Shanluo Zhou
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Ya Liu
- College
of Life Sciences, Northwest University, Xi’an 710069, China
| | - Simin Zhu
- College
of Life Sciences, Northwest University, Xi’an 710069, China
| | - Xuelian Jia
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Zihan Lu
- College
of Life Sciences, Northwest University, Xi’an 710069, China
| | - Yufan Zhang
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Wenhui Zhang
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Zhou Ye
- Applied
Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong 999077, S.A.R., China
| | - Bolei Cai
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Liang Kong
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Fuwei Liu
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| |
Collapse
|
3
|
Mohanty S, Roy S. Bioactive Hydrogels Inspired by Laminin: An Emerging Biomaterial for Tissue Engineering Applications. Macromol Biosci 2024; 24:e2400207. [PMID: 39172212 DOI: 10.1002/mabi.202400207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/01/2024] [Indexed: 08/23/2024]
Abstract
Tissue or organ damage due to severe injuries or chronic diseases can adversely affect the quality of life. Current treatments rely on organ or tissue transplantation which has limitations including unavailability of donors, ethical issues, or immune rejection after transplantations. These limitations can be addressed by tissue regeneration which involves the development of bioactive scaffolds closely mimicking the extracellular matrix (ECM). One of the major components of ECM is the laminin protein which supports several tissues associated with important organs. In this direction, peptide-based hydrogels can effectively mimic the essential characteristics of laminin. While several reports have discussed the structure of laminin, the potential of laminin-derived peptide hydrogels as effective biomaterial for tissue engineering applications is yet to be discussed. In this context, the current review focuses on the structure of laminin and its role as an essential ECM protein. Further, the potential of short peptide hydrogels in mimicking the crucial properties of laminin is proposed. The review further highlights the significance of bioactive hydrogels inspired by laminin - in addressing numerous tissue engineering applications including angiogenesis, neural, skeletal muscle, liver, and adipose tissue regeneration along with a brief outlook on the future applications of these laminin-based hydrogels.
Collapse
Affiliation(s)
- Sweta Mohanty
- Institute of Nano Science and Technology (INST), Sector 81, Knowledge City, Mohali, Punjab, 140306, India
| | - Sangita Roy
- Institute of Nano Science and Technology (INST), Sector 81, Knowledge City, Mohali, Punjab, 140306, India
| |
Collapse
|
4
|
Alonso-Puyo J, Izagirre-Fernandez O, Crende O, Valdivia A, García-Gallastegui P, Sanz B. Experimental models as a tool for research on sarcopenia: A narrative review. Ageing Res Rev 2024; 101:102534. [PMID: 39369798 DOI: 10.1016/j.arr.2024.102534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Sarcopenia is a musculoskeletal disorder related to muscle mass and function; as the worldwide population ages, its growing prevalence means a decline in quality of life and an increased burden for public health systems. As sarcopenia is a reversible condition, its early diagnosis is of utmost importance. Consensus definitions and diagnosis protocols for sarcopenia have been evolving for a long time, and the identification of molecular pathways subjacent to sarcopenia is a growing research area. The use of liquid biopsies to identify circulating molecules does not provide information about specific regulatory pathways or biomarkers in relevant tissue, and the use of skeletal muscle biopsies from older people has many limitations. Complementary tools are therefore necessary to advance the knowledge of relevant molecular aspects. The development of experimental models, such as animal, cellular, or bioengineered tissue, together with knock-in or knock-out strategies, could therefore be of great interest. This narrative review will explore experimental models of healthy muscle and aged muscle cells as a tool for research on sarcopenia. We will summarize the literature and present relevant experimental models in terms of their advantages and disadvantages. All of the presented approaches could potentially contribute to the accurate and early diagnosis, follow-up, and possible treatment of sarcopenia.
Collapse
Affiliation(s)
- Janire Alonso-Puyo
- Physiology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., Leioa 48940, Spain
| | - Oihane Izagirre-Fernandez
- Physiology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., Leioa 48940, Spain
| | - Olatz Crende
- Cell Biology and Histology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., Leioa 48940, Spain
| | - Asier Valdivia
- Cell Biology and Histology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., Leioa 48940, Spain
| | - Patricia García-Gallastegui
- Physiology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., Leioa 48940, Spain.
| | - Begoña Sanz
- Physiology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., Leioa 48940, Spain; Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia 48903, Spain.
| |
Collapse
|
5
|
Singh R, Rawat H, Kumar A, Gandhi Y, Kumar V, Mishra SK, Narasimhaji CV. Graphene and its hybrid nanocomposite: A Metamorphoses elevation in the field of tissue engineering. Heliyon 2024; 10:e33542. [PMID: 39040352 PMCID: PMC11261797 DOI: 10.1016/j.heliyon.2024.e33542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 06/06/2024] [Accepted: 06/23/2024] [Indexed: 07/24/2024] Open
Abstract
In this discourse, we delve into the manifold applications of graphene-based nanomaterials (GBNs) in the realm of biomedicine. Graphene, characterized by its two-dimensional planar structure, superconductivity, mechanical robustness, chemical inertness, extensive surface area, and propitious biocompatibility, stands as an exemplary candidate for diverse biomedical utility. Graphene include various distinctive characteristics of its two-dimensional planar structure, enormous surface area, mechanical and chemical stability, high conductivity, and exceptional biocompatibility. We investigate graphene and its diverse derivatives, which include reduced graphene oxides (rGOs), graphene oxides (GOs), and graphene composites, with a focus on elucidating the unique attributes relevant to their biomedical utility. In this review article it highlighted the unique properties of graphene, synthesis methods of graphene and functionalization methods of graphene. In the quest for novel materials to advance regenerative medicine, researchers have increasingly turned their attention to graphene-based materials, which have emerged as a prominent innovation in recent years. Notably, it highlights their applications in the regeneration of various tissues, including nerves, skeletal muscle, bones, skin, cardiac tissue, cartilage, and adipose tissue, as well as their influence on induced pluripotent stem cells, marking significant breakthroughs in the field of regenerative medicine. Additionally, this review article explores future prospects in this evolving area of study.
Collapse
Affiliation(s)
- Rajesh Singh
- Department of Chemistry, Central Ayurveda Research Institute Jhansi, U.P, 284003, India
| | - Hemant Rawat
- Department of Chemistry, Central Ayurveda Research Institute Jhansi, U.P, 284003, India
| | - Ashwani Kumar
- Department of Heterogeneous Catalysis, Max-Planck-Institut für Kohlenforschung, Kaiser-Wilhelm-Platz 1, 45470, Mülheim an der Ruhr, Germany
| | - Yashika Gandhi
- Department of Chemistry, Central Ayurveda Research Institute Jhansi, U.P, 284003, India
| | - Vijay Kumar
- Department of Chemistry, Central Ayurveda Research Institute Jhansi, U.P, 284003, India
| | - Sujeet K. Mishra
- Department of Chemistry, Central Ayurveda Research Institute Jhansi, U.P, 284003, India
| | | |
Collapse
|
6
|
Jo HJ, Kang MS, Heo HJ, Jang HJ, Park R, Hong SW, Kim YH, Han DW. Skeletal muscle regeneration with 3D bioprinted hyaluronate/gelatin hydrogels incorporating MXene nanoparticles. Int J Biol Macromol 2024; 265:130696. [PMID: 38458288 DOI: 10.1016/j.ijbiomac.2024.130696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/31/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024]
Abstract
There has been significant progress in the field of three-dimensional (3D) bioprinting technology, leading to active research on creating bioinks capable of producing structurally and functionally tissue-mimetic constructs. Ti3C2Tx MXene nanoparticles (NPs), promising two-dimensional nanomaterials, are being investigated for their potential in muscle regeneration due to their unique physicochemical properties. In this study, we integrated MXene NPs into composite hydrogels made of gelatin methacryloyl (GelMA) and hyaluronic acid methacryloyl (HAMA) to develop bioinks (namely, GHM bioink) that promote myogenesis. The prepared GHM bioinks were found to offer excellent printability with structural integrity, cytocompatibility, and microporosity. Additionally, MXene NPs within the 3D bioprinted constructs encouraged the differentiation of C2C12 cells into skeletal muscle cells without additional support of myogenic agents. Genetic analysis indicated that representative myogenic markers both for early and late myogenesis were significantly up-regulated. Moreover, animal studies demonstrated that GHM bioinks contributed to enhanced regeneration of skeletal muscle while reducing immune responses in mice models with volumetric muscle loss (VML). Our results suggest that the GHM hydrogel can be exploited to craft a range of strategies for the development of a novel bioink to facilitate skeletal muscle regeneration because these MXene-incorporated composite materials have the potential to promote myogenesis.
Collapse
Affiliation(s)
- Hyo Jung Jo
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea
| | - Moon Sung Kang
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea
| | - Hye Jin Heo
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Hee Jeong Jang
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea
| | - Rowoon Park
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea
| | - Suck Won Hong
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea; Engineering Research Center for Color-Modulated Extra-Sensory Perception Technology, Pusan National University, Busan 46241, Republic of Korea
| | - Yun Hak Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; Periodontal Disease Signaling Network Research Center & Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea.
| | - Dong-Wook Han
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea; BIO-IT Fusion Technology Research Institute, Pusan National University, Busan 46241, Republic of Korea.
| |
Collapse
|
7
|
Xu Z, Arkudas A, Munawar MA, Schubert DW, Fey T, Weisbach V, Mengen LM, Horch RE, Cai A. Schwann Cells Do Not Promote Myogenic Differentiation in the EPI Loop Model. Tissue Eng Part A 2024; 30:244-256. [PMID: 38063005 DOI: 10.1089/ten.tea.2023.0215] [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: 01/12/2024] Open
Abstract
In skeletal muscle tissue engineering, innervation and vascularization play an essential role in the establishment of functional skeletal muscle. For adequate three-dimensional assembly, biocompatible aligned nanofibers are beneficial as matrices for cell seeding. The aim of this study was to analyze the impact of Schwann cells (SC) on myoblast (Mb) and adipogenic mesenchymal stromal cell (ADSC) cocultures on poly-ɛ-caprolactone (PCL)-collagen I-nanofibers in vivo. Human Mb/ADSC cocultures, as well as Mb/ADSC/SC cocultures, were seeded onto PCL-collagen I-nanofiber scaffolds and implanted into the innervated arteriovenous loop model (EPI loop model) of immunodeficient rats for 4 weeks. Histological staining and gene expression were used to compare their capacity for vascularization, immunological response, myogenic differentiation, and innervation. After 4 weeks, both Mb/ADSC and Mb/ADSC/SC coculture systems showed similar amounts and distribution of vascularization, as well as immunological activity. Myogenic differentiation could be observed in both groups through histological staining (desmin, myosin heavy chain) and gene expression (MYOD, MYH3, ACTA1) without significant difference between groups. Expression of CHRNB and LAMB2 also implied neuromuscular junction formation. Our study suggests that the addition of SC did not significantly impact myogenesis and innervation in this model. The implanted motor nerve branch may have played a more significant role than the presence of SC.
Collapse
Affiliation(s)
- Zhou Xu
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Thyroid and Breast Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Andreas Arkudas
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Muhammad Azeem Munawar
- Department of Materials Science and Engineering, Institute of Polymer Materials, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Dirk W Schubert
- Department of Materials Science and Engineering, Institute of Polymer Materials, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Tobias Fey
- Department of Materials Science and Engineering, Institute of Glass and Ceramics, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Volker Weisbach
- Department of Transfusion Medicine, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lilly M Mengen
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Raymund E Horch
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Aijia Cai
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
8
|
Kong J, Ju X, Qi G, Wang J, Diao X, Wang B, Zhang C, Li J, Jin Y. "Light-On" Fluorescent Nanoprobes for Monitoring Dynamic Distribution of Cellular Nucleolin During Pyroptosis. Anal Chem 2024; 96:926-933. [PMID: 38158373 DOI: 10.1021/acs.analchem.3c05122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Nucleolin (NCL) is a multifunctional nuclear protein that plays significant roles in regulating physiological activities of the cells. However, it remains a challenge to monitor the dynamic distribution and expression of nucleolin within living cells during cell stress processes directly. Here, we designed "turn-on" fluorescent nanoprobes composed of specific AS1411 aptamer and nucleus-targeting peptide on gold nanoparticles (AuNPs) to effectively capture and track the NCL distribution and expression during pyroptosis triggered by electrical stimulation (ES). The distribution of nucleolin in the cell membrane and nucleus can be easily observed by simply changing the particle size of the nanoprobes. The present strategy exhibits obvious advantages such as simple operation, low cost, time saving, and suitability for living cell imaging. The ES can induce cancer cell pyroptosis controllably and selectively, with less harm to the viability of normal cells. The palpable cell nuclear stress responses of cancerous cells, including nucleus wrinkling and nucleolus fusion after ES at 1.0 V were obviously observed. Compared with normal cells (MCF-10A), NCL is overexpressed within cancerous cells (MCF-7 cells) using the as-designed nanoprobes, and the ES can effectively inhibit NCL expression within cancerous cells. The developed NCL sensing platform and ES-based methods hold great potential for cellular studies of cancer-related diseases.
Collapse
Affiliation(s)
- Jiao Kong
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin,P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Xingkai Ju
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin,P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Guohua Qi
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin,P. R. China
| | - Jiafeng Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin,P. R. China
- Department of Endodontics, School and Hospital of Stomatology, Jilin University, Changchun 130021, Jilin,P. R. China
| | - Xingkang Diao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin,P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Bo Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin,P. R. China
| | - Chenyu Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin,P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Jing Li
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin,P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Yongdong Jin
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin,P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
- Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, P. R. China
| |
Collapse
|
9
|
Abalymov A, Pinchasik BE, Akasov RA, Lomova M, Parakhonskiy BV. Strategies for Anisotropic Fibrillar Hydrogels: Design, Cell Alignment, and Applications in Tissue Engineering. Biomacromolecules 2023; 24:4532-4552. [PMID: 37812143 DOI: 10.1021/acs.biomac.3c00503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Efficient cellular alignment in biomaterials presents a considerable challenge, demanding the refinement of appropriate material morphologies, while ensuring effective cell-surface interactions. To address this, biomaterials are continuously researched with diverse coatings, hydrogels, and polymeric surfaces. In this context, we investigate the influence of physicochemical parameters on the architecture of fibrillar hydrogels that significantly orient the topography of flexible hydrogel substrates, thereby fostering cellular adhesion and spatial organization. Our Review comprehensively assesses various techniques for aligning polymer fibrils within hydrogels, specifically interventions applied during and after the cross-linking process. These methodologies include mechanical strains, precise temperature modulation, controlled fluidic dynamics, and chemical modulators, as well as the use of magnetic and electric fields. We highlight the intrinsic appeal of these methodologies in fabricating cell-aligning interfaces and discuss their potential implications within the fields of biomaterials and tissue engineering, particularly concerning the pursuit of optimal cellular alignment.
Collapse
Affiliation(s)
- Anatolii Abalymov
- Science Medical Center, Saratov State University, 410012 Saratov, Russia
| | - Bat-El Pinchasik
- School of Mechanical Engineering, Faculty of Engineering, Tel-Aviv University, 69978 Tel-Aviv, Israel
| | - Roman A Akasov
- Sechenov University and Federal Scientific Research Centre "Crystallography and Photonics" of Russian Academy of Sciences, 101000 Moscow, Russia
| | - Maria Lomova
- Science Medical Center, Saratov State University, 410012 Saratov, Russia
| | - Bogdan V Parakhonskiy
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
10
|
Tzimorotas D, Solberg NT, Andreassen RC, Moutsatsou P, Bodiou V, Pedersen ME, Rønning SB. Expansion of bovine skeletal muscle stem cells from spinner flasks to benchtop stirred-tank bioreactors for up to 38 days. Front Nutr 2023; 10:1192365. [PMID: 37609488 PMCID: PMC10442166 DOI: 10.3389/fnut.2023.1192365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 07/10/2023] [Indexed: 08/24/2023] Open
Abstract
Introduction Successful long-term expansion of skeletal muscle satellite cells (MuSCs) on a large scale is fundamental for cultivating animal cells for protein production. Prerequisites for efficient cell expansion include maintaining essential native cell activities such as cell adhesion, migration, proliferation, and differentiation while ensuring consistent reproducibility. Method This study investigated the growth of bovine MuSC culture using low-volume spinner flasks and a benchtop stirred-tank bioreactor (STR). Results and discussion Our results showed for the first time the expansion of primary MuSCs for 38 days in a bench-top STR run with low initial seeding density and FBS reduction, supported by increased expression of the satellite cell marker PAX7 and reduced expression of differentiation-inducing genes like MYOG, even without adding p38-MAPK inhibitors. Moreover, the cells retained their ability to proliferate, migrate, and differentiate after enzymatic dissociation from the microcarriers. We also showed reproducible results in a separate biological benchtop STR run.
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
|
Miranda Alarcón YS, Jazwinska D, Lymon T, Khalili A, Browe D, Newton B, Pellegrini M, Cohen RI, Shreiber DI, Freeman JW. The Use of Collagen Methacrylate in Actuating Polyethylene Glycol Diacrylate-Acrylic Acid Scaffolds for Muscle Regeneration. Ann Biomed Eng 2023; 51:1165-1180. [PMID: 36853478 DOI: 10.1007/s10439-023-03139-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 01/03/2023] [Indexed: 03/01/2023]
Abstract
After muscle loss or injury, skeletal muscle tissue has the ability to regenerate and return its function. However, large volume defects in skeletal muscle tissue pose a challenge to regenerate due to the absence of regenerative elements such as biophysical and biochemical cues, making the development of new treatments necessary. One potential solution is to utilize electroactive polymers that can change size or shape in response to an external electric field. Poly(ethylene glycol) diacrylate (PEGDA) is one such polymer, which holds great potential as a scaffold for muscle tissue regeneration due to its mechanical properties. In addition, the versatile chemistry of this polymer allows for the conjugation of new functional groups to enhance its electroactive properties and biocompatibility. Herein, we have developed an electroactive copolymer of PEGDA and acrylic acid (AA) in combination with collagen methacrylate (CMA) to promote cell adhesion and proliferation. The electroactive properties of the CMA + PEGDA:AA constructs were investigated through actuation studies. Furthermore, the biological properties of the hydrogel were investigated in a 14-day in vitro study to evaluate myosin light chain (MLC) expression and metabolic activity of C2C12 mouse myoblast cells. The addition of CMA improved some aspects of material bioactivity, such as MLC expression in C2C12 mouse myoblast cells. However, the incorporation of CMA in the PEGDA:AA hydrogels reduced the sample movement when placed under an electric field, possibly due to steric hindrance from the CMA. Further research is needed to optimize the use of CMA in combination with PEGDA:AA as a potential scaffold for skeletal muscle tissue engineering.
Collapse
Affiliation(s)
| | - Dorota Jazwinska
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Terrence Lymon
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Amin Khalili
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Daniel Browe
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Brandon Newton
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Michael Pellegrini
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Rick I Cohen
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - David I Shreiber
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Joseph W Freeman
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA.
| |
Collapse
|
13
|
Rosa E, de Mello L, Castelletto V, Dallas ML, Accardo A, Seitsonen J, Hamley IW. Cell Adhesion Motif-Functionalized Lipopeptides: Nanostructure and Selective Myoblast Cytocompatibility. Biomacromolecules 2023; 24:213-224. [PMID: 36520063 PMCID: PMC9832505 DOI: 10.1021/acs.biomac.2c01068] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The conformation and self-assembly of four lipopeptides, peptide amphiphiles comprising peptides conjugated to lipid chains, in aqueous solution have been examined. The peptide sequence in all four lipopeptides contains the integrin cell adhesion RGDS motif, and the cytocompatibility of the lipopeptides is also analyzed. Lipopeptides have either tetradecyl (C14, myristyl) or hexadecyl (C16, palmitoyl) lipid chains and peptide sequence WGGRGDS or GGGRGDS, that is, with either a tryptophan-containing WGG or triglycine GGG tripeptide spacer between the bioactive peptide motif and the alkyl chain. All four lipopeptides self-assemble above a critical aggregation concentration (CAC), determined through several comparative methods using circular dichroism (CD) and fluorescence. Spectroscopic methods [CD and Fourier transform infrared (FTIR) spectroscopy] show the presence of β-sheet structures, consistent with the extended nanotape, helical ribbon, and nanotube structures observed by cryogenic transmission electron microscopy (cryo-TEM). The high-quality cryo-TEM images clearly show the coexistence of helically twisted ribbon and nanotube structures for C14-WGGRGDS, which highlight the mechanism of nanotube formation by the closure of the ribbons. Small-angle X-ray scattering shows that the nanotapes comprise highly interdigitated peptide bilayers, which are also present in the walls of the nanotubes. Hydrogel formation was observed at sufficiently high concentrations or could be induced by a heat/cool protocol at lower concentrations. Birefringence due to nematic phase formation was observed for several of the lipopeptides, along with spontaneous flow alignment of the lyotropic liquid crystal structure in capillaries. Cell viability assays were performed using both L929 fibroblasts and C2C12 myoblasts to examine the potential uses of the lipopeptides in tissue engineering, with a specific focus on application to cultured (lab-grown) meat, based on myoblast cytocompatibility. Indeed, significantly higher cytocompatibility of myoblasts was observed for all four lipopeptides compared to that for fibroblasts, in particular at a lipopeptide concentration below the CAC. Cytocompatibility could also be improved using hydrogels as cell supports for fibroblasts or myoblasts. Our work highlights that precision control of peptide sequences using bulky aromatic residues within "linker sequences" along with alkyl chain selection can be used to tune the self-assembled nanostructure. In addition, the RGDS-based lipopeptides show promise as materials for tissue engineering, especially those of muscle precursor cells.
Collapse
Affiliation(s)
- Elisabetta Rosa
- School
of Chemistry, Pharmacy and Food Biosciences, University of Reading, Whiteknights,
Reading, Berkshire RG6 6AD, U.K.,Department
of Pharmacy and Research Centre on Bioactive Peptides (CIRPeB), University of Naples “Federico II”, Via Domenico Montesano 49, Naples 80131, Italy
| | - Lucas de Mello
- School
of Chemistry, Pharmacy and Food Biosciences, University of Reading, Whiteknights,
Reading, Berkshire RG6 6AD, U.K.,Departamento
de Biofísica, Universidade Federal
de São Paulo, São
Paulo 04023-062, Brazil
| | - Valeria Castelletto
- School
of Chemistry, Pharmacy and Food Biosciences, University of Reading, Whiteknights,
Reading, Berkshire RG6 6AD, U.K.
| | - Mark L. Dallas
- School
of Chemistry, Pharmacy and Food Biosciences, University of Reading, Whiteknights,
Reading, Berkshire RG6 6AD, U.K.
| | - Antonella Accardo
- Department
of Pharmacy and Research Centre on Bioactive Peptides (CIRPeB), University of Naples “Federico II”, Via Domenico Montesano 49, Naples 80131, Italy
| | - Jani Seitsonen
- Nanomicroscopy
Center, Aalto University, Puumiehenkuja 2, Espoo FIN-02150, Finland
| | - Ian W. Hamley
- School
of Chemistry, Pharmacy and Food Biosciences, University of Reading, Whiteknights,
Reading, Berkshire RG6 6AD, U.K.,
| |
Collapse
|
14
|
Başımoğlu Koca Y, Koca S, Öztel Z, Balcan E. Determination of histopathological effects and myoglobin, periostin gene-protein expression levels in Danio rerio muscle tissue after acaricide yoksorrun-5EC (hexythiazox) application. Drug Chem Toxicol 2023; 46:50-58. [PMID: 34879781 DOI: 10.1080/01480545.2021.2007945] [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: 02/02/2023]
Abstract
Although pesticides are essential agrochemicals to annihilate harmful organisms in agriculture, their uncontrolled use has become an important threat to environmental health. Exposure to pesticides can affect many biological systems including immune system, endocrine system, and nervous system. However, the potential side effects of pesticides to skeletal muscle system remain unclear. Present study has focused on the evaluation of this issue by using an acaricide, yoksorrun-5EC (hexythiazox), in an aquatic model organism, Danio rerio. The histological analyses revealed that increased concentrations of the acaricide cause degradation of skeletal muscle along with increased necrosis and atrophy in myocytes, intercellular edema, and increased infiltrations between perimysium sheaths of muscle fibers. The effects of acaricide on myoglobin and periostin, which are associated with oxygen transport and muscle regeneration, respectively, were investigated at the gene and protein levels. RT-PCR results suggested that high concentration yoksorrun-5EC (hexythiazox) can induce myoglobin and periostin genes. Similar results were also obtained in the protein levels of these genes by western blotting analysis. These results suggested that yoksorrun-5EC (hexythiazox)-dependent disruption of skeletal muscle architecture is closely associated with the expression levels of myoglobin and periostin genes in Danio rerio model.
Collapse
Affiliation(s)
- Yücel Başımoğlu Koca
- Department of Biology, Zoology Section, Faculty of Science and Art, Adnan Menderes University, Aydin, Turkey
| | - Serdar Koca
- Department of Biology, General Biology Section, Faculty of Science and Art, Adnan Menderes University, Aydin, Turkey
| | - Zübeyde Öztel
- Department of Biology, Molecular Biology Section, Faculty of Science and Art, Manisa Celal Bayar University, Manisa, Turkey
| | - Erdal Balcan
- Department of Biology, Molecular Biology Section, Faculty of Science and Art, Manisa Celal Bayar University, Manisa, Turkey
| |
Collapse
|
15
|
Norris SCP, Kawecki NS, Davis AR, Chen KK, Rowat AC. Emulsion-templated microparticles with tunable stiffness and topology: Applications as edible microcarriers for cultured meat. Biomaterials 2022; 287:121669. [PMID: 35853359 PMCID: PMC9834440 DOI: 10.1016/j.biomaterials.2022.121669] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 06/27/2022] [Accepted: 07/02/2022] [Indexed: 01/16/2023]
Abstract
Cultured meat has potential to diversify methods for protein production, but innovations in production efficiency will be required to make cultured meat a feasible protein alternative. Microcarriers provide a strategy to culture sufficient volumes of adherent cells in a bioreactor that are required for meat products. However, cell culture on inedible microcarriers involves extra downstream processing to dissociate cells prior to consumption. Here, we present edible microcarriers that can support the expansion and differentiation of myogenic cells in a single bioreactor system. To fabricate edible microcarriers with a scalable process, we used water-in-oil emulsions as templates for gelatin microparticles. We also developed a novel embossing technique to imprint edible microcarriers with grooved topology in order to test if microcarriers with striated surface texture can promote myoblast proliferation and differentiation in suspension culture. In this proof-of-concept demonstration, we showed that edible microcarriers with both smooth and grooved surface topologies supported the proliferation and differentiation of mouse myogenic C2C12 cells in a suspension culture. The grooved edible microcarriers showed a modest increase in the proliferation and alignment of myogenic cells compared to cells cultured on smooth, spherical microcarriers. During the expansion phase, we also observed the formation of cell-microcarrier aggregates or 'microtissues' for cells cultured on both smooth and grooved microcarriers. Myogenic microtissues cultured with smooth and grooved microcarriers showed similar characteristics in terms of myotube length, myotube volume fraction, and expression of myogenic markers. To establish feasibility of edible microcarriers for cultured meat, we showed that edible microcarriers supported the production of myogenic microtissue from C2C12 or bovine satellite muscle cells, which we harvested by centrifugation into a cookable meat patty that maintained its shape and exhibited browning during cooking. These findings demonstrate the potential of edible microcarriers for the scalable production of cultured meat in a single bioreactor.
Collapse
Affiliation(s)
- Sam C P Norris
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - N Stephanie Kawecki
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ashton R Davis
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kathleen K Chen
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Amy C Rowat
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Broad Stem Cell Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
16
|
Zhao G, Zhou H, Jin G, Jin B, Geng S, Luo Z, Ge Z, Xu F. Rational Design of Electrically Conductive Biomaterials toward Excitable Tissues Regeneration. Prog Polym Sci 2022. [DOI: 10.1016/j.progpolymsci.2022.101573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
17
|
Tomasch J, Maleiner B, Heher P, Rufin M, Andriotis OG, Thurner PJ, Redl H, Fuchs C, Teuschl-Woller AH. Changes in Elastic Moduli of Fibrin Hydrogels Within the Myogenic Range Alter Behavior of Murine C2C12 and Human C25 Myoblasts Differently. Front Bioeng Biotechnol 2022; 10:836520. [PMID: 35669058 PMCID: PMC9164127 DOI: 10.3389/fbioe.2022.836520] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Fibrin hydrogels have proven highly suitable scaffold materials for skeletal muscle tissue engineering in the past. Certain parameters of those types of scaffolds, however, greatly affect cellular mechanobiology and therefore the myogenic outcome. The aim of this study was to identify the influence of apparent elastic properties of fibrin scaffolds in 2D and 3D on myoblasts and evaluate if those effects differ between murine and human cells. Therefore, myoblasts were cultured on fibrin-coated multiwell plates ("2D") or embedded in fibrin hydrogels ("3D") with different elastic moduli. Firstly, we established an almost linear correlation between hydrogels' fibrinogen concentrations and apparent elastic moduli in the range of 7.5 mg/ml to 30 mg/ml fibrinogen (corresponds to a range of 7.7-30.9 kPa). The effects of fibrin hydrogel elastic modulus on myoblast proliferation changed depending on culture type (2D vs 3D) with an inhibitory effect at higher fibrinogen concentrations in 3D gels and vice versa in 2D. The opposite effect was evident in differentiating myoblasts as shown by gene expression analysis of myogenesis marker genes and altered myotube morphology. Furthermore, culture in a 3D environment slowed down proliferation compared to 2D, with a significantly more pronounced effect on human myoblasts. Differentiation potential was also substantially impaired upon incorporation into 3D gels in human, but not in murine, myoblasts. With this study, we gained further insight in the influence of apparent elastic modulus and culture type on cellular behavior and myogenic outcome of skeletal muscle tissue engineering approaches. Furthermore, the results highlight the need to adapt parameters of 3D culture setups established for murine cells when applied to human cells.
Collapse
Affiliation(s)
- Janine Tomasch
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Babette Maleiner
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Philipp Heher
- Ludwig Randall Centre for Cell and Molecular Biophysics, King’s College London, Guy’s Campus, London, United Kingdom
| | - Manuel Rufin
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Institute of Lightweight Design and Structural Biomechanics, TU Wien, Vienna, Austria
| | - Orestis G. Andriotis
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Institute of Lightweight Design and Structural Biomechanics, TU Wien, Vienna, Austria
| | - Philipp J. Thurner
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Institute of Lightweight Design and Structural Biomechanics, TU Wien, Vienna, Austria
| | - Heinz Redl
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
| | - Christiane Fuchs
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Wellman Center for Photomedicine, MGH, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Andreas H. Teuschl-Woller
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
18
|
Silk Fibroin-Based Biomaterials for Tissue Engineering Applications. Molecules 2022; 27:molecules27092757. [PMID: 35566110 PMCID: PMC9103528 DOI: 10.3390/molecules27092757] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/08/2022] [Accepted: 04/21/2022] [Indexed: 12/21/2022] Open
Abstract
Tissue engineering (TE) involves the combination of cells with scaffolding materials and appropriate growth factors in order to regenerate or replace damaged and degenerated tissues and organs. The scaffold materials serve as templates for tissue formation and play a vital role in TE. Among scaffold materials, silk fibroin (SF), a naturally occurring protein, has attracted great attention in TE applications due to its excellent mechanical properties, biodegradability, biocompatibility, and bio-absorbability. SF is usually dissolved in an aqueous solution and can be easily reconstituted into different forms, including films, mats, hydrogels, and sponges, through various fabrication techniques, including spin coating, electrospinning, freeze drying, and supercritical CO2-assisted drying. Furthermore, to facilitate the fabrication of more complex SF-based scaffolds, high-precision techniques such as micro-patterning and bio-printing have been explored in recent years. These processes contribute to the diversity of surface area, mean pore size, porosity, and mechanical properties of different silk fibroin scaffolds and can be used in various TE applications to provide appropriate morphological and mechanical properties. This review introduces the physicochemical and mechanical properties of SF and looks into a range of SF-based scaffolds that have recently been developed. The typical applications of SF-based scaffolds for TE of bone, cartilage, teeth and mandible tissue, cartilage, skeletal muscle, and vascular tissue are highlighted and discussed followed by a discussion of issues to be addressed in future studies.
Collapse
|
19
|
Romagnoli C, Iantomasi T, Brandi ML. Available In Vitro Models for Human Satellite Cells from Skeletal Muscle. Int J Mol Sci 2021; 22:ijms222413221. [PMID: 34948017 PMCID: PMC8706222 DOI: 10.3390/ijms222413221] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle accounts for almost 40% of the total adult human body mass. This tissue is essential for structural and mechanical functions such as posture, locomotion, and breathing, and it is endowed with an extraordinary ability to adapt to physiological changes associated with growth and physical exercise, as well as tissue damage. Moreover, skeletal muscle is the most age-sensitive tissue in mammals. Due to aging, but also to several diseases, muscle wasting occurs with a loss of muscle mass and functionality, resulting from disuse atrophy and defective muscle regeneration, associated with dysfunction of satellite cells, which are the cells responsible for maintaining and repairing adult muscle. The most established cell lines commonly used to study muscle homeostasis come from rodents, but there is a need to study skeletal muscle using human models, which, due to ethical implications, consist primarily of in vitro culture, which is the only alternative way to vertebrate model organisms. This review will survey in vitro 2D/3D models of human satellite cells to assess skeletal muscle biology for pre-clinical investigations and future directions.
Collapse
Affiliation(s)
- Cecilia Romagnoli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (C.R.); (T.I.)
| | - Teresa Iantomasi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (C.R.); (T.I.)
| | - Maria Luisa Brandi
- F.I.R.M.O. Italian Foundation for the Research on Bone Diseases, Via Reginaldo Giuliani 195/A, 50141 Florence, Italy
- Correspondence:
| |
Collapse
|
20
|
Beckerman M, Harel C, Michael I, Klip A, Bilan PJ, Gallagher EJ, LeRoith D, Lewis EC, Karnieli E, Levenberg S. GLUT4-overexpressing engineered muscle constructs as a therapeutic platform to normalize glycemia in diabetic mice. SCIENCE ADVANCES 2021; 7:eabg3947. [PMID: 34644106 PMCID: PMC8514095 DOI: 10.1126/sciadv.abg3947] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 08/23/2021] [Indexed: 05/29/2023]
Abstract
Skeletal muscle insulin resistance is a main defect in type 2 diabetes (T2D), which is associated with impaired function and content of glucose transporter type 4 (GLUT4). GLUT4 overexpression in skeletal muscle tissue can improve glucose homeostasis. Therefore, we created an engineered muscle construct (EMC) composed of GLUT4-overexpressing (OEG4) cells. The ability of the engineered implants to reduce fasting glucose levels was tested in diet-induced obesity mice. Decrease and stabilization of basal glucose levels were apparent up to 4 months after implantation. Analysis of the retrieved constructs showed elevated expression of myokines and proteins related to metabolic processes. In addition, we validated the efficiency of OEG4-EMCs in insulin-resistant mice. Following high glucose load administration, mice showed improved glucose tolerance. Our data indicate that OEG4-EMC implant is an efficient mode for restoring insulin sensitivity and improving glucose homeostasis in diabetic mice. Such procedure is a potential innovative modality for T2D therapy.
Collapse
Affiliation(s)
- Margarita Beckerman
- Faculty of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
- Rina and Avner Schneur Center of Diabetes Research, Technion—Israel Institute of Technology, Haifa, Israel
| | - Chava Harel
- Rina and Avner Schneur Center of Diabetes Research, Technion—Israel Institute of Technology, Haifa, Israel
- Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Inbal Michael
- Faculty of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| | - Amira Klip
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Philip J. Bilan
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Emily J. Gallagher
- Division of Endocrinology, Diabetes and Bone Diseases, Samuel Bronfman Department of Medicine, Ichan School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Diseases, Samuel Bronfman Department of Medicine, Ichan School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Eli C. Lewis
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eddy Karnieli
- Rina and Avner Schneur Center of Diabetes Research, Technion—Israel Institute of Technology, Haifa, Israel
- Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Shulamit Levenberg
- Faculty of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
- Rina and Avner Schneur Center of Diabetes Research, Technion—Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
21
|
Du X, Xu F, Qiao H, Liu W, He X, Yan J, Qin X, Ou G. Global Evolution of Skeletal Muscle Tissue Engineering: A Scientometric Research. Tissue Eng Part C Methods 2021; 27:497-511. [PMID: 34445889 DOI: 10.1089/ten.tec.2021.0156] [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: 11/12/2022] Open
Abstract
Skeletal muscle tissue engineering (SMTE) is of great significance in the study of skeletal muscle physiology and pathology, which could be used in skeletal muscle graft. The scientometric analysis of SMTE can help researchers to quickly understand the evolutive history, status, novelties, and trend of this field. In this study, we performed a scientometric study that can be used to construct and visualize networks of SMTE using VOSviewer. A total of 1384 documents published between 1994 and 2020 were retrieved and analyzed. Our results showed that number of publications in SMTE has increased slowly from 1994 to 2014 and has increased rapidly from 2015 to 2020. The geographical distribution of publications in terms of total publications about SMTE is concentrated in Europe and the United States. The most productive institution was University of Michigan, while Harvard University and the University of Pittsburgh were ranked the second and third places. SMTE influenced a wide spectrum of disciplines, including Biology and Medicine and Physical Sciences. In addition, the research hotspot of SMTE was expanding from seed cells to the combination with advanced strategies (electrostatic spinning, bioprinting, and materials) for emulating the highly bionic engineered skeletal muscle tissues. This study provided a unique perspective for understanding the history and trends of SMTE, which could help to promote the rapid development of the field. Impact statement Skeletal muscle tissue engineering (SMTE), which acts as an important branch of tissue engineering, hold a great promise in the study of skeletal muscle physiology and pathology. The field of SMTE has developed rapidly in recent decades while still lacking studies based on scientometric methods. This article provided the first scientometric study of SMTE from development trends and evolution of the field. The results indicated that the field of SMTE was experiencing rapid growth and had a significant impact on multiple fields, particularly in Biology and Medicine and Physical Sciences.
Collapse
Affiliation(s)
- Xincheng Du
- School of Physical Education, China University of Geosciences, Wuhan, P.R. China
| | - Fang Xu
- Department of Biomedical Engineering, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China
| | - Haowen Qiao
- Department of Biomedical Engineering, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China
| | - Wenwen Liu
- Department of Stomatology, Handan Central Hospital, Handan, P.R. China
| | - Xingdao He
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, P.R. China
| | - Jiayu Yan
- School of Physical Education, China University of Geosciences, Wuhan, P.R. China
| | - Xingping Qin
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Gaozhi Ou
- School of Physical Education, China University of Geosciences, Wuhan, P.R. China
| |
Collapse
|
22
|
Gao H, Xiao J, Wei Y, Wang H, Wan H, Liu S. Regulation of Myogenic Differentiation by Topologically Microgrooved Surfaces for Skeletal Muscle Tissue Engineering. ACS OMEGA 2021; 6:20931-20940. [PMID: 34423201 PMCID: PMC8374903 DOI: 10.1021/acsomega.1c02347] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/29/2021] [Indexed: 05/05/2023]
Abstract
Inspired by the natural topological structure of skeletal muscle tissue, the topological surface construction of bionic scaffolds for skeletal muscle repair has attracted great interest. Many previous studies have focused on the effects of the topological structure on myoblasts. However, these studies used only specific repeating sizes and shapes to achieve the myoblast alignment and myotube formation; moreover, the regulatory effects of the size of a topological structure on myogenic differentiation are often neglected, leading to a lack of guidance for the design of scaffolds for skeletal muscle tissue engineering. In this study, we fabricated a series of microgroove topographies with various widths and depths via a combination of soft lithography and melt-casting and studied their effects on the behaviors of skeletal muscle cells, especially myogenic differentiation, in detail. Microgrooved poly(lactic-co-glycolic acid) substrates were found to effectively regulate the proliferation, myogenic differentiation, and myotube formation of C2C12 cells, and the degree of myogenic differentiation was significantly dependent on signals in response to the size of the microgroove structure. Compared with their depth, the width of the microgroove structures can more strongly affect the myogenic differentiation of C2C12 cells, and the degree of myoblast differentiation was enhanced with increasing groove width. Microgroove structures with relatively large groove widths and small groove depths promoted the myogenic differentiation of C2C12 cells. In addition, the integrin-mediated focal adhesion kinase signaling pathway and MAPK signaling pathway were activated in cells in response to the external topological structure, and the size of the topological structure of the material surface effectively regulated the degree of the cellular response to the external topological structure. These results can guide the design of scaffolds for skeletal muscle tissue engineering and the construction of effective bionic scaffold surfaces for skeletal muscle regeneration.
Collapse
Affiliation(s)
- Huichang Gao
- School
of Medicine, South China University of Technology, Guangzhou 510006, China
- A
National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou 510006, China
| | - Jin Xiao
- Department
of Orthopedics, Guangdong Provincial People’s Hospital Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Yingqi Wei
- The
Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China
| | - Hao Wang
- School
of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Hongxia Wan
- School
of Food Science and Health Preserving, Guangzhou
City Polytechnic, Guangzhou 510230, China
| | - Shan Liu
- School
of Medicine, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
23
|
Mohamed MA, Shahini A, Rajabian N, Caserto J, El-Sokkary AM, Akl MA, Andreadis ST, Cheng C. Fast photocurable thiol-ene elastomers with tunable biodegradability, mechanical and surface properties enhance myoblast differentiation and contractile function. Bioact Mater 2021; 6:2120-2133. [PMID: 33511311 PMCID: PMC7810627 DOI: 10.1016/j.bioactmat.2020.12.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 01/04/2023] Open
Abstract
Biodegradable elastomers are important emerging biomaterials for biomedical applications, particularly in the area of soft-tissue engineering in which scaffolds need to match the physicochemical properties of native tissues. Here, we report novel fast photocurable elastomers with readily tunable mechanical properties, surface wettability, and degradability. These elastomers are prepared by a 5-min UV-irradiation of thiol-ene reaction systems of glycerol tripentenoate (GTP; a triene) or the combination of GTP and 4-pentenyl 4-pentenoate (PP; a diene) with a carefully chosen series of di- or tri-thiols. In the subsequent application study, these elastomers were found to be capable of overcoming delamination of myotubes, a technical bottleneck limiting the in vitro growth of mature functional myofibers. The glycerol-based elastomers supported the proliferation of mouse and human myoblasts, as well as myogenic differentiation into contractile myotubes. More notably, while beating mouse myotubes detached from conventional tissue culture plates, they remain adherent on the elastomer surface. The results suggest that these elastomers as novel biomaterials may provide a promising platform for engineering functional soft tissues with potential applications in regenerative medicine or pharmacological testing.
Collapse
Affiliation(s)
- Mohamed Alaa Mohamed
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
- Chemistry Department, College of Science, Mansoura University, Mansoura, 35516, Egypt
| | - Aref Shahini
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Nika Rajabian
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Julia Caserto
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Ahmed M.A. El-Sokkary
- Chemistry Department, College of Science, Mansoura University, Mansoura, 35516, Egypt
| | - Magda A. Akl
- Chemistry Department, College of Science, Mansoura University, Mansoura, 35516, Egypt
| | - Stelios T. Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
- Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY, 14263, USA
| | - Chong Cheng
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| |
Collapse
|
24
|
Bilge S, Ergene E, Talak E, Gokyer S, Donar YO, Sınağ A, Yilgor Huri P. Recycled algae-based carbon materials as electroconductive 3D printed skeletal muscle tissue engineering scaffolds. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:73. [PMID: 34152502 PMCID: PMC8217022 DOI: 10.1007/s10856-021-06534-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 05/28/2021] [Indexed: 05/03/2023]
Abstract
Skeletal muscle is an electrically and mechanically active tissue that contains highly oriented, densely packed myofibrils. The tissue has self-regeneration capacity upon injury, which is limited in the cases of volumetric muscle loss. Several regenerative therapies have been developed in order to enhance this capacity, as well as to structurally and mechanically support the defect site during regeneration. Among them, biomimetic approaches that recapitulate the native microenvironment of the tissue in terms of parallel-aligned structure and biophysical signals were shown to be effective. In this study, we have developed 3D printed aligned and electrically active scaffolds in which the electrical conductivity was provided by carbonaceous material (CM) derived from algae-based biomass. The synthesis of this conductive and functional CM consisted of eco-friendly synthesis procedure such as pre-carbonization and multi-walled carbon nanotube (MWCNT) catalysis. CM obtained from biomass via hydrothermal carbonization (CM-03) and its ash form (CM-03K) were doped within poly(ɛ-caprolactone) (PCL) matrix and 3D printed to form scaffolds with aligned fibers for structural biomimicry. Scaffolds were seeded with C2C12 mouse myoblasts and subjected to electrical stimulation during the in vitro culture. Enhanced myotube formation was observed in electroactive groups compared to their non-conductive counterparts and it was observed that myotube formation and myotube maturity were significantly increased for CM-03 group after electrical stimulation. The results have therefore showed that the CM obtained from macroalgae biomass is a promising novel source for the production of the electrically conductive scaffolds for skeletal muscle tissue engineering.
Collapse
Affiliation(s)
- Selva Bilge
- Department of Chemistry, Ankara University Faculty of Science, Ankara, Turkey
| | - Emre Ergene
- Department of Biomedical Engineering, Ankara University Faculty of Engineering, Ankara, Turkey
- Ankara University Biotechnology Institute, Ankara, Turkey
| | - Ebru Talak
- Department of Biomedical Engineering, Ankara University Faculty of Engineering, Ankara, Turkey
| | - Seyda Gokyer
- Department of Biomedical Engineering, Ankara University Faculty of Engineering, Ankara, Turkey
| | - Yusuf Osman Donar
- Department of Chemistry, Ankara University Faculty of Science, Ankara, Turkey
| | - Ali Sınağ
- Department of Chemistry, Ankara University Faculty of Science, Ankara, Turkey.
| | - Pinar Yilgor Huri
- Department of Biomedical Engineering, Ankara University Faculty of Engineering, Ankara, Turkey.
| |
Collapse
|
25
|
Dessauge F, Schleder C, Perruchot MH, Rouger K. 3D in vitro models of skeletal muscle: myopshere, myobundle and bioprinted muscle construct. Vet Res 2021; 52:72. [PMID: 34011392 PMCID: PMC8136231 DOI: 10.1186/s13567-021-00942-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/15/2021] [Indexed: 12/25/2022] Open
Abstract
Typical two-dimensional (2D) culture models of skeletal muscle-derived cells cannot fully recapitulate the organization and function of living muscle tissues, restricting their usefulness in in-depth physiological studies. The development of functional 3D culture models offers a major opportunity to mimic the living tissues and to model muscle diseases. In this respect, this new type of in vitro model significantly increases our understanding of the involvement of the different cell types present in the formation of skeletal muscle and their interactions, as well as the modalities of response of a pathological muscle to new therapies. This second point could lead to the identification of effective treatments. Here, we report the significant progresses that have been made the last years to engineer muscle tissue-like structures, providing useful tools to investigate the behavior of resident cells. Specifically, we interest in the development of myopshere- and myobundle-based systems as well as the bioprinting constructs. The electrical/mechanical stimulation protocols and the co-culture systems developed to improve tissue maturation process and functionalities are presented. The formation of these biomimetic engineered muscle tissues represents a new platform to study skeletal muscle function and spatial organization in large number of physiological and pathological contexts.
Collapse
|
26
|
Xiong F, Cheng X, Zhang C, Klar RM, He T. Optimizations for identifying reference genes in bone and cartilage bioengineering. BMC Biotechnol 2021; 21:25. [PMID: 33731065 PMCID: PMC7972220 DOI: 10.1186/s12896-021-00685-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 03/08/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR) remains one of the best-established techniques to assess gene expression patterns. However, appropriate reference gene(s) selection remains a critical and challenging subject in which inappropriate reference gene selction can distort results leading to false interpretations. To date, mixed opinions still exist in how to choose the most optimal reference gene sets in accodrance to the Minimum Information for Publication of Quantitative Real-Time PCR Experiments (MIQE) guideline. Therefore, the purpose of this study was to investigate which schemes were the most feasible for the identification of reference genes in a bone and cartilage bioengineering experimental setting. In this study, rat bone mesenchymal stem cells (rBMSCs), skeletal muscle tissue and adipose tissue were utilized, undergoing either chondrogenic or osteogenic induction, to investigate the optimal reference gene set identification scheme that would subsequently ensure stable and accurate interpretation of gene expression in bone and cartilage bioengineering. RESULTS The stability and pairwise variance of eight candidate reference genes were analyzed using geNorm. The V0.15- vs. Vmin-based normalization scheme in rBMSCs had no significant effect on the eventual normalization of target genes. In terms of the muscle tissue, the results of the correlation of NF values between the V0.15 and Vmin schemes and the variance of target genes expression levels generated by these two schemes showed that different schemes do indeed have a significant effect on the eventual normalization of target genes. Three selection schemes were adopted in terms of the adipose tissue, including the three optimal reference genes (Opt3), V0.20 and Vmin schemes, and the analysis of NF values with eventual normalization of target genes showed that the different selection schemes also have a significant effect on the eventual normalization of target genes. CONCLUSIONS Based on these results, the proposed cut-off value of Vn/n + 1 under 0.15, according to the geNorm algorithm, should be considered with caution. For cell only experiments, at least rBMSCs, a Vn/n + 1 under 0.15 is sufficient in RT-qPCR studies. However, when using certain tissue types such as skeletal muscle and adipose tissue the minimum Vn/n + 1 should be used instead as this provides a far superior mode of generating accurate gene expression results. We thus recommended that when the stability and variation of a candidate reference genes in a specific study is unclear the minimum Vn/n + 1 should always be used as this ensures the best and most accurate gene expression value is achieved during RT-qPCR assays.
Collapse
Affiliation(s)
- Fei Xiong
- Department of Sports Medicine, Wuxi 9th People's Hospital affiliated to Soochow University, Wuxi, Jiangsu, China
| | - Xiangyun Cheng
- Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Chao Zhang
- Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Roland Manfred Klar
- Department of Orthopedics, Physical Medicine and Rehabilitation, University Hospital of Munich (LMU), Munich, Germany.
| | - Tao He
- Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
27
|
Zhang L, Zhang W, Hu Y, Fei Y, Liu H, Huang Z, Wang C, Ruan D, Heng BC, Chen W, Shen W. Systematic Review of Silk Scaffolds in Musculoskeletal Tissue Engineering Applications in the Recent Decade. ACS Biomater Sci Eng 2021; 7:817-840. [PMID: 33595274 DOI: 10.1021/acsbiomaterials.0c01716] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
During the past decade, various novel tissue engineering (TE) strategies have been developed to maintain, repair, and restore the biomechanical functions of the musculoskeletal system. Silk fibroins are natural polymers with numerous advantageous properties such as good biocompatibility, high mechanical strength, and low degradation rate and are increasingly being recognized as a scaffolding material of choice in musculoskeletal TE applications. This current systematic review examines and summarizes the latest research on silk scaffolds in musculoskeletal TE applications within the past decade. Scientific databases searched include PubMed, Web of Science, Medline, Cochrane library, and Embase. The following keywords and search terms were used: musculoskeletal, tendon, ligament, intervertebral disc, muscle, cartilage, bone, silk, and tissue engineering. Our Review was limited to articles on musculoskeletal TE, which were published in English from 2010 to September 2019. The eligibility of the articles was assessed by two reviewers according to prespecified inclusion and exclusion criteria, after which an independent reviewer performed data extraction and a second independent reviewer validated the data obtained. A total of 1120 articles were reviewed from the databases. According to inclusion and exclusion criteria, 480 articles were considered as relevant for the purpose of this systematic review. Tissue engineering is an effective modality for repairing or replacing injured or damaged tissues and organs with artificial materials. This Review is intended to reveal the research status of silk-based scaffolds in the musculoskeletal system within the recent decade. In addition, a comprehensive translational research route for silk biomaterial from bench to bedside is described in this Review.
Collapse
Affiliation(s)
- Li Zhang
- Department of Orthopedic Surgery of The Second Affiliated Hospital and Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.,Department of Orthopaedics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wei Zhang
- School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China.,Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, Jiangsu 210096, China
| | - Yejun Hu
- Department of Orthopedic Surgery of The Second Affiliated Hospital and Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.,Orthopaedics Research Institute, Zhejiang Univerisity, Hangzhou, Zhejiang 310000, China
| | - Yang Fei
- Department of Orthopedic Surgery of The Second Affiliated Hospital and Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.,Orthopaedics Research Institute, Zhejiang Univerisity, Hangzhou, Zhejiang 310000, China
| | - Haoyang Liu
- School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China.,Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, Jiangsu 210096, China
| | - Zizhan Huang
- Department of Orthopedic Surgery of The Second Affiliated Hospital and Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.,Orthopaedics Research Institute, Zhejiang Univerisity, Hangzhou, Zhejiang 310000, China
| | - Canlong Wang
- Department of Orthopedic Surgery of The Second Affiliated Hospital and Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.,Orthopaedics Research Institute, Zhejiang Univerisity, Hangzhou, Zhejiang 310000, China
| | - Dengfeng Ruan
- Department of Orthopedic Surgery of The Second Affiliated Hospital and Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.,Orthopaedics Research Institute, Zhejiang Univerisity, Hangzhou, Zhejiang 310000, China
| | | | - Weishan Chen
- Department of Orthopedic Surgery of The Second Affiliated Hospital and Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.,Orthopaedics Research Institute, Zhejiang Univerisity, Hangzhou, Zhejiang 310000, China
| | - Weiliang Shen
- Department of Orthopedic Surgery of The Second Affiliated Hospital and Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.,Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Sports System Disease Research and Accurate Diagnosis and Treatment of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Orthopaedics Research Institute, Zhejiang Univerisity, Hangzhou, Zhejiang 310000, China.,China Orthopaedic Regenerative Medicine (CORMed), Chinese Medical Association, Hangzhou, Zhejiang, China
| |
Collapse
|
28
|
Abstract
Tissue engineering refers to the attempt to create functional human tissue from cells in a laboratory. This is a field that uses living cells, biocompatible materials, suitable biochemical and physical factors, and their combinations to create tissue-like structures. To date, no tissue engineered skeletal muscle implants have been developed for clinical use, but they may represent a valid alternative for the treatment of volumetric muscle loss in the near future. Herein, we reviewed the literature and showed different techniques to produce synthetic tissues with the same architectural, structural and functional properties as native tissues.
Collapse
|
29
|
Boularaoui S, Al Hussein G, Khan KA, Christoforou N, Stefanini C. An overview of extrusion-based bioprinting with a focus on induced shear stress and its effect on cell viability. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.bprint.2020.e00093] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
30
|
Silk fibroin as a natural polymeric based bio-material for tissue engineering and drug delivery systems-A review. Int J Biol Macromol 2020; 163:2145-2161. [DOI: 10.1016/j.ijbiomac.2020.09.057] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/06/2020] [Accepted: 09/09/2020] [Indexed: 12/13/2022]
|
31
|
Zidarič T, Milojević M, Vajda J, Vihar B, Maver U. Cultured Meat: Meat Industry Hand in Hand with Biomedical Production Methods. FOOD ENGINEERING REVIEWS 2020. [DOI: 10.1007/s12393-020-09253-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
32
|
Zhao S, Mehta AS, Zhao M. Biomedical applications of electrical stimulation. Cell Mol Life Sci 2020; 77:2681-2699. [PMID: 31974658 PMCID: PMC7954539 DOI: 10.1007/s00018-019-03446-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/12/2019] [Accepted: 12/27/2019] [Indexed: 12/14/2022]
Abstract
This review provides a comprehensive overview on the biomedical applications of electrical stimulation (EStim). EStim has a wide range of direct effects on both biomolecules and cells. These effects have been exploited to facilitate proliferation and functional development of engineered tissue constructs for regenerative medicine applications. They have also been tested or used in clinics for pain mitigation, muscle rehabilitation, the treatment of motor/consciousness disorders, wound healing, and drug delivery. However, the research on fundamental mechanism of cellular response to EStim has fell behind its applications, which has hindered the full exploitation of the clinical potential of EStim. Moreover, despite the positive outcome from the in vitro and animal studies testing the efficacy of EStim, existing clinical trials failed to establish strong, conclusive supports for the therapeutic efficacy of EStim for most of the clinical applications mentioned above. Two potential directions of future research to improve the clinical utility of EStim are presented, including the optimization and standardization of the stimulation protocol and the development of more tissue-matching devices.
Collapse
Affiliation(s)
- Siwei Zhao
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, 985965 Nebraska Medical Center, Omaha, NE, 68198, USA.
- Department of Surgery, University of Nebraska Medical Center, Nebraska Medical Center 985965, Omaha, NE, 68198, USA.
| | - Abijeet Singh Mehta
- Department of Dermatology, University of California, Davis, CA, USA
- Department of Ophthalmology & Vision Science, Institute for Regenerative Cures, Center for Neuroscience, University of California at Davis, School of Medicine, Suite 1630, Room 1617, 2921 Stockton Blvd., Sacramento, CA, 95817, USA
| | - Min Zhao
- Department of Dermatology, University of California, Davis, CA, USA
- Department of Ophthalmology & Vision Science, Institute for Regenerative Cures, Center for Neuroscience, University of California at Davis, School of Medicine, Suite 1630, Room 1617, 2921 Stockton Blvd., Sacramento, CA, 95817, USA
| |
Collapse
|
33
|
Mostafavi E, Medina-Cruz D, Kalantari K, Taymoori A, Soltantabar P, Webster TJ. Electroconductive Nanobiomaterials for Tissue Engineering and Regenerative Medicine. Bioelectricity 2020; 2:120-149. [PMID: 34471843 PMCID: PMC8370325 DOI: 10.1089/bioe.2020.0021] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Regenerative medicine aims to engineer tissue constructs that can recapitulate the functional and structural properties of native organs. Most novel regenerative therapies are based on the recreation of a three-dimensional environment that can provide essential guidance for cell organization, survival, and function, which leads to adequate tissue growth. The primary motivation in the use of conductive nanomaterials in tissue engineering has been to develop biomimetic scaffolds to recapitulate the electrical properties of the natural extracellular matrix, something often overlooked in numerous tissue engineering materials to date. In this review article, we focus on the use of electroconductive nanobiomaterials for different biomedical applications, particularly, very recent advancements for cardiovascular, neural, bone, and muscle tissue regeneration. Moreover, this review highlights how electroconductive nanobiomaterials can facilitate cell to cell crosstalk (i.e., for cell growth, migration, proliferation, and differentiation) in different tissues. Thoughts on what the field needs for future growth are also provided.
Collapse
Affiliation(s)
- Ebrahim Mostafavi
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - David Medina-Cruz
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Katayoon Kalantari
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Ada Taymoori
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Pooneh Soltantabar
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas, USA
| | - Thomas J. Webster
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
34
|
Orellana N, Sánchez E, Benavente D, Prieto P, Enrione J, Acevedo CA. A New Edible Film to Produce In Vitro Meat. Foods 2020; 9:foods9020185. [PMID: 32069986 PMCID: PMC7073543 DOI: 10.3390/foods9020185] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 02/06/2023] Open
Abstract
In vitro meat is a novel concept of food science and biotechnology. Methods to produce in vitro meat employ muscle cells cultivated on a scaffold in a serum-free medium using a bioreactor. The microstructure of the scaffold is a key factor, because muscle cells must be oriented to generate parallel alignments of fibers. This work aimed to develop a new scaffold (microstructured film) to grow muscle fibers. The microstructured edible films were made using micromolding technology. A micromold was tailor-made using a laser cutting machine to obtain parallel fibers with a diameter in the range of 70-90 µm. Edible films were made by means of solvent casting using non-mammalian biopolymers. Myoblasts were cultured on flat and microstructured films at three cell densities. Cells on the microstructured films grew with a muscle fiber morphology, but in the case of using the flat film, they only produced unorganized cell proliferation. Myogenic markers were assessed using quantitative polymerase chain reaction. After 14 days, the expression of desmin, myogenin, and myosin heavy chain were significantly higher in microstructured films compared to the flat films. The formation of fiber morphology and the high expression of myogenic markers indicated that a microstructured edible film can be used for the production of in vitro meat.
Collapse
Affiliation(s)
- Nicole Orellana
- Centro de Biotecnología, Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile; (N.O.); (E.S.)
| | - Elizabeth Sánchez
- Centro de Biotecnología, Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile; (N.O.); (E.S.)
| | - Diego Benavente
- Departamento de Ingeniería en Diseño, Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile; (D.B.); (P.P.)
| | - Pablo Prieto
- Departamento de Ingeniería en Diseño, Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile; (D.B.); (P.P.)
| | - Javier Enrione
- Biopolymer Research and Engineering Lab, Facultad de Medicina, Universidad de Los Andes, Monseñor Álvaro del Portillo 12455, Las Condes, Santiago 7550000, Chile;
| | - Cristian A. Acevedo
- Centro de Biotecnología, Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile; (N.O.); (E.S.)
- Departamento de Física, Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile
- Correspondence:
| |
Collapse
|
35
|
|
36
|
Ribeiro S, Puckert C, Ribeiro C, Gomes AC, Higgins MJ, Lanceros-Méndez S. Surface Charge-Mediated Cell-Surface Interaction on Piezoelectric Materials. ACS APPLIED MATERIALS & INTERFACES 2020; 12:191-199. [PMID: 31825193 DOI: 10.1021/acsami.9b17222] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Cell-material interactions play an essential role in the development of scaffold-based tissue engineering strategies. Cell therapies are still limited in treating injuries when severe damage causes irreversible loss of muscle cells. Electroactive biomaterials and, in particular, piezoelectric materials offer new opportunities for skeletal muscle tissue engineering since these materials have demonstrated suitable electroactive microenvironments for tissue development. In this study, the influence of the surface charge of piezoelectric poly(vinylidene fluoride) (PVDF) on cell adhesion was investigated. The cytoskeletal organization of C2C12 myoblast cells grown on different PVDF samples was studied by immunofluorescence staining, and the interactions between single live cells and PVDF were analyzed using an atomic force microscopy (AFM) technique termed single-cell force spectroscopy. It was demonstrated that C2C12 myoblast cells seeded on samples with net surface charge present a more elongated morphology, this effect being dependent on the surface charge but independent of the poling direction (negative or positive surface charge). It was further shown that the cell deadhesion forces of individual C2C12 cells were higher on PVDF samples with an overall negative surface charge (8.92 ± 0.45 nN) compared to those on nonpoled substrates (zero overall surface charge) (4.06 ± 0.20 nN). These findings explicitly demonstrate that the polarization/surface charge is an important parameter to determine cell fate as it affects C2C12 cell adhesion, which in turn will influence cell behavior, namely, cell proliferation and differentiation.
Collapse
Affiliation(s)
- Sylvie Ribeiro
- Centro/Departamento de Física , Universidade do Minho , 4710-057 Braga , Portugal
| | - Christina Puckert
- ARC Centre for Electromaterials Science (ACES), Innovation Campus , University of Wollongong , Squires Way , Wollongong , NSW 2500 , Australia
| | - Clarisse Ribeiro
- Centro/Departamento de Física , Universidade do Minho , 4710-057 Braga , Portugal
| | | | - Michael J Higgins
- ARC Centre for Electromaterials Science (ACES), Innovation Campus , University of Wollongong , Squires Way , Wollongong , NSW 2500 , Australia
| | - Senentxu Lanceros-Méndez
- BCMaterials, Basque Centre for Materials, Applications and Nanostructures , UPV/EHU Science Park , 48940 Leioa , Spain
- IKERBASQUE , Basque Foundation for Science , 48013 Bilbao , Spain
| |
Collapse
|
37
|
Strateva M, Penchev G. HISTOLOGICAL, PHYSICOCHEMICAL AND MICROBIOLOGICAL CHANGES IN FRESH AND FROZEN/THAWED FISH. BULGARIAN JOURNAL OF VETERINARY MEDICINE 2020. [DOI: 10.15547/tjs.2020.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The production and supply of fish as food is constantly growing worldwide. Various methods are applied to extend its shelf life, one of them being freezing. According to European Union legislation, the state of the food and its treatment must be indicated on the label. If the food had been frozen prior to marketing and then sold thawed, this information must be provided to the consumer by labelling it. Otherwise, this is considered a fraud to the consumer since freezing significantly degrades the quality of fish. Histological, physicochemical and microbiological changes in the muscle tissue of frozen fish occur. Different methods may be applied to distinguish between fresh and frozen and them thawed fish, of which histological examination is a reliable method.
Collapse
|
38
|
Hong J, Yeo M, Yang GH, Kim G. Cell-Electrospinning and Its Application for Tissue Engineering. Int J Mol Sci 2019; 20:E6208. [PMID: 31835356 PMCID: PMC6940787 DOI: 10.3390/ijms20246208] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/05/2019] [Accepted: 12/05/2019] [Indexed: 12/27/2022] Open
Abstract
Electrospinning has gained great interest in the field of regenerative medicine, due to its fabrication of a native extracellular matrix-mimicking environment. The micro/nanofibers generated through this process provide cell-friendly surroundings which promote cellular activities. Despite these benefits of electrospinning, a process was introduced to overcome the limitations of electrospinning. Cell-electrospinning is based on the basic process of electrospinning for producing viable cells encapsulated in the micro/nanofibers. In this review, the process of cell-electrospinning and the materials used in this process will be discussed. This review will also discuss the applications of cell-electrospun structures in tissue engineering. Finally, the advantages, limitations, and future perspectives will be discussed.
Collapse
Affiliation(s)
| | | | | | - GeunHyung Kim
- Department of Biomechatronic Engineering, Sungkyunkwan University (SKKU), Suwon-si, Gyeonggi-do 16419, Korea; (J.H.); (M.Y.); (G.H.Y.)
| |
Collapse
|
39
|
Enhanced Host Neovascularization of Prevascularized Engineered Muscle Following Transplantation into Immunocompetent versus Immunocompromised Mice. Cells 2019; 8:cells8121472. [PMID: 31757007 PMCID: PMC6953003 DOI: 10.3390/cells8121472] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 11/10/2019] [Accepted: 11/18/2019] [Indexed: 01/03/2023] Open
Abstract
: Engineering of functional tissue, by combining either autologous or allogeneic cells with biomaterials, holds promise for the treatment of various diseases and injuries. Prevascularization of the engineered tissue was shown to enhance and improve graft integration and neovascularization post-implantation in immunocompromised mice. However, the neovascularization and integration processes of transplanted engineered tissues have not been widely studied in immunocompetent models. Here, we fabricated a three-dimensional (3D) vascularized murine muscle construct that was transplanted into immunocompetent and immunocompromised mice. Intravital imaging demonstrated enhanced neovascularization in immunocompetent mice compared to immunocompromised mice, 18 days post-implantation, indicating the advantageous effect of an intact immune system on neovascularization. Moreover, construct prevascularization enhanced neovascularization, integration, and myogenesis in both animal models. These findings demonstrate the superiority of implantation into immunocompetent over immunocompromised mice and, therefore, suggest that using autologous cells might be beneficial compared to allogeneic cells and subsequent immunosuppression. Taken together, these observations have the potential to advance the field of regenerative medicine and tissue engineering, ultimately reducing the need for donor organs and tissues.
Collapse
|
40
|
Saberi A, Jabbari F, Zarrintaj P, Saeb MR, Mozafari M. Electrically Conductive Materials: Opportunities and Challenges in Tissue Engineering. Biomolecules 2019; 9:E448. [PMID: 31487913 PMCID: PMC6770812 DOI: 10.3390/biom9090448] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/26/2019] [Accepted: 08/28/2019] [Indexed: 01/09/2023] Open
Abstract
Tissue engineering endeavors to regenerate tissues and organs through appropriate cellular and molecular interactions at biological interfaces. To this aim, bio-mimicking scaffolds have been designed and practiced to regenerate and repair dysfunctional tissues by modifying cellular activity. Cellular activity and intracellular signaling are performances given to a tissue as a result of the function of elaborated electrically conductive materials. In some cases, conductive materials have exhibited antibacterial properties; moreover, such materials can be utilized for on-demand drug release. Various types of materials ranging from polymers to ceramics and metals have been utilized as parts of conductive tissue engineering scaffolds, having conductivity assortments from a range of semi-conductive to conductive. The cellular and molecular activity can also be affected by the microstructure; therefore, the fabrication methods should be evaluated along with an appropriate selection of conductive materials. This review aims to address the research progress toward the use of electrically conductive materials for the modulation of cellular response at the material-tissue interface for tissue engineering applications.
Collapse
Affiliation(s)
- Azadeh Saberi
- Nanotechnology and Advanced Materials Department, Materials and Energy Research Center (MERC), P.O. Box: 31787-316 Tehran, Iran.
| | - Farzaneh Jabbari
- Nanotechnology and Advanced Materials Department, Materials and Energy Research Center (MERC), P.O. Box: 31787-316 Tehran, Iran.
| | - Payam Zarrintaj
- Polymer Engineering Department, Faculty of Engineering, Urmia University, P.O. Box: 5756151818-165 Urmia, Iran.
| | - Mohammad Reza Saeb
- Department of Resin and Additives, Institute for Color Science and Technology, P.O. Box: 16765-654 Tehran, Iran.
| | - Masoud Mozafari
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), P.O Box: 14665-354 Tehran, Iran.
| |
Collapse
|
41
|
Kankala RK, Zhao J, Liu CG, Song XJ, Yang DY, Zhu K, Wang SB, Zhang YS, Chen AZ. Highly Porous Microcarriers for Minimally Invasive In Situ Skeletal Muscle Cell Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1901397. [PMID: 31066236 PMCID: PMC6750270 DOI: 10.1002/smll.201901397] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 04/12/2019] [Indexed: 05/19/2023]
Abstract
Microscale cell carriers have recently garnered enormous interest in repairing tissue defects by avoiding substantial open surgeries using implants for tissue regeneration. In this study, the highly open porous microspheres (HOPMs) are fabricated using a microfluidic technique for harboring proliferating skeletal myoblasts and evaluating their feasibility toward cell delivery application in situ. These biocompatible HOPMs with particle sizes of 280-370 µm possess open pores of 10-80 µm and interconnected paths. Such structure of the HOPMs conveniently provide a favorable microenvironment, where the cells are closely arranged in elongated shapes with the deposited extracellular matrix, facilitating cell adhesion and proliferation, as well as augmented myogenic differentiation. Furthermore, in vivo results in mice confirm improved cell retention and vascularization, as well as partial myoblast differentiation. These modular cell-laden microcarriers potentially allow for in situ tissue construction after minimally invasive delivery providing a convenient means for regeneration medicine.
Collapse
Affiliation(s)
- Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, P. R. China
| | - Jia Zhao
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, P. R. China
| | - Chen-Guang Liu
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, P. R. China
| | - Xiao-Jie Song
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, P. R. China
| | - Da-Yun Yang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, P. R. China
| | - Kai Zhu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Shi-Bin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, P. R. China
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, P. R. China
| |
Collapse
|
42
|
Research Progress on Conducting Polymer-Based Biomedical Applications. APPLIED SCIENCES-BASEL 2019. [DOI: 10.3390/app9061070] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Conducting polymers (CPs) have attracted significant attention in a variety of research fields, particularly in biomedical engineering, because of the ease in controlling their morphology, their high chemical and environmental stability, and their biocompatibility, as well as their unique optical and electrical properties. In particular, the electrical properties of CPs can be simply tuned over the full range from insulator to metal via a doping process, such as chemical, electrochemical, charge injection, and photo-doping. Over the past few decades, remarkable progress has been made in biomedical research including biosensors, tissue engineering, artificial muscles, and drug delivery, as CPs have been utilized as a key component in these fields. In this article, we review CPs from the perspective of biomedical engineering. Specifically, representative biomedical applications of CPs are briefly summarized: biosensors, tissue engineering, artificial muscles, and drug delivery. The motivation for use of and the main function of CPs in these fields above are discussed. Finally, we highlight the technical and scientific challenges regarding electrical conductivity, biodegradability, hydrophilicity, and the loading capacity of biomolecules that are faced by CPs for future work. This is followed by several strategies to overcome these drawbacks.
Collapse
|
43
|
Huang T, Xiao J, Wang S, Liao Z, Huang T, Gu R, Li J, Wu G, Liao H. The thickness of poly-phenoxyethyl methacrylate brush interferes with cellular behavior and function of myofibers. J Biomed Mater Res A 2019; 107:1264-1272. [PMID: 30724032 DOI: 10.1002/jbm.a.36636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 12/19/2018] [Accepted: 12/30/2018] [Indexed: 11/07/2022]
Abstract
Introducing or grafting molecules onto biomaterial surfaces to regulate muscle cell destination via biophysical cues is one of the important steps for biomaterial design in muscle tissue engineering. Therefore, it is important to understand the interaction between myoblasts and myofibers with substrates modified by biomimetic layer with different thicknesses. In this study, we used a surface-induced atom transfer radical polymerization method to synthetize and graft poly-phenoxyethyl methacrylate (PHEMA) brushes having different lengths on the glass substrates. C2C12 myoblasts were seeded on the PHEMA brushes and differentiated using horse serum, for analyzing the sensibility of muscle cells to feel environment changing, and further investigating whether the depths of grafting layer on the biomaterial surface are important factors in regulating muscle cell behaviors. Our results demonstrated that on the thicker PHEMA brushes surface (200 and 450 nm), C2C12 myoblasts showed a better survival and proliferation and were favorable for cell fusion and myotube formation. Furthermore, myofibers survived on the thicker brushes were more functional and upregulated cytoskeleton proteins (tubulin, vimentin, and vinculin) and FAK levels, and enhanced the expression levels for mechanical stress molecules (HGF, NOS-1, and c-Met). These results suggest that grafting thickness of PHEMA layer on the substrate led to the myoblasts/myofiber behavior change, which would be valuable for the design and preparation of the modified layer on muscle tissue engineering scaffolds. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1264-1272, 2019.
Collapse
Affiliation(s)
- Tao Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China
| | - Jiangwei Xiao
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shuhao Wang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
| | - Zhaohong Liao
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Tao Huang
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ruicai Gu
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Junhua Li
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Gang Wu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
| | - Hua Liao
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
44
|
Chen H, Zhong J, Wang J, Huang R, Qiao X, Wang H, Tan Z. Enhanced growth and differentiation of myoblast cells grown on E-jet 3D printed platforms. Int J Nanomedicine 2019; 14:937-950. [PMID: 30787608 PMCID: PMC6366362 DOI: 10.2147/ijn.s193624] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Skeletal muscle tissue engineering often involves the prefabrication of muscle tissues in vitro by differentiation and maturation of muscle precursor cells on a platform which provides an environment that facilitates the myogenic differentiation of the seeded cells. METHODS Poly lactic-co-glycolic acid (PLGA) 3D printed scaffolds, which simulate the highly complex structure of extracellular matrix (ECM), were fabricated by E-jet 3D printing in this study. The scaffolds were used as platforms, providing environment that aids in growth, differentiation and other properties of C2C12 myoblast cells. RESULTS The C2C12 myoblast cells grown on the PLGA 3D printed platforms had enhanced cell adhesion and proliferation. Moreover, the platforms were able to induce myogenic differentiation of the myoblast cells by promoting the formation of myotubes and up-regulating the expressions of myogenic genes (MyHC and MyOG). CONCLUSION The fabricated 3D printed platforms have excellent biocompatibility, thereby can potentially be used as functional cell culture platforms in skeletal tissue engineering and regeneration.
Collapse
Affiliation(s)
- Haoxiang Chen
- College of Biology, Hunan University, Changsha, Hunan 410082, China,
| | - Juchang Zhong
- College of Biology, Hunan University, Changsha, Hunan 410082, China,
| | - Jian Wang
- College of Biology, Hunan University, Changsha, Hunan 410082, China,
| | - Ruiying Huang
- College of Biology, Hunan University, Changsha, Hunan 410082, China,
| | - Xiaoyin Qiao
- College of Biology, Hunan University, Changsha, Hunan 410082, China,
| | - Honghui Wang
- College of Biology, Hunan University, Changsha, Hunan 410082, China,
| | - Zhikai Tan
- College of Biology, Hunan University, Changsha, Hunan 410082, China,
| |
Collapse
|
45
|
Kadavil H, Zagho M, Elzatahry A, Altahtamouni T. Sputtering of Electrospun Polymer-Based Nanofibers for Biomedical Applications: A Perspective. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E77. [PMID: 30626067 PMCID: PMC6359597 DOI: 10.3390/nano9010077] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/08/2018] [Accepted: 11/13/2018] [Indexed: 12/22/2022]
Abstract
Electrospinning has gained wide attention recently in biomedical applications. Electrospun biocompatible scaffolds are well-known for biomedical applications such as drug delivery, wound dressing, and tissue engineering applications. In this review, the synthesis of polymer-based fiber composites using an electrospinning technique is discussed. Formerly, metal particles were then deposited on the surface of electrospun fibers using sputtering technology. Key nanometals for biomedical applications including silver and copper nanoparticles are discussed throughout this review. The formulated scaffolds were found to be suitable candidates for biomedical uses such as antibacterial coatings, surface modification for improving biocompatibility, and tissue engineering. This review briefly mentions the characteristics of the nanostructures while focusing on how nanostructures hold potential for a wide range of biomedical applications.
Collapse
Affiliation(s)
- Hana Kadavil
- Materials Science and Technology Program, College of Arts and Sciences, Qatar University, P.O. Box 2713, Doha, Qatar.
| | - Moustafa Zagho
- Materials Science and Technology Program, College of Arts and Sciences, Qatar University, P.O. Box 2713, Doha, Qatar.
| | - Ahmed Elzatahry
- Materials Science and Technology Program, College of Arts and Sciences, Qatar University, P.O. Box 2713, Doha, Qatar.
| | - Talal Altahtamouni
- Materials Science and Technology Program, College of Arts and Sciences, Qatar University, P.O. Box 2713, Doha, Qatar.
| |
Collapse
|
46
|
Kaufman T, Kaplan B, Perry L, Shandalov Y, Landau S, Srugo I, Ad-El D, Levenberg S. Innervation of an engineered muscle graft for reconstruction of muscle defects. Am J Transplant 2019; 19:37-47. [PMID: 29856531 DOI: 10.1111/ajt.14957] [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] [Received: 12/05/2017] [Revised: 05/24/2018] [Accepted: 05/27/2018] [Indexed: 01/25/2023]
Abstract
Autologous muscle flaps are commonly used to reconstruct defects that involve muscle impairment. To maintain viability and functionality of these flaps, they must be properly vascularized and innervated. Tissue-engineered muscles could potentially replace autologous muscle tissue, but still require establishment of sufficient innervation to ensure functionality. In this study, we explored the possibility of innervating engineered muscle grafts transplanted to an abdominal wall defect in mice, by transferring the native femoral nerve to the graft. Six weeks posttransplantation, nerve conduction studies and electromyography demonstrated increased innervation in engineered grafts neurotized with the femoral nerve, as compared to non-neurotized grafts. Histologic assessments revealed axonal penetration and formation of neuromuscular junctions within the grafts. The innervation process described here may advance the fabrication of a fully functional engineered muscle graft that will be of utility in clinical settings.
Collapse
Affiliation(s)
- Tal Kaufman
- Department of Plastic Surgery, Rabin Medical Center, Petach Tikva, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ben Kaplan
- Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel.,Bruce Rapaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Luba Perry
- Bruce Rapaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.,Inter-departmental Program in Biotechnology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Yulia Shandalov
- Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Shira Landau
- Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | | | - Dean Ad-El
- Department of Plastic Surgery, Rabin Medical Center, Petach Tikva, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shulamit Levenberg
- Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
47
|
Zhang Z, Klausen LH, Chen M, Dong M. Electroactive Scaffolds for Neurogenesis and Myogenesis: Graphene-Based Nanomaterials. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1801983. [PMID: 30264534 DOI: 10.1002/smll.201801983] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 07/28/2018] [Indexed: 05/24/2023]
Abstract
One of the major issues in tissue engineering is constructing a functional scaffold to support cell growth and also provide proper synergistic guidance cues. Graphene-based nanomaterials have emerged as biocompatible and electroactive scaffolds for neurogenesis and myogenesis, due to their excellent tunable chemical, physical, and mechanical properties. This review first assesses the recent investigations focusing on the fabrication and applications of graphene-based nanomaterials for neurogenesis and myogenesis, in the form of either 2D films, 3D scaffolds, or composite architectures. Besides, because of their outstanding electrical properties, graphene family materials are particularly suitable for designing electroactive scaffolds that could provide proper electrical stimulation (i.e., electrical or photo stimuli) to promote the regeneration of excitable neurons and muscle cells. Therefore, the effects and mechanism of electrical and/or photo stimulations on neurogenesis and myogenesis are followed. Furthermore, studies on their biocompatibilities and toxicities especially to neural and muscle cells are evaluated. Finally, the future challenges and perspectives in facilitating the development of clinical translation of graphene-family nanomaterials in treating neurodegenerative and muscle diseases are discussed.
Collapse
Affiliation(s)
- Zhongyang Zhang
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, DK-8000, Aarhus C, Denmark
| | | | - Menglin Chen
- Department of Engineering, Aarhus University, DK-8000, Aarhus C, Denmark
| | - Mingdong Dong
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, DK-8000, Aarhus C, Denmark
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
48
|
Roveimiab Z, Lin F, Anderson JE. Emerging Development of Microfluidics-Based Approaches to Improve Studies of Muscle Cell Migration. TISSUE ENGINEERING PART B-REVIEWS 2018; 25:30-45. [PMID: 30073911 DOI: 10.1089/ten.teb.2018.0181] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
IMPACT STATEMENT The essential interactions between and among cells in the three types of muscle tissue in development, wound healing, and regeneration of tissues, are underpinned by the ability of cardiac, smooth, and skeletal muscle cells to migrate in maintaining functional capacity after pathologies such as myocardial infarction, tissue grafting, and traumatic and postsurgical injury. Microfluidics-based devices now offer significant enhancement over conventional approaches to studying cell chemotaxis and haptotaxis that are inherent in migration. Advances in experimental approaches to muscle cell movement and tissue formation will contribute to innovations in tissue engineering for patching wound repair and muscle tissue replacement.
Collapse
Affiliation(s)
- Ziba Roveimiab
- 1 Department of Biological Sciences and University of Manitoba, Winnipeg, Canada.,2 Department of Physics and Astronomy, University of Manitoba, Winnipeg, Canada
| | - Francis Lin
- 1 Department of Biological Sciences and University of Manitoba, Winnipeg, Canada.,2 Department of Physics and Astronomy, University of Manitoba, Winnipeg, Canada
| | - Judy E Anderson
- 1 Department of Biological Sciences and University of Manitoba, Winnipeg, Canada
| |
Collapse
|
49
|
Ether-Oxygen Containing Electrospun Microfibrous and Sub-Microfibrous Scaffolds Based on Poly(butylene 1,4-cyclohexanedicarboxylate) for Skeletal Muscle Tissue Engineering. Int J Mol Sci 2018; 19:ijms19103212. [PMID: 30336625 PMCID: PMC6214009 DOI: 10.3390/ijms19103212] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/10/2018] [Accepted: 10/11/2018] [Indexed: 01/29/2023] Open
Abstract
We report the study of novel biodegradable electrospun scaffolds from poly(butylene 1,4-cyclohexandicarboxylate-co-triethylene cyclohexanedicarboxylate) (P(BCE-co-TECE)) as support for in vitro and in vivo muscle tissue regeneration. We demonstrate that chemical composition, i.e., the amount of TECE co-units (constituted of polyethylene glycol-like moieties), and fibre morphology, i.e., aligned microfibrous or sub-microfibrous scaffolds, are crucial in determining the material biocompatibility. Indeed, the presence of ether linkages influences surface wettability, mechanical properties, hydrolytic degradation rate, and density of cell anchoring points of the studied materials. On the other hand, electrospun scaffolds improve cell adhesion, proliferation, and differentiation by favouring cell alignment along fibre direction (fibre morphology), also allowing for better cell infiltration and oxygen and nutrient diffusion (fibre size). Overall, C2C12 myogenic cells highly differentiated into mature myotubes when cultured on microfibres realised with the copolymer richest in TECE co-units (micro-P73 mat). Lastly, when transplanted in the tibialis anterior muscles of healthy, injured, or dystrophic mice, micro-P73 mat appeared highly vascularised, colonised by murine cells and perfectly integrated with host muscles, thus confirming the suitability of P(BCE-co-TECE) scaffolds as substrates for skeletal muscle tissue engineering.
Collapse
|
50
|
Genetically engineered human muscle transplant enhances murine host neovascularization and myogenesis. Commun Biol 2018; 1:161. [PMID: 30320229 PMCID: PMC6172230 DOI: 10.1038/s42003-018-0161-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 08/24/2018] [Indexed: 11/30/2022] Open
Abstract
Engineered tissues are a promising tool for addressing the growing need for tissues and organs in surgical reconstructions. Prevascularization of implanted tissues is expected to enhance survival prospects post transplantation and minimize deficiencies and/or hypoxia deeper in the tissue. Here, we fabricate a three-dimensional, prevascularized engineered muscle containing human myoblasts, genetically modified endothelial cells secreting angiopoietin 1 (ANGPT1) and genetically modified smooth muscle cells secreting vascular endothelial growth factor (VEGF). The genetically engineered human muscle shows enhanced host neovascularization and myogenesis following transplantation into a mouse host, compared to the non-secreting control. The vascular, genetically modified cells have been cleared for clinical trials and can be used to construct autologous vascularized tissues. Therefore, the described genetically engineered vascularized muscle has the potential to be fully translated to the clinical setting to overcome autologous tissue shortage and to accelerate host neovascularization and integration of engineered grafts following transplantation. Luba Perry et al. report transplantation of engineered prevascularized human muscle into mice to repair an abdominal muscle defect. They show that genetically engineering smooth muscle cells to secrete VEGF and endothelial cells to secrete ANGPT1 significantly improves host neovascularization and myogenesis.
Collapse
|