1
|
Liao W, Shi Y, Li Z, Yin X. Advances in 3D printing combined with tissue engineering for nerve regeneration and repair. J Nanobiotechnology 2025; 23:5. [PMID: 39754257 PMCID: PMC11697815 DOI: 10.1186/s12951-024-03052-9] [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: 09/20/2024] [Accepted: 11/29/2024] [Indexed: 01/06/2025] Open
Abstract
The repair of nerve damage has long posed a challenge owing to limited self-repair capacity and the highly differentiated nature of nerves. While new therapeutic and pharmacologic interventions have emerged in neurology, their regenerative efficacy remains limited. Tissue engineering offers a promising avenue for overcoming the limitations of conventional treatments and increasing the outcomes of regenerative repair. By implanting scaffolds into damaged nerve tissue sites, the repair and functional reconstruction of nerve injuries can be significantly facilitated. The integration of three-dimensional (3D) printing technology introduces a novel approach for accurate simulation and scalably fabricating neural tissue structures. Tissue-engineered scaffolds developed through 3D printing technology are expected to be a viable therapeutic option for nerve injuries, with broad applicability and continued development. This review systematically examines recent advances in 3D printing and tissue engineering for nerve regeneration and repair. It details the basic principles and construction strategies of neural tissue engineering and explores the crucial role of 3D printing technology. Additionally, it elucidates specific applications and technical challenges associated with this integrated approach, thereby providing valuable insights into innovative strategies and pragmatic implementation within this field.
Collapse
Affiliation(s)
- Weifang Liao
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, China
| | - Yuying Shi
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, China
| | - Zuguang Li
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Yin
- Department of Neurology, Affiliated Hospital of Jiujiang University, No. 57 East Xunyang Road, Jiujiang, Jiangxi, 332005, China.
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, China.
| |
Collapse
|
2
|
Caiado Decarli M, Ferreira HP, Sobreiro‐Almeida R, Teixeira FC, Correia TR, Babilotte J, Olijve J, Custódio CA, Gonçalves IC, Mota C, Mano JF, Moroni L. Embedding Bioprinting of Low Viscous, Photopolymerizable Blood-Based Bioinks in a Crystal Self-Healing Transparent Supporting Bath. SMALL METHODS 2025; 9:e2400857. [PMID: 38970553 PMCID: PMC11740956 DOI: 10.1002/smtd.202400857] [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: 06/08/2024] [Indexed: 07/08/2024]
Abstract
Protein-based hydrogels have great potential to be used as bioinks for biofabrication-driven tissue regeneration strategies due to their innate bioactivity. Nevertheless, their use as bioinks in conventional 3D bioprinting is impaired due to their intrinsic low viscosity. Using embedding bioprinting, a liquid bioink is printed within a support that physically holds the patterned filament. Inspired by the recognized microencapsulation technique complex coacervation, crystal self-healing embedding bioprinting (CLADDING) is introduced based on a highly transparent crystal supporting bath. The suitability of distinct classes of gelatins is evaluated (i.e., molecular weight distribution, isoelectric point, and ionic content), as well as the formation of gelatin-gum arabic microparticles as a function of pH, temperature, solvent, and mass ratios. Characterizing and controlling this parametric window resulted in high yields of support bath with ideal self-healing properties for interaction with protein-based bioinks. This support bath achieved transparency, which boosted light permeation within the bath. Bioprinted constructs fully composed of platelet lysates encapsulating a co-culture of human mesenchymal stromal cells and endothelial cells are obtained, demonstrating a high-dense cellular network with excellent cell viability and stability over a month. CLADDING broadens the spectrum of photocrosslinkable materials with extremely low viscosity that can now be bioprinted with sensitive cells without any additional support.
Collapse
Affiliation(s)
- Monize Caiado Decarli
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
- Department of Biomaterials & Biomedical TechnologyUniversity Medical Center Groningen/University of GroningenA. Deusinglaan 1GroningenAV 9713The Netherlands
| | - Helena P. Ferreira
- i3S – Instituto de Investigação e Inovação em Saúde/INEB – Instituto de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen 208Porto4200‐180Portugal
- ICBAS – Instituto de Ciências Biomédicas Abel SalazarUniversidade do PortoPorto4050‐313Portugal
| | - Rita Sobreiro‐Almeida
- CICECO – Department of ChemistryAveiro Institute of MaterialsUniversity of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
| | - Filipa C. Teixeira
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Tiago R. Correia
- CICECO – Department of ChemistryAveiro Institute of MaterialsUniversity of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
| | - Joanna Babilotte
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Jos Olijve
- Rousselot BiomedicalExpertise CenterMeulestedekaai 81Ghent9000Belgium
| | - Catarina A. Custódio
- CICECO – Department of ChemistryAveiro Institute of MaterialsUniversity of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
- Metatissue, PCICreative Science Park Aveiro RegionVia do ConhecimentoÍlhavo3830‐352Portugal
| | - Inês C. Gonçalves
- i3S – Instituto de Investigação e Inovação em Saúde/INEB – Instituto de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen 208Porto4200‐180Portugal
- ICBAS – Instituto de Ciências Biomédicas Abel SalazarUniversidade do PortoPorto4050‐313Portugal
| | - Carlos Mota
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - João F. Mano
- CICECO – Department of ChemistryAveiro Institute of MaterialsUniversity of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
| | - Lorenzo Moroni
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| |
Collapse
|
3
|
Iwai T, Ikeguchi R, Aoyama T, Noguchi T, Yoshimoto K, Sakamoto D, Fujita K, Miyazaki Y, Akieda S, Nagamura-Inoue T, Nagamura F, Nakayama K, Matsuda S. Nerve regeneration using a Bio 3D conduit derived from umbilical cord-Derived mesenchymal stem cells in a rat sciatic nerve defect model. PLoS One 2024; 19:e0310711. [PMID: 39715170 DOI: 10.1371/journal.pone.0310711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 09/05/2024] [Indexed: 12/25/2024] Open
Abstract
Human umbilical cord-derived mesenchymal stromal cells (UC-MSCs), which can be prepared in advance and are presumed to be advantageous for nerve regeneration, have potential as a cell source for Bio 3D conduits. The purpose of this study was to evaluate the nerve regeneration ability of Bio 3D conduits made from UC-MSCs using a rat sciatic nerve defect model. METHODS A Bio 3D conduit was fabricated using a Bio 3D printer by placing UC-MSC spheroids into thin needles according to predesigned 3D data. The conduit was transplanted to bridge the 5-mm gaps of Lewis rat sciatic nerve, and nerve regeneration was evaluated at 8 weeks (Bio 3D group). Transplantation of autologous nerve segments (autograft) and silicone tubes represented the positive and negative control groups, respectively. In a second experiment, immunological reactions were evaluated in Bio 3D, autograft, and allograft groups by histochemical staining of transplanted segments in Brown Norway rats. RESULTS The mean angle of attack value in the kinematic analysis was significantly better in the Bio 3D group (‒20.1 ± 0.5°) than in the silicone group (‒33.7 ± 1.5°) 8 weeks after surgery. The average diameters of myelinated axons were significantly larger in the Bio 3D group (3.61 ± 0.15 μm) than in the silicone group (3.07 ± 0.12 μm), and the number of myelinated axons was significantly higher in the Bio 3D group (11,201 ± 980) than in the silicone group (8117 ± 646). Histological findings (hematoxylin and eosin [HE] staining and anti-CD3 fluorescent immunostaining) showed that rejection was suppressed in the Bio 3D group compared to the allograft group. Based on macroscopic findings and histological findings (anti-human mitochondrial fluorescent immunostaining), UC-MSCs in the Bio 3D conduit disappeared gradually from week 1 to week 8. CONCLUSIONS The Bio 3D conduit prepared from UC-MSCs was superior to the silicone tube and achieved comparable nerve regeneration to the autologous (autograft) group. Rejection was suppressed in the Bio 3D group compared to the allograft group. Although this study used a xenograft model, we speculate that rejection was low due to the characteristics of UC-MSCs. UC-MSCs are a useful cell source for Bio 3D conduits.
Collapse
Affiliation(s)
- Terunobu Iwai
- Department of Orthopaedic Surgery, Kyoto University, Kyoto, Japan
| | - Ryosuke Ikeguchi
- Department of Orthopaedic Surgery, Kyoto University, Kyoto, Japan
- Department of Rehabilitation Medicine, Kyoto University, Kyoto, Japan
| | - Tomoki Aoyama
- Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Noguchi
- Department of Orthopaedic Surgery, Kyoto University, Kyoto, Japan
| | - Koichi Yoshimoto
- Department of Orthopaedic Surgery, Kyoto University, Kyoto, Japan
| | - Daichi Sakamoto
- Department of Orthopaedic Surgery, Kyoto University, Kyoto, Japan
| | - Kazuaki Fujita
- Department of Orthopaedic Surgery, Kyoto University, Kyoto, Japan
| | | | | | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, The Institute of Medical Science, IMSUT CORD, Research Hospital, The University of Tokyo, Tokyo, Japan
| | - Fumitaka Nagamura
- Division of Advanced Medicine Promotion, The Advanced Clinical Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Koichi Nakayama
- Department of Regenerative Medicine and Biomedical Engineering, Faculty of Medicine, Saga University, Saga, Japan
| | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Kyoto University, Kyoto, Japan
| |
Collapse
|
4
|
Gholami F, Hajiheidari A, Barkhidarian B, Soveid N, Yekaninejad MS, Karimi Z, Bahrampour N, Keshavarz SA, Javdan G, Mirzaei K. A comparison of principal component analysis, reduced-rank regression, and partial least-squares in the identification of dietary patterns associated with cardiometabolic risk factors in Iranian overweight and obese women. BMC Med Res Methodol 2024; 24:215. [PMID: 39333898 PMCID: PMC11428567 DOI: 10.1186/s12874-024-02298-z] [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: 04/29/2023] [Accepted: 07/25/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND According to epidemiological studies, unhealthy dietary patterns and lifestyle lead to rising obesity and cardiometabolic diseases in Iran. Hybrid techniques were used to identify a dietary pattern characterized by fiber, folic acid, and carotenoid intake due to their association with cardiometabolic risk factors such as anthropometric measurements, blood pressure, lipid profile, C-Reactive Protein (CRP), Plasminogen Activator Inhibitor (PAI), Homeostatic Model Assessment Index (HOMA Index), cardiometabolic index (CMI), and monocyte chemoattractant protein (MCP-1). So, the objective of the recent study is to compare the reduced-rank regression (RRR) and partial least-squares (PLS) approaches to principal component analysis (PCA) for estimating diet-cardiometabolic risk factor correlations in Iranian obese women. METHODS Data on dietary intake was gathered from 376 healthy overweight and obese females aged 18 to 65 years using a 147-item food frequency questionnaire (FFQ). In this cross-sectional study, participants were referred to health centers of Tehran. Dietary patterns were developed using PCA, PLS, and RRR, and their outputs were assessed to identify reasonable patterns connected to cardiometabolic risk factors. The response variables for PLS and RRR were fiber, folic acid, and carotenoid intake. RESULTS In this study, 3 dietary patterns were identified by the PCA method, 2 dietary patterns by the PLS method, and one dietary pattern by the RRR method. High adherence to the plant-based dietary pattern identified by all methods were associated with higher fat free mass index (FFMI) (P < 0.05). Women in the highest tertile of the plant-based dietary pattern identified by PLS had 0.06 mmol/L (95% CI: 0.007,0.66, P = 0.02), 0.36 mmHg (95% CI: 0.14,0.88, P = 0.02), and 0.46 mg/l (95% CI: 0.25,0.82, P < 0.001), lower FBS, DBP, and CRP respectively than women in the first tertile. Also, PLS and RRR-derived patterns explained greater variance in the outcome (PCA: 1.05%; PLS: 11.62%; RRR: 25.28%), while the PCA dietary patterns explained greater variance in the food groups (PCA: 22.81%; PLS: 14.54%; RRR: 1.59%). CONCLUSION PLS was found to be more appropriate in determining dietary patterns associated with cardiometabolic-related risk factors. Nevertheless, the advantage of PLS over PCA and RRR must be confirmed in future longitudinal studies with extended follow-up in different settings, population groups, and response variables.
Collapse
Affiliation(s)
- Fatemeh Gholami
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), P.O Box 6446, 14155, Tehran, Iran.
| | | | - Bahareh Barkhidarian
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), P.O Box 6446, 14155, Tehran, Iran
| | - Neda Soveid
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), P.O Box 6446, 14155, Tehran, Iran
| | - Mir Saeid Yekaninejad
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Science, Tehran, Iran
| | - Zahra Karimi
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Science, Tehran, Iran
| | - Niki Bahrampour
- Department of Nutrition, Science and Research Branch, Islamic Azad University (SRBIAU), Tehran, Iran
| | - Seyed Ali Keshavarz
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamali Javdan
- Food Health Research Center, Hormozgan University of Medical Sciences, Bandar 'Abbas, Iran
| | - Khadijeh Mirzaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), P.O Box 6446, 14155, Tehran, Iran.
| |
Collapse
|
5
|
Drewry MD, Shi D, Dailey MT, Rothermund K, Trbojevic S, Almarza AJ, Cui XT, Syed-Picard FN. Enhancing facial nerve regeneration with scaffold-free conduits engineered using dental pulp stem cells and their endogenous, aligned extracellular matrix. J Neural Eng 2024; 21:056015. [PMID: 39197480 PMCID: PMC11406051 DOI: 10.1088/1741-2552/ad749d] [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: 01/30/2024] [Revised: 07/02/2024] [Accepted: 08/28/2024] [Indexed: 09/01/2024]
Abstract
Objective. Engineered nerve conduits must simultaneously enhance axon regeneration and orient axon extension to effectively restore function of severely injured peripheral nerves. The dental pulp contains a population of stem/progenitor cells that endogenously express neurotrophic factors (NTFs), growth factors known to induce axon repair. We have previously generated scaffold-free dental pulp stem/progenitor cell (DPSC) sheets comprising an aligned extracellular matrix (ECM). Through the intrinsic NTF expression of DPSCs and the topography of the aligned ECM, these sheets both induce and guide axon regeneration. Here, the capacity of bioactive conduits generated using these aligned DPSC sheets to restore function in critical-sized nerve injuries in rodents was evaluated.Approach. Scaffold-free nerve conduits were formed by culturing DPSCs on a substrate with aligned microgrooves, inducing the cells to align and deposit an aligned ECM. The sheets were then detached from the substrate and assembled into scaffold-free cylindrical tissues.Main results. In vitroanalyses confirmed that scaffold-free DPSC conduits maintained an aligned ECM and had uniformly distributed NTF expression. Implanting the aligned DPSC conduits across critical-sized defects in the buccal branch of rat facial nerves resulted in the regeneration of a fascicular nerve-like structure and myelinated axon extension across the injury site. Furthermore, compound muscle action potential and stimulated whisker movement measurements revealed that the DPSC conduit treatment promoted similar functional recovery compared to the clinical standard of care, autografts. Significance. This study demonstrates that scaffold-free aligned DPSC conduits supply trophic and guidance cues, key design elements needed to successfully promote and orient axon regeneration. Consequently, these conduits restore function in nerve injuries to similar levels as autograft treatments. These conduits offer a novel bioactive approach to nerve repair capable of improving clinical outcomes and patient quality of life.
Collapse
Affiliation(s)
- Michelle D Drewry
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Delin Shi
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Matthew T Dailey
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Oral and Maxillofacial Surgery, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Kristi Rothermund
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Sara Trbojevic
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Alejandro J Almarza
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Xinyan T Cui
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States of America
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Fatima N Syed-Picard
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| |
Collapse
|
6
|
Fakhr MJ, Kavakebian F, Ababzadeh S, Rezapour A. Challenges and Advances in Peripheral Nerve Tissue Engineering Critical Factors Affecting Nerve Regeneration. J Tissue Eng Regen Med 2024; 2024:8868411. [PMID: 40225756 PMCID: PMC11918807 DOI: 10.1155/2024/8868411] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 08/20/2024] [Indexed: 04/15/2025]
Abstract
Peripheral neuropathy is painful and can cause a considerable decline in quality of life. Surgery and autograft are the current approaches and clinical standards for restoring function after nerve damage. However, they usually result in unacceptable clinical results, so we need modern peripheral nerve defect treatment approaches. Tissue engineering techniques have been developed as a promising approach, but there are some considerations for translational application. Clinical application of novel tissue engineering methods is related to combining the appropriate cell and scaffold type to introduce safe and efficient bioscaffolds. Efficient nerve regeneration occurs by mimicking the extracellular matrix and combining topographical, biochemical, mechanical, and conductive signs via different cells, biomolecules, and polymers. In brief, ideal engineered biomaterial scaffolds will have to cover all characteristics of nerve tissue, such as nerve number, myelin, and axon thickness. Nerve regeneration has a highly sensitive response to its surrounding microenvironment. For designing a suitable construct, matching the regenerative potential of the autograft as the golden standard is essential. This review article examines the newest advancements in peripheral nerve tissue engineering. Specifically, the discussion will focus on incorporating innovative cues, biological modification, biomaterials, techniques, and concepts in this area of research.
Collapse
Affiliation(s)
- Massoumeh Jabbari Fakhr
- Department of Tissue Engineering and Applied Cell SciencesSchool of MedicineQom University of Medical Sciences, Qom, Iran
| | - Fatemeh Kavakebian
- Department of Tissue Engineering and Applied Cell SciencesSchool of MedicineQom University of Medical Sciences, Qom, Iran
| | - Shima Ababzadeh
- Department of Tissue Engineering and Applied Cell SciencesSchool of MedicineQom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research CentreQom University of Medical Sciences, Qom, Iran
| | - Alireza Rezapour
- Department of Tissue Engineering and Applied Cell SciencesSchool of MedicineQom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research CentreQom University of Medical Sciences, Qom, Iran
| |
Collapse
|
7
|
Redolfi Riva E, Özkan M, Contreras E, Pawar S, Zinno C, Escarda-Castro E, Kim J, Wieringa P, Stellacci F, Micera S, Navarro X. Beyond the limiting gap length: peripheral nerve regeneration through implantable nerve guidance conduits. Biomater Sci 2024; 12:1371-1404. [PMID: 38363090 DOI: 10.1039/d3bm01163a] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Peripheral nerve damage results in the loss of sensorimotor and autonomic functions, which is a significant burden to patients. Furthermore, nerve injuries greater than the limiting gap length require surgical repair. Although autografts are the preferred clinical choice, their usage is impeded by their limited availability, dimensional mismatch, and the sacrifice of another functional donor nerve. Accordingly, nerve guidance conduits, which are tubular scaffolds engineered to provide a biomimetic environment for nerve regeneration, have emerged as alternatives to autografts. Consequently, a few nerve guidance conduits have received clinical approval for the repair of short-mid nerve gaps but failed to regenerate limiting gap damage, which represents the bottleneck of this technology. Thus, it is still necessary to optimize the morphology and constituent materials of conduits. This review summarizes the recent advances in nerve conduit technology. Several manufacturing techniques and conduit designs are discussed, with emphasis on the structural improvement of simple hollow tubes, additive manufacturing techniques, and decellularized grafts. The main objective of this review is to provide a critical overview of nerve guidance conduit technology to support regeneration in long nerve defects, promote future developments, and speed up its clinical translation as a reliable alternative to autografts.
Collapse
Affiliation(s)
- Eugenio Redolfi Riva
- The Biorobotic Institute, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy.
- Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy
| | - Melis Özkan
- Institute of Materials, école Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
- Bertarelli Foundation Chair in Translational Neural Engineering, Center for Neuroprosthetics and Institute of Bioengineering, école Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Estefania Contreras
- Integral Service for Laboratory Animals (SIAL), Faculty of Veterinary, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Spain.
| | - Sujeet Pawar
- Institute of Materials, école Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Ciro Zinno
- The Biorobotic Institute, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy.
- Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy
| | - Enrique Escarda-Castro
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Jaehyeon Kim
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Paul Wieringa
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Francesco Stellacci
- Institute of Materials, école Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
- Institute of Materials, Department of Bioengineering and Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Station 12, CH-1015 Lausanne, Switzerland
| | - Silvestro Micera
- The Biorobotic Institute, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy.
- Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy
- Bertarelli Foundation Chair in Translational Neural Engineering, Center for Neuroprosthetics and Institute of Bioengineering, école Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Xavier Navarro
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Institute Guttmann Foundation, Hospital of Neurorehabilitation, Badalona, Spain
| |
Collapse
|
8
|
Ikeguchi R, Aoyama T, Noguchi T, Ushimaru M, Amino Y, Nakakura A, Matsuyama N, Yoshida S, Nagai-Tanima M, Matsui K, Arai Y, Torii Y, Miyazaki Y, Akieda S, Matsuda S. Peripheral nerve regeneration following scaffold-free conduit transplant of autologous dermal fibroblasts: a non-randomised safety and feasibility trial. COMMUNICATIONS MEDICINE 2024; 4:12. [PMID: 38278956 PMCID: PMC10817910 DOI: 10.1038/s43856-024-00438-6] [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: 04/27/2023] [Accepted: 01/15/2024] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND The use of Bio 3D nerve conduits is a promising approach for peripheral nerve reconstruction. This study aimed to assess their safety in three patients with peripheral nerve defects in their hands. METHODS We describe a single institution, non-blinded, non-randomised control trial conducted at Kyoto University Hospital. Eligibility criteria included severed peripheral nerve injuries or a defect in the region distal to the wrist joint not caused by a congenital anomaly; a defect with a length of ≤20 mm in a nerve with a diameter ≤2 mm; failed results of sensory functional tests; ability to register in the protocol within 6 months from the day of injury; refusal of artificial nerve or autologous nerve transplantation; age 20-60 years; and willingness to participate and provide informed written consent. Six weeks before transplantation, skin was harvested, dermal fibroblasts were isolated and expanded, and Bio 3D nerve conduits were created using a Bio 3D printer. Bio 3D nerve conduits were transplanted into the patients' nerve defects. The safety of Bio 3D nerve conduits in patients with a peripheral nerve injury in the distal part of the wrist joint were assessed over a 48-week period after transplantation. RESULTS No adverse events related to the use of Bio 3D nerve conduits were observed in any patient, and all three patients completed the trial. CONCLUSIONS Bio 3D nerve conduits were successfully used for clinical nerve reconstruction without adverse events and are a possible treatment option for peripheral nerve injuries.
Collapse
Affiliation(s)
- Ryosuke Ikeguchi
- Department of Rehabilitation Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Tomoki Aoyama
- Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Noguchi
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mika Ushimaru
- Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| | - Yoko Amino
- Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| | - Akiyoshi Nakakura
- Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| | - Noriko Matsuyama
- Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| | - Shiori Yoshida
- Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| | - Momoko Nagai-Tanima
- Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keiko Matsui
- Center for Research and Application of Cellular Therapy, Kyoto University Hospital, Kyoto, Japan
| | - Yasuyuki Arai
- Center for Research and Application of Cellular Therapy, Kyoto University Hospital, Kyoto, Japan
| | | | | | | | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
9
|
Maekawa T. [Contribution to the world of 3D cell products created by bio 3D printing technology and to the life science field]. Nihon Yakurigaku Zasshi 2024; 159:144-149. [PMID: 38692876 DOI: 10.1254/fpj.23049] [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: 05/03/2024]
Abstract
We have been making 3D tissues consist of cells only, based on the corporate philosophy of "contributing to dramatic advances in medical care through the practical application of innovative 3D cell stacking technology." Currently, in the field of regenerative medicine, we are working toward obtaining approval from the Ministry of Health, Labor and Welfare and commercializing large artificial organs that are made from patients' own cells and have functions such as nerve regeneration, osteochondral regeneration, and blood vessels. On the other hand, this three-dimensional cell stacking technology can be extended to technology for culturing cells in an environment similar to the human body, and is expected to serve as a new methodology for evaluating the effects of new products in various fields on living organisms. Therefore, we are planning a business to provide developers of pharmaceuticals, foods, cosmetics, etc. with a small device called "Functional Cell Device (FCD)" that reproduces some of the functions of human organs outside the body. As the first step, we have developed a three-dimensional liver construct (3D mini-liver). The in vitro human liver model has a wide range of usage, such as evaluation of hepatotoxicity of drugs, elucidation of drug metabolism mechanism, and model of liver disease. In this report, we will outline it together with actual examples in regenerative medicine.
Collapse
|
10
|
Hoveizi E. Enhancement of nerve regeneration through schwann cell-mediated healing in a 3D printed polyacrylonitrile conduit incorporating hydrogel and graphene quantum dots: a study on rat sciatic nerve injury model. Biomed Mater 2023; 19:015012. [PMID: 38091624 DOI: 10.1088/1748-605x/ad1576] [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: 09/09/2023] [Accepted: 12/13/2023] [Indexed: 12/22/2023]
Abstract
Despite recent technological advancements, effective healing from sciatic nerve damage remains inadequate. Cell-based therapies offer a promising alternative to autograft restoration for peripheral nerve injuries, and 3D printing techniques can be used to manufacture conduits with controlled diameter and size. In this study, we investigated the potential of Wharton's jelly-derived mesenchymal stem cells (WJMSCs) differentiated into schwann cells, using a polyacrylonitrile (PAN) conduit filled with fibrin hydrogel and graphene quantum dots (GQDs) to promote nerve regeneration in a rat sciatic nerve injury model. We investigated the potential of WJMSCs, extracted from the umbilical cord, to differentiate into schwann cells and promote nerve regeneration in a rat sciatic nerve injury model. WJMSCs were 3D cultured and differentiated into schwann cells within fibrin gel for two weeks. A 3 mm defect was created in the sciatic nerve of the rat model, which was then regenerated using a conduit/fibrin, conduit covered with schwann cells in fibrin/GQDs, GQDs in fibrin, and a control group without any treatment (n= 6/group). At 10 weeks after transplantation, motor and sensory functions and histological improvement were assessed. The WJMSCs were extracted, identified, and differentiated. The differentiated cells expressed typical schwann cell markers, S100 and P75.In vivoinvestigations established the durability and efficacy of the conduit to resist the pressures over two months of implantation. Histological measurements showed conduit efficiency, schwann cell infiltration, and association within the fibrin gel and lumen. Rats treated with the composite hydrogel-filled PAN conduit with GQDs showed significantly higher sensorial recovery than the other groups. Histological results showed that this group had significantly more axon numbers and remyelination than others. Our findings suggest that the conduit/schwann approach has the potential to improve nerve regeneration in peripheral nerve injuries, with future therapeutic implications.
Collapse
Affiliation(s)
- Elham Hoveizi
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| |
Collapse
|
11
|
Lv H, Deng G, Lai J, Yu Y, Chen F, Yao J. Advances in 3D Bioprinting of Biomimetic and Engineered Meniscal Grafts. Macromol Biosci 2023; 23:e2300199. [PMID: 37436941 DOI: 10.1002/mabi.202300199] [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: 05/08/2023] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 07/14/2023]
Abstract
The meniscus plays a crucial role in loads distribution and protection of articular cartilage. Meniscal injury can result in cartilage degeneration, loss of mechanical stability in the knee joint and ultimately lead to arthritis. Surgical interventions provide only short-term pain relief but fail to repair or regenerate the injured meniscus. Emerging tissue engineering approaches based on 3D bioprinting provide alternatives to current surgical methods for meniscus repair. In this review, the current bioprinting techniques employed in developing engineered meniscus grafts are summarized and discuss the latest strategies for mimicking the gradient structure, composition, and viscoelastic properties of native meniscus. Recent progress is highlighted in gene-activated matrices for meniscus regeneration as well. Finally, a perspective is provided on the future development of 3D bioprinting for meniscus repair, emphasizing the potential of this technology to revolutionize meniscus regeneration and improve patient outcomes.
Collapse
Affiliation(s)
- Haiyuan Lv
- Department of Bone and Joint Surgery & Guangxi Key Laboratory of Regenerative Medicine, International Joint Laboratory on Regeneration of Bone and Soft Tissue, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Guotao Deng
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jiaqi Lai
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yin Yu
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Fei Chen
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jun Yao
- Department of Bone and Joint Surgery & Guangxi Key Laboratory of Regenerative Medicine, International Joint Laboratory on Regeneration of Bone and Soft Tissue, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| |
Collapse
|
12
|
Meng Q, Burrell JC, Zhang Q, Le AD. Potential Application of Orofacial MSCs in Tissue Engineering Nerve Guidance for Peripheral Nerve Injury Repair. Stem Cell Rev Rep 2023; 19:2612-2631. [PMID: 37642899 DOI: 10.1007/s12015-023-10609-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2023] [Indexed: 08/31/2023]
Abstract
Injury to the peripheral nerve causes potential loss of sensory and motor functions, and peripheral nerve repair (PNR) remains a challenging endeavor. The current clinical methods of nerve repair, such as direct suture, autografts, and acellular nerve grafts (ANGs), exhibit their respective disadvantages like nerve tension, donor site morbidity, size mismatch, and immunogenicity. Even though commercially available nerve guidance conduits (NGCs) have demonstrated some clinical successes, the overall clinical outcome is still suboptimal, especially for nerve injuries with a large gap (≥ 3 cm) due to the lack of biologics. In the last two decades, the combination of advanced tissue engineering technologies, stem cell biology, and biomaterial science has significantly advanced the generation of a new generation of NGCs incorporated with biological factors or supportive cells, including mesenchymal stem cells (MSCs), which hold great promise to enhance peripheral nerve repair/regeneration (PNR). Orofacial MSCs are emerging as a unique source of MSCs for PNR due to their neural crest-origin and easy accessibility. In this narrative review, we have provided an update on the pathophysiology of peripheral nerve injury and the properties and biological functions of orofacial MSCs. Then we have highlighted the application of orofacial MSCs in tissue engineering nerve guidance for PNR in various preclinical models and the potential challenges and future directions in this field.
Collapse
Affiliation(s)
- Qingyu Meng
- Department of Oral & Maxillofacial Surgery & Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40Th Street, Philadelphia, PA, 19104, USA
| | - Justin C Burrell
- Department of Oral & Maxillofacial Surgery & Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40Th Street, Philadelphia, PA, 19104, USA
| | - Qunzhou Zhang
- Department of Oral & Maxillofacial Surgery & Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40Th Street, Philadelphia, PA, 19104, USA.
| | - Anh D Le
- Department of Oral & Maxillofacial Surgery & Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40Th Street, Philadelphia, PA, 19104, USA.
- Department of Oral & Maxillofacial Surgery, Penn Medicine Hospital of the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA.
| |
Collapse
|
13
|
Rattanapan Y, Narkpetch S, Chareonsirisuthigul T. Upregulation of miR-20a-5p as the Potential MicroRNA Marker in Red Blood Cell Storage Lesion. BIOMED RESEARCH INTERNATIONAL 2023; 2023:5598590. [PMID: 37829050 PMCID: PMC10567411 DOI: 10.1155/2023/5598590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/29/2023] [Accepted: 09/25/2023] [Indexed: 10/14/2023]
Abstract
Background Packed red blood cells (PRBCs) can be preserved for 42 days, and stored PRBCs have slow, dangerous changes over time during storage. miRNA is approximately 22 nucleotides long, a small single-stranded noncoding RNA molecule. miRNA guides by pairing bases with their downstream target mRNA to regulate negative expression. They are essential in many life processes, including cell differentiation, proliferation, and apoptosis. Therefore, miRNA alterations may represent possible biomarkers of PRBC storage lesions. This study is aimed at validating the miR-20a-5p in PRBC storage. Study Design and Methods. A total of 20 PRBC samples were divided into day 1 and day 20 storage groups. Total miRNA was extracted and quantified by probe-based RT-qPCR assays to explore the potential role of miRNAs in PRBC storage lesions. Results Upregulated miR-20a-5p in PRBC storage on day 20 compared to day 1. MiR-20a-5p promoted cell survival, which may affect the downstream regulation and decrease PRBC viability in prolonged storage. Conclusion On this basis, this detection may help to assess the quality of stored PRBCs.
Collapse
Affiliation(s)
- Yanisa Rattanapan
- Medical Technology, School of Allied Health Sciences, Walailak University, Tha Sala, Nakhon Si Thammarat, Thailand
- Hematology and Transfusion Science Research Center, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Sodsai Narkpetch
- Blood Bank, Maharaj Nakhon Si Thammarat Hospital, Nakhon Si Thammarat 80000, Thailand
| | - Takol Chareonsirisuthigul
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
14
|
Yeheyis T, Lemma K, Nuramo A, Musema M, Dolmolo A, Aynalem A, Mekonnen S. Level of modern health-seeking behavior for common childhood illnesses and its associated factors among mothers of under-five children in southern Ethiopia: A community based study. Heliyon 2023; 9:e20121. [PMID: 37810091 PMCID: PMC10559870 DOI: 10.1016/j.heliyon.2023.e20121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 09/12/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
Background Health-seeking behavior is an action taken by an individual who perceives to have a health problem. Many childhood morbidities and mortalities are associated with a low level of a mother's healthcare-seeking behavior. However, there are limited studies about modern health-seeking behavior among mothers of ill under-five children in the study area. Objective To assess the level of modern health-seeking behavior of mothers/caregivers and associated factors for childhood illness in Hawassa city, Sidama, Ethiopia 2021. Methods A community-based cross-sectional study was conducted from November 15 to December 15, 2021, in Hawassa City. Eight kebeles were selected by using simple random sampling methods. A total of 366 mothers with children less than five years were included in this study and an interviewer-administered questionnaire was used to collect data. Data entry, cleaning, and analysis were done by using Statistical Package for Social sciences version 24 and logistic regression was used to determine the presence of association, and significance was declared at p-value <0.05. Result The study found that 70.2% of mothers/caregivers seek modern health care for their child's illnesses. Number of Antenatal care follow up ([AOR(Adjusted Odds Ratio) = 2.106; 95% CI(Confidence Interval) (1.097-4.042)), urban residence ([AOR = 2.688; 95% CI (1.403-5.149)), perceived severity of illness ([AOR2.832; 95% CI1.101-7.290)), four or above birth order (5.501; 95% CI (1.761-17.184)) and symptoms guiding severity of illness ([AOR = 4.664; 95% CI (1.918-11.342)) were associated with modern health-seeking behavior. Conclusion The overall modern health-seeking behaviors of mothers of under-five children are higher than in previous studies. However, a still significant proportion of mothers do not seek modern health care for their ill children. mothers/caregivers' residence, birth order, number of Antenatal care follow-ups, perceived severity of childhood illness, and perceived guiding symptoms of severity are the significant predictors of mothers' healthcare-seeking behavior.
Collapse
Affiliation(s)
- Tomas Yeheyis
- School of Nursing, College of Medicine and Health Sciences, Hawassa University, Ethiopia
| | - Kidanemihret Lemma
- School of Nursing, College of Medicine and Health Sciences, Hawassa University, Ethiopia
| | - Asebech Nuramo
- School of Nursing, College of Medicine and Health Sciences, Hawassa University, Ethiopia
| | - Merema Musema
- School of Nursing, College of Medicine and Health Sciences, Hawassa University, Ethiopia
| | - Abdo Dolmolo
- School of Nursing, College of Medicine and Health Sciences, Hawassa University, Ethiopia
| | - Amdehiwot Aynalem
- School of Nursing, College of Medicine and Health Sciences, Hawassa University, Ethiopia
| | - Shewangizaw Mekonnen
- School of Nursing, College of Medicine and Health Sciences, Hawassa University, Ethiopia
| |
Collapse
|
15
|
Kawai H, Ito A, Kawaguchi A, Nagai-Tanima M, Nakahara R, Xu S, Kuroki H. Ultrasound therapy for a week promotes regeneration and reduces pro-inflammatory macrophages in a rat sciatic nerve autograft model. Sci Rep 2023; 13:11494. [PMID: 37460651 DOI: 10.1038/s41598-023-38630-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 07/12/2023] [Indexed: 07/20/2023] Open
Abstract
Peripheral nerve injury causes long-term motor dysfunction. Ultrasound (US) therapy is expected to accelerate peripheral nerve regeneration. However, its optimal usage and effects on macrophage phenotypes during peripheral nerve regeneration remain unknown. In this study, we investigated the optimal duration of US therapy and its effects on macrophage phenotype. Twenty-seven rats with autologous sciatic nerve grafting were divided into three groups: two received US therapy (1 MHz frequency, intensity of 140 mW/cm2, 20% duty cycle, 5 min/day) for one (US1) or 4 weeks (US4), and one group received sham stimulation. Immunohistochemistry was performed 3 and 7 days after injury in another set of 12 rats. Eight weeks after the injury, the compound muscle action potential amplitude of the gastrocnemius in the US1 and US4 groups was significantly higher than that in the sham group. The toe-spreading test showed functional recovery, whereas the gait pattern during treadmill walking did not recover. There were no significant differences in motor function, histomorphometry, or muscle weight between groups. Immunohistochemistry showed that US therapy decreased the number of pro-inflammatory macrophages seven days after injury. Therefore, US therapy for both one or 4 weeks can similarly promote reinnervation and reduce proinflammatory macrophages in autograft model rats.
Collapse
Affiliation(s)
- Hideki Kawai
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Japan Society for the Promotion of Science, Tokyo, Japan
| | - Akira Ito
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Asuka Kawaguchi
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Momoko Nagai-Tanima
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ryo Nakahara
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Shixuan Xu
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiroshi Kuroki
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
16
|
Otsuka K, Takata T, Sasaki H, Shikano M. Horizon Scanning in Tissue Engineering Using Citation Network Analysis. Ther Innov Regul Sci 2023; 57:810-822. [PMID: 37204641 PMCID: PMC10276778 DOI: 10.1007/s43441-023-00529-x] [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: 09/15/2022] [Accepted: 04/28/2023] [Indexed: 05/20/2023]
Abstract
BACKGROUND Establishing a horizon scanning method is critical for identifying technologies that require new guidelines or regulations. We studied the application of bibliographic citation network analysis to horizon scanning. OBJECTIVE The possibility of applying the proposed method to interdisciplinary fields was investigated with the emphasis on tissue engineering and its example, three-dimensional bio-printing. METHODOLOGY AND RESULTS In all, 233,968 articles on tissue engineering, regenerative medicine, biofabrication, and additive manufacturing published between January 1, 1900 and November 3, 2021 were obtained from the Web of Science Core Collection. The citation network of the articles was analyzed for confirmation that the evolution of 3D bio-printing is reflected by tracking the key articles in the field. However, the results revealed that the major articles on the clinical application of 3D bio-printed products are located in clusters other than that of 3D bio-printers. We investigated the research trends in this field by analyzing the articles published between 2019 and 2021 and detected various basic technologies constituting tissue engineering, including microfluidics and scaffolds such as electrospinning and conductive polymers. The results suggested that the research trend of technologies required for product development and future clinical applications of the product are sometimes detected independently by bibliographic citation network analysis, particularly for interdisciplinary fields. CONCLUSION This method can be applied to the horizon scanning of an interdisciplinary field. However, identifying basic technologies of the targeted field and following the progress of research and the integration process of each component of technology are critical.
Collapse
Affiliation(s)
- Kouhei Otsuka
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Tokyo, Japan
| | - Takuya Takata
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Tokyo, Japan
| | - Hajime Sasaki
- Institute for Future Initiatives, The University of Tokyo, Tokyo, Japan
| | - Mayumi Shikano
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Tokyo, Japan.
| |
Collapse
|
17
|
Takeuchi H, Sakamoto A, Ikeguchi R, Ohta S, Noguchi T, Ando M, Yoshimoto K, Sakamoto D, Matsuda S. Muscle Grafts with Doxorubicin Pretreatment Produce "Empty Tubes" in the Basal Laminae, Promote Contentious Maturation of the Regenerated Axons, and Bridge 20-mm Sciatic Nerve Defects in Rats. J Reconstr Microsurg 2023; 39:120-130. [PMID: 35850137 DOI: 10.1055/s-0042-1750082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND We newly developed a muscle graft that employs a doxorubicin pretreatment technique. The aims of this study were to reveal the biological and morphological features of the muscle tissue in the second week (Study I), to reveal the regeneration outcomes of functional and kinematic assessments of longer-term follow-up (16 weeks, Study II), and to make assessments of the muscle graft with doxorubicin pretreatment in the critical-sized nerve defect model (20 mm, Study III). METHODS A total of 26 adult rats were used in this study. Doxorubicin treatment was accomplished by immersion in a doxorubicin solution for 10 minutes followed by a rinsing procedure. The rats were divided into three groups: the muscle graft with and without doxorubicin pretreatment (M-graft-w-Dox and M-graft-w/o-Dox) groups and the autologous nerve graft (N-graft) group. Assays of apoptosis, immunofluorescent histochemistry including CD68 (macrophage marker), scanning electron microscopy (SEM), morphometrical studies of the regenerated axons, nerve conduction studies, and kinematic studies were performed. RESULTS The M-graft-w-Dox group contained significantly larger numbers of apoptotic cells and CD68-positive cells. SEM revealed the existence of the basal lamina, so called "empty tubes," in the M-graft-w-Dox group. Study II showed contentious maturation of the regenerated axons, especially in the compound muscle action potentials. Study III showed that even at 20 mm, the M-graft-w-Dox group promoted axonal regeneration and functional regeneration. CONCLUSION The M-graft-w-Dox group showed superior regeneration results, and this easy and short-term procedure can expand the muscle graft clinical indication for the treatment of peripheral nerve defects.
Collapse
Affiliation(s)
- Hisataka Takeuchi
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akio Sakamoto
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryosuke Ikeguchi
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Souichi Ohta
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Noguchi
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Maki Ando
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koichi Yoshimoto
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Daichi Sakamoto
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
18
|
Soleymani-Goloujeh M, Hosseini S, Baghaban Eslaminejad M. Advanced Nanotechnology Approaches as Emerging Tools in Cellular-Based Technologies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1409:127-144. [PMID: 35816248 DOI: 10.1007/5584_2022_725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Stem cells are valuable tools in regenerative medicine because they can generate a wide variety of cell types and tissues that can be used to treat or replace damaged tissues and organs. However, challenges related to the application of stem cells in the scope of regenerative medicine have urged scientists to utilize nanomedicine as a prerequisite to circumvent some of these hurdles. Nanomedicine plays a crucial role in this process and manipulates surface biology, the fate of stem cells, and biomaterials. Many attempts have been made to modify cellular behavior and improve their regenerative ability using nano-based strategies. Notably, nanotechnology applications in regenerative medicine and cellular therapies are controversial because of ethical and legal considerations. Therefore, this review describes nanotechnology in cell-based applications and focuses on newly proposed nano-based approaches. Cutting-edge strategies to engineer biological tissues and the ethical, legal, and social considerations of nanotechnology in regenerative nanomedicine applications are also discussed.
Collapse
Affiliation(s)
- Mehdi Soleymani-Goloujeh
- Department of Applied Cell Sciences, Faculty of Basic Sciences and Advanced Medical Technologies, Royan Institute, ACECR, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Samaneh Hosseini
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
19
|
Jo Y, Hwang DG, Kim M, Yong U, Jang J. Bioprinting-assisted tissue assembly to generate organ substitutes at scale. Trends Biotechnol 2023; 41:93-105. [PMID: 35907704 DOI: 10.1016/j.tibtech.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/29/2022] [Accepted: 07/01/2022] [Indexed: 12/27/2022]
Abstract
Various external cues can guide cellular behavior and maturation during developmental processes. Recent studies on bioprinting-assisted tissue engineering have considered this a practical, versatile, and flexible way to provide external cues to developing engineered tissues. An ensemble of multiple external cues can improve the speed and capability of morphogenesis. In this review, we discuss how bioprinting and biomaterials provide multiple guidance to generate micro-sized building blocks with specific shapes and also highlight their applications in tissue assembly toward volumetric tissue and organ generation. Furthermore, we discuss our perspectives on the future translation of bioprinting technologies integrated with artificial intelligence (AI) and robot-assisted apparatus to promote automation, standardization, and clinical translation of bioprinted tissues.
Collapse
Affiliation(s)
- Yeonggwon Jo
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Dong Gyu Hwang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Myungji Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Uijung Yong
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Jinah Jang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea; Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea; Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea; Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
20
|
Griffin KH, Fok SW, Kent Leach J. Strategies to capitalize on cell spheroid therapeutic potential for tissue repair and disease modeling. NPJ Regen Med 2022; 7:70. [PMID: 36494368 PMCID: PMC9734656 DOI: 10.1038/s41536-022-00266-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
Cell therapies offer a tailorable, personalized treatment for use in tissue engineering to address defects arising from trauma, inefficient wound repair, or congenital malformation. However, most cell therapies have achieved limited success to date. Typically injected in solution as monodispersed cells, transplanted cells exhibit rapid cell death or insufficient retention at the site, thereby limiting their intended effects to only a few days. Spheroids, which are dense, three-dimensional (3D) aggregates of cells, enhance the beneficial effects of cell therapies by increasing and prolonging cell-cell and cell-matrix signaling. The use of spheroids is currently under investigation for many cell types. Among cells under evaluation, spheroids formed of mesenchymal stromal cells (MSCs) are particularly promising. MSC spheroids not only exhibit increased cell survival and retained differentiation, but they also secrete a potent secretome that promotes angiogenesis, reduces inflammation, and attracts endogenous host cells to promote tissue regeneration and repair. However, the clinical translation of spheroids has lagged behind promising preclinical outcomes due to hurdles in their formation, instruction, and use that have yet to be overcome. This review will describe the current state of preclinical spheroid research and highlight two key examples of spheroid use in clinically relevant disease modeling. It will highlight techniques used to instruct the phenotype and function of spheroids, describe current limitations to their use, and offer suggestions for the effective translation of cell spheroids for therapeutic treatments.
Collapse
Affiliation(s)
- Katherine H Griffin
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, 95817, USA
- School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Shierly W Fok
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, 95817, USA
| | - J Kent Leach
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, 95817, USA.
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, 95616, USA.
| |
Collapse
|
21
|
Banerjee D, Singh YP, Datta P, Ozbolat V, O'Donnell A, Yeo M, Ozbolat IT. Strategies for 3D bioprinting of spheroids: A comprehensive review. Biomaterials 2022; 291:121881. [DOI: 10.1016/j.biomaterials.2022.121881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/04/2022] [Accepted: 10/23/2022] [Indexed: 11/17/2022]
|
22
|
Namhongsa M, Daranarong D, Sriyai M, Molloy R, Ross S, Ross GM, Tuantranont A, Tocharus J, Sivasinprasasn S, Topham PD, Tighe B, Punyodom W. Surface-Modified Polypyrrole-Coated PLCL and PLGA Nerve Guide Conduits Fabricated by 3D Printing and Electrospinning. Biomacromolecules 2022; 23:4532-4546. [PMID: 36169096 DOI: 10.1021/acs.biomac.2c00626] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The efficiency of nerve guide conduits (NGCs) in repairing peripheral nerve injury is not high enough yet to be a substitute for autografts and is still insufficient for clinical use. To improve this efficiency, 3D electrospun scaffolds (3D/E) of poly(l-lactide-co-ε-caprolactone) (PLCL) and poly(l-lactide-co-glycolide) (PLGA) were designed and fabricated by the combination of 3D printing and electrospinning techniques, resulting in an ideal porous architecture for NGCs. Polypyrrole (PPy) was deposited on PLCL and PLGA scaffolds to enhance biocompatibility for nerve recovery. The designed pore architecture of these "PLCL-3D/E" and "PLGA-3D/E" scaffolds exhibited a combination of nano- and microscale structures. The mean pore size of PLCL-3D/E and PLGA-3D/E scaffolds were 289 ± 79 and 287 ± 95 nm, respectively, which meets the required pore size for NGCs. Furthermore, the addition of PPy on the surfaces of both PLCL-3D/E (PLCL-3D/E/PPy) and PLGA-3D/E (PLGA-3D/E/PPy) led to an increase in their hydrophilicity, conductivity, and noncytotoxicity compared to noncoated PPy scaffolds. Both PLCL-3D/E/PPy and PLGA-3D/E/PPy showed conductivity maintained at 12.40 ± 0.12 and 10.50 ± 0.08 Scm-1 for up to 15 and 9 weeks, respectively, which are adequate for the electroconduction of neuron cells. Notably, the PLGA-3D/E/PPy scaffold showed superior cytocompatibility when compared with PLCL-3D/E/PPy, as evident via the viability assay, proliferation, and attachment of L929 and SC cells. Furthermore, analysis of cell health through membrane leakage and apoptotic indices showed that the 3D/E/PPy scaffolds displayed significant decreases in membrane leakage and reductions in necrotic tissue. Our finding suggests that these 3D/E/PPy scaffolds have a favorable design architecture and biocompatibility with potential for use in peripheral nerve regeneration applications.
Collapse
Affiliation(s)
- Manasanan Namhongsa
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand.,Graduate School, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Donraporn Daranarong
- Science and Technology Research Institute, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Montira Sriyai
- Bioplastics Production Laboratory for Medical Applications, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand.,Center of Excellence in Materials Science and Technology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Robert Molloy
- Center of Excellence in Materials Science and Technology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sukunya Ross
- Center of Excellence in Biomaterials, Department of Chemistry, Faculty of Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Gareth M Ross
- Center of Excellence in Biomaterials, Department of Chemistry, Faculty of Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Adisorn Tuantranont
- National Security and Dual-Use Technology Center, National Science and Technology Development Agency, Khlong Luang 12120, Thailand
| | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sivanan Sivasinprasasn
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Paul D Topham
- Aston Institute of Materials Research, Aston University, Birmingham B4 7ET, United Kingdom
| | - Brian Tighe
- Aston Institute of Materials Research, Aston University, Birmingham B4 7ET, United Kingdom
| | - Winita Punyodom
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand.,Center of Excellence in Materials Science and Technology, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
23
|
Sabrina N, Rizal M, Nurkolis F, Hardinsyah H, Tanner MJ, Gunawan WB, Handoko MN, Mayulu N, Taslim NA, Puspaningtyas DS, Noor SL, Yusuf VM, Permatasari HK, Radu S. Bioactive peptides identification and nutritional status ameliorating properties on malnourished rats of combined eel and soy-based tempe flour. Front Nutr 2022; 9:963065. [PMID: 36245543 PMCID: PMC9554436 DOI: 10.3389/fnut.2022.963065] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/29/2022] [Indexed: 12/23/2022] Open
Abstract
Background and aims A combined eel and soy-based tempe (CEST) flour is rich in nutrients, especially its high amino acid content in which bioactive peptides (BPs) are expected to be found. Hence, this research aimed to identify the BPs of CEST flour and CEST supplementation’s effect on improving nutritional status biomarkers by ameliorating serum protein, hemoglobin, and IGF-1 of malnourished rats. Methods CEST flour with a ratio of eel and soy-based tempe of 1:3.5 was produced by applying the oven drying method. Amino acid sequences from six BPs were analyzed using a protein sequencer and spectrometer-electrospray ionization (MS-ESI). A total of thirty malnourished male Rattus norvegicus aged 3–4 weeks were given low-protein (LP; 4% w/w protein) diet treatment for 4 weeks. Afterward, rats were divided into 3 groups of 10 rats. Group A and B remained on a low-protein diet for 4 weeks, receiving an LP diet and getting doses of CEST of 100 and 200 mg/kg BW, respectively, via oral. Group C or control was given a Normal-protein (NP) diet (23% w/w of protein) and was allowed to feed ad libitum during the trial period without a dose of CEST. Results Six bioactive peptides were found, with WMGPY being the most abundant, along with a DPPH radical scavenging activity of 5.0 mg/mL. The results showed that serum protein, hemoglobin, and IGF-1 of group B were significantly higher compared to groups A and C (p = 0.0021). CEST dose of 200 mg/kg BW was more effective to increase serum levels of protein (p = 0.0052), hemoglobin, and IGF-1 (p < 0.0001) compared to a 100 mg/kg BW dose. Conclusion This indicates that the CEST flour has six bioactive peptides, which may contribute to the improvement of nutritional status biomarkers. To establish its potential impact, a human clinical study is urgently needed.
Collapse
Affiliation(s)
- Nindy Sabrina
- Department of Nutrition, Dietetics, and Food, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, VIC, Australia
- Nutrition Program, Faculty of Food Technology and Health, Sahid University of Jakarta, South Jakarta, Indonesia
| | - Mochammad Rizal
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Fahrul Nurkolis
- Department of Biological Sciences, Faculty of Sciences and Technology, State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga Yogyakarta), Yogyakarta, Indonesia
- *Correspondence: Fahrul Nurkolis,
| | | | - Melvin Junior Tanner
- Department of Nutrition, Faculty of Public Health, University of Indonesia, Depok, Indonesia
| | - William Ben Gunawan
- Department of Nutrition Science, Faculty of Medicine, Diponegoro University, Semarang, Indonesia
| | | | - Nelly Mayulu
- Nutrition and Food, Faculty of Medicine, Sam Ratulangi University, Manado, Indonesia
| | | | - Dwi Sari Puspaningtyas
- Human Nutrition, Bloomberg School of Public Health, The Johns Hopkins University, Baltimore, MD, United States
| | - Sutamara Lasurdi Noor
- Clinical and Public Health Nutrition Programme, University College London, London, United Kingdom
| | - Vincentius Mario Yusuf
- Department of Biochemistry and Biomolecular, Faculty of Medicine, Brawijaya University, Malang, Indonesia
| | - Happy Kurnia Permatasari
- Department of Biochemistry and Biomolecular, Faculty of Medicine, Brawijaya University, Malang, Indonesia
| | - Son Radu
- Department of Food Sciences, Universiti Putra Malaysia, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
24
|
Nerve regeneration using the Bio 3D nerve conduit fabricated with spheroids. J Artif Organs 2022; 25:289-297. [PMID: 35970971 DOI: 10.1007/s10047-022-01358-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/06/2022] [Indexed: 10/15/2022]
Abstract
Autologous nerve grafting is the gold standard method for peripheral nerve injury with defects. Artificial nerve conduits have been developed to prevent morbidity at the harvest site. However, the artificial conduit regeneration capacity is not sufficient. A Bio 3D printer is technology that creates three-dimensional tissue using only cells. Using this technology, a three-dimensional nerve conduit (Bio 3D nerve conduit) was created from several cell spheroids. We reported the first application of the Bio 3D nerve conduit for peripheral nerve injury. A Bio 3D nerve conduit that was created from several cells promotes peripheral nerve regeneration. The Bio 3D nerve conduit may be useful clinically to treat peripheral nerve defects.
Collapse
|
25
|
Tafti MF, Aghamollaei H, Moghaddam MM, Jadidi K, Alio JL, Faghihi S. Emerging tissue engineering strategies for the corneal regeneration. J Tissue Eng Regen Med 2022; 16:683-706. [PMID: 35585479 DOI: 10.1002/term.3309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 04/16/2022] [Accepted: 04/19/2022] [Indexed: 11/10/2022]
Abstract
Cornea as the outermost layer of the eye is at risk of various genetic and environmental diseases that can damage the cornea and impair vision. Corneal transplantation is among the most applicable surgical procedures for repairing the defected tissue. However, the scarcity of healthy tissue donations as well as transplantation failure has remained as the biggest challenges in confront of corneal grafting. Therefore, alternative approaches based on stem-cell transplantation and classic regenerative medicine have been developed for corneal regeneration. In this review, the application and limitation of the recently-used advanced approaches for regeneration of cornea are discussed. Additionally, other emerging powerful techniques such as 5D printing as a new branch of scaffold-based technologies for construction of tissues other than the cornea are highlighted and suggested as alternatives for corneal reconstruction. The introduced novel techniques may have great potential for clinical applications in corneal repair including disease modeling, 3D pattern scheming, and personalized medicine.
Collapse
Affiliation(s)
- Mahsa Fallah Tafti
- Stem Cell and Regenerative Medicine Group, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Hossein Aghamollaei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Khosrow Jadidi
- Vision Health Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Jorge L Alio
- Department of Research and Development, VISSUM, Alicante, Spain.,Cornea, Cataract and Refractive Surgery Department, VISSUM, Alicante, Spain.,Department of Pathology and Surgery, Division of Ophthalmology, Faculty of Medicine, Miguel Hernández University, Alicante, Spain
| | - Shahab Faghihi
- Stem Cell and Regenerative Medicine Group, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
26
|
Yan Y, Yao R, Zhao J, Chen K, Duan L, Wang T, Zhang S, Guan J, Zheng Z, Wang X, Liu Z, Li Y, Li G. Implantable nerve guidance conduits: Material combinations, multi-functional strategies and advanced engineering innovations. Bioact Mater 2022; 11:57-76. [PMID: 34938913 PMCID: PMC8665266 DOI: 10.1016/j.bioactmat.2021.09.030] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/17/2021] [Accepted: 09/26/2021] [Indexed: 01/15/2023] Open
Abstract
Nerve guidance conduits (NGCs) have attracted much attention due to their great necessity and applicability in clinical use for the peripheral nerve repair. Great efforts in recent years have been devoted to the development of high-performance NGCs using various materials and strategies. The present review provides a comprehensive overview of progress in the material innovation, structural design, advanced engineering technologies and multi functionalization of state-of-the-art nerve guidance conduits NGCs. Abundant advanced engineering technologies including extrusion-based system, laser-based system, and novel textile forming techniques in terms of weaving, knitting, braiding, and electrospinning techniques were also analyzed in detail. Findings arising from this review indicate that the structural mimetic NGCs combined with natural and synthetic materials using advanced manufacturing technologies can make full use of their complementary advantages, acquiring better biomechanical properties, chemical stability and biocompatibility. Finally, the existing challenges and future opportunities of NGCs were put forward aiming for further research and applications of NGCs.
Collapse
Affiliation(s)
- Yixin Yan
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, 215123, China
- Department of Materials, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Ruotong Yao
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, 215123, China
| | - Jingyuan Zhao
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, 215123, China
| | - Kaili Chen
- Department of Materials, Imperial College London, SW7 2AZ, UK
| | - Lirong Duan
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, 215123, China
| | - Tian Wang
- Wilson College of Textiles, North Carolina State University, Raleigh, 27695, USA
| | - Shujun Zhang
- Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Jinping Guan
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, 215123, China
| | - Zhaozhu Zheng
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, 215123, China
| | - Xiaoqin Wang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, 215123, China
| | - Zekun Liu
- Department of Materials, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Yi Li
- Department of Materials, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Gang Li
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, 215123, China
| |
Collapse
|
27
|
Liu K, Yan L, Li R, Song Z, Ding J, Liu B, Chen X. 3D Printed Personalized Nerve Guide Conduits for Precision Repair of Peripheral Nerve Defects. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103875. [PMID: 35182046 PMCID: PMC9036027 DOI: 10.1002/advs.202103875] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 12/25/2021] [Indexed: 05/07/2023]
Abstract
The treatment of peripheral nerve defects has always been one of the most challenging clinical practices in neurosurgery. Currently, nerve autograft is the preferred treatment modality for peripheral nerve defects, while the therapy is constantly plagued by the limited donor, loss of donor function, formation of neuroma, nerve distortion or dislocation, and nerve diameter mismatch. To address these clinical issues, the emerged nerve guide conduits (NGCs) are expected to offer effective platforms to repair peripheral nerve defects, especially those with large or complex topological structures. Up to now, numerous technologies are developed for preparing diverse NGCs, such as solvent casting, gas foaming, phase separation, freeze-drying, melt molding, electrospinning, and three-dimensional (3D) printing. 3D printing shows great potential and advantages because it can quickly and accurately manufacture the required NGCs from various natural and synthetic materials. This review introduces the application of personalized 3D printed NGCs for the precision repair of peripheral nerve defects and predicts their future directions.
Collapse
Affiliation(s)
- Kai Liu
- Department of Hand and Foot SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Lesan Yan
- Biomedical Materials and Engineering Research Center of Hubei ProvinceState Key Laboratory of Advanced Technology for Materials Synthesis and ProcessingWuhan University of Technology122 Luoshi RoadWuhan430070P. R. China
| | - Ruotao Li
- Department of Hand and Foot SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Zhiming Song
- Department of Sports MedicineThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
- State Key Laboratory of Molecular Engineering of PolymersFudan University220 Handan RoadShanghai200433P. R. China
| | - Bin Liu
- Department of Hand and Foot SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| |
Collapse
|
28
|
Stocco E, Porzionato A, De Rose E, Barbon S, Caro RD, Macchi V. Meniscus regeneration by 3D printing technologies: Current advances and future perspectives. J Tissue Eng 2022; 13:20417314211065860. [PMID: 35096363 PMCID: PMC8793124 DOI: 10.1177/20417314211065860] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/24/2021] [Indexed: 01/10/2023] Open
Abstract
Meniscal tears are a frequent orthopedic injury commonly managed by conservative
strategies to avoid osteoarthritis development descending from altered
biomechanics. Among cutting-edge approaches in tissue engineering, 3D printing
technologies are extremely promising guaranteeing for complex biomimetic
architectures mimicking native tissues. Considering the anisotropic
characteristics of the menisci, and the ability of printing over structural
control, it descends the intriguing potential of such vanguard techniques to
meet individual joints’ requirements within personalized medicine. This
literature review provides a state-of-the-art on 3D printing for meniscus
reconstruction. Experiences in printing materials/technologies, scaffold types,
augmentation strategies, cellular conditioning have been compared/discussed;
outcomes of pre-clinical studies allowed for further considerations. To date,
translation to clinic of 3D printed meniscal devices is still a challenge:
meniscus reconstruction is once again clear expression of how the integration of
different expertise (e.g., anatomy, engineering, biomaterials science, cell
biology, and medicine) is required to successfully address native tissues
complexities.
Collapse
Affiliation(s)
- Elena Stocco
- Department of Neuroscience, Section of Human Anatomy, University of Padova, Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria, Padova, Italy
| | - Andrea Porzionato
- Department of Neuroscience, Section of Human Anatomy, University of Padova, Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria, Padova, Italy
| | - Enrico De Rose
- Department of Neuroscience, Section of Human Anatomy, University of Padova, Padova, Italy
| | - Silvia Barbon
- Department of Neuroscience, Section of Human Anatomy, University of Padova, Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria, Padova, Italy
| | - Raffaele De Caro
- Department of Neuroscience, Section of Human Anatomy, University of Padova, Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria, Padova, Italy
| | - Veronica Macchi
- Department of Neuroscience, Section of Human Anatomy, University of Padova, Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria, Padova, Italy
| |
Collapse
|
29
|
Establishment of large canine hepatocyte spheroids by mixing vascular endothelial cells and canine adipose-derived mesenchymal stem cells. Regen Ther 2022; 19:1-8. [PMID: 35024388 PMCID: PMC8728312 DOI: 10.1016/j.reth.2021.11.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/05/2021] [Accepted: 11/24/2021] [Indexed: 11/20/2022] Open
Abstract
Introduction Differentiation of hepatocytes and culture methods have been investigated in dogs as a tool to establish liver transplant and drug metabolism examination systems. However, mass culture techniques for canine hepatocytes (cHep) have not been investigated, and it is necessary to construct a suitable culture system. Recently, a protocol called Bud production has attracted attention, and a mixed culture of human and mouse hepatocytes, stem cells, and artificial blood vessels significantly improved the size and formation ratio of spheroids. The purpose of this study was to investigate and improve the in vitro culture of cHep by mixing canine adipose-derived mesenchymal stem cells (cASCs) and human umbilical vein endothelial cells (HUVECs). Methods Spheroid formation ratio and histological examination were evaluated among four culture methods, including cHep alone, two-mix (cHep + cASCs and cHep + HUVEC), and three-mix (cHep + HUVEC + cASCs), on days 0, 4, and 7. Expression levels of liver-related genes (ALB, AFP, α1-AT, CDH1, CYP2E1, CYP3A12, and TAT) were evaluated by quantitative real-time polymerase chain reaction (RT-PCR). Protein expression of albumin, vimentin, and von Willebrand Factor (vWF) was investigated to confirm the location of the hepatocytes. Results The ratio of spheroid formation was 60.2% in the three-mix culture and was significantly improved compared with cHep alone (5.9%) and two-mix; cHep + cASCs (36.2%) and cHep + HUVEC (26.4%) (P < 0.001). Histological evaluation revealed that the three-mix spheroids formed large canine hepatocyte spheroids (LcHS), and hepatocytes were distributed in the center of the spheroids. Quantitative gene expression analysis of LcHS showed that liver-related genes expression were maintained the same levels with that of a culture of cHep alone from days 4-7. Conclusion These results revealed that the three-mix culture method using cHep, HUVECs, and cASCs was capable of promoting LcHS without impairing liver function in cHep, suggesting that LcHS could be used for the application of high-volume culture techniques in dogs.
Collapse
Key Words
- 3D, three-dimensions
- AFP, α-fetoprotein
- ALB, albumin
- CD, cluster of differentiation
- CDH1, cadherin-1 (epithelital-cadherin)
- CYP, cytochrome
- Canine
- DILI, drug induced liver injury
- EGM, endothelial cell growth medium
- FBS, fetal bovine serum
- HGM, hepatocyte growth medium
- HTM, hepatocyte thawing medium
- HUVEC, human umbilical vein endothelial cells
- Hepatocyte
- LcHS, large canine hepatocyte spheroids
- MSCGM, mesenchymal stem cell growth medium
- Mesenchymal stem cells
- PBS, phosphate-buffered saline
- SF-HGM, hepatocyte growth medium for spheroid formation
- Spheroids
- TAT, tyrosine aminotransferase
- Three-dimensions
- cASC, canine adipose-derived mesenchymal stem cells
- cHep, canine primary-cultured hepatocytes
- hFGF, human fibroblast growth factor
- iPSC, induced pluripotent stem cells
- rf-HGF, recombinant ferine hepatocyte growth factor
- α1-AT, α1-antitrypsin
Collapse
|
30
|
Hibbitts AJ, Kočí Z, Kneafsey S, Matsiko A, Žilić L, Dervan A, Hinton P, Chen G, Cavanagh B, Dowling J, McCoy C, Buckley CT, Archibald SJ, O'Brien FJ. Multi-Factorial Nerve Guidance Conduit Engineering Improves Outcomes in Inflammation, Angiogenesis and Large Defect Nerve Repair. Matrix Biol 2022; 106:34-57. [PMID: 35032612 DOI: 10.1016/j.matbio.2022.01.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 11/13/2021] [Accepted: 01/05/2022] [Indexed: 12/20/2022]
Abstract
Nerve guidance conduits (NGCs) are sub-optimal for long-distance injuries with inflammation and poor vascularization related to poor axonal repair. This study used a multi-factorial approach to create an optimized biomaterial NGC to address each of these issues. Through stepwise optimization, a collagen-chondroitin-6-sulphate (Coll-CS) biomaterial was functionalized with extracellular matrix (ECM) components; fibronectin, laminin 1 and laminin 2 (FibL1L2) in specific ratios. A snap-cooled freeze-drying process was then developed with optimal pore architecture and alignment to guide axonal bridging. Culture of adult rat dorsal root ganglia on NGCs demonstrated significant improvements in inflammation, neurogenesis and angiogenesis in the specific Fib:L1:L2 ratio of 1:4:1. In clinically relevant, large 15 mm rat sciatic nerve defects, FibL1L2-NGCs demonstrated significant improvements in axonal density and angiogenesis compared to unmodified NGCs with functional equivalence to autografts. Therefore, a multiparameter ECM-driven strategy can significantly improve axonal repair across large defects, without exogenous cells or growth factors.
Collapse
Affiliation(s)
- Alan J Hibbitts
- Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Zuzana Kočí
- Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Simone Kneafsey
- Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Amos Matsiko
- Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Leyla Žilić
- Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Adrian Dervan
- Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Paige Hinton
- Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Gang Chen
- Department of Physiology and Medical Physics, Centre for the Study of Neurological Disorders, Microsurgical Research and Training Facility (MRTF), RCSI, Dublin, Ireland
| | | | - Jennifer Dowling
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, D02 YN77 Dublin, Ireland
| | - Claire McCoy
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, D02 YN77 Dublin, Ireland
| | - Conor T Buckley
- Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | | | - Fergal J O'Brien
- Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| |
Collapse
|
31
|
Jeon S, Lee SH, Ahmed SB, Han J, Heo SJ, Kang HW. 3D cell aggregate printing technology and its applications. Essays Biochem 2021; 65:467-480. [PMID: 34223609 PMCID: PMC11293493 DOI: 10.1042/ebc20200128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/14/2021] [Accepted: 06/08/2021] [Indexed: 12/27/2022]
Abstract
Various cell aggregate culture technologies have been developed and actively applied to tissue engineering and organ-on-a-chip. However, the conventional culture technologies are labor-intensive, and their outcomes are highly user dependent. In addition, the technologies cannot be used to produce three-dimensional (3D) complex tissues. In this regard, 3D cell aggregate printing technology has attracted increased attention from many researchers owing to its 3D processability. The technology allows the fabrication of 3D freeform constructs using multiple types of cell aggregates in an automated manner. Technological advancement has resulted in the development of a printing technology with a high resolution of approximately 20 μm in 3D space. A high-speed printing technology that can print a cell aggregate in milliseconds has also been introduced. The developed aggregate printing technologies are being actively applied to produce various types of engineered tissues. Although various types of high-performance printing technologies have been developed, there are still some technical obstacles in the fabrication of engineered tissues that mimic the structure and function of native tissues. This review highlights the central importance and current technical level of 3D cell aggregate printing technology, and their applications to tissue/disease models, artificial tissues, and drug-screening platforms. The paper also discusses the remaining hurdles and future directions of the printing processes.
Collapse
Affiliation(s)
- Seunggyu Jeon
- School of Life Sciences, Ulsan National Institute of Science and Technology, 50, UNIST-gil, Ulju-gun 44919, Ulsan, South Korea
| | - Se-Hwan Lee
- School of Life Sciences, Ulsan National Institute of Science and Technology, 50, UNIST-gil, Ulju-gun 44919, Ulsan, South Korea
- Department of Orthopaedic Surgery, University of Pennsylvania, 36 Street and Hamilton Walk, Philadelphia, PA 19104, U.S.A
| | - Saeed B. Ahmed
- Department of Orthopaedic Surgery, University of Pennsylvania, 36 Street and Hamilton Walk, Philadelphia, PA 19104, U.S.A
| | - Jonghyeuk Han
- School of Life Sciences, Ulsan National Institute of Science and Technology, 50, UNIST-gil, Ulju-gun 44919, Ulsan, South Korea
| | - Su-Jin Heo
- Department of Orthopaedic Surgery, University of Pennsylvania, 36 Street and Hamilton Walk, Philadelphia, PA 19104, U.S.A
| | - Hyun-Wook Kang
- School of Life Sciences, Ulsan National Institute of Science and Technology, 50, UNIST-gil, Ulju-gun 44919, Ulsan, South Korea
| |
Collapse
|
32
|
Arai K, Kitsuka T, Nakayama K. Scaffold-based and scaffold-free cardiac constructs for drug testing. Biofabrication 2021; 13. [PMID: 34233316 DOI: 10.1088/1758-5090/ac1257] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 07/07/2021] [Indexed: 12/24/2022]
Abstract
The safety and therapeutic efficacy of new drugs are tested in experimental animals. However, besides being a laborious, costly process, differences in drug responses between humans and other animals and potential cardiac adverse effects lead to the discontinued development of new drugs. Thus, alternative approaches to animal tests are needed. Cardiotoxicity and responses of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to drugs are conventionally evaluated by cell seeding and two-dimensional (2D) culture, which allows measurements of field potential duration and the action potentials of CMs using multielectrode arrays. However, 2D-cultured hiPSC-CMs lack 3D spatial adhesion, and have fewer intercellular and extracellular matrix interactions, as well as different contractile behavior from CMsin vivo. This issue has been addressed using tissue engineering to fabricate three-dimensional (3D) cardiac constructs from hiPSC-CMs culturedin vitro. Tissue engineering can be categorized as scaffold-based and scaffold-free. In scaffold-based tissue engineering, collagen and fibrin gel scaffolds comprise a 3D culture environment in which seeded cells exhibit cardiac-specific functions and drug responses, whereas 3D cardiac constructs fabricated by tissue engineering without a scaffold have high cell density and form intercellular interactions. This review summarizes the characteristics of scaffold-based and scaffold-free cardiac tissue engineering and discusses the applications of fabricated cardiac constructs to drug screening.
Collapse
Affiliation(s)
- Kenichi Arai
- Center for Regenerative Medicine Research, Faculty of Medicine, Saga University, Saga, Japan.,Department of Clinical Biomaterial Applied Science, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Takahiro Kitsuka
- Department of Cardiovascular Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Koichi Nakayama
- Center for Regenerative Medicine Research, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
33
|
Yurie H, Ikeguchi R, Aoyama T, Tanaka M, Oda H, Takeuchi H, Mitsuzawa S, Ando M, Yoshimoto K, Noguchi T, Akieda S, Nakayama K, Matsuda S. Bio 3D Conduits Derived from Bone Marrow Stromal Cells Promote Peripheral Nerve Regeneration. Cell Transplant 2021; 29:963689720951551. [PMID: 32830545 PMCID: PMC7784509 DOI: 10.1177/0963689720951551] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We previously reported that a nerve conduit created from fibroblasts promotes nerve regeneration in a rat sciatic nerve model. This study aims to determine whether a nerve conduit created from bone marrow stromal cells (BMSCs) can promote nerve regeneration. Primary BMSCs were isolated from femur bone marrow of two Lewis rats, and cells at passages 4–7 were used. We created seven Bio 3D nerve conduits from BMSCs using a Bio-3D Printer. The conduits were transplanted to other Lewis rats to bridge 5-mm right sciatic nerve gaps (Bio 3D group, n = 7). We created two control groups: a silicone group (S group, n = 5) in which the same nerve gap was bridged with a silicone tube, and a silicone cell group (SC group, n = 5) in which the gap was bridged with a BMSC injection. Twelve weeks after transplantation, nerve regeneration was evaluated functionally and morphologically. In addition, PKH26-labeled BMSCs were used to fabricate a Bio 3D conduit that was transplanted for cell trafficking analysis. Electrophysiological study, kinematic analysis, wet muscle weight, and morphological parameters showed significantly better nerve regeneration in the Bio 3D group than in the S group or SC group. In immunohistochemical studies, sections from the Bio 3D group contained abundant S-100-positive cells. In cell trafficking analysis, PKH26-positive cells stained positive for the Schwann cell markers S-100, p75NTR, and GFAP. Bio 3D nerve conduits created from BMSCs can promote peripheral nerve regeneration in a rat sciatic nerve model through BMSC differentiation into Schwann-like cells.
Collapse
Affiliation(s)
- Hirofumi Yurie
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryosuke Ikeguchi
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomoki Aoyama
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mai Tanaka
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroki Oda
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hisataka Takeuchi
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sadaki Mitsuzawa
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Maki Ando
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koichi Yoshimoto
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takashi Noguchi
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Koichi Nakayama
- Department of Regenerative Medicine and Biomedical Engineering, 13030Saga University, Saga, Japan
| | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
34
|
3D bioprinting in tissue engineering and regenerative medicine. Cell Tissue Bank 2021; 23:199-212. [PMID: 34021839 DOI: 10.1007/s10561-021-09936-6] [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: 04/07/2021] [Accepted: 05/14/2021] [Indexed: 10/21/2022]
Abstract
This review paper is primarily focused on bioprinting technology for biomedical applications. Bioprinting can be utilized for fabrication of wide range of tissue, based on which this chapter describes in detail its application in tissue regeneration. Further, the difficulties and potential in developing a construct for tissue regeneration are discussed herein. In this review paper, application of 3D bioprinting in tissue regeneration will be discussed in depth.
Collapse
|
35
|
Selim OA, Lakhani S, Midha S, Mosahebi A, Kalaskar DM. Three-Dimensional Engineered Peripheral Nerve: Toward a New Era of Patient-Specific Nerve Repair Solutions. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:295-335. [PMID: 33593147 DOI: 10.1089/ten.teb.2020.0355] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Reconstruction of peripheral nerve injuries (PNIs) with substance loss remains challenging because of limited treatment solutions and unsatisfactory patient outcomes. Currently, nerve autografting is the first-line management choice for bridging critical-sized nerve defects. The procedure, however, is often complicated by donor site morbidity and paucity of nerve tissue, raising a quest for better alternatives. The application of other treatment surrogates, such as nerve guides, remains questionable, and it is inefficient in irreducible nerve gaps. More importantly, these strategies lack customization for personalized patient therapy, which is a significant drawback of these nerve repair options. This negatively impacts the fascicle-to-fascicle regeneration process, critical to restoring the physiological axonal pathway of the disrupted nerve. Recently, the use of additive manufacturing (AM) technologies has offered major advancements to the bioengineering solutions for PNI therapy. These techniques aim at reinstating the native nerve fascicle pathway using biomimetic approaches, thereby augmenting end-organ innervation. AM-based approaches, such as three-dimensional (3D) bioprinting, are capable of biofabricating 3D-engineered nerve graft scaffolds in a patient-specific manner with high precision. Moreover, realistic in vitro models of peripheral nerve tissues that represent the physiologically and functionally relevant environment of human organs could also be developed. However, the technology is still nascent and faces major translational hurdles. In this review, we spotlighted the clinical burden of PNIs and most up-to-date treatment to address nerve gaps. Next, a summarized illustration of the nerve ultrastructure that guides research solutions is discussed. This is followed by a contrast of the existing bioengineering strategies used to repair peripheral nerve discontinuities. In addition, we elaborated on the most recent advances in 3D printing and biofabrication applications in peripheral nerve modeling and engineering. Finally, the major challenges that limit the evolution of the field along with their possible solutions are also critically analyzed.
Collapse
Affiliation(s)
- Omar A Selim
- Department of Surgical Biotechnology, Division of Surgery and Interventional Sciences, Royal Free Hospital, University College London (UCL), London, United Kingdom
| | - Saad Lakhani
- Department of Surgical Biotechnology, Division of Surgery and Interventional Sciences, Royal Free Hospital, University College London (UCL), London, United Kingdom
| | - Swati Midha
- Department of Surgical Biotechnology, Division of Surgery and Interventional Sciences, Royal Free Hospital, University College London (UCL), London, United Kingdom.,Department of Surgical Biotechnology, Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi, India
| | - Afshin Mosahebi
- Department of Plastic Surgery, Royal Free Hospital, University College London (UCL), London, United Kingdom
| | - Deepak M Kalaskar
- Department of Surgical Biotechnology, Division of Surgery and Interventional Sciences, Royal Free Hospital, University College London (UCL), London, United Kingdom.,Department of Surgical Biotechnology, Institute of Orthopaedics and Musculoskeletal Science, Royal National Orthopaedic Hospital, University College London (UCL), Stanmore, United Kingdom
| |
Collapse
|
36
|
Recent advances in bioprinting technologies for engineering different cartilage-based tissues. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 123:112005. [PMID: 33812625 DOI: 10.1016/j.msec.2021.112005] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/19/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023]
Abstract
Inadequate self-repair and regenerative efficiency of the cartilage tissues has motivated the researchers to devise advanced and effective strategies to resolve this issue. Introduction of bioprinting to tissue engineering has paved the way for fabricating complex biomimetic engineered constructs. In this context, the current review gears off with the discussion of standard and advanced 3D/4D printing technologies and their implications for the repair of different cartilage tissues, namely, articular, meniscal, nasoseptal, auricular, costal, and tracheal cartilage. The review is then directed towards highlighting the current stem cell opportunities. On a concluding note, associated critical issues and prospects for future developments, particularly in this sphere of personalized medicines have been discussed.
Collapse
|
37
|
Hamada T, Nakamura A, Soyama A, Sakai Y, Miyoshi T, Yamaguchi S, Hidaka M, Hara T, Kugiyama T, Takatsuki M, Kamiya A, Nakayama K, Eguchi S. Bile duct reconstruction using scaffold-free tubular constructs created by Bio-3D printer. Regen Ther 2021; 16:81-89. [PMID: 33732817 PMCID: PMC7921183 DOI: 10.1016/j.reth.2021.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/16/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
Introduction Biliary strictures after bile duct injury or duct-to-duct biliary reconstruction are serious complications that markedly reduce patients’ quality of life because their treatment involves periodic stent replacements. This study aimed to create a scaffold-free tubular construct as an interposition graft to treat biliary complications. Methods Scaffold-free tubular constructs of allogeneic pig fibroblasts, that is, fibroblast tubes, were created using a Bio-3D Printer and implanted into pigs as interposition grafts for duct-to-duct biliary reconstruction. Results Although the fibroblast tube was weaker than the native bile duct, it was sufficiently strong to enable suturing. The pigs' serum hepatobiliary enzyme levels remained stable during the experimental period. Micro-computed tomography showed no biliary strictures, no biliary leakages, and no intrahepatic bile duct dilations. The tubular structure was retained in all resected specimens, and the fibroblasts persisted at the graft sites. Immunohistochemical analyses revealed angiogenesis in the fibroblast tube and absence of extensions of the biliary epithelium into the fibroblast tube's lumen. Conclusions This study's findings demonstrated successful reconstruction of the extrahepatic bile duct with a scaffold-free tubular construct created from pig fibroblasts using a novel Bio-3D Printer. This construct could provide a novel regenerative treatment for patients with hepatobiliary diseases.
Collapse
Key Words
- ALP, alkaline phosphatase
- ALT, alanine aminotransferase
- AST, aspartate aminotransferase
- Artificial bile duct
- Bio-3D printer
- Cr, creatinine
- DMEM, Dulbecco's Modified Eagle's Medium
- EDTA, trypsin-ethylenediaminetetraacetic acid
- FBS, fetal bovine serum
- IBDI, iatrogenic bile duct injury
- KCL, potassium chloride
- LDLT, living donor liver transplantation
- PBS, phosphate-buffered saline
- QOL, quality of life
- Reconstruction
- Scaffold-free tubular construct
- T-Bil, total bilirubin
- γ-GTP, γ-glutamyl transpeptidase
Collapse
Affiliation(s)
- Takashi Hamada
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Japan
| | - Anna Nakamura
- Department of Regenerative Medicine and Biomedical Engineering, Faculty of Medicine, Saga University, Japan
| | - Akihiko Soyama
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Japan
| | - Yusuke Sakai
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Japan.,Department of Chemical Engineering, Faculty of Engineering, Graduate School, Kyushu University, Japan
| | - Takayuki Miyoshi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Japan
| | - Shun Yamaguchi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Japan
| | - Masaaki Hidaka
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Japan
| | - Takanobu Hara
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Japan
| | - Tota Kugiyama
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Japan
| | - Mitsuhisa Takatsuki
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Japan
| | - Akihide Kamiya
- Department of Molecular Life Sciences, Tokai University School of Medicine, Japan
| | - Koichi Nakayama
- Department of Regenerative Medicine and Biomedical Engineering, Faculty of Medicine, Saga University, Japan
| | - Susumu Eguchi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Japan
| |
Collapse
|
38
|
Ono T, Tomokiyo A, Ipposhi K, Yamashita K, Alhasan MA, Miyazaki Y, Kunitomi Y, Tsuchiya A, Ishikawa K, Maeda H. Generation of biohybrid implants using a multipotent human periodontal ligament cell line and bioactive core materials. J Cell Physiol 2021; 236:6742-6753. [PMID: 33604904 DOI: 10.1002/jcp.30336] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/15/2022]
Abstract
We aimed to generate periodontal ligament (PDL) tissue-like structures from a multipotent human PDL cell line using three-dimensional (3D) bioprinting technology and to incorporate these structures with bioactive core materials to develop a new biohybrid implant system. After 3D bioprinting, single-cell spheroids were able to form 3D tubular structures (3DTBs). We established three types of complexes using 3DTBs and different core materials: 3DTB-titanium core (TIC), 3DTB-hydroxyapatite core (HAC), and 3DTB without a core material (WOC). The expressions of PDL-, angiogenesis-, cementum-, and bone-related genes were significantly increased in the three complexes compared with monolayer-cultured cells. Abundant collagen fibers and cells positive for the above markers were confirmed in the three complexes. However, more positive cells were detected in HAC than in WOC or TIC. The present results suggest that 3D-bioprinted structures and hydroxyapatite core materials can function similarly to the PDL and may be useful for the development of a new biohybrid implant system.
Collapse
Affiliation(s)
- Taiga Ono
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan.,Department of Endodontology, Kyushu University Hospital, Fukuoka, Japan
| | - Atsushi Tomokiyo
- Department of Endodontology, Kyushu University Hospital, Fukuoka, Japan
| | - Keita Ipposhi
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Kozue Yamashita
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - M Anas Alhasan
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | | | | | - Akira Tsuchiya
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Kunio Ishikawa
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Hidefumi Maeda
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan.,Department of Endodontology, Kyushu University Hospital, Fukuoka, Japan
| |
Collapse
|
39
|
Yoshimatsu M, Ohnishi H, Zhao C, Hayashi Y, Kuwata F, Kaba S, Okuyama H, Kawai Y, Hiwatashi N, Kishimoto Y, Sakamoto T, Ikeya M, Omori K. In vivo regeneration of rat laryngeal cartilage with mesenchymal stem cells derived from human induced pluripotent stem cells via neural crest cells. Stem Cell Res 2021; 52:102233. [PMID: 33607469 DOI: 10.1016/j.scr.2021.102233] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/21/2021] [Accepted: 02/04/2021] [Indexed: 12/18/2022] Open
Abstract
The laryngotracheal cartilage is a cardinal framework for the maintenance of the airway for breathing, which occasionally requires reconstruction. Because hyaline cartilage has a poor intrinsic regenerative ability, various regenerative approaches have been attempted to regenerate laryngotracheal cartilage. The use of autologous mesenchymal stem cells (MSCs) for cartilage regeneration has been widely investigated. However, long-term culture may limit proliferative capacity. Human-induced pluripotent stem cell-derived MSCs (iMSCs) can circumvent this problem due to their unlimited proliferative capacity. This study aimed to investigate the efficacy of iMSCs in the regeneration of thyroid cartilage in immunodeficient rats. Herein, we induced iMSCs through neural crest cell intermediates. For the relevance to prospective future clinical application, induction was conducted under xeno-free/serum-free conditions. Then, clumps fabricated from an iMSC/extracellular matrix complex (C-iMSC) were transplanted into thyroid cartilage defects in immunodeficient rats. Histological examinations revealed cartilage-like regenerated tissue and human nuclear antigen (HNA)-positive surviving transplanted cells in the regenerated lesion. HNA-positive cells co-expressed SOX9, and type II collagen was identified around HNA-positive cells. These results indicated that the transplanted C-iMSCs promoted thyroid cartilage regeneration and some of the iMSCs differentiated into chondrogenic lineage cells. Induced MSCs may be a promising candidate cell therapy for human laryngotracheal reconstruction.
Collapse
Affiliation(s)
- Masayoshi Yoshimatsu
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Hiroe Ohnishi
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Chengzhu Zhao
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Yasuyuki Hayashi
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Fumihiko Kuwata
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinji Kaba
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hideaki Okuyama
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshitaka Kawai
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nao Hiwatashi
- Department of Otolaryngology, Kyoto-Katsura Hospital, Kyoto, Japan
| | - Yo Kishimoto
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Tatsunori Sakamoto
- Department of Otorhinolaryngology, Shimane University Faculty of Medicine, Shimane, Japan
| | - Makoto Ikeya
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Koichi Omori
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
40
|
Ando M, Ikeguchi R, Aoyama T, Tanaka M, Noguchi T, Miyazaki Y, Akieda S, Nakayama K, Matsuda S. Long-Term Outcome of Sciatic Nerve Regeneration Using Bio3D Conduit Fabricated from Human Fibroblasts in a Rat Sciatic Nerve Model. Cell Transplant 2021; 30:9636897211021357. [PMID: 34105391 PMCID: PMC8193652 DOI: 10.1177/09636897211021357] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 11/17/2022] Open
Abstract
Previously, we developed a Bio3D conduit fabricated from human fibroblasts and reported a significantly better outcome compared with artificial nerve conduit in the treatment of rat sciatic nerve defect. The purpose of this study is to investigate the long-term safety and nerve regeneration of Bio3D conduit compared with treatments using artificial nerve conduit and autologous nerve transplantation.We used 15 immunodeficient rats and randomly divided them into three groups treated with Bio3D (n = 5) conduit, silicon tube (n = 5), and autologous nerve transplantation (n = 5). We developed Bio3D conduits composed of human fibroblasts and bridged the 5 mm nerve gap created in the rat sciatic nerve. The same procedures were performed to bridge the 5 mm gap with a silicon tube. In the autologous nerve group, we removed the 5 mm sciatic nerve segment and transplanted it. We evaluated the nerve regeneration 24 weeks after surgery.Toe dragging was significantly better in the Bio3D group (0.20 ± 0.28) than in the silicon group (0.6 ± 0.24). The wet muscle weight ratios of the tibial anterior muscle of the Bio3D group (79.85% ± 5.47%) and the autologous nerve group (81.74% ± 2.83%) were significantly higher than that of the silicon group (66.99% ± 3.51%). The number of myelinated axons and mean myelinated axon diameter was significantly higher in the Bio3D group (14708 ± 302 and 5.52 ± 0.44 μm) and the autologous nerve group (14927 ± 5089 and 6.04 ± 0.85 μm) than the silicon group (7429 ± 1465 and 4.36 ± 0.21 μm). No tumors were observed in any of the rats in the Bio3D group at 24 weeks after surgery.The Bio3D group showed significantly better nerve regeneration and there was no significant difference between the Bio3D group and the nerve autograft group in all endpoints.
Collapse
Affiliation(s)
| | - Ryosuke Ikeguchi
- Ryosuke Ikeguchi, Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Mota C, Camarero-Espinosa S, Baker MB, Wieringa P, Moroni L. Bioprinting: From Tissue and Organ Development to in Vitro Models. Chem Rev 2020; 120:10547-10607. [PMID: 32407108 PMCID: PMC7564098 DOI: 10.1021/acs.chemrev.9b00789] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Indexed: 02/08/2023]
Abstract
Bioprinting techniques have been flourishing in the field of biofabrication with pronounced and exponential developments in the past years. Novel biomaterial inks used for the formation of bioinks have been developed, allowing the manufacturing of in vitro models and implants tested preclinically with a certain degree of success. Furthermore, incredible advances in cell biology, namely, in pluripotent stem cells, have also contributed to the latest milestones where more relevant tissues or organ-like constructs with a certain degree of functionality can already be obtained. These incredible strides have been possible with a multitude of multidisciplinary teams around the world, working to make bioprinted tissues and organs more relevant and functional. Yet, there is still a long way to go until these biofabricated constructs will be able to reach the clinics. In this review, we summarize the main bioprinting activities linking them to tissue and organ development and physiology. Most bioprinting approaches focus on mimicking fully matured tissues. Future bioprinting strategies might pursue earlier developmental stages of tissues and organs. The continuous convergence of the experts in the fields of material sciences, cell biology, engineering, and many other disciplines will gradually allow us to overcome the barriers identified on the demanding path toward manufacturing and adoption of tissue and organ replacements.
Collapse
Affiliation(s)
- Carlos Mota
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Sandra Camarero-Espinosa
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Matthew B. Baker
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Paul Wieringa
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Lorenzo Moroni
- Department of Complex Tissue Regeneration,
MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6211 LK Maastricht, The Netherlands
| |
Collapse
|
42
|
Yurie H, Ikeguchi R, Aoyama T, Ito A, Tanaka M, Noguchi T, Oda H, Takeuchi H, Mitsuzawa S, Ando M, Yoshimoto K, Akieda S, Nakayama K, Matsuda S. Mechanism of Peripheral Nerve Regeneration Using a Bio 3D Conduit Derived from Normal Human Dermal Fibroblasts. J Reconstr Microsurg 2020; 37:357-364. [PMID: 32957155 DOI: 10.1055/s-0040-1716855] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND We previously reported the development of a scaffold-free Bio three-dimensional (3D) nerve conduit from normal human dermal fibroblasts (NHDFs). The aim of this study was to investigate the regenerative mechanism of peripheral nerve cells using a Bio 3D conduit in a rat sciatic nerve defect model. METHODS Bio 3D conduits composed of NHDFs were developed, and cell viability was evaluated using a LIVE/DEAD cell viability assay immediately before transplantation and 1-week post-surgery. Tracking analysis using PKH26-labeled NHDFs was performed to assess the distribution of NHDFs within the regenerated nerve and the differentiation of NHDFs into functional Schwann cells (SCs). RESULTS The assessment of the viability of cells within the Bio 3D conduit showed high cell viability both immediately before transplantation and 1-week post-surgery (88.56 ± 1.70 and 87.58 ± 9.11, respectively). A modified Masson's trichrome staining of the Bio 3D conduit revealed the formation of a prominent extracellular matrix (ECM) in between the cells. We observed, via tracking analysis, that the tube-like distribution of the NHDFs remained stable, the majority of the regenerated axons had penetrated this structure and PKH26-labeled cells were also positive for S-100. CONCLUSION Abundant ECM formation resulted in a stable tube-like structure of the Bio 3D conduit with high cell viability. NHDFs in the Bio 3D conduit have the potential to differentiate into SCs-like cells.
Collapse
Affiliation(s)
- Hirofumi Yurie
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryosuke Ikeguchi
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomoki Aoyama
- Department of Physical Therapy, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Ito
- Department of Physical Therapy, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mai Tanaka
- Department of Physical Therapy, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Noguchi
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroki Oda
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hisataka Takeuchi
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sadaki Mitsuzawa
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Maki Ando
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koichi Yoshimoto
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Koichi Nakayama
- Department of Regenerative Medicine and Biomedical Engineering Faculty of Medicine, Saga University, Saga, Japan
| | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
43
|
Murata D, Arai K, Nakayama K. Scaffold-Free Bio-3D Printing Using Spheroids as "Bio-Inks" for Tissue (Re-)Construction and Drug Response Tests. Adv Healthc Mater 2020; 9:e1901831. [PMID: 32378363 DOI: 10.1002/adhm.201901831] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/21/2020] [Accepted: 03/04/2020] [Indexed: 02/06/2023]
Abstract
In recent years, scaffold-free bio-3D printing using cell aggregates (spheroids) as "bio-inks" has attracted increasing attention as a method for 3D cell construction. Bio-3D printing uses a technique called the Kenzan method, wherein spheroids are placed one-by-one in a microneedle array (the "Kenzan") using a bio-3D printer. The bio-3D printer is a machine that was developed to perform bio-3D printing automatically. Recently, it has been reported that cell constructs can be produced by a bio-3D printer using spheroids composed of many types of cells and that this can contribute to tissue (re-)construction. This progress report summarizes the production and effectiveness of various cell constructs prepared using bio-3D printers. It also considers the future issues and prospects of various cell constructs obtained by using this method for further development of scaffold-free 3D cell constructions.
Collapse
Affiliation(s)
- Daiki Murata
- Center for Regenerative Medicine ResearchFaculty of MedicineSaga University Honjo‐machi Saga 840‐8502 Japan
| | - Kenichi Arai
- Center for Regenerative Medicine ResearchFaculty of MedicineSaga University Honjo‐machi Saga 840‐8502 Japan
| | - Koichi Nakayama
- Center for Regenerative Medicine ResearchFaculty of MedicineSaga University Honjo‐machi Saga 840‐8502 Japan
| |
Collapse
|
44
|
Pro-angiogenic scaffold-free Bio three-dimensional conduit developed from human induced pluripotent stem cell-derived mesenchymal stem cells promotes peripheral nerve regeneration. Sci Rep 2020; 10:12034. [PMID: 32694698 PMCID: PMC7374629 DOI: 10.1038/s41598-020-68745-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/26/2020] [Indexed: 02/07/2023] Open
Abstract
Although autologous nerve grafting is widely accepted as the gold standard treatment for segmental nerve defects, harvesting autologous nerves is highly invasive and leads to functional loss of the ablated part. In response, artificial nerve conduits made of artificial materials have been reported, but the efficacy of the nerve regeneration still needs improvement. The purpose of this study is to investigate the efficacy and mechanism of the Bio three-dimensional (3D) conduit composed of xeno-free human induced pluripotent stem cell–derived mesenchymal stem cells (iMSCs). The 5-mm nerve gap of the sciatic nerve in immunodeficient rats was bridged with the Bio 3D conduit or silicone tube. Functional and histological recovery were assessed at 8 weeks after surgery. The regenerated nerve in the Bio 3D group was significantly superior to that in the silicone group based on morphology, kinematics, electrophysiology, and wet muscle weight. Gene expression analyses demonstrated neurotrophic and angiogenic factors. Macroscopic observation revealed neovascularization both inside and on the surface of the Bio 3D conduit. Upon their subcutaneous implantation, iMSCs could induce angiogenesis. The Bio 3D conduit fabricated from iMSCs are an effective strategy for nerve regeneration in animal model. This technology will be useful in future clinical situations.
Collapse
|
45
|
Askarzadeh N, Nazarpak MH, Mansoori K, Farokhi M, Gholami M, Mohammadi J, Mottaghitalab F. Bilayer Cylindrical Conduit Consisting of Electrospun Polycaprolactone Nanofibers and DSC Cross-Linked Sodium Alginate Hydrogel to Bridge Peripheral Nerve Gaps. Macromol Biosci 2020; 20:e2000149. [PMID: 32627956 DOI: 10.1002/mabi.202000149] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/13/2020] [Indexed: 12/15/2022]
Abstract
Herein, a bilayer cylindrical conduit (P-CA) is presented consisting of electrospun polycaprolactone (PCL) nanofibers and sodium alginate hydrogel covalently cross-linked with N,N'-disuccinimidyl carbonate (DSC). The bilayer P-CA conduit is developed by combining the electrospinning and outer-inner layer methods. Using DSC, as a covalent crosslinker, increases the degradation time of the sodium alginate hydrogel up to 2 months. The swelling ratio of the hydrogel is also 503% during the first 8 h. The DSC cross-linked sodium alginate in the inner layer of the conduit promotes the adhesion and proliferation of nerve cells, while the electrospun PCL nanofibers in the outer layer provide maximum tensile strength of the conduit during surgery. P-CA conduit promotes the migration of Schwann cells along the axon in a rat model based on functional and histological evidences. In conclusion, P-CA conduit will be a promising construct for repairing sciatic nerves in a rat model.
Collapse
Affiliation(s)
- Neshat Askarzadeh
- Faculty of New Sciences and Technologies, Department of Life Science Engineering, University of Tehran, Tehran, 1439957131, Iran
| | | | - Korosh Mansoori
- Physical medicine and Rehabilitation Neuromusculoskeletal Research Center, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Mehdi Farokhi
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Mahdi Gholami
- Faculty of Pharmacy and Pharmaceutical Science Research Center, Tehran University of Medical Sciences, Tehran, 141556451, Iran
| | - Javad Mohammadi
- Faculty of New Sciences and Technologies, Department of Life Science Engineering, University of Tehran, Tehran, 1439957131, Iran
| | - Fatemeh Mottaghitalab
- Nanotechnology Research Centre, Tehran University of Medical Sciences, Tehran, 141556451, Iran
| |
Collapse
|
46
|
Cai H, Wu Z, Ao Z, Nunez A, Chen B, Jiang L, Bondesson M, Guo F. Trapping cell spheroids and organoids using digital acoustofluidics. Biofabrication 2020; 12:035025. [DOI: 10.1088/1758-5090/ab9582] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
47
|
Ito A, Wang T, Nakahara R, Kawai H, Nishitani K, Aoyama T, Kuroki H. Ultrasound therapy with optimal intensity facilitates peripheral nerve regeneration in rats through suppression of pro-inflammatory and nerve growth inhibitor gene expression. PLoS One 2020; 15:e0234691. [PMID: 32555658 PMCID: PMC7299378 DOI: 10.1371/journal.pone.0234691] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 05/29/2020] [Indexed: 02/07/2023] Open
Abstract
Background Therapeutic ultrasound (US) is a promising physical therapy modality for peripheral nerve regeneration. However, it is necessary to identify the most effective US parameters and clarify the underlying mechanisms before its clinical application. The intensity of US is one of the most important parameters. However, the optimum intensity for the promotion of peripheral nerve regeneration has yet to be determined. Objectives To identify the optimum intensity of US necessary for the promotion of peripheral nerve regeneration after crush injuries in rats and to clarify the underlying mechanisms of US by mRNA expression analysis. Methods We inflicted sciatic nerve crush injuries on adult Lewis rats and performed ultrasound irradiation using 4 different US intensities: 0 (sham stimulation), 30, 140, and 250 mW/cm2 with frequency (5 days/week) and duration (5 min/day). We evaluated peripheral nerve regeneration by quantitative real-time PCR one week after injury. Histomorphometric analyses and motor function analysis were evaluated 3 weeks after injury. Results US stimulation enhanced re-myelination as well as sprouting of axons, especially at an intensity of 140 mW/cm2. mRNA expression revealed that US suppressed the expression of the inflammatory cytokines TNF and IL-6 and the axonal growth inhibitors SEMA3A and GSK3β. Conclusions An intensity of 140 mW/cm2 was optimal to support regeneration of the sciatic nerve after a crush injury in rats by, in part, the suppression of pro-inflammatory and nerve growth inhibitor gene expression.
Collapse
Affiliation(s)
- Akira Ito
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tianshu Wang
- Department of Development and Rehabilitation of Motor Function, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryo Nakahara
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hideki Kawai
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kohei Nishitani
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoki Aoyama
- Department of Development and Rehabilitation of Motor Function, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Kuroki
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
48
|
Drug response analysis for scaffold-free cardiac constructs fabricated using bio-3D printer. Sci Rep 2020; 10:8972. [PMID: 32487993 PMCID: PMC7265390 DOI: 10.1038/s41598-020-65681-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 05/05/2020] [Indexed: 12/04/2022] Open
Abstract
Cardiac constructs fabricated using human induced pluripotent stem cells-derived cardiomyocytes (iPSCs-CMs) are useful for evaluating the cardiotoxicity of and cardiac response to new drugs. Previously, we fabricated scaffold-free three-dimensional (3D) tubular cardiac constructs using a bio-3D printer, which can load cardiac spheroids onto a needle array. In this study, we developed a method to measure the contractile force and to evaluate the drug response in cardiac constructs. Specifically, we measured the movement of the needle tip upon contraction of the cardiac constructs on the needle array. The contractile force and beating rate of the cardiac constructs were evaluated by analysing changes in the movement of the needle tip. To evaluate the drug response, contractile properties were measured following treatment with isoproterenol, propranolol, or blebbistatin, in which the movement of the needle tip was increased following isoproterenol treatment, but was decreased following propranolol or blebbistain, treatments. To evaluate cardiotoxicity, contraction and cell viability of the cardiac constructs were measured following doxorubicin treatment. Cell viability was found to decrease with decreasing movement of the needle tip following doxorubicin treatment. Collectively, our results show that this method can aid in evaluating the contractile force of cardiac constructs.
Collapse
|
49
|
Brito ACNDL, Santos SEV, Martins WA, Queiroz PCDS, Sougey WWD, Alves PKN, Ribeiro KL, de Oliveira MDL, de Moraes SRA. Efficacy of tubing technique with biomaterials compared to direct coaptation technique after peripheral neurotmesis in nerve healing and return to functionality in young adult rats: a systematic review protocol. Syst Rev 2020; 9:118. [PMID: 32460835 PMCID: PMC7254672 DOI: 10.1186/s13643-020-01388-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/12/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Peripheral nerves are constant targets of traumatic injury which may result in neurotmesis and which invariably requires surgical treatment. In view of this, tissue engineering studies developed biomaterials which were first tested in animal models and used as a guide for nerve stumps in the procedure in order to speed up the healing process. Therefore, the aim of this study is to evaluate the efficacy of biomaterials used in tubing technique on healing and histological and functional recovery after peripheral nerve neurotmesis in rats. METHODS We will search PubMed/MEDLINE, Embase, Web of Science, LILACS, and CENTRAL (from inception onwards). Grey literature will be identified through searching dissertation databases, guidelines, policy documents, and reports. We will include randomized and non-randomized trials conducted in young adult rats with peripheral neurometsis undergoing surgical repair through tubing technique with biomaterials. Primary outcomes will be histomorphometry, immunohistochemistry of the nerve tissue, and sciatic functional index. Secondary outcome will be nerve macroscopic evaluation. Two reviewers will independently screen all citations, full-text articles, and abstract data. Potential conflicts will be resolved through discussion. The methodological quality (or risk of bias) of individual studies will be appraised using an appropriate tool. If feasible, we will conduct random effects meta-analysis. DISCUSSION This systematic review of animal studies will identify, evaluate, and synthetize the evidence on the the efficacy of tubing technique with biomaterials compared to direct coaptation technique after peripheral neurotmesis in nerve healing and return to functionality. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42018106042.
Collapse
Affiliation(s)
- Ana Camila Nobre de Lacerda Brito
- Neuropsychiatry and Behavioral Science Program, Federal University of Pernambuco, Avenida Professor Moraes Rego, 1235, Recife, 50670-901, Pernambuco, Brazil. .,Department of Anatomy, Neuromuscular Plasticity Laboratory, Federal University of Pernambuco, Recife, Pernambuco, Brazil.
| | | | - Wilayane Alves Martins
- Department of Physiotherapy, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | | | | | | | | | | | - Sílvia Regina Arruda de Moraes
- Department of Anatomy, Neuromuscular Plasticity Laboratory, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| |
Collapse
|
50
|
Cryopreservation method for spheroids and fabrication of scaffold-free tubular constructs. PLoS One 2020; 15:e0230428. [PMID: 32240195 PMCID: PMC7117714 DOI: 10.1371/journal.pone.0230428] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 02/28/2020] [Indexed: 12/24/2022] Open
Abstract
Cryopreservation is a method used for preserving living cells by cooling them to very low temperatures. Although cryopreservation methods for oocytes and embryos have been developed for use in reproductive medicine, there are no established methods yet for preserving cell aggregates (spheroids) in regenerative medicine. We have developed a bio-three-dimensional (3D) printer that can fabricate scaffold-free 3D constructs by loading spheroids onto a needle array. We fabricated several constructs such as blood vessels, liver, diaphragm, and a conduit for nerves by using this method. These constructs have the potential to be applied in patients. However, the process of fabricating tissue constructs (harvesting cells, expanding cells, making spheroids using cultured cells, printing constructs, and maturing constructs) is time-consuming. Therefore, cryopreservation methods for spheroids or constructs should be developed to increase the efficiency of this method for clinical use. Here, we developed a method for cryopreserving spheroids, which were then used to fabricate constructs. Fibroblast cell-based spheroids were cryopreserved in phosphate-buffered saline or cryopreservation solution at −80°C for 1 week. After thawing, spheroids in cryopreservation solution began to fuse on day 1. Cryopreserved spheroids were printed onto a needle array to fabricate a scaffold-free tubular construct using a bio-3D printer. After 7 days, the printed spheroids fused and formed scaffold-free constructs. We confirmed the viability of cells in the cryopreserved spheroids and fabricated tubular constructs. Our results indicate that spheroids can be cryopreserved and used to prepare scaffold-free constructs for clinical use.
Collapse
|