1
|
Wang Z, Liang W, Ao R, An Y. Adipose Decellularized Matrix: A Promising Skeletal Muscle Tissue Engineering Material for Volume Muscle Loss. Biomater Res 2025; 29:0174. [PMID: 40248249 PMCID: PMC12003953 DOI: 10.34133/bmr.0174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 02/06/2025] [Accepted: 03/07/2025] [Indexed: 04/19/2025] Open
Abstract
Volume muscle loss is a severe injury often caused by trauma, fracture, tumor resection, or degenerative disease, leading to long-term dysfunction or disability. The current gold-standard treatment is autologous muscle tissue transplantation, with limitations due to donor site restrictions, complications, and low regeneration efficiency. Tissue engineering shows potential to overcome these challenges and achieve optimal muscle regeneration, vascularization, nerve repair, and immunomodulation. In the field of muscle tissue engineering, skeletal muscle decellularized matrices are regarded as an ideal material due to their similarity to the defect site environment, yet they suffer from difficulties in preparation, severe fibrosis, and inconsistent experimental findings. Adipose decellularized matrices (AdECMs) have demonstrated consistent efficacy in promoting muscle regeneration, and their ease of preparation and abundant availability make them even more attractive. The full potential of AdECMs for muscle regeneration remains to be explored. The aim of this review is to summarize the relevant studies on using AdECMs to promote muscle regeneration, to summarize the preparation methods of various applied forms, and to analyze their advantages and shortcomings, as well as to further explore their mechanisms and to propose possible improvements, so as to provide new ideas for the clinical solution of the problem of volume muscle loss.
Collapse
Affiliation(s)
| | - Wei Liang
- Address correspondence to: (W.L.); (Y.A.)
| | - Rigele Ao
- Department of Plastic Surgery,
Peking University Third Hospital, Beijing 100191, China
| | - Yang An
- Department of Plastic Surgery,
Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
2
|
Heisser RH, Bawa M, Shah J, Bu A, Raman R. Soft Biological Actuators for Meter-Scale Homeostatic Biohybrid Robots. Chem Rev 2025; 125:3976-4007. [PMID: 40138615 DOI: 10.1021/acs.chemrev.4c00785] [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: 03/29/2025]
Abstract
Skeletal muscle's elegant protein-based architecture powers motion throughout the animal kingdom, with its constituent actomyosin complexes driving intra- and extra-cellular motion. Classical motors and recently developed soft actuators cannot match the packing density and contractility of individual muscle fibers that scale to power the motion of ants and elephants alike. Accordingly, the interdisciplinary fields of robotics and tissue engineering have combined efforts to build living muscle actuators that can power a new class of robots to be more energy-efficient, dexterous, and safe than existing motor-powered and hydraulic paradigms. Doing so ethically and at scale─creating meter-scale tissue constructs from sustainable muscle progenitor cell lines─has inspired innovations in biomaterials and tissue culture methodology. We weave discussions of muscle cell biology, materials chemistry, tissue engineering, and biohybrid design to review the state of the art in soft actuator biofabrication. Looking forward, we outline a vision for meter-scale biohybrid robotic systems and tie discussions of recent progress to long-term research goals.
Collapse
Affiliation(s)
- Ronald H Heisser
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
| | - Maheera Bawa
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
| | - Jessica Shah
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 45 Carleton St., Cambridge, Massachusetts 02142, United States of America
| | - Angel Bu
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
| | - Ritu Raman
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
| |
Collapse
|
3
|
Li Y, Kawamura G, Dong Q, Li Q, Ozawa T. Near-Infrared Bioluminescence Assays for Protein-Protein Interactions and Cellular Membrane Fusion in Deep Tissues Using Split Akaluc Reconstitution. Anal Chem 2025; 97:6182-6191. [PMID: 40062634 DOI: 10.1021/acs.analchem.4c06986] [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: 03/26/2025]
Abstract
Bioluminescence analysis using luciferase is an essential tool for studying biological processes in different cells. Split luciferase reconstitution is a technique that enables the analysis of biological events through the monitoring of protein-protein interactions. However, effective detection of cellular events in vivo remains challenging due to the limitation of light penetration into deep tissues and optical sensitivity. To address this, we developed a novel split luciferase reconstitution method using a near-infrared-emitting luciferase, Akaluc, and applied it to monitor two important biological events: G protein-coupled receptor (GPCR)/β-arrestin interactions and myogenic cell fusion in vivo. The developed split Akaluc reconstitution system demonstrated high sensitivity in detecting GPCR/β-arrestin interactions as well as myogenic cell fusion in vitro, enabling real-time insights into their temporal dynamics. Moreover, in vivo bioluminescence imaging successfully monitored GPCR/β-arrestin interactions in the mouse lung and the progression of myogenesis during mouse leg muscle regeneration. The split Akaluc reconstitution method will be a versatile tool for both in vitro and in vivo analyses of protein-protein interactions and cell fusion events. This system holds significant potential for advancing drug development, especially in the screening of GPCR-targeted therapeutic and myogenesis-promoting compounds in animal models.
Collapse
Affiliation(s)
- Yiling Li
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Genki Kawamura
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Qi Dong
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Qiaojing Li
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takeaki Ozawa
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
4
|
Barajaa MA, Ghosh D, Laurencin CT. Decellularized Extracellular Matrix-Derived Hydrogels: a Powerful Class of Biomaterials for Skeletal Muscle Regenerative Engineering Applications. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2025; 11:39-63. [PMID: 40201194 PMCID: PMC11978403 DOI: 10.1007/s40883-023-00328-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/18/2023] [Accepted: 11/28/2023] [Indexed: 04/10/2025]
Abstract
Purpose The extracellular matrix (ECM) is a complicated milieu consisting of structural and functional molecules secreted by the resident cells that provides an optimal microenvironmental niche for enhanced cell adhesion, growth, differentiation, and tissue formation and maturation. For decades, ECM bio-scaffolds prepared from decellularized tissues have been used to promote skeletal muscle regeneration; however, it was recently discovered that these decellularized ECM (dECM) materials can be further processed into hydrogels, thus expanding the potential applications of dECM materials in skeletal muscle regenerative engineerisng (SMRE). This review article highlights the recent advances in dECM-derived hydrogels toward skeletal muscle regeneration and repair. Method We screened articles in PubMed and bibliographic search using a combination of keywords. Relevant and high-cited articles were chosen for inclusion in this narrative review. Results Here, we discuss the skeletal muscle ECM's structure, function, and biochemical composition with emphasis on the role of the ECM during skeletal muscle embryogenesis, growth, development, and repair. Furthermore, we review various hydrogels used to promote skeletal muscle regeneration. We also review the current applications of dECM-derived hydrogels toward SMRE. Finally, we discuss the clinical translation potential of dECM-derived hydrogels for skeletal muscle regeneration and repair and their potential clinical considerations in the future. Conclusion Although much progress has been made in the field of dECM-derived hydrogels toward SMRE, it is still in its nascent stage. We believe improving and standardizing the methods of decellularization, lowering the immunogenicity of dECMs, and carrying out in vivo investigations in large animal models would advance their future clinical applications. Lay Summary Researchers have discovered an effective way to turn tissue materials into jelly-like substances known as extracellular matrix (ECM)-derived hydrogels. These ECM-derived hydrogels can help muscles heal better after serious injuries. They can be injected into gaps or used to guide muscle growth in the lab or body. This review article explains how these ECM-derived hydrogels are made and how they can be used to improve muscle healing. It also discusses their possible use in clinics and what needs to be considered before using them for medical treatments.
Collapse
Affiliation(s)
- Mohammed A. Barajaa
- Department of Biomedical Engineering, College of Engineering, Imam Abdulrahman Bin Faisal University, 34212 Dammam, Saudi Arabia
| | - Debolina Ghosh
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
| | - Cato T. Laurencin
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT 06030, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Chemical & Bimolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
5
|
Bu A, Afghah F, Castro N, Bawa M, Kohli S, Shah K, Rios B, Butty V, Raman R. Actuating Extracellular Matrices Decouple the Mechanical and Biochemical Effects of Muscle Contraction on Motor Neurons. Adv Healthc Mater 2025; 14:e2403712. [PMID: 39523700 PMCID: PMC11874633 DOI: 10.1002/adhm.202403712] [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: 09/28/2024] [Revised: 10/03/2024] [Indexed: 11/16/2024]
Abstract
Emerging in vivo evidence suggests that repeated muscle contraction, or exercise, impacts peripheral nerves. However, the difficulty of isolating the muscle-specific impact on motor neurons in vivo, as well as the inability to decouple the biochemical and mechanical impacts of muscle contraction in this setting, motivates investigating this phenomenon in vitro. This study demonstrates that tuning the mechanical properties of fibrin enables longitudinal culture of highly contractile skeletal muscle monolayers, enabling functional characterization of and long-term secretome harvesting from exercised tissues. Motor neurons stimulated with exercised muscle-secreted factors significantly upregulate neurite outgrowth and migration, with an effect size dependent on muscle contraction intensity. Actuating magnetic microparticles embedded within fibrin hydrogels enable dynamically stretching motor neurons and non-invasively mimicking the mechanical effects of muscle contraction. Interestingly, axonogenesis is similarly upregulated in both mechanically and biochemically stimulated motor neurons, but RNA sequencing reveals different transcriptomic signatures between groups, with biochemical stimulation having a greater impact on cell signaling related to axonogenesis and synapse maturation. This study leverages actuating extracellular matrices to robustly validate a previously hypothesized role for muscle contraction in regulating motor neuron growth and maturation from the bottom-up through both mechanical and biochemical signaling.
Collapse
Affiliation(s)
- Angel Bu
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Ferdows Afghah
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Nicolas Castro
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Maheera Bawa
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Sonika Kohli
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Karina Shah
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Brandon Rios
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Vincent Butty
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Ritu Raman
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
6
|
Lee Y, Song J, Batjargal U, Kim MS, Lee G, Kim G, Lee T, Kang R, Kim Y, Kim HJ, Lee J. Pseudo-3D Topological Alignments Regulate Mechanotransduction and Maturation of Smooth Muscle Cells. Adv Healthc Mater 2025; 14:e2402492. [PMID: 39363803 DOI: 10.1002/adhm.202402492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 08/30/2024] [Indexed: 10/05/2024]
Abstract
Smooth muscle cells (SMCs) sense and respond to mechanical stimuli in their extracellular microenvironments (ECMs), playing a crucial role in muscle tissue engineering. Increasing evidence from topological cues-mediated mechanotransduction of SMCs in ECMs has suggested some potential underlying mechanisms of how SMC functions and maturation are regulated by their mechanosensing leading to transduction. However, how the expression of yes-associated protein 1 (YAP) influences the phenotypic shift from synthetic to contractile is still controversial. Here, pseudo-3D topological alignments mimicking native muscle tissues are generated using laser-cutter engraving to explore the influence of topological cues on SMC mechanotransduction and maturation. The analysis of topological cue-mediated mechanotransduction and maturation marker expression revealed YAP is involved in mechanotransduction for SMCs cultured on cross-patterned substrates in the presence of cell-cell interactions. Moreover, these SMCs with YAP-linked mechanosensing showed higher expression of calponin, indicating a shift toward contractile phenotypes in vitro and in vivo. Furthermore, it showed skeletal muscle cells has different mechanosensing and maturation mechanisms compared to SMCs, revealing muscle type-dependent different sensing of topological cues, and converting into maturation-associated signaling cascades. This study provides insights into the regulation of SMC mechanotransduction and maturation by topological cues, suggesting the involvement of YAP-linked signaling pathways in this process.
Collapse
Affiliation(s)
- Yeji Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 790-784, Republic of Korea
| | - Jihyeon Song
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 790-784, Republic of Korea
| | | | - Min-Seok Kim
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea
| | - Geonho Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 790-784, Republic of Korea
| | - Geonwoo Kim
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 790-784, Republic of Korea
| | - Taehoon Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 790-784, Republic of Korea
| | - RaeHui Kang
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 790-784, Republic of Korea
| | - Younggyun Kim
- Department of Bioengineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Han-Jun Kim
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea
| | - Junmin Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 790-784, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Incheon, 21983, Republic of Korea
| |
Collapse
|
7
|
Kamaraj M, Rezayof O, Barer A, Kim H, Moghimi N, Joshi A, Dokmeci MR, Khademhosseini A, Alambeigi F, John JV. Development of silk microfiber-reinforced bioink for muscle tissue engineering and in situ printing by a handheld 3D printer. BIOMATERIALS ADVANCES 2025; 166:214057. [PMID: 39366204 PMCID: PMC11560616 DOI: 10.1016/j.bioadv.2024.214057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/17/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Volumetric muscle loss (VML) presents a significant challenge in tissue engineering due to the irreparable nature of extensive muscle injuries. In this study, we propose a novel approach for VML treatment using a bioink composed of silk microfiber-reinforced silk fibroin (SF) hydrogel. The engineered scaffolds are predesigned to provide structural support and fiber alignment to promote tissue regeneration in situ. We also validated our custom-made handheld 3D printer performance and showcased its potential applications for in situ printing using robotics. The fiber contents of SF and gelatin ink were varied from 1 to 5 %. Silk fibroin microfibers reinforced ink offered increased viscosity of the gel, which enhanced the shape fidelity and mechanical strength of the bulk scaffold. The fiber-reinforced bioink also demonstrated better cell-biomaterial interaction upon printing. The handheld 3D printer enabled the precise and on-demand fabrication of scaffolds directly at the defect site, for personalized and minimally invasive treatment. This innovative approach holds promise for addressing the challenges associated with VML treatment and advancing the field of regenerative medicine.
Collapse
Affiliation(s)
| | - Omid Rezayof
- Walker Department of Mechanical Engineering and Texas Robotics, The University of Texas at Austin, TX, USA
| | - Alison Barer
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, USA
| | - Hansoul Kim
- Walker Department of Mechanical Engineering and Texas Robotics, The University of Texas at Austin, TX, USA
| | - Nafiseh Moghimi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, USA
| | - Akshat Joshi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, USA
| | - Mehmet R Dokmeci
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, USA
| | | | - Farshid Alambeigi
- Walker Department of Mechanical Engineering and Texas Robotics, The University of Texas at Austin, TX, USA
| | - Johnson V John
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, USA.
| |
Collapse
|
8
|
Lee MC, Jodat YA, Endo Y, Rodríguez-delaRosa A, Zhang T, Karvar M, Tanoury ZA, Quint J, Kamperman T, Kiaee K, Ochoa SL, Shi K, Huang Y, Rosales MP, Lee H, Kim J, Ceron EL, Reyes IG, Panayi AC, Wang X, Kim KT, Moon JI, Park SG, Lee K, Calabrese MA, Lee J, Tamayol A, Lee L, Pourquié O, Kim WJ, Sinha I, Shin SR. Engineering large-scale hiPSC-derived vessel-integrated muscle-like lattices for enhanced volumetric muscle regeneration. Trends Biotechnol 2024; 42:1715-1744. [PMID: 39306493 PMCID: PMC11625013 DOI: 10.1016/j.tibtech.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 12/08/2024]
Abstract
Engineering biomimetic tissue implants with human induced pluripotent stem cells (hiPSCs) holds promise for repairing volumetric tissue loss. However, these implants face challenges in regenerative capability, survival, and geometric scalability at large-scale injury sites. Here, we present scalable vessel-integrated muscle-like lattices (VMLs), containing dense and aligned hiPSC-derived myofibers alongside passively perfusable vessel-like microchannels inside an endomysium-like supporting matrix using an embedded multimaterial bioprinting technology. The contractile and millimeter-long myofibers are created in mechanically tailored and nanofibrous extracellular matrix-based hydrogels. Incorporating vessel-like lattice enhances myofiber maturation in vitro and guides host vessel invasion in vivo, improving implant integration. Consequently, we demonstrate successful de novo muscle formation and muscle function restoration through a combinatorial effect between improved graft-host integration and its increased release of paracrine factors within volumetric muscle loss injury models. The proposed modular bioprinting technology enables scaling up to centimeter-sized prevascularized hiPSC-derived muscle tissues with custom geometries for next-generation muscle regenerative therapies.
Collapse
Affiliation(s)
- Myung Chul Lee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Medicinal Materials Research Center, Korea Institute of Science and Technology, Seoul, 02792 Republic of Korea
| | - Yasamin A. Jodat
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Yori Endo
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Alejandra Rodríguez-delaRosa
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Ting Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
| | - Mehran Karvar
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ziad Al Tanoury
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Jacob Quint
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE, 68588, USA
| | - Tom Kamperman
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Kiavash Kiaee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Sofia Lara Ochoa
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Kun Shi
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Yike Huang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Montserrat Pineda Rosales
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Hyeseon Lee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Jiseong Kim
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Eder Luna Ceron
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Isaac Garcia Reyes
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Adriana C. Panayi
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xichi Wang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Ki-Tae Kim
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Dental Multi-omics Center, Seoul National University, Seoul, 08826, Republic of Korea
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jae-I Moon
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Dental Multi-omics Center, Seoul National University, Seoul, 08826, Republic of Korea
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Gwa Park
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Dental Multi-omics Center, Seoul National University, Seoul, 08826, Republic of Korea
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kangju Lee
- Department of Healthcare and Medical Engineering, Chonnam National University, Yeosu 59626, South Korea
| | - Michelle A. Calabrese
- Chemical Engineering and Materials Science Department, University of Minnesota, Minneapolis, MN 55455, USA
| | - Junmin Lee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE, 68588, USA
| | - Luke Lee
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Korea
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Korea
| | - Olivier Pourquié
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Woo-Jin Kim
- Correspondence: (I.S.), (W.J.K.), (S.R.S.), Twitter: Yasamin A. Jodat: @YasaminJodat
| | - Indranil Sinha
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Lead contact
| |
Collapse
|
9
|
Lee SJ, Jeong W, Atala A. 3D Bioprinting for Engineered Tissue Constructs and Patient-Specific Models: Current Progress and Prospects in Clinical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2408032. [PMID: 39420757 PMCID: PMC11875024 DOI: 10.1002/adma.202408032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/24/2024] [Indexed: 10/19/2024]
Abstract
Advancements in bioprinting technology are driving the creation of complex, functional tissue constructs for use in tissue engineering and regenerative medicine. Various methods, including extrusion, jetting, and light-based bioprinting, have their unique advantages and drawbacks. Over the years, researchers and industry leaders have made significant progress in enhancing bioprinting techniques and materials, resulting in the production of increasingly sophisticated tissue constructs. Despite this progress, challenges still need to be addressed in achieving clinically relevant, human-scale tissue constructs, presenting a hurdle to widespread clinical translation. However, with ongoing interdisciplinary research and collaboration, the field is rapidly evolving and holds promise for personalized medical interventions. Continued development and refinement of bioprinting technologies have the potential to address complex medical needs, enabling the development of functional, transplantable tissues and organs, as well as advanced in vitro tissue models.
Collapse
Affiliation(s)
| | | | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, United States
| |
Collapse
|
10
|
Kang JH, Kim DH, Yoo J, Shin JH, Kim JH, Lee JW, Shin SH. Sinapine suppresses ROS-induced C2C12 myoblast cell death through MAPK and autophagy pathways. Food Sci Biotechnol 2024; 33:3629-3637. [PMID: 39493388 PMCID: PMC11525351 DOI: 10.1007/s10068-024-01718-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/05/2024] [Accepted: 09/18/2024] [Indexed: 11/05/2024] Open
Abstract
Oxidative stress in skeletal muscle can lead to muscle atrophy through reactive oxygen species (ROS)-induced damage and cell death. tert-Butyl hydroperoxide (TBHP), an exogenous ROS generator, induces oxidative stress and cell death in various cells. Sinapine from cruciferous plants possesses beneficial effects, but its role in protecting skeletal muscle cells against ROS-induced cell death remains unclear. This study demonstrates that sinapine pretreatment significantly reduced TBHP-induced cell death and ROS accumulation in a dose-dependent manner. TBHP activated mitogen-activated protein kinase (MAPK) pathways including Akt, p38, and JNK, and triggered autophagy. Sinapine suppressed the phosphorylation of Akt, MEK3/6, p38, MEK4, and JNK, and modulated key autophagy markers. Notably, the co-treatment of MAPK inhibitors attenuated TBHP-induced cell death and LC3B-II accumulation. These findings suggest that sinapine is a promising phytochemical for mitigating oxidative stress-mediated muscle injury, offering potential therapeutic strategies for maintaining skeletal muscle homeostasis and addressing muscle-related pathologies.
Collapse
Affiliation(s)
- Jung Hyun Kang
- Department of Food and Nutrition, Gyeongsang National University, Jinju, 52828 South Korea
| | - Dong Hwan Kim
- Department of Bio & Medical Bigdata (BK4 Program), Gyeongsang National University, Jinju, 52828 South Korea
| | - Jin Yoo
- Department of Food and Nutrition, Gyeongsang National University, Jinju, 52828 South Korea
| | - Jun Hong Shin
- Department of Food and Nutrition, Gyeongsang National University, Jinju, 52828 South Korea
| | - Ju Hyun Kim
- Department of Food and Nutrition, Gyeongsang National University, Jinju, 52828 South Korea
| | - Ji Won Lee
- Department of Food and Nutrition, Gyeongsang National University, Jinju, 52828 South Korea
| | - Seung Ho Shin
- Department of Food and Nutrition, Gyeongsang National University, Jinju, 52828 South Korea
- Department of Bio & Medical Bigdata (BK4 Program), Gyeongsang National University, Jinju, 52828 South Korea
| |
Collapse
|
11
|
Volpi M, Paradiso A, Walejewska E, Gargioli C, Costantini M, Swieszkowski W. Automated Microfluidics-Assisted Hydrogel-Based Wet-Spinning for the Biofabrication of Biomimetic Engineered Myotendinous Junction. Adv Healthc Mater 2024; 13:e2402075. [PMID: 39313990 DOI: 10.1002/adhm.202402075] [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: 06/05/2024] [Revised: 09/08/2024] [Indexed: 09/25/2024]
Abstract
The muscle-tendon junction (MTJ) plays a pivotal role in efficiently converting the muscular contraction into a controlled skeletal movement through the tendon. Given its complex biomechanical intricacy, the biofabrication of such tissue interface represents a significant challenge in the field of musculoskeletal tissue engineering. Herein, a novel method to produce MTJ-like hydrogel yarns using a microfluidics-assisted 3D rotary wet-spinning strategy is developed. Optimization of flow rates, rotational speed, and delivery time of bioinks enables the production of highly compartmentalized scaffolds that recapitulate the muscle, tendon, and the transient MTJ-like region. Additionally, such biofabrication parameters are validated in terms of cellular response by promoting an optimal uniaxial alignment for both muscle and tendon precursor cells. By sequentially wet-spinning C2C12 myoblasts and NIH 3T3 fibroblasts, a gradient-patterned cellular arrangement mirroring the intrinsic biological heterogeneity of the MTJ is successfully obtained. The immunofluorescence assessment further reveals the localized expression of tissue-specific markers, including myosin heavy chain and collagen type I/III, which demonstrate muscle and tenogenic tissue maturation, respectively. Remarkably, the muscle-tendon transition zone exhibits finger-like projection of the multinucleated myotubes in the tenogenic compartment, epitomizing the MTJ signature architecture.
Collapse
Affiliation(s)
- Marina Volpi
- Faculty of Materials Science and Engineering, Warsaw University of Technology, Warsaw, 02-507, Poland
| | - Alessia Paradiso
- Faculty of Materials Science and Engineering, Warsaw University of Technology, Warsaw, 02-507, Poland
| | - Ewa Walejewska
- Faculty of Materials Science and Engineering, Warsaw University of Technology, Warsaw, 02-507, Poland
| | - Cesare Gargioli
- Department of Biology, University of Rome Tor Vergata, Rome, 00133, Italy
| | - Marco Costantini
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, 01-224, Poland
| | - Wojciech Swieszkowski
- Faculty of Materials Science and Engineering, Warsaw University of Technology, Warsaw, 02-507, Poland
| |
Collapse
|
12
|
Kwon J, Eom S, Kong JS, Cho DW, Kim DS, Kim J. Engineered Regenerative Isolated Peripheral Nerve Interface for Targeted Reinnervation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2406652. [PMID: 39051516 DOI: 10.1002/adma.202406652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/24/2024] [Indexed: 07/27/2024]
Abstract
A regenerative peripheral nerve interface (RPNI) offers a therapeutic solution for nerve injury through reconstruction of the target muscle. However, implanting a transected peripheral nerve into an autologous skeletal muscle graft in RPNI causes donor-site morbidity, highlighting the need for tissue-engineered skeletal muscle constructs. Here, an engineered regenerative isolated peripheral nerve interface (eRIPEN) is developed using 3D skeletal cell printing combined with direct electrospinning to create a nanofiber membrane envelop for host nerve implantation. In this in vivo study, after over 8 months of RPNI surgery, the eRIPEN exhibits a minimum Feret diameter of 15-20 µm with a cross-sectional area of 100-500 µm2, representing the largest distribution of myofibers. Furthermore, neuromuscular junction formation and muscle contraction with a force of ≈28 N are observed. Notably, the decreased hypersensitivity to mechanical/thermal stimuli and an improved tibial functional index from -77 to -56 are found in the eRIPEN group. The present novel concept of eRIPEN paves the way for the utilization and application of tissue-engineered constructs in RPNI, ultimately realizing neuroprosthesis control through synaptic connections.
Collapse
Affiliation(s)
- Jinju Kwon
- Department of Health Science, Graduate School, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
- Transdisciplinary Major in Learning Health Systems, Department of Healthcare Sciences, Graduate School, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Seongsu Eom
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Jeong Sik Kong
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk, 37673, Republic of Korea
- POSTECH-Catholic Biomedical Engineering Institute, POSTECH, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk, 37673, Republic of Korea
- POSTECH-Catholic Biomedical Engineering Institute, POSTECH, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Dong Sung Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk, 37673, Republic of Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk, 37673, Republic of Korea
- Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk, 37673, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, 50, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Junesun Kim
- Department of Health Science, Graduate School, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
- Department of Health and Environmental Science, Undergraduate School, College of Health Science, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| |
Collapse
|
13
|
Goswami AB, Rybchyn MS, Walsh W, le Coutre J. Obtaining source material for cellular agriculture. Heliyon 2024; 10:e38006. [PMID: 39364244 PMCID: PMC11447359 DOI: 10.1016/j.heliyon.2024.e38006] [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: 06/22/2024] [Revised: 09/10/2024] [Accepted: 09/16/2024] [Indexed: 10/05/2024] Open
Abstract
Cellular Agriculture (CellAg) is an attractive concept for innovative technology with the intent to provide food and nutrition complementary to existing supply streams. The past decade has seen considerable progress in the field with advancement of cellular technology that delivers the initial building blocks for meaningful implementation. The availability of natural cell-based material that can serve as nutrient-filled source for human consumption at low cost is a critical challenge for the emerging cellular agriculture industry. Therefore, here the isolation of bovine myofibroblasts of the Black Angus breed has been pursued and accomplished together with its characterisation by using RNA sequencing and proteomics through western blotting. To transition CellAg from a concept to a game changing technology for the industry, multiple challenges need to be overcome. The field requires powerful initial material, i.e., dedicated cells that can proliferate and differentiate robustly at scale. The methodology described allows for the production of healthy cells, which have been unequivocally characterized as clonal representatives of a stable myofibroblast cell line using transcriptomics and proteomics validation. Stringent and rigorous live cell monitoring of a nascent cell line derived from healthy muscle tissue allowed for stable cell growth. In this research article, a simple and precise methodology is presented for creating a bovine myofibroblast cell line (Bov.mia). Our work puts forward a low-tech use of materials and expertise that is devoid of transgenic approaches, thus creating a reliable approach for lab-scale research.
Collapse
Affiliation(s)
- Apeksha Bharatgiri Goswami
- School of Chemical Engineering, University of New South Wales, Sydney, New South Wales, Sydney, Australia
| | - Mark S. Rybchyn
- School of Chemical Engineering, University of New South Wales, Sydney, New South Wales, Sydney, Australia
| | - W.R. Walsh
- School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Johannes le Coutre
- School of Chemical Engineering, University of New South Wales, Sydney, New South Wales, Sydney, Australia
- Australian Human Rights Institute, University of New South Wales, Sydney, New South Wales, Sydney, Australia
| |
Collapse
|
14
|
Yadav S, Joshi R, Majumder A. Concave/Convex Curvature of Anisotropic Grooves Differentially Alters Cellular Morphology, Adhesion, and Proliferation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:17590-17600. [PMID: 39132850 DOI: 10.1021/acs.langmuir.4c01896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Curvature is an integral part of the complex in vivo tissue architecture across various length scales. Therefore, several in vitro models with a patterned curvature in different length scales have been developed to understand the role of this in cellular behavior. At the subcellular scale, wavy patterns have been reported wherein concave and convex grooves are adjacently present. However, the independent effect of continuous subcellular concave and convex shapes has not been reported, mainly owing to the limitations in fabricating such patterns. In this study, we developed continuous concave and convex grooves on polydimethylsiloxane (PDMS) using a Dracaena sanderiana (bamboo) leaf as a template. The first (negative) replica from the abaxial side of the bamboo leaf, which imparted concave grooves on PDMS, was subsequently used as a template to fabricate a positive replica of the leaf, resulting in convex grooves of the same size and arrangement as the concave grooves. We examined the influence of the groove curvature on the morphology of bone marrow-derived human mesenchymal stem cells (BM-hMSCs) and skeletal muscle cells (C2C12). BM-hMSCs and C2C12 cells aligned on both concave and convex grooves as compared to the random orientation on a flat substrate. The significant difference was observed in the morphology of both cells, in terms of area, aspect ratio, number, and length of protrusions on concave and convex patterns. We found that the number of protrusions was also dependent on the ratio of cell to pattern length scale for convex-shaped grooves but independent of length scale for concave-shaped grooves. The proliferation of BM-hMSCs was also found to be different on concave and convex shapes. Therefore, this study shows the importance of (1) convex and concave curvatures of the subcellular length scale in cellular response, (2) dependence on the ratio of cell and curvature length scale, and (3) use of natural templates for overcoming fabrication challenges.
Collapse
Affiliation(s)
- Shital Yadav
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai400076,India
| | - Rohit Joshi
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai400076,India
| | - Abhijit Majumder
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai400076,India
| |
Collapse
|
15
|
Nguyen ML, Demri N, Lapin B, Di Federico F, Gropplero G, Cayrac F, Hennig K, Gomes ER, Wilhelm C, Roman W, Descroix S. Studying the impact of geometrical and cellular cues on myogenesis with a skeletal muscle-on-chip. LAB ON A CHIP 2024; 24:4147-4160. [PMID: 39072529 DOI: 10.1039/d4lc00417e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
In the skeletal muscle tissue, cells are organized following an anisotropic architecture, which is both required during myogenesis when muscle precursor cells fuse to generate myotubes and for its contractile function. To build an in vitro skeletal muscle tissue, it is therefore essential to develop methods to organize cells in an anisotropic fashion, which can be particularly challenging, especially in 3D. In this study, we present a versatile muscle-on-chip system with adjustable collagen hollow tubes that can be seeded with muscle precursor cells. The collagen acts both as a tube-shaped hollow mold and as an extracellular matrix scaffold that can house other cell types for co-culture. We found that the diameter of the channel affects the organization of the muscle cells and that proper myogenesis was obtained at a diameter of 75 μm. In these conditions, muscle precursor cells fused into long myotubes aligned along these collagen channels, resulting in a fascicle-like structure. These myotubes exhibited actin striations and upregulation of multiple myogenic genes, reflecting their maturation. Moreover, we showed that our chip allowed muscle tissue culture and maturation over a month, with the possibility of fibroblast co-culture embedding in the collagen matrix.
Collapse
Affiliation(s)
- M-L Nguyen
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - N Demri
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - B Lapin
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - F Di Federico
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - G Gropplero
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - F Cayrac
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - K Hennig
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Edgar R Gomes
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - C Wilhelm
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - W Roman
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Australian Regenerative Medicine Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - S Descroix
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| |
Collapse
|
16
|
Bonetti L, Scalet G. 4D fabrication of shape-changing systems for tissue engineering: state of the art and perspectives. PROGRESS IN ADDITIVE MANUFACTURING 2024; 10:1913-1943. [PMID: 40125451 PMCID: PMC11926060 DOI: 10.1007/s40964-024-00743-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/30/2024] [Indexed: 03/25/2025]
Abstract
In recent years, four-dimensional (4D) fabrication has emerged as a powerful technology capable of revolutionizing the field of tissue engineering. This technology represents a shift in perspective from traditional tissue engineering approaches, which generally rely on static-or passive-structures (e.g., scaffolds, constructs) unable of adapting to changes in biological environments. In contrast, 4D fabrication offers the unprecedented possibility of fabricating complex designs with spatiotemporal control over structure and function in response to environment stimuli, thus mimicking biological processes. In this review, an overview of the state of the art of 4D fabrication technology for the obtainment of cellularized constructs is presented, with a focus on shape-changing soft materials. First, the approaches to obtain cellularized constructs are introduced, also describing conventional and non-conventional fabrication techniques with their relative advantages and limitations. Next, the main families of shape-changing soft materials, namely shape-memory polymers and shape-memory hydrogels are discussed and their use in 4D fabrication in the field of tissue engineering is described. Ultimately, current challenges and proposed solutions are outlined, and valuable insights into future research directions of 4D fabrication for tissue engineering are provided to disclose its full potential.
Collapse
Affiliation(s)
- Lorenzo Bonetti
- Department of Civil Engineering and Architecture (DICAr), University of Pavia, Via Ferrata 3, 27100 Pavia, Italy
| | - Giulia Scalet
- Department of Civil Engineering and Architecture (DICAr), University of Pavia, Via Ferrata 3, 27100 Pavia, Italy
| |
Collapse
|
17
|
Nurul Alam AMM, Kim CJ, Kim SH, Kumari S, Lee EY, Hwang YH, Joo ST. Scaffolding fundamentals and recent advances in sustainable scaffolding techniques for cultured meat development. Food Res Int 2024; 189:114549. [PMID: 38876607 DOI: 10.1016/j.foodres.2024.114549] [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: 01/03/2024] [Revised: 02/26/2024] [Accepted: 05/25/2024] [Indexed: 06/16/2024]
Abstract
In cultured meat (CM) production, Scaffolding plays an important role by aiding cell adhesion, growth, differentiation, and alignment. The existence of fibrous microstructure in connective and muscle tissues has attracted considerable interest in the realm of tissue engineering and triggered the interest of researchers to implement scaffolding techniques. A wide array of research efforts is ongoing in scaffolding technologies for achieving the real meat structure on the principality of biomedical research and to replace serum free CM production. Scaffolds made of animal-derived biomaterials are found efficient in replicating the extracellular matrix (ECM), thus focus should be paid to utilize animal byproducts for this purpose. Proper identification and utilization of plant-derived scaffolding biomaterial could be helpful to add diversified options in addition to animal derived sources and reduce in cost of CM production through scaffolds. Furthermore, techniques like electrospinning, modified electrospinning and 3D bioprinting should be focused on to create 3D porous scaffolds to mimic the ECM of the muscle tissue and form real meat-like structures. This review discusses recent advances in cutting edge scaffolding techniques and edible biomaterials related to structured CM production.
Collapse
Affiliation(s)
- A M M Nurul Alam
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52852, Republic of Korea.
| | - Chan-Jin Kim
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52852, Republic of Korea.
| | - So-Hee Kim
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52852, Republic of Korea
| | - Swati Kumari
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52852, Republic of Korea
| | - Eun-Yeong Lee
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52852, Republic of Korea
| | - Young-Hwa Hwang
- Institute of Agriculture & Life Science, Gyeongsang National University, Jinju 52852, Republic of Korea.
| | - Seon-Tea Joo
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52852, Republic of Korea; Institute of Agriculture & Life Science, Gyeongsang National University, Jinju 52852, Republic of Korea.
| |
Collapse
|
18
|
Rich J, Bennaroch M, Notel L, Patalakh P, Alberola J, Issa F, Opolon P, Bawa O, Rondof W, Marchais A, Dessen P, Meurice G, Le-Gall M, Polrot M, Ser-Le Roux K, Mamchaoui K, Droin N, Raslova H, Maire P, Geoerger B, Pirozhkova I. DiPRO1 distinctly reprograms muscle and mesenchymal cancer cells. EMBO Mol Med 2024; 16:1840-1885. [PMID: 39009887 PMCID: PMC11319797 DOI: 10.1038/s44321-024-00097-z] [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: 01/10/2023] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 07/17/2024] Open
Abstract
We have recently identified the uncharacterized ZNF555 protein as a component of a productive complex involved in the morbid function of the 4qA locus in facioscapulohumeral dystrophy. Subsequently named DiPRO1 (Death, Differentiation, and PROliferation related PROtein 1), our study provides substantial evidence of its role in the differentiation and proliferation of human myoblasts. DiPRO1 operates through the regulatory binding regions of SIX1, a master regulator of myogenesis. Its relevance extends to mesenchymal tumors, such as rhabdomyosarcoma (RMS) and Ewing sarcoma, where DiPRO1 acts as a repressor via the epigenetic regulators TIF1B and UHRF1, maintaining methylation of cis-regulatory elements and gene promoters. Loss of DiPRO1 mimics the host defense response to virus, awakening retrotransposable repeats and the ZNF/KZFP gene family. This enables the eradication of cancer cells, reprogramming the cellular decision balance towards inflammation and/or apoptosis by controlling TNF-α via NF-kappaB signaling. Finally, our results highlight the vulnerability of mesenchymal cancer tumors to si/shDiPRO1-based nanomedicines, positioning DiPRO1 as a potential therapeutic target.
Collapse
Affiliation(s)
- Jeremy Rich
- UMR8126 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Melanie Bennaroch
- UMR8126 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Laura Notel
- UMR8126 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Polina Patalakh
- UMR8126 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Julien Alberola
- UMR8126 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Fayez Issa
- INSERM U1016, CNRS UMR 8104, Institut Cochin, Université Paris-Cité, Paris, France
| | - Paule Opolon
- Pathology and Cytology Section, UMS AMMICA, CNRS, INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Olivia Bawa
- Pathology and Cytology Section, UMS AMMICA, CNRS, INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Windy Rondof
- Bioinformatics Platform, UMS AMMICA, CNRS, INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer campus, INSERM U1015, Université Paris-Saclay, Villejuif, France
| | - Antonin Marchais
- Bioinformatics Platform, UMS AMMICA, CNRS, INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer campus, INSERM U1015, Université Paris-Saclay, Villejuif, France
| | - Philippe Dessen
- Bioinformatics Platform, UMS AMMICA, CNRS, INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Guillaume Meurice
- Bioinformatics Platform, UMS AMMICA, CNRS, INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Morgane Le-Gall
- Proteom'IC facility, Université Paris Cité, CNRS, INSERM, Institut Cochin, F-75014, Paris, France
| | - Melanie Polrot
- Pre-clinical Evaluation Unit (PFEP), INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Karine Ser-Le Roux
- Pre-clinical Evaluation Unit (PFEP), INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Kamel Mamchaoui
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, F-75013, Paris, France
| | - Nathalie Droin
- Genomic Platform, UMS AMMICA US 23 INSERM UAR 3655 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
- UMR1287 INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Hana Raslova
- UMR1287 INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Pascal Maire
- INSERM U1016, CNRS UMR 8104, Institut Cochin, Université Paris-Cité, Paris, France
| | - Birgit Geoerger
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer campus, INSERM U1015, Université Paris-Saclay, Villejuif, France
| | - Iryna Pirozhkova
- UMR8126 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France.
- INSERM U1016, CNRS UMR 8104, Institut Cochin, Université Paris-Cité, Paris, France.
| |
Collapse
|
19
|
Luo W, Zhang H, Wan R, Cai Y, Liu Y, Wu Y, Yang Y, Chen J, Zhang D, Luo Z, Shang X. Biomaterials-Based Technologies in Skeletal Muscle Tissue Engineering. Adv Healthc Mater 2024; 13:e2304196. [PMID: 38712598 DOI: 10.1002/adhm.202304196] [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: 11/28/2023] [Revised: 04/26/2024] [Indexed: 05/08/2024]
Abstract
For many clinically prevalent severe injuries, the inherent regenerative capacity of skeletal muscle remains inadequate. Skeletal muscle tissue engineering (SMTE) seeks to meet this clinical demand. With continuous progress in biomedicine and related technologies including micro/nanotechnology and 3D printing, numerous studies have uncovered various intrinsic mechanisms regulating skeletal muscle regeneration and developed tailored biomaterial systems based on these understandings. Here, the skeletal muscle structure and regeneration process are discussed and the diverse biomaterial systems derived from various technologies are explored in detail. Biomaterials serve not merely as local niches for cell growth, but also as scaffolds endowed with structural or physicochemical properties that provide tissue regenerative cues such as topographical, electrical, and mechanical signals. They can also act as delivery systems for stem cells and bioactive molecules that have been shown as key participants in endogenous repair cascades. To achieve bench-to-bedside translation, the typical effect enabled by biomaterial systems and the potential underlying molecular mechanisms are also summarized. Insights into the roles of biomaterials in SMTE from cellular and molecular perspectives are provided. Finally, perspectives on the advancement of SMTE are provided, for which gene therapy, exosomes, and hybrid biomaterials may hold promise to make important contributions.
Collapse
Affiliation(s)
- Wei Luo
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Hanli Zhang
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Renwen Wan
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yuxi Cai
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yinuo Liu
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Yang Wu
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yimeng Yang
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Jiani Chen
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, 999077, Hong Kong
| | - Zhiwen Luo
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Xiliang Shang
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| |
Collapse
|
20
|
Tamo AK, Djouonkep LDW, Selabi NBS. 3D Printing of Polysaccharide-Based Hydrogel Scaffolds for Tissue Engineering Applications: A Review. Int J Biol Macromol 2024; 270:132123. [PMID: 38761909 DOI: 10.1016/j.ijbiomac.2024.132123] [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: 12/05/2023] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/20/2024]
Abstract
In tissue engineering, 3D printing represents a versatile technology employing inks to construct three-dimensional living structures, mimicking natural biological systems. This technology efficiently translates digital blueprints into highly reproducible 3D objects. Recent advances have expanded 3D printing applications, allowing for the fabrication of diverse anatomical components, including engineered functional tissues and organs. The development of printable inks, which incorporate macromolecules, enzymes, cells, and growth factors, is advancing with the aim of restoring damaged tissues and organs. Polysaccharides, recognized for their intrinsic resemblance to components of the extracellular matrix have garnered significant attention in the field of tissue engineering. This review explores diverse 3D printing techniques, outlining distinctive features that should characterize scaffolds used as ideal matrices in tissue engineering. A detailed investigation into the properties and roles of polysaccharides in tissue engineering is highlighted. The review also culminates in a profound exploration of 3D polysaccharide-based hydrogel applications, focusing on recent breakthroughs in regenerating different tissues such as skin, bone, cartilage, heart, nerve, vasculature, and skeletal muscle. It further addresses challenges and prospective directions in 3D printing hydrogels based on polysaccharides, paving the way for innovative research to fabricate functional tissues, enhancing patient care, and improving quality of life.
Collapse
Affiliation(s)
- Arnaud Kamdem Tamo
- Institute of Microsystems Engineering IMTEK, University of Freiburg, 79110 Freiburg, Germany; Freiburg Center for Interactive Materials and Bioinspired Technologies FIT, University of Freiburg, 79110 Freiburg, Germany; Freiburg Materials Research Center FMF, University of Freiburg, 79104 Freiburg, Germany; Ingénierie des Matériaux Polymères (IMP), Université Claude Bernard Lyon 1, INSA de Lyon, Université Jean Monnet, CNRS, UMR 5223, 69622 Villeurbanne CEDEX, France.
| | - Lesly Dasilva Wandji Djouonkep
- College of Petroleum Engineering, Yangtze University, Wuhan 430100, China; Key Laboratory of Drilling and Production Engineering for Oil and Gas, Wuhan 430100, China
| | - Naomie Beolle Songwe Selabi
- Institute of Advanced Materials and Nanotechnology, Wuhan University of Science and Technology, Wuhan 430081, China
| |
Collapse
|
21
|
Farhang Doost N, Srivastava SK. A Comprehensive Review of Organ-on-a-Chip Technology and Its Applications. BIOSENSORS 2024; 14:225. [PMID: 38785699 PMCID: PMC11118005 DOI: 10.3390/bios14050225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/09/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024]
Abstract
Organ-on-a-chip (OOC) is an emerging technology that simulates an artificial organ within a microfluidic cell culture chip. Current cell biology research focuses on in vitro cell cultures due to various limitations of in vivo testing. Unfortunately, in-vitro cell culturing fails to provide an accurate microenvironment, and in vivo cell culturing is expensive and has historically been a source of ethical controversy. OOC aims to overcome these shortcomings and provide the best of both in vivo and in vitro cell culture research. The critical component of the OOC design is utilizing microfluidics to ensure a stable concentration gradient, dynamic mechanical stress modeling, and accurate reconstruction of a cellular microenvironment. OOC also has the advantage of complete observation and control of the system, which is impossible to recreate in in-vivo research. Multiple throughputs, channels, membranes, and chambers are constructed in a polydimethylsiloxane (PDMS) array to simulate various organs on a chip. Various experiments can be performed utilizing OOC technology, including drug delivery research and toxicology. Current technological expansions involve multiple organ microenvironments on a single chip, allowing for studying inter-tissue interactions. Other developments in the OOC technology include finding a more suitable material as a replacement for PDMS and minimizing artefactual error and non-translatable differences.
Collapse
Affiliation(s)
| | - Soumya K. Srivastava
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA;
| |
Collapse
|
22
|
de Melo LF, Almeida GHDR, Azarias FR, Carreira ACO, Astolfi-Ferreira C, Ferreira AJP, Pereira EDSBM, Pomini KT, Marques de Castro MV, Silva LMD, Maria DA, Rici REG. Decellularized Bovine Skeletal Muscle Scaffolds: Structural Characterization and Preliminary Cytocompatibility Evaluation. Cells 2024; 13:688. [PMID: 38667303 PMCID: PMC11048772 DOI: 10.3390/cells13080688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Skeletal muscle degeneration is responsible for major mobility complications, and this muscle type has little regenerative capacity. Several biomaterials have been proposed to induce muscle regeneration and function restoration. Decellularized scaffolds present biological properties that allow efficient cell culture, providing a suitable microenvironment for artificial construct development and being an alternative for in vitro muscle culture. For translational purposes, biomaterials derived from large animals are an interesting and unexplored source for muscle scaffold production. Therefore, this study aimed to produce and characterize bovine muscle scaffolds to be applied to muscle cell 3D cultures. Bovine muscle fragments were immersed in decellularizing solutions for 7 days. Decellularization efficiency, structure, composition, and three-dimensionality were evaluated. Bovine fetal myoblasts were cultured on the scaffolds for 10 days to attest cytocompatibility. Decellularization was confirmed by DAPI staining and DNA quantification. Histological and immunohistochemical analysis attested to the preservation of main ECM components. SEM analysis demonstrated that the 3D structure was maintained. In addition, after 10 days, fetal myoblasts were able to adhere and proliferate on the scaffolds, attesting to their cytocompatibility. These data, even preliminary, infer that generated bovine muscular scaffolds were well structured, with preserved composition and allowed cell culture. This study demonstrated that biomaterials derived from bovine muscle could be used in tissue engineering.
Collapse
Affiliation(s)
- Luana Félix de Melo
- Graduate Program in Anatomy of Domestic and Wild Animals, University of São Paulo, São Paulo 03828-000, Brazil; (L.F.d.M.); (A.C.O.C.); (R.E.G.R.)
| | | | - Felipe Rici Azarias
- Graduate Program of Medical Sciences, College of Medicine, University of São Paulo, São Paulo 03828-000, Brazil;
| | - Ana Claudia Oliveira Carreira
- Graduate Program in Anatomy of Domestic and Wild Animals, University of São Paulo, São Paulo 03828-000, Brazil; (L.F.d.M.); (A.C.O.C.); (R.E.G.R.)
- Center of Human and Natural Sciences, Federal University of ABC, Santo André 09210-170, Brazil
| | - Claudete Astolfi-Ferreira
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 03828-000, Brazil; (C.A.-F.); (A.J.P.F.)
| | - Antônio José Piantino Ferreira
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 03828-000, Brazil; (C.A.-F.); (A.J.P.F.)
| | - Eliana de Souza Bastos Mazuqueli Pereira
- Graduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil; (E.d.S.B.M.P.); (K.T.P.); (M.V.M.d.C.); (L.M.D.S.)
| | - Karina Torres Pomini
- Graduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil; (E.d.S.B.M.P.); (K.T.P.); (M.V.M.d.C.); (L.M.D.S.)
| | - Marcela Vialogo Marques de Castro
- Graduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil; (E.d.S.B.M.P.); (K.T.P.); (M.V.M.d.C.); (L.M.D.S.)
| | - Laira Mireli Dias Silva
- Graduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil; (E.d.S.B.M.P.); (K.T.P.); (M.V.M.d.C.); (L.M.D.S.)
| | | | - Rose Eli Grassi Rici
- Graduate Program in Anatomy of Domestic and Wild Animals, University of São Paulo, São Paulo 03828-000, Brazil; (L.F.d.M.); (A.C.O.C.); (R.E.G.R.)
- Graduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil; (E.d.S.B.M.P.); (K.T.P.); (M.V.M.d.C.); (L.M.D.S.)
| |
Collapse
|
23
|
Taale M, Schamberger B, Monclus MA, Dolle C, Taheri F, Mager D, Eggeler YM, Korvink JG, Molina‐Aldareguia JM, Selhuber‐Unkel C, Lantada AD, Islam M. Microarchitected Compliant Scaffolds of Pyrolytic Carbon for 3D Muscle Cell Growth. Adv Healthc Mater 2024; 13:e2303485. [PMID: 38150609 PMCID: PMC11469158 DOI: 10.1002/adhm.202303485] [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: 11/21/2023] [Indexed: 12/29/2023]
Abstract
The integration of additive manufacturing technologies with the pyrolysis of polymeric precursors enables the design-controlled fabrication of architected 3D pyrolytic carbon (PyC) structures with complex architectural details. Despite great promise, their use in cellular interaction remains unexplored. This study pioneers the utilization of microarchitected 3D PyC structures as biocompatible scaffolds for the colonization of muscle cells in a 3D environment. PyC scaffolds are fabricated using micro-stereolithography, followed by pyrolysis. Furthermore, an innovative design strategy using revolute joints is employed to obtain novel, compliant structures of architected PyC. The pyrolysis process results in a pyrolysis temperature- and design-geometry-dependent shrinkage of up to 73%, enabling the geometrical features of microarchitected compatible with skeletal muscle cells. The stiffness of architected PyC varies with the pyrolysis temperature, with the highest value of 29.57 ± 0.78 GPa for 900 °C. The PyC scaffolds exhibit excellent biocompatibility and yield 3D cell colonization while culturing skeletal muscle C2C12 cells. They further induce good actin fiber alignment along the compliant PyC construction. However, no conclusive myogenic differentiation is observed here. Nevertheless, these results are highly promising for architected PyC scaffolds as multifunctional tissue implants and encourage more investigations in employing compliant architected PyC structures for high-performance tissue engineering applications.
Collapse
Affiliation(s)
- Mohammadreza Taale
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityIm Neuenheimer Feld 22569120HeidelbergGermany
| | - Barbara Schamberger
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityIm Neuenheimer Feld 22569120HeidelbergGermany
| | | | - Christian Dolle
- Microscopy of Nanoscale Structures and Mechanisms (MNM)Laboratory for Electron Microscopy (LEM)Karlsruhe Institute of TechnologyEngesserstr. 7D‐76131KarlsruheGermany
| | - Fereydoon Taheri
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityIm Neuenheimer Feld 22569120HeidelbergGermany
| | - Dario Mager
- Institute of Microstructure TechnologyKarlsruhe Institute of TechnologyHermann‐von‐Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
| | - Yolita M. Eggeler
- Microscopy of Nanoscale Structures and Mechanisms (MNM)Laboratory for Electron Microscopy (LEM)Karlsruhe Institute of TechnologyEngesserstr. 7D‐76131KarlsruheGermany
| | - Jan G. Korvink
- Institute of Microstructure TechnologyKarlsruhe Institute of TechnologyHermann‐von‐Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
| | - Jon M. Molina‐Aldareguia
- IMDEA Materials InstituteEric Kandel, 2Getafe28906Spain
- Department of Mechanical EngineeringUniversidad Politécnica de MadridJosé Gutierréz Abascal, 2Madrid28006Spain
| | - Christine Selhuber‐Unkel
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityIm Neuenheimer Feld 22569120HeidelbergGermany
| | - Andrés Díaz Lantada
- Department of Mechanical EngineeringUniversidad Politécnica de MadridJosé Gutierréz Abascal, 2Madrid28006Spain
| | - Monsur Islam
- IMDEA Materials InstituteEric Kandel, 2Getafe28906Spain
- Institute of Microstructure TechnologyKarlsruhe Institute of TechnologyHermann‐von‐Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
| |
Collapse
|
24
|
Spedicati M, Zoso A, Mortati L, Chiono V, Marcello E, Carmagnola I. Three-Dimensional Microfibrous Scaffold with Aligned Topography Produced via a Combination of Melt-Extrusion Additive Manufacturing and Porogen Leaching for In Vitro Skeletal Muscle Modeling. Bioengineering (Basel) 2024; 11:332. [PMID: 38671754 PMCID: PMC11047940 DOI: 10.3390/bioengineering11040332] [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: 12/30/2023] [Revised: 03/13/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
Skeletal muscle tissue (SMT) has a highly hierarchical and anisotropic morphology, featuring aligned and parallel structures at multiple levels. Various factors, including trauma and disease conditions, can compromise the functionality of skeletal muscle. The in vitro modeling of SMT represents a useful tool for testing novel drugs and therapies. The successful replication of SMT native morphology demands scaffolds with an aligned anisotropic 3D architecture. In this work, a 3D PCL fibrous scaffold with aligned morphology was developed through the synergistic combination of Melt-Extrusion Additive Manufacturing (MEAM) and porogen leaching, utilizing PCL as the bulk material and PEG as the porogen. PCL/PEG blends with different polymer ratios (60/40, 50/50, 40/60) were produced and characterized through a DSC analysis. The MEAM process parameters and porogen leaching in bi-distilled water allowed for the development of a micrometric anisotropic fibrous structure with fiber diameters ranging from 10 to 100 µm, depending on PCL/PEG blend ratios. The fibrous scaffolds were coated with Gelatin type A to achieve a biomimetic coating for an in vitro cell culture and mechanically characterized via AFM. The 40/60 PCL/PEG scaffolds yielded the most homogeneous and smallest fibers and the greatest physiological stiffness. In vitro cell culture studies were performed by seeding C2C12 cells onto a selected scaffold, enabling their attachment, alignment, and myotube formation along the PCL fibers during a 14-day culture period. The resultant anisotropic scaffold morphology promoted SMT-like cell conformation, establishing a versatile platform for developing in vitro models of tissues with anisotropic morphology.
Collapse
Affiliation(s)
- Mattia Spedicati
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Torino, Italy; (M.S.); (A.Z.); (V.C.)
- POLITO BioMedLab, Politecnico di Torino, 10129 Torino, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, 56122 Pisa, Italy
| | - Alice Zoso
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Torino, Italy; (M.S.); (A.Z.); (V.C.)
- POLITO BioMedLab, Politecnico di Torino, 10129 Torino, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, 56122 Pisa, Italy
| | - Leonardo Mortati
- Istituto Nazionale di Ricerca Metrologica (INRIM), 10135 Torino, Italy;
| | - Valeria Chiono
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Torino, Italy; (M.S.); (A.Z.); (V.C.)
- POLITO BioMedLab, Politecnico di Torino, 10129 Torino, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, 56122 Pisa, Italy
| | - Elena Marcello
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Torino, Italy; (M.S.); (A.Z.); (V.C.)
- POLITO BioMedLab, Politecnico di Torino, 10129 Torino, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, 56122 Pisa, Italy
| | - Irene Carmagnola
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Torino, Italy; (M.S.); (A.Z.); (V.C.)
- POLITO BioMedLab, Politecnico di Torino, 10129 Torino, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, 56122 Pisa, Italy
| |
Collapse
|
25
|
Slepičková Kasálková N, Juřicová V, Fajstavr D, Frýdlová B, Rimpelová S, Švorčík V, Slepička P. Plasma-Activated Polydimethylsiloxane Microstructured Pattern with Collagen for Improved Myoblast Cell Guidance. Int J Mol Sci 2024; 25:2779. [PMID: 38474025 DOI: 10.3390/ijms25052779] [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: 01/27/2024] [Revised: 02/22/2024] [Accepted: 02/25/2024] [Indexed: 03/14/2024] Open
Abstract
We focused on polydimethylsiloxane (PDMS) as a substrate for replication, micropatterning, and construction of biologically active surfaces. The novelty of this study is based on the combination of the argon plasma exposure of a micropatterned PDMS scaffold, where the plasma served as a strong tool for subsequent grafting of collagen coatings and their application as cell growth scaffolds, where the standard was significantly exceeded. As part of the scaffold design, templates with a patterned microstructure of different dimensions (50 × 50, 50 × 20, and 30 × 30 μm2) were created by photolithography followed by pattern replication on a PDMS polymer substrate. Subsequently, the prepared microstructured PDMS replicas were coated with a type I collagen layer. The sample preparation was followed by the characterization of material surface properties using various analytical techniques, including scanning electron microscopy (SEM), energy-dispersive X-ray spectroscopy (EDS), and X-ray photoelectron spectroscopy (XPS). To evaluate the biocompatibility of the produced samples, we conducted studies on the interactions between selected polymer replicas and micro- and nanostructures and mammalian cells. Specifically, we utilized mouse myoblasts (C2C12), and our results demonstrate that we achieved excellent cell alignment in conjunction with the development of a cytocompatible surface. Consequently, the outcomes of this research contribute to an enhanced comprehension of surface properties and interactions between structured polymers and mammalian cells. The use of periodic microstructures has the potential to advance the creation of novel materials and scaffolds in tissue engineering. These materials exhibit exceptional biocompatibility and possess the capacity to promote cell adhesion and growth.
Collapse
Affiliation(s)
- Nikola Slepičková Kasálková
- Department of Solid State Engineering, The University of Chemistry and Technology Prague, 166 28 Prague, Czech Republic
| | - Veronika Juřicová
- Department of Solid State Engineering, The University of Chemistry and Technology Prague, 166 28 Prague, Czech Republic
| | - Dominik Fajstavr
- Department of Solid State Engineering, The University of Chemistry and Technology Prague, 166 28 Prague, Czech Republic
| | - Bára Frýdlová
- Department of Solid State Engineering, The University of Chemistry and Technology Prague, 166 28 Prague, Czech Republic
| | - Silvie Rimpelová
- Department of Biochemistry and Microbiology, The University of Chemistry and Technology Prague, 166 28 Prague, Czech Republic
| | - Václav Švorčík
- Department of Solid State Engineering, The University of Chemistry and Technology Prague, 166 28 Prague, Czech Republic
| | - Petr Slepička
- Department of Solid State Engineering, The University of Chemistry and Technology Prague, 166 28 Prague, Czech Republic
| |
Collapse
|
26
|
Kang MS, Yu Y, Park R, Heo HJ, Lee SH, Hong SW, Kim YH, Han DW. Highly Aligned Ternary Nanofiber Matrices Loaded with MXene Expedite Regeneration of Volumetric Muscle Loss. NANO-MICRO LETTERS 2024; 16:73. [PMID: 38175358 PMCID: PMC10767178 DOI: 10.1007/s40820-023-01293-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/16/2023] [Indexed: 01/05/2024]
Abstract
Current therapeutic approaches for volumetric muscle loss (VML) face challenges due to limited graft availability and insufficient bioactivities. To overcome these limitations, tissue-engineered scaffolds have emerged as a promising alternative. In this study, we developed aligned ternary nanofibrous matrices comprised of poly(lactide-co-ε-caprolactone) integrated with collagen and Ti3C2Tx MXene nanoparticles (NPs) (PCM matrices), and explored their myogenic potential for skeletal muscle tissue regeneration. The PCM matrices demonstrated favorable physicochemical properties, including structural uniformity, alignment, microporosity, and hydrophilicity. In vitro assays revealed that the PCM matrices promoted cellular behaviors and myogenic differentiation of C2C12 myoblasts. Moreover, in vivo experiments demonstrated enhanced muscle remodeling and recovery in mice treated with PCM matrices following VML injury. Mechanistic insights from next-generation sequencing revealed that MXene NPs facilitated protein and ion availability within PCM matrices, leading to elevated intracellular Ca2+ levels in myoblasts through the activation of inducible nitric oxide synthase (iNOS) and serum/glucocorticoid regulated kinase 1 (SGK1), ultimately promoting myogenic differentiation via the mTOR-AKT pathway. Additionally, upregulated iNOS and increased NO- contributed to myoblast proliferation and fiber fusion, thereby facilitating overall myoblast maturation. These findings underscore the potential of MXene NPs loaded within highly aligned matrices as therapeutic agents to promote skeletal muscle tissue recovery.
Collapse
Affiliation(s)
- Moon Sung Kang
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea
| | - Yeuni Yu
- Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Rowoon Park
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea
| | - Hye Jin Heo
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Seok Hyun Lee
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea
- Osstem Implant Inc., Seoul, 07789, Republic of Korea
| | - Suck Won Hong
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea.
- Engineering Research Center for Color‑Modulated Extra‑Sensory Perception Technology, Pusan National University, Busan, 46241, Republic of Korea.
| | - Yun Hak Kim
- Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea.
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea.
- Periodontal Disease Signaling Network Research Center and Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea.
| | - Dong-Wook Han
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea.
- BIO-IT Fusion Technology Research Institute, Pusan National University, Busan, 46241, Republic of Korea.
| |
Collapse
|
27
|
Alam AMMN, Kim CJ, Kim SH, Kumari S, Lee SY, Hwang YH, Joo ST. Trends in Hybrid Cultured Meat Manufacturing Technology to Improve Sensory Characteristics. Food Sci Anim Resour 2024; 44:39-50. [PMID: 38229861 PMCID: PMC10789553 DOI: 10.5851/kosfa.2023.e76] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/26/2023] [Accepted: 11/20/2023] [Indexed: 01/18/2024] Open
Abstract
The projected growth of global meat production over the next decade is attributed to rising income levels and population expansion. One potentially more pragmatic approach to mitigating the adverse externalities associated with meat production involves implementing alterations to the production process, such as transitioning to cultured meat, hybrid cultured meat, and meat alternatives. Cultured meat (CM) is derived from animal stem cells and undergoes a growth and division process that closely resembles the natural in vivo cellular development. CM is emerging as a widely embraced substitute for traditional protein sources, with the potential to alleviate the future strain on animal-derived meat production. To date, the primary emphasis of cultured meat research and production has predominantly been around the ecological advantages and ethical considerations pertaining to animal welfare. However, there exists substantial study potential in exploring consumer preferences with respect to the texture, color, cuts, and sustainable methodologies associated with cultured meat. The potential augmentation of cultured meat's acceptance could be facilitated through the advancement of a wider range of cuts to mimic real muscle fibers. This review examines the prospective commercial trends of hybrid cultured meat. Subsequently, the present state of research pertaining to the advancement of scaffolding, coloration, and muscle fiber development in hybrid cultured meat, encompassing plant-based alternatives designed to emulate authentic meat, has been deliberated. However, this discussion highlights the obstacles that have arisen in current procedures and proposes future research directions for the development of sustainable cultured meat and meat alternatives, such as plant-based meat production.
Collapse
Affiliation(s)
- AMM Nurul Alam
- Division of Applied Life Science (BK21
Four), Gyeongsang National University, Jinju 52828,
Korea
| | - Chan-Jin Kim
- Division of Applied Life Science (BK21
Four), Gyeongsang National University, Jinju 52828,
Korea
| | - So-Hee Kim
- Division of Applied Life Science (BK21
Four), Gyeongsang National University, Jinju 52828,
Korea
| | - Swati Kumari
- Division of Applied Life Science (BK21
Four), Gyeongsang National University, Jinju 52828,
Korea
| | - Seung-Yun Lee
- Division of Animal Science, Gyeongsang
National University, Jinju 52828, Korea
| | - Young-Hwa Hwang
- Institute of Agriculture & Life
Science, Gyeongsang National University, Jinju 52828,
Korea
| | - Seon-Tea Joo
- Division of Applied Life Science (BK21
Four), Gyeongsang National University, Jinju 52828,
Korea
- Division of Animal Science, Gyeongsang
National University, Jinju 52828, Korea
- Institute of Agriculture & Life
Science, Gyeongsang National University, Jinju 52828,
Korea
| |
Collapse
|
28
|
Taborda M, Catalan KN, Orellana N, Bezjak D, Enrione J, Acevedo CA, Corrales TP. Micropatterned Nanofiber Scaffolds of Salmon Gelatin, Chitosan, and Poly(vinyl alcohol) for Muscle Tissue Engineering. ACS OMEGA 2023; 8:47883-47896. [PMID: 38144088 PMCID: PMC10733945 DOI: 10.1021/acsomega.3c06436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 12/26/2023]
Abstract
The development of scaffolds that mimic the aligned fibrous texture of the extracellular matrix has become an important requirement in muscle tissue engineering. Electrospinning is a widely used technique to fabricate biomimetic scaffolds. Therefore, a biopolymer blend composed of salmon gelatin (SG), chitosan (Ch), and poly(vinyl alcohol) (PVA) was developed by electrospinning onto a micropatterned (MP) collector, resulting in a biomimetic scaffold for seeding muscle cells. Rheology and surface tension studies were performed to determine the optimum solution concentration and viscosity for electrospinning. The scaffold microstructure was analyzed using SEM to determine the nanofiber's diameter and orientation. Blends of SG/Ch/PVA exhibited better electrospinnability and handling properties than pure PVA. The resulting scaffolds consist of a porous surface (∼46%), composed of a random fiber distribution, for a flat collector and scaffolds with regions of aligned nanofibers for the MP collector. The nanofiber diameters are 141 ± 2 and 151 ± 2 nm for the flat and MP collector, respectively. In vitro studies showed that myoblasts cultured on scaffold SG/Ch/PVA presented a high rate of cell growth. Furthermore, the aligned nanofibers on the SG/Ch/PVA scaffold provide a suitable platform for myoblast alignment.
Collapse
Affiliation(s)
- María
I. Taborda
- Centro
de Biotecnología, Universidad Técnica
Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile
- Programa
de doctorado en Biotecnología, Pontificia
Universidad Católica de Valparaíso−Universidad
Técnica Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile
| | - Karina N. Catalan
- Departamento
de Física, Universidad Técnica
Federico Santa María, Av. España 1680, Valparaíso 2340000, Chile
| | - Nicole Orellana
- Centro
de Biotecnología, Universidad Técnica
Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile
| | - Dragica Bezjak
- Centro
de Biotecnología, Universidad Técnica
Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile
- Programa
de doctorado en Biotecnología, Pontificia
Universidad Católica de Valparaíso−Universidad
Técnica Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile
| | - Javier Enrione
- Escuela
de Nutrición y Dietética, 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
- Departamento
de Física, Universidad Técnica
Federico Santa María, Av. España 1680, Valparaíso 2340000, Chile
- Centro
Científico Tecnológico de Valparaíso (CCTVAL), Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile
| | - Tomas P. Corrales
- Centro
de Biotecnología, Universidad Técnica
Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile
- Departamento
de Física, Universidad Técnica
Federico Santa María, Av. España 1680, Valparaíso 2340000, Chile
- Millenium
Nucleus in NanoBioPhysics (NNBP), Valparaíso 2340000, Chile
| |
Collapse
|
29
|
Stephens DC, Mungai M, Crabtree A, Beasley HK, Garza-Lopez E, Vang L, Neikirk K, Vue Z, Vue N, Marshall AG, Turner K, Shao JQ, Sarker B, Murray S, Gaddy JA, Davis J, Damo SM, Hinton AO. Protocol for isolating mice skeletal muscle myoblasts and myotubes via differential antibody validation. STAR Protoc 2023; 4:102591. [PMID: 37938976 PMCID: PMC10663959 DOI: 10.1016/j.xpro.2023.102591] [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: 05/22/2023] [Revised: 08/06/2023] [Accepted: 09/01/2023] [Indexed: 11/10/2023] Open
Abstract
Isolation of skeletal muscles allows for the exploration of many complex diseases. Here, we present a protocol for isolating mice skeletal muscle myoblasts and myotubes that have been differentiated through antibody validation. We describe steps for collecting and preparing murine skeletal tissue, myoblast cell maintenance, plating, and cell differentiation. We then detail procedures for cell incubation, immunostaining, slide preparation and storage, and imaging for immunofluorescence validation.
Collapse
Affiliation(s)
- Dominique C Stephens
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA; Department of Life and Physical Sciences, Fisk University, Nashville, TN 37232, USA
| | - Margaret Mungai
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Amber Crabtree
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Heather K Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Edgar Garza-Lopez
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Neng Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Andrea G Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Kyrin Turner
- Department of Life and Physical Sciences, Fisk University, Nashville, TN 37232, USA
| | - Jian-Qiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA 52242, USA
| | - Bishnu Sarker
- School of Applied Computational Sciences, Meharry Medical College, Nashville, TN 37232, USA
| | - Sandra Murray
- Department of Cell Biology, College of Medicine, University of Pittsburgh, Pittsburgh, TN 15260, USA
| | - Jennifer A Gaddy
- Division of Infectious Diseases, Vanderbilt University School of Medicine, Nashville, TN, USA; Tennessee Valley Healthcare Systems, U.S. Department of Veterans Affairs, Nashville, TN, USA
| | - Jamaine Davis
- Department of Biochemistry and Cancer Biology. Meharry Medical College, Nashville, TN, USA
| | - Steven M Damo
- Department of Life and Physical Sciences, Fisk University, Nashville, TN 37232, USA.
| | - Antentor O Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
30
|
Zhang YC, Yang YX, Liu Y, Liu XJ, Dai JH, Gao RS, Hu YY, Fei WY. Combining Porous Se@SiO 2 Nanocomposites and dECM Enhances the Myogenic Differentiation of Adipose-Derived Stem Cells. Int J Nanomedicine 2023; 18:7661-7676. [PMID: 38111844 PMCID: PMC10726970 DOI: 10.2147/ijn.s436081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 12/05/2023] [Indexed: 12/20/2023] Open
Abstract
Background Volumetric Muscle Loss (VML) denotes the traumatic loss of skeletal muscle, a condition that can result in chronic functional impairment and even disability. While the body can naturally repair injured skeletal muscle within a limited scope, patients experiencing local and severe muscle loss due to VML surpass the compensatory capacity of the muscle itself. Currently, clinical treatments for VML are constrained and demonstrate minimal efficacy. Selenium, a recognized antioxidant, plays a crucial role in regulating cell differentiation, anti-inflammatory responses, and various other physiological functions. Methods We engineered a porous Se@SiO2 nanocomposite (SeNPs) with the purpose of releasing selenium continuously and gradually. This nanocomposite was subsequently combined with a decellularized extracellular matrix (dECM) to explore their collaborative protective and stimulatory effects on the myogenic differentiation of adipose-derived mesenchymal stem cells (ADSCs). The influence of dECM and NPs on the myogenic level, reactive oxygen species (ROS) production, and mitochondrial respiratory chain (MRC) activity of ADSCs was evaluated using Western Blot, ELISA, and Immunofluorescence assay. Results Our findings demonstrate that the concurrent application of SeNPs and dECM effectively mitigates the apoptosis and intracellular ROS levels in ADSCs. Furthermore, the combination of dECM with SeNPs significantly upregulated the expression of key myogenic markers, including MYOD, MYOG, Desmin, and myosin heavy chain in ADSCs. Notably, this combination also led to an increase in both the number of mitochondria and the respiratory chain activity in ADSCs. Conclusion The concurrent application of SeNPs and dECM effectively diminishes ROS production, boosts mitochondrial function, and stimulates the myogenic differentiation of ADSCs. This study lays the groundwork for future treatments of VML utilizing the combination of SeNPs and dECM.
Collapse
Affiliation(s)
- Yu-Cheng Zhang
- Clinical Medical College, Dalian Medical University, Dalian, 116044, People’s Republic of China
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People’s Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, People’s Republic of China
| | - Yu-Xia Yang
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People’s Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, People’s Republic of China
- Clinical Medical College, Yangzhou University, Yangzhou, 225001, People’s Republic of China
| | - Yu Liu
- Department of Orthopedics, Wuxi Ninth People’s Hospital Affiliated to Soochow University, Wuxi, 214062, People’s Republic of China
| | - Xi-Jian Liu
- School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, People’s Republic of China
| | - Ji-Hang Dai
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People’s Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, People’s Republic of China
| | - Rang-Shan Gao
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People’s Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, People’s Republic of China
| | - Yang-Yang Hu
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People’s Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, People’s Republic of China
| | - Wen-Yong Fei
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People’s Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, People’s Republic of China
| |
Collapse
|
31
|
de Barros NR, Darabi MA, Ma X, Diltemiz SE, Ermis M, Hassani Najafabasi A, Nadine S, Banton EA, Mandal K, Abbasgholizadeh R, Falcone N, Mano JF, Nasiri R, Herculano RD, Zhu Y, Ostrovidov S, Lee J, Kim HJ, Hosseini V, Dokmeci MR, Ahadian S, Khademhosseini A. Enhanced Maturation of 3D Bioprinted Skeletal Muscle Tissue Constructs Encapsulating Soluble Factor-Releasing Microparticles. Macromol Biosci 2023; 23:e2300276. [PMID: 37534566 PMCID: PMC10837326 DOI: 10.1002/mabi.202300276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Indexed: 08/04/2023]
Abstract
Several microfabrication technologies have been used to engineer native-like skeletal muscle tissues. However, the successful development of muscle remains a significant challenge in the tissue engineering field. Muscle tissue engineering aims to combine muscle precursor cells aligned within a highly organized 3D structure and biological factors crucial to support cell differentiation and maturation into functional myotubes and myofibers. In this study, the use of 3D bioprinting is proposed for the fabrication of muscle tissues using gelatin methacryloyl (GelMA) incorporating sustained insulin-like growth factor-1 (IGF-1)-releasing microparticles and myoblast cells. This study hypothesizes that functional and mature myotubes will be obtained more efficiently using a bioink that can release IGF-1 sustainably for in vitro muscle engineering. Synthesized microfluidic-assisted polymeric microparticles demonstrate successful adsorption of IGF-1 and sustained release of IGF-1 at physiological pH for at least 21 days. Incorporating the IGF-1-releasing microparticles in the GelMA bioink assisted in promoting the alignment of myoblasts and differentiation into myotubes. Furthermore, the myotubes show spontaneous contraction in the muscle constructs bioprinted with IGF-1-releasing bioink. The proposed bioprinting strategy aims to improve the development of new therapies applied to the regeneration and maturation of muscle tissues.
Collapse
Affiliation(s)
| | - Mohammad Ali Darabi
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Xin Ma
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Sibel Emir Diltemiz
- Department of Chemistry, Eskisehir Technical University, Eskisehir, 26470, Turkey
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | | | - Sara Nadine
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
- Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Ethan A. Banton
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | | | - Natashya Falcone
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - João F. Mano
- Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Rohollah Nasiri
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | | | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Serge Ostrovidov
- Department of Radiological Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Junmin Lee
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Han-Jun Kim
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Vahid Hosseini
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Mehmet R. Dokmeci
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Samad Ahadian
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Radiological Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
32
|
Melzener L, Spaans S, Hauck N, Pötgens AJG, Flack JE, Post MJ, Doğan A. Short-Stranded Zein Fibers for Muscle Tissue Engineering in Alginate-Based Composite Hydrogels. Gels 2023; 9:914. [PMID: 37999004 PMCID: PMC10671123 DOI: 10.3390/gels9110914] [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: 10/14/2023] [Revised: 11/04/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023] Open
Abstract
Cultivated meat is a nascent technology that aims to create an environmentally and animal-friendly alternative to conventional meat. Producing skeletal muscle tissue in an animal-free system allowing for high levels of myofusion and maturation is important for the nutritional and sensorial value of cultivated meat. Alginate is an attractive biomaterial to support muscle formation as it is food-safe, sustainable and cheap and can be crosslinked using non-toxic methods. Although alginate can be functionalized to promote cell attachment, limitations in its mechanical properties, including form, viscosity, and stress relaxation, hinder the cellular capacity for myogenic differentiation and maturation in alginate-based hydrogels. Here, we show that the addition of electrospun short-stranded zein fibers increased hydrogel degradation, resulting in faster compaction, improved cell-gel interaction, and enhanced alignment of bovine muscle precursor cells. We conclude that fiber-hydrogel composites are a promising approach to support optimal formation of 3D constructs, by improving tissue stability and thus prolonging culture duration. Together, this improves muscle-related protein content by facilitating myogenic differentiation and priming muscle organoids for maturation.
Collapse
Affiliation(s)
- Lea Melzener
- Department of Physiology, Maastricht University, 6200 MD Maastricht, The Netherlands; (L.M.); (M.J.P.)
- Mosa Meat B.V., 6229 PM Maastricht, The Netherlands; (S.S.); (A.J.G.P.); (J.E.F.)
| | - Sergio Spaans
- Mosa Meat B.V., 6229 PM Maastricht, The Netherlands; (S.S.); (A.J.G.P.); (J.E.F.)
| | - Nicolas Hauck
- Mosa Meat B.V., 6229 PM Maastricht, The Netherlands; (S.S.); (A.J.G.P.); (J.E.F.)
| | - André J. G. Pötgens
- Mosa Meat B.V., 6229 PM Maastricht, The Netherlands; (S.S.); (A.J.G.P.); (J.E.F.)
| | - Joshua E. Flack
- Mosa Meat B.V., 6229 PM Maastricht, The Netherlands; (S.S.); (A.J.G.P.); (J.E.F.)
| | - Mark J. Post
- Department of Physiology, Maastricht University, 6200 MD Maastricht, The Netherlands; (L.M.); (M.J.P.)
- Mosa Meat B.V., 6229 PM Maastricht, The Netherlands; (S.S.); (A.J.G.P.); (J.E.F.)
| | - Arın Doğan
- Mosa Meat B.V., 6229 PM Maastricht, The Netherlands; (S.S.); (A.J.G.P.); (J.E.F.)
| |
Collapse
|
33
|
Philips C, Terrie L, Muylle E, Van Ginderdeuren R, Vereecke E, Mombaerts I, Thorrez L. The Immunoarchitecture of Human Extraocular Muscles. Invest Ophthalmol Vis Sci 2023; 64:23. [PMID: 37975851 PMCID: PMC10664723 DOI: 10.1167/iovs.64.14.23] [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: 08/14/2023] [Accepted: 10/22/2023] [Indexed: 11/19/2023] Open
Abstract
Purpose The purpose of this study was to describe the immunoarchitecture of normal extraocular muscles (EOMs) in terms of presence, distribution, and organization of various immune cells. Methods We performed unilateral orbital exenterations in six fresh human cadavers from elderly patients, followed by dissection of the medial, lateral, superior and inferior rectus, superior and inferior oblique, and superior palpebral levator muscle in their entirety. We further cross sectioned each EOM in an anterior, central, and posterior third. After immunohistochemical staining for CD3, CD8, CD20, CD138, CD68, and podoplanin, quantitative analysis was performed. Results We found all EOMs (rectus, oblique, and levator muscles) to harbor both T- and B-lymphocytes, with a B-lymphocyte dominance and an absence of plasma cells. The highest prevalence of immune cells was seen in the muscle bellies, with, on average, 488 ± 63 CD3+ T-lymphocytes and 44 ± 110 CD20+ B-lymphocytes per mm2, and significant differences from the anterior (T-lymphocytes) and posterior (T- and B-lymphocytes) thirds. T- and B-lymphocytes were primarily organized in hotspots in the vicinity of blood vessels. In addition, a small resident population of macrophages scattered throughout the specimens was detected. No lymphatic vessels were found in any of the EOMs. Conclusions These findings can serve as a reference dataset in the assessment of EOM biopsies in the diagnostic process of inflammatory orbital and systemic disorders. Moreover, from a regenerative perspective, our results highlight the importance of taking into account the presence of a resident immune cell population when studying the host immune response on transplanted tissues or engineered constructs.
Collapse
Affiliation(s)
- Charlot Philips
- Tissue Engineering Lab, Department of Development and Regeneration, Catholic University Leuven campus Kulak, Kortrijk, Belgium
| | - Lisanne Terrie
- Tissue Engineering Lab, Department of Development and Regeneration, Catholic University Leuven campus Kulak, Kortrijk, Belgium
| | - Ewout Muylle
- Tissue Engineering Lab, Department of Development and Regeneration, Catholic University Leuven campus Kulak, Kortrijk, Belgium
| | | | - Evie Vereecke
- Jan Palfijn Anatomy Lab, Department of Development and Regeneration, Catholic University Leuven campus Kulak, Kortrijk, Belgium
| | - Ilse Mombaerts
- Department of Ophthalmology, University Hospitals Leuven, Leuven, Belgium
- Department of Neurosciences, Catholic University Leuven, Leuven, Belgium
| | - Lieven Thorrez
- Tissue Engineering Lab, Department of Development and Regeneration, Catholic University Leuven campus Kulak, Kortrijk, Belgium
| |
Collapse
|
34
|
Brooks AK, Ramsey RG, Zhang N, Yadavalli VK. Tunable Light-Actuated Interpenetrating Networks of Silk Fibroin and Gelatin for Tissue Engineering and Flexible Biodevices. ACS Biomater Sci Eng 2023; 9:5793-5803. [PMID: 37698556 DOI: 10.1021/acsbiomaterials.3c00741] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
Soft materials with tunable properties are valuable for applications such as tissue engineering, electronic skins, and human-machine interfaces. Materials that are nature-derived offer additional advantages such as biocompatibility, biodegradability, low-cost sourcing, and sustainability. However, these materials often have contrasting properties that limit their use. For example, silk fibroin (SF) has high mechanical strength but lacks processability and cell-adhesive domains. Gelatin, derived from collagen, has excellent biological properties, but is fragile and lacks stability. To overcome these limitations, composites of gelatin and SF have been explored. However, mechanically robust self-supported matrices and electrochemically active or micropatterned substrates were not demonstrated. In this study, we present a composite of photopolymerizable SF and photogelatin, termed photofibrogel (PFG). By incorporating photoreactive properties in both SF and gelatin, control over material properties can be achieved. The PFG composite can be easily and rapidly formed into free-standing, high-resolution architectures with tunable properties. By optimizing the ratio of SF to gelatin, properties such as swelling, mechanical behavior, enzymatic degradation, and patternability are tailored. The PFG composite allows for macroscale and microscale patterning without significant swelling, enabling the fabrication of structures using photolithography and laser cutting techniques. PFG can be patterned with electrically conductive materials, making it suitable for cell guidance and stimulation. The versatility, mechanical robustness, bioactivity, and electrochemical properties of PFG are shown for skeletal muscle tissue engineering using C2C12 cells as a model. Overall, such composite biomaterials with tunable properties have broad potential in flexible bioelectronics, wound healing, regenerative medicine, and food systems.
Collapse
|
35
|
Xiong F, Wei S, Wu S, Jiang W, Li B, Xuan H, Xue Y, Yuan H. Aligned Electroactive Electrospun Fibrous Scaffolds for Peripheral Nerve Regeneration. ACS APPLIED MATERIALS & INTERFACES 2023; 15:41385-41402. [PMID: 37606339 DOI: 10.1021/acsami.3c09237] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
Effective repair and functional recovery of large peripheral nerve deficits are urgent clinical needs. A biofunctional electroactive scaffold typically acts as a "bridge" for the repair of large nerve defects. In this study, we constructed a biomimetic piezoelectric and conductive aligned polypyrrole (PPy)/polydopamine (PDA)/poly-l-lactic acid (PLLA) electrospun fibrous scaffold to improve the hydrophilicity and cellular compatibility of PLLA and restore the weakened piezoelectric effect of PDA, which is beneficial in promoting Schwann cell differentiation and dorsal root ganglion neuronal extension and alignment. The aligned PPy/PDA/PLLA fibrous scaffold bridged the sciatic nerve of Sprague-Dawley rats with a 10 mm deficit, prevented autotomy, and promoted nerve regeneration and functional recovery, thereby activating the calcium and AMP-activated protein kinase signaling pathways. Therefore, electroactive fibrous scaffolds exhibit great potential for neural tissue regeneration.
Collapse
Affiliation(s)
- Feng Xiong
- School of Life Sciences, Nantong University, 226019 Nantong, China
| | - Shuo Wei
- School of Life Sciences, Nantong University, 226019 Nantong, China
| | - Shuyuan Wu
- School of Life Sciences, Nantong University, 226019 Nantong, China
| | - Wei Jiang
- School of Life Sciences, Nantong University, 226019 Nantong, China
| | - Biyun Li
- School of Life Sciences, Nantong University, 226019 Nantong, China
| | - Hongyun Xuan
- School of Life Sciences, Nantong University, 226019 Nantong, China
| | - Ye Xue
- School of Life Sciences, Nantong University, 226019 Nantong, China
| | - Huihua Yuan
- School of Life Sciences, Nantong University, 226019 Nantong, China
| |
Collapse
|
36
|
Wang Z, Chen T, Li X, Guo B, Liu P, Zhu Z, Xu RX. Oxygen-releasing biomaterials for regenerative medicine. J Mater Chem B 2023; 11:7300-7320. [PMID: 37427691 DOI: 10.1039/d3tb00670k] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Oxygen is critical to the survival, function and fate of mammalian cells. Oxygen tension controls cellular behavior through metabolic programming, which in turn controls tissue regeneration. A variety of biomaterials with oxygen-releasing capabilities have been developed to provide oxygen supply to ensure cell survival and differentiation for therapeutic efficacy, and to prevent hypoxia-induced tissue damage and cell death. However, controlling the oxygen release with spatial and temporal accuracy is still technically challenging. In this review, we provide a comprehensive overview of organic and inorganic materials available as oxygen sources, including hemoglobin-based oxygen carriers (HBOCs), perfluorocarbons (PFCs), photosynthetic organisms, solid and liquid peroxides, and some of the latest materials such as metal-organic frameworks (MOFs). Additionally, we introduce the corresponding carrier materials and the oxygen production methods and present state-of-the-art applications and breakthroughs of oxygen-releasing materials. Furthermore, we discuss the current challenges and the future perspectives in the field. After reviewing the recent progress and the future perspectives of oxygen-releasing materials, we predict that smart material systems that combine precise detection of oxygenation and adaptive control of oxygen delivery will be the future trend for oxygen-releasing materials in regenerative medicine.
Collapse
Affiliation(s)
- Zhaojun Wang
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215000, China.
| | - Tianao Chen
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230026, China.
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xin Li
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215000, China.
| | - Buyun Guo
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230026, China.
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Peng Liu
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215000, China.
| | - Zhiqiang Zhu
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230026, China.
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Ronald X Xu
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215000, China.
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230026, China.
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
37
|
Chen Y, Li L, Chen L, Shao W, Chen X, Fan X, Liu Y, Ding S, Xu X, Zhou G, Feng X. Gellan gum-gelatin scaffolds with Ca 2+ crosslinking for constructing a structured cell cultured meat model. Biomaterials 2023; 299:122176. [PMID: 37253307 DOI: 10.1016/j.biomaterials.2023.122176] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 05/03/2023] [Accepted: 05/20/2023] [Indexed: 06/01/2023]
Abstract
As an emerging technology to obtain protein by culturing animal-derived cells in vitro, it is crucial to construct 3D edible scaffolds to prepare structured cell cultured meat products. In this study, a scaffold based on gellan gum (GG)-gelatin (Gel) was prepared and further cross-linked with Ca2+. FTIR confirmed the electrostatic interaction between GG and Gel and the ionic cross-linking of Ca2+ and carboxyl groups, and SEM images showed the porous structure of the scaffolds. The staining results showed that scaffolds with high concentrations of Ca2+ had higher biocompatibility than scaffolds with low concentrations of Ca2+ and non-crosslinked scaffolds, and scaffolds Ca2+-GG2-Gel3-0.5 adhered to more cells and were more conducive to cell spreading. The immunofluorescence staining, SEM images, Western blot, and RT-qPCR showed that the scaffolds supported the proliferation and myogenic differentiation of chicken skeletal muscle satellite cells (CSMSCs) and myotubes were formed on the scaffolds. Finally, the scaffolds were stained and fried after culturing. The results of the textural and chromatic analysis showed that the texture and color of the scaffolds were similar to fresh meat and meat products. These results showed that ionically crosslinked GG-Gel scaffolds are biocompatible and stable for structured cell cultured meat models.
Collapse
Affiliation(s)
- Yan Chen
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi, 712100, China
| | - Linzi Li
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi, 712100, China
| | - Lin Chen
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi, 712100, China
| | - Wei Shao
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi, 712100, China
| | - Xiaohong Chen
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi, 712100, China
| | - Xiaojing Fan
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi, 712100, China
| | - Yaping Liu
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi, 712100, China
| | - Shijie Ding
- Lab of Meat Processing and Quality Control of EDU, College of Food Science and Technology, Synergetic Innovation Center of Food Safety and Nutrition, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Xinglian Xu
- Lab of Meat Processing and Quality Control of EDU, College of Food Science and Technology, Synergetic Innovation Center of Food Safety and Nutrition, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Guanghong Zhou
- Lab of Meat Processing and Quality Control of EDU, College of Food Science and Technology, Synergetic Innovation Center of Food Safety and Nutrition, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Xianchao Feng
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
38
|
Lee DK, Kim M, Jeong J, Lee YS, Yoon JW, An MJ, Jung HY, Kim CH, Ahn Y, Choi KH, Jo C, Lee CK. Unlocking the potential of stem cells: Their crucial role in the production of cultivated meat. Curr Res Food Sci 2023; 7:100551. [PMID: 37575132 PMCID: PMC10412782 DOI: 10.1016/j.crfs.2023.100551] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/05/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
Cellular agriculture is an emerging research field of agribiotechnology that aims to produce agricultural products using stem cells, without sacrificing animals or cultivating crops. Cultivated meat, as a representative cellular product of cellular agriculture, is being actively researched due to global food insecurity, environmental, and ethical concerns. This review focuses on the application of stem cells, which are the seeds of cellular agriculture, for the production of cultivated meat, with emphasis on deriving and culturing muscle and adipose stem cells for imitating fresh meat. Establishing standards and safety regulations for culturing stem cells is crucial for the market entry of cultured muscle tissue-based biomaterials. Understanding stem cells is a prerequisite for creating reliable cultivated meat and other cellular agricultural biomaterials. The techniques and regulations from the cultivated meat industry could pave the way for new cellular agriculture industries in the future.
Collapse
Affiliation(s)
- Dong-Kyung Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Research and Development Center, Space F Corporation, Hwasung, 18471, Gyeonggi-do, Republic of Korea
| | - Minsu Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jinsol Jeong
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Young-Seok Lee
- Research and Development Center, Space F Corporation, Hwasung, 18471, Gyeonggi-do, Republic of Korea
| | - Ji Won Yoon
- Research and Development Center, Space F Corporation, Hwasung, 18471, Gyeonggi-do, Republic of Korea
| | - Min-Jeong An
- Research and Development Center, Space F Corporation, Hwasung, 18471, Gyeonggi-do, Republic of Korea
| | - Hyun Young Jung
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Cho Hyun Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yelim Ahn
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kwang-Hwan Choi
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Research and Development Center, Space F Corporation, Hwasung, 18471, Gyeonggi-do, Republic of Korea
| | - Cheorun Jo
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Center for Food and Bioconvergence, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, 25354, Gangwon-do, Republic of Korea
| | - Chang-Kyu Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, 25354, Gangwon-do, Republic of Korea
| |
Collapse
|
39
|
Chen B, Wang Y, Hou D, Zhang Y, Zhang B, Niu Y, Ji H, Tian Y, Liu X, Kang X, Cai H, Li Z. Transcriptome-Based Identification of the Muscle Tissue-Specific Expression Gene CKM and Its Regulation of Proliferation, Apoptosis and Differentiation in Chicken Primary Myoblasts. Animals (Basel) 2023; 13:2316. [PMID: 37508090 PMCID: PMC10376263 DOI: 10.3390/ani13142316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/11/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Skeletal muscle is an essential tissue in meat-producing animals, and meat-producing traits have been a hot topic in chicken genetic breeding research. Current research shows that creatine kinase M-type-like (CKM) is one of the most abundant proteins in skeletal muscle and plays an important role in the growth and development of skeletal muscle, but its role in the development of chicken skeletal muscle is still unclear. Via RNA sequencing (RNA-seq), we found that CKM was highly expressed in chicken breast muscle tissue. In this study, the expression profile of CKM was examined by quantitative real-time PCR (qPCR), and overexpression and RNA interference techniques were used to explore the functions of CKM in the proliferation, apoptosis and differentiation of chicken primary myoblasts (CPMs). It was shown that CKM was specifically highly expressed in breast muscle and leg muscle and was highly expressed in stage 16 embryonic muscle, while CKM inhibited proliferation, promoted the apoptosis and differentiation of CPMs and was involved in regulating chicken myogenesis. Transcriptome sequencing was used to identify genes that were differentially expressed in CPMs after CKM disruption, and bioinformatics analysis showed that CKM was involved in regulating chicken myogenesis. In summary, CKM plays an important role in skeletal muscle development during chicken growth and development.
Collapse
Affiliation(s)
- Bingjie Chen
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450001, China
| | - Yanxing Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450001, China
| | - Dan Hou
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450001, China
| | - Yushi Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450001, China
| | - Bochun Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450001, China
| | - Yufang Niu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450001, China
| | - Haigang Ji
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450001, China
| | - Yadong Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450001, China
- Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Zhengzhou 450001, China
| | - Xiaojun Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450001, China
- Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Zhengzhou 450001, China
| | - Xiangtao Kang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450001, China
- Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Zhengzhou 450001, China
| | - Hanfang Cai
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450001, China
| | - Zhuanjian Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450001, China
- Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Zhengzhou 450001, China
| |
Collapse
|
40
|
Burattini M, Lippens R, Baleine N, Gerard M, Van Meerssche J, Geeroms C, Odent J, Raquez JM, Van Vlierberghe S, Thorrez L. Ionically Modified Gelatin Hydrogels Maintain Murine Myogenic Cell Viability and Fusion Capacity. Macromol Biosci 2023; 23:e2300019. [PMID: 37059590 DOI: 10.1002/mabi.202300019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/23/2023] [Indexed: 04/16/2023]
Abstract
For tissue engineering of skeletal muscles, there is a need for biomaterials which do not only allow cell attachment, proliferation, and differentiation, but also support the physiological conditions of the tissue. Next to the chemical nature and structure of the biomaterial, its response to the application of biophysical stimuli, such as mechanical deformation or application of electrical pulses, can impact in vitro tissue culture. In this study, gelatin methacryloyl (GelMA) is modified with hydrophilic 2-acryloxyethyltrimethylammonium chloride (AETA) and 3-sulfopropyl acrylate potassium (SPA) ionic comonomers to obtain a piezoionic hydrogel. Rheology, mass swelling, gel fraction, and mechanical characteristics are determined. The piezoionic properties of the SPA and AETA-modified GelMA are confirmed by a significant increase in ionic conductivity and an electrical response as a function of mechanical stress. Murine myoblasts display a viability of >95% after 1 week on the piezoionic hydrogels, confirming their biocompatibility. The GelMA modifications do not influence the fusion capacity of the seeded myoblasts or myotube width after myotube formation. These results describe a novel functionalization providing new possibilities to exploit piezo-effects in the tissue engineering field.
Collapse
Affiliation(s)
- Margherita Burattini
- Tissue Engineering Lab, Dep. Development and Regeneration, KU Leuven Kulak, Kortrijk, 8500, Belgium
- Dep. Of Surgical Sciences, Dentistry and Maternity, University of Verona, Verona, 37129, Italy
| | - Robrecht Lippens
- Polymer Chemistry & Biomaterials Group, Center of Macromolecular Chemistry (CMaC), Dep. Of Organic and Macromolecular Chemistry, Ghent University (UGent), Ghent, 9000, Belgium
| | - Nicolas Baleine
- Laboratory of Polymeric and Composite Materials (LPCM), Center of Innovation and Research in Materials and Polymers (CIRMAP), University of Mons (UMONS), Place du Parc 20, Mons, 7000, Belgium
| | - Melanie Gerard
- Tissue Engineering Lab, Dep. Development and Regeneration, KU Leuven Kulak, Kortrijk, 8500, Belgium
| | - Joeri Van Meerssche
- Polymer Chemistry & Biomaterials Group, Center of Macromolecular Chemistry (CMaC), Dep. Of Organic and Macromolecular Chemistry, Ghent University (UGent), Ghent, 9000, Belgium
| | - Chloë Geeroms
- Polymer Chemistry & Biomaterials Group, Center of Macromolecular Chemistry (CMaC), Dep. Of Organic and Macromolecular Chemistry, Ghent University (UGent), Ghent, 9000, Belgium
| | - Jérémy Odent
- Laboratory of Polymeric and Composite Materials (LPCM), Center of Innovation and Research in Materials and Polymers (CIRMAP), University of Mons (UMONS), Place du Parc 20, Mons, 7000, Belgium
| | - Jean-Marie Raquez
- Laboratory of Polymeric and Composite Materials (LPCM), Center of Innovation and Research in Materials and Polymers (CIRMAP), University of Mons (UMONS), Place du Parc 20, Mons, 7000, Belgium
| | - Sandra Van Vlierberghe
- Polymer Chemistry & Biomaterials Group, Center of Macromolecular Chemistry (CMaC), Dep. Of Organic and Macromolecular Chemistry, Ghent University (UGent), Ghent, 9000, Belgium
| | - Lieven Thorrez
- Tissue Engineering Lab, Dep. Development and Regeneration, KU Leuven Kulak, Kortrijk, 8500, Belgium
| |
Collapse
|
41
|
Filippi M, Yasa O, Giachino J, Graf R, Balciunaite A, Stefani L, Katzschmann RK. Perfusable Biohybrid Designs for Bioprinted Skeletal Muscle Tissue. Adv Healthc Mater 2023; 12:e2300151. [PMID: 36911914 PMCID: PMC11468554 DOI: 10.1002/adhm.202300151] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Indexed: 03/14/2023]
Abstract
Engineered, centimeter-scale skeletal muscle tissue (SMT) can mimic muscle pathophysiology to study development, disease, regeneration, drug response, and motion. Macroscale SMT requires perfusable channels to guarantee cell survival, and support elements to enable mechanical cell stimulation and uniaxial myofiber formation. Here, stable biohybrid designs of centimeter-scale SMT are realized via extrusion-based bioprinting of an optimized polymeric blend based on gelatin methacryloyl and sodium alginate, which can be accurately coprinted with other inks. A perfusable microchannel network is designed to functionally integrate with perfusable anchors for insertion into a maturation culture template. The results demonstrate that i) coprinted synthetic structures display highly coherent interfaces with the living tissue, ii) perfusable designs preserve cells from hypoxia all over the scaffold volume, iii) constructs can undergo passive mechanical tension during matrix remodeling, and iv) the constructs can be used to study the distribution of drugs. Extrusion-based multimaterial bioprinting with the inks and design realizes in vitro matured biohybrid SMT for biomedical applications.
Collapse
Affiliation(s)
- Miriam Filippi
- Soft Robotics LaboratoryETH ZurichTannenstrasse 3Zurich8092Switzerland
| | - Oncay Yasa
- Soft Robotics LaboratoryETH ZurichTannenstrasse 3Zurich8092Switzerland
| | - Jan Giachino
- Soft Robotics LaboratoryETH ZurichTannenstrasse 3Zurich8092Switzerland
| | - Reto Graf
- Soft Robotics LaboratoryETH ZurichTannenstrasse 3Zurich8092Switzerland
| | - Aiste Balciunaite
- Soft Robotics LaboratoryETH ZurichTannenstrasse 3Zurich8092Switzerland
| | - Lisa Stefani
- Soft Robotics LaboratoryETH ZurichTannenstrasse 3Zurich8092Switzerland
| | | |
Collapse
|
42
|
Cosentino M, Nicoletti C, Valenti V, Schirone L, Di Nonno F, Apa L, Zouhair M, Genovese D, Madaro L, Dinarelli S, Rossi M, Del Prete Z, Sciarretta S, Frati G, Rizzuto E, Musarò A. Remodeled eX vivo muscle engineered tissue improves heart function after chronic myocardial ischemia. Sci Rep 2023; 13:10370. [PMID: 37365262 DOI: 10.1038/s41598-023-37553-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 06/23/2023] [Indexed: 06/28/2023] Open
Abstract
The adult heart displays poor reparative capacities after injury. Cell transplantation and tissue engineering approaches have emerged as possible therapeutic options. Several stem cell populations have been largely used to treat the infarcted myocardium. Nevertheless, transplanted cells displayed limited ability to establish functional connections with the host cardiomyocytes. In this study, we provide a new experimental tool, named 3D eX vivo muscle engineered tissue (X-MET), to define the contribution of mechanical stimuli in triggering functional remodeling and to rescue cardiac ischemia. We revealed that mechanical stimuli trigger a functional remodeling of the 3D skeletal muscle system toward a cardiac muscle-like structure. This was supported by molecular and functional analyses, demonstrating that remodeled X-MET expresses relevant markers of functional cardiomyocytes, compared to unstimulated and to 2D- skeletal muscle culture system. Interestingly, transplanted remodeled X-MET preserved heart function in a murine model of chronic myocardial ischemia and increased survival of transplanted injured mice. X-MET implantation resulted in repression of pro-inflammatory cytokines, induction of anti-inflammatory cytokines, and reduction in collagen deposition. Altogether, our findings indicate that biomechanical stimulation induced a cardiac functional remodeling of X-MET, which showed promising seminal results as a therapeutic product for the development of novel strategies for regenerative medicine.
Collapse
Affiliation(s)
- Marianna Cosentino
- Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14, 00161, Rome, Italy
| | - Carmine Nicoletti
- Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14, 00161, Rome, Italy
| | - Valentina Valenti
- Department of Cardiology, Ospedale Santa Maria Goretti, 04100, Latina, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Leonardo Schirone
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | | | - Ludovica Apa
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, 00184, Rome, Italy
| | - Mariam Zouhair
- Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14, 00161, Rome, Italy
| | - Desiree Genovese
- Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14, 00161, Rome, Italy
| | - Luca Madaro
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
| | - Simone Dinarelli
- Department of Basic and Applied Sciences for Engineering, Sapienza University of Rome, 00161, Rome, Italy
| | - Marco Rossi
- Department of Basic and Applied Sciences for Engineering, Sapienza University of Rome, 00161, Rome, Italy
| | - Zaccaria Del Prete
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, 00184, Rome, Italy
| | - Sebastiano Sciarretta
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- IRCCS Neuromed, Pozzilli (IS), Italy
| | - Giacomo Frati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- IRCCS Neuromed, Pozzilli (IS), Italy
| | - Emanuele Rizzuto
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, 00184, Rome, Italy
| | - Antonio Musarò
- Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14, 00161, Rome, Italy.
- Scuola Superiore di Studi Avanzati Sapienza (SSAS), Sapienza University of Rome, 00185, Rome, Italy.
| |
Collapse
|
43
|
An C, Chen Y, Wu Y, Hu Z, Zhang H, Liu R, Zhou Y, Cen L. Manipulation of porous poly(l-lactide-co-ε-caprolactone) microcarriers via microfluidics for C2C12 expansion. Int J Biol Macromol 2023; 242:124625. [PMID: 37146858 DOI: 10.1016/j.ijbiomac.2023.124625] [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: 02/27/2023] [Revised: 04/14/2023] [Accepted: 04/23/2023] [Indexed: 05/07/2023]
Abstract
The growth and repair of skeletal muscle are due in part to activation of muscle precursor cells, commonly known as satellite cells or myoblasts. In order to acquire enough cells for neoskeletal muscle regeneration, it is urgent to develop microcarriers for skeletal myoblasts proliferation with a considerable efficiency. The current study was thus proposed to develop a microfluidic technology to manufacture porous poly(l-lactide-co-ε-caprolactone) (PLCL) microcarriers of high uniformity, and porosity was manipulated via camphene to suit the proliferation of C2C12 cells. A co-flow capillary microfluidic device was first designed to obtain PLCL microcarriers with different porosity. The attachment and proliferation of C2C12 cells on these microcarriers were evaluated and the differentiation potential of expanded cells were verified. The obtained porous microcarriers were all uniform in size with a high mono-dispersity (CV < 5 %). The content of camphene rendered effects on the size, porosity, and pore size of microcarriers, and porous structure addition produced a softening of their mechanical properties. The one of 10 % camphene (PM-10) exhibited the superior expansion for C2C12 cells with the number of cells after 5 days of culture reached 9.53 times of the adherent cells on the first day. The expanded cells from PM-10 still retained excellent myogenic differentiation performance as the expressions of MYOD, Desmin and MYH2 were intensively enhanced. Hence, the current developed porous PLCL microcarriers could offer as a promising type of substrates not only for in vitro muscular precursor cells expansion without compromising any multipotency but also have the potential as injectable constructs to mediate muscle regeneration.
Collapse
Affiliation(s)
- Chenjing An
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, Department of Product Engineering, School of Chemical Engineering, East China University of Science and Technology. No.130 Mei Long Road, Shanghai 200237, China
| | - Yawen Chen
- State Key Laboratory of Bioreactor Engineering, School of Bioengineering, East China University of Science and Technology. No.130 Mei Long Road, Shanghai 200237, China
| | - Yanfei Wu
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, Department of Product Engineering, School of Chemical Engineering, East China University of Science and Technology. No.130 Mei Long Road, Shanghai 200237, China
| | - Zhihuan Hu
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, Department of Product Engineering, School of Chemical Engineering, East China University of Science and Technology. No.130 Mei Long Road, Shanghai 200237, China
| | - Huan Zhang
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, Department of Product Engineering, School of Chemical Engineering, East China University of Science and Technology. No.130 Mei Long Road, Shanghai 200237, China
| | - Ruilai Liu
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, Department of Product Engineering, School of Chemical Engineering, East China University of Science and Technology. No.130 Mei Long Road, Shanghai 200237, China
| | - Yan Zhou
- State Key Laboratory of Bioreactor Engineering, School of Bioengineering, East China University of Science and Technology. No.130 Mei Long Road, Shanghai 200237, China.
| | - Lian Cen
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, Department of Product Engineering, School of Chemical Engineering, East China University of Science and Technology. No.130 Mei Long Road, Shanghai 200237, China.
| |
Collapse
|
44
|
Tavares-Negrete JA, Pedroza-González SC, Frías-Sánchez AI, Salas-Ramírez ML, de Santiago-Miramontes MDLÁ, Luna-Aguirre CM, Alvarez MM, Trujillo-de Santiago G. Supplementation of GelMA with Minimally Processed Tissue Promotes the Formation of Densely Packed Skeletal-Muscle-Like Tissues. ACS Biomater Sci Eng 2023. [PMID: 37126642 DOI: 10.1021/acsbiomaterials.2c01521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
We present a simple and cost-effective strategy for developing gelatin methacryloyl (GelMA) hydrogels supplemented with minimally processed tissue (MPT) to fabricate densely packed skeletal-muscle-like tissues. MPT powder was prepared from skeletal muscle by freeze-drying, grinding, and sieving. Cell-culture experiments showed that the incorporation of 0.5-2.0% (w/v) MPT into GelMA hydrogels enhances the proliferation of murine myoblasts (C2C12 cells) compared to proliferation in pristine GelMA hydrogels and GelMA supplemented with decellularized skeletal-muscle tissues (DCTs). MPT-supplemented constructs also preserved their three-dimensional (3D) integrity for 28 days. By contrast, analogous pristine GelMA constructs only maintained their structure for 14 days or less. C2C12 cells embedded in MPT-supplemented constructs exhibited a higher degree of cell alignment and reached a significantly higher density than cells loaded in pristine GelMA constructs. Our results suggest that the addition of MPT incorporates a rich source of biochemical and topological cues, such as growth factors, glycosaminoglycans (GAGs), and structurally preserved proteins (e.g., collagen). In addition, GelMA supplemented with MPT showed suitable rheological properties for use as bioinks for extrusion bioprinting. We envision that this simple and cost-effective strategy of hydrogel supplementation will evolve into an exciting spectrum of applications for tissue engineers, primarily in the biofabrication of relevant microtissues for in vitro models and cultured meat and ultimately for the biofabrication of transplant materials using autologous MPT.
Collapse
Affiliation(s)
- Jorge A Tavares-Negrete
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, 64849 Monterrey, México
- Departamento de Ingeniería Mecatrónica y Eléctrica, Tecnológico de Monterrey, 64849 Monterrey, México
| | - Sara Cristina Pedroza-González
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, 64849 Monterrey, México
- Departamento de Ingeniería Mecatrónica y Eléctrica, Tecnológico de Monterrey, 64849 Monterrey, México
| | - Ada I Frías-Sánchez
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, 64849 Monterrey, México
- Departamento de Ingeniería Mecatrónica y Eléctrica, Tecnológico de Monterrey, 64849 Monterrey, México
| | - Miriam L Salas-Ramírez
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, 64849 Monterrey, México
- Departamento de Ingeniería Mecatrónica y Eléctrica, Tecnológico de Monterrey, 64849 Monterrey, México
| | | | - Claudia Maribel Luna-Aguirre
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, 64849 Monterrey, México
- Departamento de Ingeniería Mecatrónica y Eléctrica, Tecnológico de Monterrey, 64849 Monterrey, México
| | - Mario M Alvarez
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, 64849 Monterrey, México
- Departamento de Ingeniería Mecatrónica y Eléctrica, Tecnológico de Monterrey, 64849 Monterrey, México
| | - Grissel Trujillo-de Santiago
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, 64849 Monterrey, México
- Departamento de Ingeniería Mecatrónica y Eléctrica, Tecnológico de Monterrey, 64849 Monterrey, México
| |
Collapse
|
45
|
Lee H, Kim SH, Lee JS, Lee YJ, Lee OJ, Ajiteru O, Sultan MT, Lee SW, Park CH. Functional Skeletal Muscle Regeneration Using Muscle Mimetic Tissue Fabricated by Microvalve-Assisted Coaxial 3D Bioprinting. Adv Healthc Mater 2023; 12:e2202664. [PMID: 36469728 DOI: 10.1002/adhm.202202664] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/23/2022] [Indexed: 12/12/2022]
Abstract
3D-printed artificial skeletal muscle, which mimics the structural and functional characteristics of native skeletal muscle, is a promising treatment method for muscle reconstruction. Although various fabrication techniques for skeletal muscle using 3D bio-printers are studied, it is still challenging to build a functional muscle structure. A strategy using microvalve-assisted coaxial 3D bioprinting in consideration of functional skeletal muscle fabrication is reported. The unit (artificial muscle fascicle: AMF) of muscle mimetic tissue is composed of a core filled with medium-based C2C12 myoblast aggregates as a role of muscle fibers and a photo cross-linkable hydrogel-based shell as a role of connective tissue in muscles that enhances printability and cell adhesion and proliferation. Especially, a microvalve system is applied for the core part with even cell distribution and strong cell-cell interaction. This system enhances myotube formation and consequently shows spontaneous contraction. A multi-printed AMF (artificial muscle tissue: AMT) as a piece of muscle is implanted into the anterior tibia (TA) muscle defect site of immunocompromised rats. As a result, the TA-implanted AMT responds to electrical stimulation and represents histologically regenerated muscle tissue. This microvalve-assisted coaxial 3D bioprinting shows a significant step forward to mimicking native skeletal muscle tissue.
Collapse
Affiliation(s)
- Hanna Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Soon Hee Kim
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Ji Seung Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Young Jin Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Ok Joo Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Olatunji Ajiteru
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Md Tipu Sultan
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Suk Woo Lee
- Department of Obstetrics and Gynecology, Hallym University Sacred Heart Hospital, Anyang, 14068, Republic of Korea
| | - Chan Hum Park
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea.,Department of Otorhinolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hospital, School of Medicine, Hallym University, Chuncheon, 24252, Republic of Korea
| |
Collapse
|
46
|
Chae S, Cho DW. Biomaterial-based 3D bioprinting strategy for orthopedic tissue engineering. Acta Biomater 2023; 156:4-20. [PMID: 35963520 DOI: 10.1016/j.actbio.2022.08.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 07/05/2022] [Accepted: 08/02/2022] [Indexed: 02/02/2023]
Abstract
The advent of three-dimensional (3D) bioprinting has enabled impressive progress in the development of 3D cellular constructs to mimic the structural and functional characteristics of natural tissues. Bioprinting has considerable translational potential in tissue engineering and regenerative medicine. This review highlights the rational design and biofabrication strategies of diverse 3D bioprinted tissue constructs for orthopedic tissue engineering applications. First, we elucidate the fundamentals of 3D bioprinting techniques and biomaterial inks and discuss the basic design principles of bioprinted tissue constructs. Next, we describe the rationale and key considerations in 3D bioprinting of tissues in many different aspects. Thereafter, we outline the recent advances in 3D bioprinting technology for orthopedic tissue engineering applications, along with detailed strategies of the engineering methods and materials used, and discuss the possibilities and limitations of different 3D bioprinted tissue products. Finally, we summarize the current challenges and future directions of 3D bioprinting technology in orthopedic tissue engineering and regenerative medicine. This review not only delineates the representative 3D bioprinting strategies and their tissue engineering applications, but also provides new insights for the clinical translation of 3D bioprinted tissues to aid in prompting the future development of orthopedic implants. STATEMENT OF SIGNIFICANCE: 3D bioprinting has driven major innovations in the field of tissue engineering and regenerative medicine; aiming to develop a functional viable tissue construct that provides an alternative regenerative therapy for musculoskeletal tissue regeneration. 3D bioprinting-based biofabrication strategies could open new clinical possibilities for creating equivalent tissue substitutes with the ability to customize them to meet patient demands. In this review, we summarize the significance and recent advances in 3D bioprinting technology and advanced bioinks. We highlight the rationale for biofabrication strategies using 3D bioprinting for orthopedic tissue engineering applications. Furthermore, we offer ample perspective and new insights into the current challenges and future direction of orthopedic bioprinting translation research.
Collapse
Affiliation(s)
- Suhun Chae
- Department of Mechanical Engineering, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Gyeongsangbuk-do, Pohang 37673, South Korea; EDmicBio Inc., 111 Hoegi-ro, Dongdaemun-gu, Seoul 02445, South Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Gyeongsangbuk-do, Pohang 37673, South Korea; Institute for Convergence Research and Education in Advanced Technology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea.
| |
Collapse
|
47
|
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
|
48
|
Ostrovidov S, Ramalingam M, Bae H, Orive G, Fujie T, Shi X, Kaji H. Latest developments in engineered skeletal muscle tissues for drug discovery and development. Expert Opin Drug Discov 2023; 18:47-63. [PMID: 36535280 DOI: 10.1080/17460441.2023.2160438] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION With the advances in skeletal muscle tissue engineering, new platforms have arisen with important applications in biology studies, disease modeling, and drug testing. Current developments highlight the quest for engineering skeletal muscle tissues with higher complexity . These new human skeletal muscle tissue models will be powerful tools for drug discovery and development and disease modeling. AREAS COVERED The authors review the latest advances in in vitro models of engineered skeletal muscle tissues used for testing drugs with a focus on the use of four main cell culture techniques: Cell cultures in well plates, in microfluidics, in organoids, and in bioprinted constructs. Additional information is provided on the satellite cell niche. EXPERT OPINION In recent years, more sophisticated in vitro models of skeletal muscle tissues have been fabricated. Important developments have been made in stem cell research and in the engineering of human skeletal muscle tissue. Some platforms have already started to be used for drug testing, notably those based on the parameters of hypertrophy/atrophy and the contractibility of myotubes. More developments are expected through the use of multicellular types and multi-materials as matrices . The validation and use of these models in drug testing should now increase.
Collapse
Affiliation(s)
- Serge Ostrovidov
- Department of Biomechanics, Institute of Biomaterials and Bioengineering (IBB), Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Murugan Ramalingam
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science, BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, Republic of Korea.,School of Basic Medical Science, Chengdu University, Chengdu, Sichuan, China.,Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Republic of Korea.,Department of Metallurgical and Materials Engineering, Atilim University, Ankara, Turkey
| | - Hojae Bae
- KU Convergence Science and Technology Institute, Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Hwayang-dong, Kwangjin-gu, Seoul, Republic of Korea
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain.,Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.,Biomaterials and Nanomedicine (CIBER-BBN), Biomedical Research Networking Centre in Bioengineering, Vitoria-Gasteiz, Spain
| | - Toshinori Fujie
- Department of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Xuetao Shi
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, China
| | - Hirokazu Kaji
- Department of Biomechanics, Institute of Biomaterials and Bioengineering (IBB), Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
49
|
Patient-specific 3D bioprinting for in situ tissue engineering and regenerative medicine. 3D Print Med 2023. [DOI: 10.1016/b978-0-323-89831-7.00003-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
50
|
Demri N, Dumas S, Nguyen M, Gropplero G, Abou‐Hassan A, Descroix S, Wilhelm C. Remote Magnetic Microengineering and Alignment of Spheroids into 3D Cellular Fibers. ADVANCED FUNCTIONAL MATERIALS 2022; 32. [DOI: 10.1002/adfm.202204850] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Indexed: 01/05/2025]
Abstract
AbstractDeveloping in vitro models that recapitulate the in vivo organization of living cells in a 3D microenvironment is one of the current challenges in the field of tissue engineering. In particular for anisotropic tissues where alignment of precursor cells is required for them to create functional structures. Herein, a new method is proposed that allows aligning in the direction of a uniform magnetic field both individual cells (muscle, stromal, and stem cells) or spheroids in a thermoresponsive collagen hydrogel. In an all‐in‐one approach, spheroids are generated at high throughput by magnetic engineering using microfabricated micromagnets and are used as building blocks to create 3D anisotropic tissue structures of different scales. The magnetic cells and spheroids alignment process is optimized in terms of magnetic cell labeling, concentration, and size. Anisotropic structures are induced to form fibers in the direction of the magnetic alignment, with the respective roles of the magnetic field, the mechanical stretching of hydrogel or co‐culture of the aligned cells with non‐magnetic stromal cells, being investigated. Over days, spheroids fuse into 3D tubular structures, oriented in the direction of the magnetic alignment. Moreover, in the case of the muscle cells model, multinucleated cells can be observed within the fibers.
Collapse
Affiliation(s)
- Noam Demri
- Laboratoire Physico Chimie Curie PCC CNRS UMR168 Institut Curie Sorbonne University PSL University 75005 Paris France
| | - Simon Dumas
- Laboratoire Physico Chimie Curie PCC CNRS UMR168 Institut Curie Sorbonne University PSL University 75005 Paris France
| | - Manh‐Louis Nguyen
- Laboratoire Physico Chimie Curie PCC CNRS UMR168 Institut Curie Sorbonne University PSL University 75005 Paris France
| | - Giacomo Gropplero
- Laboratoire Physico Chimie Curie PCC CNRS UMR168 Institut Curie Sorbonne University PSL University 75005 Paris France
| | - Ali Abou‐Hassan
- Institut Universitaire de France (IUF) 75231 Paris Cedex 05 France
- PHysico‐chimie des Electrolytes et Nanosystèmes InterfaciauX PHENIX CNRS UMR234 Sorbonne University 75005 Paris France
| | - Stéphanie Descroix
- Laboratoire Physico Chimie Curie PCC CNRS UMR168 Institut Curie Sorbonne University PSL University 75005 Paris France
| | - Claire Wilhelm
- Laboratoire Physico Chimie Curie PCC CNRS UMR168 Institut Curie Sorbonne University PSL University 75005 Paris France
| |
Collapse
|