1
|
Nedrelow DS, Townsend JM, Detamore MS. Osteochondral Regeneration With Anatomical Scaffold 3D-Printing-Design Considerations for Interface Integration. J Biomed Mater Res A 2025; 113:e37804. [PMID: 39387548 DOI: 10.1002/jbm.a.37804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 10/15/2024]
Abstract
There is a clinical need for osteochondral scaffolds with complex geometries for restoring articulating joint surfaces. To address that need, 3D-printing has enabled scaffolds to be created with anatomically shaped geometries and interconnected internal architectures, going beyond simple plug-shaped scaffolds that are limited to small, cylindrical, focal defects. A key challenge for restoring articulating joint surfaces with 3D-printed constructs is the mechanical loading environment, particularly to withstand delamination or mechanical failure. Although the mechanical performance of interfacial scaffolds is essential, interface strength testing has rarely been emphasized in prior studies with stratified scaffolds. In the pioneering studies where interface strength was assessed, varying methods were employed, which has made direct comparisons difficult. Therefore, the current review focused on 3D-printed scaffolds for osteochondral applications with an emphasis on interface integration and biomechanical evaluation. This 3D-printing focus included both multiphasic cylindrical scaffolds and anatomically shaped scaffolds. Combinations of different 3D-printing methods (e.g., fused deposition modeling, stereolithography, bioprinting with pneumatic extrusion of cell-laden hydrogels) have been employed in a handful of studies to integrate osteoinductive and chondroinductive regions into a single scaffold. Most 3D-printed multiphasic structures utilized either an interdigitating or a mechanical interlocking design to strengthen the construct interface and to prevent delamination during function. The most effective approach to combine phases may be to infill a robust 3D-printed osteal polymer with an interlocking chondral phase hydrogel. Mechanical interlocking is therefore recommended for scaling up multiphasic scaffold applications to larger anatomically shaped joint surface regeneration. For the evaluation of layer integration, the interface shear test is recommended to avoid artifacts or variability that may be associated with alternative approaches that require adhesives or mechanical grips. The 3D-printing literature with interfacial scaffolds provides a compelling foundation for continued work toward successful regeneration of injured or diseased osteochondral tissues in load-bearing joints such as the knee, hip, or temporomandibular joint.
Collapse
Affiliation(s)
- David S Nedrelow
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, USA
- College of Dentistry, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Jakob M Townsend
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, USA
| | - Michael S Detamore
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, USA
| |
Collapse
|
2
|
Kaboodkhani R, Mehrabani D, Moghaddam A, Salahshoori I, Khonakdar HA. Tissue engineering in otology: a review of achievements. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:1105-1153. [PMID: 38386362 DOI: 10.1080/09205063.2024.2318822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/09/2024] [Indexed: 02/23/2024]
Abstract
Tissue engineering application in otology spans a distance from the pinna to auditory nerve covered with specialized tissues and functions such as sense of hearing and aesthetics. It holds the potential to address the barriers of lack of donor tissue, poor tissue match, and transplant rejection through provision of new and healthy tissues similar to the host and possesses the capacity to renew, to regenerate, and to repair in-vivo and was shown to be a bypasses for any need to immunosuppression. This review aims to investigate the application of tissue engineering in otology and to evaluate the achievements and challenges in external, middle and inner ear sections. Since gaining the recent knowledge and training on use of different scaffolds is essential for otology specialists and who look for the recovery of ear function and aesthetics of patients, it is shown in this review how utilizing tissue engineering and cell transplantation, regenerative medicine can provide advancements in hearing and ear aesthetics to fit different patients' needs.
Collapse
Affiliation(s)
- Reza Kaboodkhani
- Otorhinolaryngology Research Center, Department of Otorhinolaryngology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Davood Mehrabani
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | | | | | - Hossein Ali Khonakdar
- Iran Polymer and Petrochemical Institute (IPPI), Tehran, Iran
- Max Bergmann Center of Biomaterials and Institute of Materials Science, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
3
|
Ding J, Wei C, Xu Y, Dai W, Chen R. 3D printing of Ceffe-infused scaffolds for tailored nipple-like cartilage development. BMC Biotechnol 2024; 24:25. [PMID: 38689309 PMCID: PMC11059701 DOI: 10.1186/s12896-024-00848-3] [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: 02/19/2024] [Accepted: 04/04/2024] [Indexed: 05/02/2024] Open
Abstract
The reconstruction of a stable, nipple-shaped cartilage graft that precisely matches the natural nipple in shape and size on the contralateral side is a clinical challenge. While 3D printing technology can efficiently and accurately manufacture customized complex structures, it faces limitations due to inadequate blood supply, which hampers the stability of nipple-shaped cartilage grafts produced using this technology. To address this issue, we employed a biodegradable biomaterial, Poly(lactic-co-glycolic acid) (PLGA), loaded with Cell-Free Fat Extract (Ceffe). Ceffe has demonstrated the ability to promote angiogenesis and cell proliferation, making it an ideal bio-ink for bioprinting precise nipple-shaped cartilage grafts. We utilized the Ceffe/PLGA scaffold to create a porous structure with a precise nipple shape. This scaffold exhibited favorable porosity and pore size, ensuring stable shape maintenance and satisfactory biomechanical properties. Importantly, it could release Ceffe in a sustained manner. Our in vitro results confirmed the scaffold's good biocompatibility and its ability to promote angiogenesis, as evidenced by supporting chondrocyte proliferation and endothelial cell migration and tube formation. Furthermore, after 8 weeks of in vivo culture, the Ceffe/PLGA scaffold seeded with chondrocytes regenerated into a cartilage support structure with a precise nipple shape. Compared to the pure PLGA group, the Ceffe/PLGA scaffold showed remarkable vascular formation, highlighting the beneficial effects of Ceffe. These findings suggest that our designed Ceffe/PLGA scaffold with a nipple shape represents a promising strategy for precise nipple-shaped cartilage regeneration, laying a foundation for subsequent nipple reconstruction.
Collapse
Affiliation(s)
- Jinghao Ding
- Department of Breast Surgery, Hainan Affiliated Hospital of Hainan Medical University (Hainan General Hospital), Haikou, China
| | - Chuanzhi Wei
- Department of Breast Surgery, Hainan Affiliated Hospital of Hainan Medical University (Hainan General Hospital), Haikou, China
| | - Yong Xu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wufei Dai
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ru Chen
- Department of Breast Surgery, Hainan Affiliated Hospital of Hainan Medical University (Hainan General Hospital), Haikou, China.
| |
Collapse
|
4
|
Donnelly H, Kurjan A, Yong LY, Xiao Y, Lemgruber L, West C, Salmeron-Sanchez M, Dalby MJ. Fibronectin matrix assembly and TGFβ1 presentation for chondrogenesis of patient derived pericytes for microtia repair. BIOMATERIALS ADVANCES 2023; 148:213370. [PMID: 36931082 DOI: 10.1016/j.bioadv.2023.213370] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 02/10/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023]
Abstract
Tissue engineered cartilage for external ear reconstruction of congenital deformities, such as microtia or resulting from trauma, remains a significant challenge for plastic and reconstructive surgeons. Current strategies involve harvesting autologous costal cartilage or expanding autologous chondrocytes ex vivo. However, these procedures often lead to donor site morbidity and a cell source with limited expansion capacity. Stromal stem cells such as perivascular stem cells (pericytes) offer an attractive alternative cell source, as they can be isolated from many human tissues, readily expanded in vitro and possess chondrogenic differentiation potential. Here, we successfully isolate CD146+ pericytes from the microtia remnant from patients undergoing reconstructive surgery (Microtia pericytes; MPs). Then we investigate their chondrogenic potential using the polymer poly(ethyl acrylate) (PEA) to unfold the extracellular matrix protein fibronectin (FN). FN unfolding exposes key growth factor (GF) and integrin binding sites on the molecule, allowing tethering of the chondrogenic GF transforming growth factor beta 1 (TGFβ1). This system leads to solid-phase, matrix-bound, GF presentation in a more physiological-like manner than that of typical chondrogenic induction media (CM) formulations that tend to lead to off-target effects. This simple and controlled material-based approach demonstrates similar chondrogenic potential to CM, while minimising proclivity toward hypertrophy, without the need for complex induction media formulations.
Collapse
Affiliation(s)
- Hannah Donnelly
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell & Systems Biology, College of Medical, Veterinary and Life Sciences, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, United Kingdom.
| | - Alina Kurjan
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell & Systems Biology, College of Medical, Veterinary and Life Sciences, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Li Yenn Yong
- MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - Yinbo Xiao
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell & Systems Biology, College of Medical, Veterinary and Life Sciences, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Leandro Lemgruber
- Glasgow Imaging Facility, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Christopher West
- MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell & Systems Biology, College of Medical, Veterinary and Life Sciences, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| |
Collapse
|
5
|
Dupard SJ, Garcia AG, Bourgine PE. Customizable 3D printed perfusion bioreactor for the engineering of stem cell microenvironments. Front Bioeng Biotechnol 2023; 10:1081145. [PMID: 36698631 PMCID: PMC9870251 DOI: 10.3389/fbioe.2022.1081145] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023] Open
Abstract
Faithful modeling of tissues and organs requires the development of systems reflecting their dynamic 3D cellular architecture and organization. Current technologies suffer from a lack of design flexibility and complex prototyping, preventing their broad adoption by the scientific community. To make 3D cell culture more available and adaptable we here describe the use of the fused deposition modeling (FDM) technology to rapid-prototype 3D printed perfusion bioreactors. Our 3D printed bioreactors are made of polylactic acid resulting in reusable systems customizable in size and shape. Following design confirmation, our bioreactors were biologically validated for the culture of human mesenchymal stromal cells under perfusion for up to 2 weeks on collagen scaffolds. Microenvironments of various size/volume (6-12 mm in diameter) could be engineered, by modulating the 3D printed bioreactor design. Metabolic assay and confocal microscopy confirmed the homogenous mesenchymal cell distribution throughout the material pores. The resulting human microenvironments were further exploited for the maintenance of human hematopoietic stem cells. Following 1 week of stromal coculture, we report the recapitulation of 3D interactions between the mesenchymal and hematopoietic fractions, associated with a phenotypic expansion of the blood stem cell populations.Our data confirm that perfusion bioreactors fit for cell culture can be generated using a 3D printing technology and exploited for the 3D modeling of complex stem cell systems. Our approach opens the gates for a more faithful investigation of cellular processes in relation to a dynamic 3D microenvironment.
Collapse
Affiliation(s)
- Steven J. Dupard
- Cell, Tissue and Organ engineering laboratory, Biomedical Centre (BMC), Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Alejandro Garcia Garcia
- Cell, Tissue and Organ engineering laboratory, Biomedical Centre (BMC), Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Paul E. Bourgine
- Cell, Tissue and Organ engineering laboratory, Biomedical Centre (BMC), Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| |
Collapse
|
6
|
Dwivedi R, Yadav PK, Pandey R, Mehrotra D. Auricular reconstruction via 3D bioprinting strategies: An update. J Oral Biol Craniofac Res 2022; 12:580-588. [PMID: 35968037 DOI: 10.1016/j.jobcr.2022.07.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/22/2022] [Accepted: 07/28/2022] [Indexed: 10/16/2022] Open
Abstract
Image 1.
Collapse
Affiliation(s)
- Ruby Dwivedi
- King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Pradeep Kumar Yadav
- Department of Oral and Maxillofacial Surgery, King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Rahul Pandey
- King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Divya Mehrotra
- Department of Oral and Maxillofacial Surgery, King George's Medical University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
7
|
Lan X, Liang Y, Vyhlidal M, Erkut EJN, Kunze M, Mulet-Sierra A, Osswald M, Ansari K, Seikaly H, Boluk Y, Adesida AB. In vitro maturation and in vivo stability of bioprinted human nasal cartilage. J Tissue Eng 2022; 13:20417314221086368. [PMID: 35599742 PMCID: PMC9122109 DOI: 10.1177/20417314221086368] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 02/23/2022] [Indexed: 11/25/2022] Open
Abstract
The removal of skin cancer lesions on the nose often results in the loss of nasal
cartilage. The cartilage loss is either surgically replaced with autologous
cartilage or synthetic grafts. However, these replacement options come with
donor-site morbidity and resorption issues. 3-dimensional (3D) bioprinting
technology offers the opportunity to engineer anatomical-shaped autologous nasal
cartilage grafts. The 3D bioprinted cartilage grafts need to embody a
mechanically competent extracellular matrix (ECM) to allow for surgical suturing
and resistance to contraction during scar tissue formation. We investigated the
effect of culture period on ECM formation and mechanical properties of 3D
bioprinted constructs of human nasal chondrocytes (hNC)-laden type I collagen
hydrogel in vitro and in vivo. Tissue-engineered nasal cartilage constructs
developed from hNC culture in clinically approved collagen type I and type III
semi-permeable membrane scaffold served as control. The resulting 3D bioprinted
engineered nasal cartilage constructs were comparable or better than the
controls both in vitro and in vivo. This study demonstrates that 3D bioprinted
constructs of engineered nasal cartilage are feasible options in nasal cartilage
reconstructive surgeries.
Collapse
Affiliation(s)
- Xiaoyi Lan
- Department of Civil and Environmental Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
| | - Yan Liang
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Margaret Vyhlidal
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Esra JN Erkut
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Melanie Kunze
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Aillette Mulet-Sierra
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Martin Osswald
- Institute for Reconstructive Sciences in Medicine, Misericordia Community Hospital, Edmonton, AB, Canada
- Division of Otolaryngology, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Khalid Ansari
- Division of Otolaryngology, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Hadi Seikaly
- Division of Otolaryngology, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Yaman Boluk
- Department of Civil and Environmental Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
| | - Adetola B Adesida
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Division of Otolaryngology, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
8
|
Bhamare N, Tardalkar K, Khadilkar A, Parulekar P, Joshi MG. Tissue engineering of human ear pinna. Cell Tissue Bank 2022; 23:441-457. [PMID: 35103863 DOI: 10.1007/s10561-022-09991-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 01/06/2022] [Indexed: 12/30/2022]
Abstract
Auricular deformities (Microtia) can cause physical, social as well as psychological impacts on a patient's wellbeing. Biofabrication of a complex structure such as ear pinna is not precise with currently available techniques. These limitations can be overcome with the help of tissue engineering. In this article, the authors presented molding and three dimensional (3D) printing to generate a flexible, human size ear pinna. The decellularization of goat ear cartilage protocol and bioink alkaline digestion protocol was followed to yield complete removal of all cellular components without changing the properties of the Extra Cellular Matrix (ECM). Decellularized scaffold used in molding technology and 3D printing technology Computer-Aided Design /Stereolithography (CAD/STL) uses bioink to construct the patient-specific ear. In vivo biocompatibility of the both ear pinnae showed demonstrable recellularization. Histology and scanning electron microscopy analysis revealed the recellularization of cartilage-specific cells and the development of ECM in molded and 3D printed ear pinna after transplantation. Both the techniques provided ideal results for mechanical properties such as elasticity. Vascular Associated Protein expression revealed specific vasculogenic pattern (angiogenesis) in transplanted molded pinna. Chondrocyte specific progenitor cells express CD90+ which highlighted newly developed chondrocytes in both the grafts which indicated that the xenograft was accepted by the rat. Transplantation of molded as well as 3D ear pinna was successful in an animal model and can be available for clinical treatments as a medical object to cure auricular deformities.
Collapse
Affiliation(s)
- Nilesh Bhamare
- Department of Stem Cells and Regenerative Medicine, D. Y. Patil Education Society (Deemed to be University), Kasaba Bawada, 416 006, Kolhapur, Maharashtra, India.
| | - Kishor Tardalkar
- Department of Stem Cells and Regenerative Medicine, D. Y. Patil Education Society (Deemed to be University), Kasaba Bawada, 416 006, Kolhapur, Maharashtra, India
| | - Archana Khadilkar
- Department of Biotechnology Engineering, KIT's College of Engineering (Autonomous), Kolhapur, India
| | - Pratima Parulekar
- Department of Biotechnology Engineering, KIT's College of Engineering (Autonomous), Kolhapur, India
| | - Meghnad G Joshi
- Department of Stem Cells and Regenerative Medicine, D. Y. Patil Education Society (Deemed to be University), Kasaba Bawada, 416 006, Kolhapur, Maharashtra, India. .,Stem Plus Biotech Pvt. Ltd.Sangli Miraj Kupwad Commercial Complex, C/S No. 1317/2, Near Shivaji Maharaj Putla, Bus Stand Road,Gaon Bhag, 416416, Sangli, MS, India.
| |
Collapse
|
9
|
Bertassoni LE. Bioprinting of Complex Multicellular Organs with Advanced Functionality-Recent Progress and Challenges Ahead. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2101321. [PMID: 35060652 PMCID: PMC10171718 DOI: 10.1002/adma.202101321] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/20/2021] [Indexed: 05/12/2023]
Abstract
Bioprinting has emerged as one of the most promising strategies for fabrication of functional organs in the lab as an alternative to transplant organs. While progress in the field has mostly been restricted to a few miniaturized tissues with minimal biological functionality until a few years ago, recent progress has advanced the concept of building three-dimensional multicellular organ complexity remarkably. This review discusses a series of milestones that have paved the way for bioprinting of tissue constructs that have advanced levels of biological and architectural functionality. Critical materials, engineering and biological challenges that are key to addressing the desirable function of engineered organs are presented. These are discussed in light of the many difficulties to replicate the heterotypic organization of multicellular solid organs, the nanoscale precision of the extracellular microenvironment in hierarchical tissues, as well as the advantages and limitations of existing bioprinting methods to adequately overcome these barriers. In summary, the advances of the field toward realistic manufacturing of functional organs have never been so extensive, and this manuscript serves as a road map for some of the recent progress and the challenges ahead.
Collapse
Affiliation(s)
- Luiz E Bertassoni
- Division of Biomaterials and Biomechanics, School of Dentistry, Oregon Health and Science University, Portland, OR, 97201, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR, 97239, USA
- Center for Regenerative Medicine, Oregon Health and Science University, Portland, OR, 97239, USA
- Cancer Early Detection Advanced Research (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97239, USA
| |
Collapse
|
10
|
Dong X, Premaratne ID, Bernstein JL, Samadi A, Lin AJ, Toyoda Y, Kim J, Bonassar LJ, Spector JA. Three-Dimensional-Printed External Scaffolds Mitigate Loss of Volume and Topography in Engineered Elastic Cartilage Constructs. Cartilage 2021; 13:1780S-1789S. [PMID: 34636646 PMCID: PMC8804786 DOI: 10.1177/19476035211049556] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE A major obstacle in the clinical translation of engineered auricular scaffolds is the significant contraction and loss of topography that occur during maturation of the soft collagen-chondrocyte matrix into elastic cartilage. We hypothesized that 3-dimensional-printed, biocompatible scaffolds would "protect" maturing hydrogel constructs from contraction and loss of topography. DESIGN External disc-shaped and "ridged" scaffolds were designed and 3D-printed using polylactic acid (PLA). Acellular type I collagen constructs were cultured in vitro for up to 3 months. Collagen constructs seeded with bovine auricular chondrocytes (BAuCs) were prepared in 3 groups and implanted subcutaneously in vivo for 3 months: preformed discs with ("Scaffolded/S") or without ("Naked/N") an external scaffold and discs that were formed within an external scaffold via injection molding ("Injection Molded/SInj"). RESULTS The presence of an external scaffold or use of injection molding methodology did not affect the acellular construct volume or base area loss. In vivo, the presence of an external scaffold significantly improved preservation of volume and base area at 3 months compared to the naked group (P < 0.05). Construct contraction was mitigated even further in the injection molded group, and topography of the ridged constructs was maintained with greater fidelity (P < 0.05). Histology verified the development of mature auricular cartilage in the constructs within external scaffolds after 3 months. CONCLUSION Custom-designed, 3D-printed, biocompatible external scaffolds significantly mitigate BAuC-seeded construct contraction and maintain complex topography. Further refinement and scaling of this approach in conjunction with construct fabrication utilizing injection molding may aid in the development of full-scale auricular scaffolds.
Collapse
Affiliation(s)
- Xue Dong
- Laboratory of Bioregenerative Medicine
& Surgery, Department of Surgery, Division of Plastic Surgery, Weill Cornell
Medical College, New York, NY, USA
- Department of Breast Surgery, Xiangya
Hospital, Central South University, Changsha, Hunan, China
| | - Ishani D. Premaratne
- Laboratory of Bioregenerative Medicine
& Surgery, Department of Surgery, Division of Plastic Surgery, Weill Cornell
Medical College, New York, NY, USA
| | - Jaime L. Bernstein
- Laboratory of Bioregenerative Medicine
& Surgery, Department of Surgery, Division of Plastic Surgery, Weill Cornell
Medical College, New York, NY, USA
| | - Arash Samadi
- Laboratory of Bioregenerative Medicine
& Surgery, Department of Surgery, Division of Plastic Surgery, Weill Cornell
Medical College, New York, NY, USA
| | - Alexandra J. Lin
- Laboratory of Bioregenerative Medicine
& Surgery, Department of Surgery, Division of Plastic Surgery, Weill Cornell
Medical College, New York, NY, USA
| | - Yoshiko Toyoda
- Laboratory of Bioregenerative Medicine
& Surgery, Department of Surgery, Division of Plastic Surgery, Weill Cornell
Medical College, New York, NY, USA
| | - Jongkil Kim
- Nancy E. and Peter C. Meinig School of
Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Lawrence J. Bonassar
- Nancy E. and Peter C. Meinig School of
Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Sibley School of Mechanical and
Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Jason A. Spector
- Laboratory of Bioregenerative Medicine
& Surgery, Department of Surgery, Division of Plastic Surgery, Weill Cornell
Medical College, New York, NY, USA
- Nancy E. and Peter C. Meinig School of
Biomedical Engineering, Cornell University, Ithaca, NY, USA
| |
Collapse
|
11
|
Harris AF, Lacombe J, Zenhausern F. The Emerging Role of Decellularized Plant-Based Scaffolds as a New Biomaterial. Int J Mol Sci 2021; 22:12347. [PMID: 34830229 PMCID: PMC8625747 DOI: 10.3390/ijms222212347] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/30/2021] [Accepted: 11/10/2021] [Indexed: 12/14/2022] Open
Abstract
The decellularization of plant-based biomaterials to generate tissue-engineered substitutes or in vitro cellular models has significantly increased in recent years. These vegetal tissues can be sourced from plant leaves and stems or fruits and vegetables, making them a low-cost, accessible, and sustainable resource from which to generate three-dimensional scaffolds. Each construct is distinct, representing a wide range of architectural and mechanical properties as well as innate vasculature networks. Based on the rapid rise in interest, this review aims to detail the current state of the art and presents the future challenges and perspectives of these unique biomaterials. First, we consider the different existing decellularization techniques, including chemical, detergent-free, enzymatic, and supercritical fluid approaches that are used to generate such scaffolds and examine how these protocols can be selected based on plant cellularity. We next examine strategies for cell seeding onto the plant-derived constructs and the importance of the different functionalization methods used to assist in cell adhesion and promote cell viability. Finally, we discuss how their structural features, such as inherent vasculature, porosity, morphology, and mechanical properties (i.e., stiffness, elasticity, etc.) position plant-based scaffolds as a unique biomaterial and drive their use for specific downstream applications. The main challenges in the field are presented throughout the discussion, and future directions are proposed to help improve the development and use of vegetal constructs in biomedical research.
Collapse
Affiliation(s)
- Ashlee F. Harris
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, AZ 85004, USA;
| | - Jerome Lacombe
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, AZ 85004, USA;
- Department of Basic Medical Sciences, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, AZ 85004, USA
| | - Frederic Zenhausern
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, AZ 85004, USA;
- Department of Basic Medical Sciences, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, AZ 85004, USA
- Department of Biomedical Engineering, College of Engineering, The University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
12
|
Tang P, Song P, Peng Z, Zhang B, Gui X, Wang Y, Liao X, Chen Z, Zhang Z, Fan Y, Li Z, Cen Y, Zhou C. Chondrocyte-laden GelMA hydrogel combined with 3D printed PLA scaffolds for auricle regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 130:112423. [PMID: 34702546 DOI: 10.1016/j.msec.2021.112423] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/21/2021] [Accepted: 09/02/2021] [Indexed: 02/05/2023]
Abstract
The current gold standard for auricular reconstruction after microtia or ear trauma is the autologous cartilage graft with an autologous skin flap overlay. Harvesting autologous cartilage requires an additional surgery that may result in donor area complications. In addition, autologous cartilage is limited and the auricular reconstruction requires complex sculpting, which requires excellent clinical skill and is very time consuming. This work explores the use of 3D printing technology to fabricate bioactive artificial auricular cartilage using chondrocyte-laden gelatin methacrylate (GelMA) and polylactic acid (PLA) for auricle reconstruction. In this study, chondrocytes were loaded within GelMA hydrogel and combined with the 3D-printed PLA scaffolds to biomimetic the biological mechanical properties and personalized shape. The printing accuracy personalized scaffolds, biomechanics and chondrocyte viability and biofunction of artificial auricle have been studied. It was found that chondrocytes were fixed in the PLA auricle scaffolds via GelMA hydrogels and exhibited good proliferative properties and cellular activity. In addition, new chondrocytes and chondrogenic matrix, as well as type II collagen were observed after 8 weeks of implantation. At the same time, the transplanted auricle complex kept full and delicate auricle shape. This study demonstrates the potential of using 3D printing technology to construct in vitro living auricle tissue. It shows a great prospect in the clinical application of auricle regeneration.
Collapse
Affiliation(s)
- Pei Tang
- Department of Burn and Plastic Surgery, West China School of Medicine, West China Hospital, Sichuan University, 610041 Chengdu, China
| | - Ping Song
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Zhiyu Peng
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Boqing Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xingyu Gui
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yixi Wang
- Department of Burn and Plastic Surgery, West China School of Medicine, West China Hospital, Sichuan University, 610041 Chengdu, China
| | - Xiaoxia Liao
- Department of Burn and Plastic Surgery, West China School of Medicine, West China Hospital, Sichuan University, 610041 Chengdu, China
| | - Zhixing Chen
- Department of Burn and Plastic Surgery, West China School of Medicine, West China Hospital, Sichuan University, 610041 Chengdu, China
| | - Zhenyu Zhang
- Department of Burn and Plastic Surgery, West China School of Medicine, West China Hospital, Sichuan University, 610041 Chengdu, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Zhengyong Li
- Department of Burn and Plastic Surgery, West China School of Medicine, West China Hospital, Sichuan University, 610041 Chengdu, China.
| | - Ying Cen
- Department of Burn and Plastic Surgery, West China School of Medicine, West China Hospital, Sichuan University, 610041 Chengdu, China
| | - Changchun Zhou
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|
13
|
Hirano N, Kusuhara H, Sueyoshi Y, Teramura T, Murthy A, Asamura S, Isogai N, Jacquet RD, Landis WJ. Ethanol treatment of nanoPGA/PCL composite scaffolds enhances human chondrocyte development in the cellular microenvironment of tissue-engineered auricle constructs. PLoS One 2021; 16:e0253149. [PMID: 34242238 PMCID: PMC8270150 DOI: 10.1371/journal.pone.0253149] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 05/24/2021] [Indexed: 11/24/2022] Open
Abstract
A major obstacle for tissue engineering ear-shaped cartilage is poorly developed tissue comprising cell-scaffold constructs. To address this issue, bioresorbable scaffolds of poly-ε-caprolactone (PCL) and polyglycolic acid nanofibers (nanoPGA) were evaluated using an ethanol treatment step before auricular chondrocyte scaffold seeding, an approach considered to enhance scaffold hydrophilicity and cartilage regeneration. Auricular chondrocytes were isolated from canine ears and human surgical samples discarded during otoplasty, including microtia reconstruction. Canine chondrocytes were seeded onto PCL and nanoPGA sheets either with or without ethanol treatment to examine cellular adhesion in vitro. Human chondrocytes were seeded onto three-dimensional bioresorbable composite scaffolds (PCL with surface coverage of nanoPGA) either with or without ethanol treatment and then implanted into athymic mice for 10 and 20 weeks. On construct retrieval, scanning electron microscopy showed canine auricular chondrocytes seeded onto ethanol-treated scaffolds in vitro developed extended cell processes contacting scaffold surfaces, a result suggesting cell-scaffold adhesion and a favorable microenvironment compared to the same cells with limited processes over untreated scaffolds. Adhesion of canine chondrocytes was statistically significantly greater (p ≤ 0.05) for ethanol-treated compared to untreated scaffold sheets. After implantation for 10 weeks, constructs of human auricular chondrocytes seeded onto ethanol-treated scaffolds were covered with glossy cartilage while constructs consisting of the same cells seeded onto untreated scaffolds revealed sparse connective tissue and cartilage regeneration. Following 10 weeks of implantation, RT-qPCR analyses of chondrocytes grown on ethanol-treated scaffolds showed greater expression levels for several cartilage-related genes compared to cells developed on untreated scaffolds with statistically significantly increased SRY-box transcription factor 5 (SOX5) and decreased interleukin-1α (inflammation-related) expression levels (p ≤ 0.05). Ethanol treatment of scaffolds led to increased cartilage production for 20- compared to 10-week constructs. While hydrophilicity of scaffolds was not assessed directly in the present findings, a possible factor supporting the summary data is that hydrophilicity may be enhanced for ethanol-treated nanoPGA/PCL scaffolds, an effect leading to improvement of chondrocyte adhesion, the cellular microenvironment and cartilage regeneration in tissue-engineered auricle constructs.
Collapse
Affiliation(s)
- Narihiko Hirano
- Department of Plastic and Reconstructive Surgery, Kindai University, Osakasayama, Japan
| | - Hirohisa Kusuhara
- Department of Plastic and Reconstructive Surgery, Kindai University, Osakasayama, Japan
| | - Yu Sueyoshi
- Department of Plastic and Reconstructive Surgery, Kindai University, Osakasayama, Japan
| | - Takeshi Teramura
- Institute of Advanced Clinical Medicine, Kindai University, Osakasayama, Japan
| | - Ananth Murthy
- Division of Plastic and Reconstructive Surgery, Children’s Hospital Medical Center, Akron, Ohio, United States of America
| | - Shinichi Asamura
- Department of Plastic and Reconstructive Surgery, Wakayama Medical College, Wakayama, Japan
| | - Noritaka Isogai
- Department of Plastic and Reconstructive Surgery, Kindai University, Osakasayama, Japan
- * E-mail: (WJL); (NI)
| | - Robin DiFeo Jacquet
- Division of Plastic and Reconstructive Surgery, Children’s Hospital Medical Center, Akron, Ohio, United States of America
- Department of Polymer Science, University of Akron, Akron, Ohio, United States of America
| | - William J. Landis
- Department of Polymer Science, University of Akron, Akron, Ohio, United States of America
- * E-mail: (WJL); (NI)
| |
Collapse
|
14
|
Yang Y, Lu Y, Zeng K, Heinze T, Groth T, Zhang K. Recent Progress on Cellulose-Based Ionic Compounds for Biomaterials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2000717. [PMID: 32270900 PMCID: PMC11469321 DOI: 10.1002/adma.202000717] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 05/06/2023]
Abstract
Glycans play important roles in all major kingdoms of organisms, such as archea, bacteria, fungi, plants, and animals. Cellulose, the most abundant polysaccharide on the Earth, plays a predominant role for mechanical stability in plants, and finds a plethora of applications by humans. Beyond traditional use, biomedical application of cellulose becomes feasible with advances of soluble cellulose derivatives with diverse functional moieties along the backbone and modified nanocellulose with versatile functional groups on the surface due to the native features of cellulose as both cellulose chains and supramolecular ordered domains as extractable nanocellulose. With the focus on ionic cellulose-based compounds involving both these groups primarily for biomedical applications, a brief introduction about glycoscience and especially native biologically active glycosaminoglycans with specific biomedical application areas on humans is given, which inspires further development of bioactive compounds from glycans. Then, both polymeric cellulose derivatives and nanocellulose-based compounds synthesized as versatile biomaterials for a large variety of biomedical applications, such as for wound dressings, controlled release, encapsulation of cells and enzymes, and tissue engineering, are separately described, regarding the diverse routes of synthesis and the established and suggested applications for these highly interesting materials.
Collapse
Affiliation(s)
- Yang Yang
- Wood Technology and Wood ChemistryUniversity of GoettingenBüsgenweg 4Göttingen37077Germany
- State Key Laboratory of Pulp and Paper EngineeringSouth China University of TechnologyWushan Road 381Guangzhou510640P. R. China
| | - Yi‐Tung Lu
- Department Biomedical MaterialsInstitute of PharmacyMartin Luther University Halle‐WittenbergHeinrich‐Damerow‐Strasse 4Halle (Saale)06120Germany
| | - Kui Zeng
- Wood Technology and Wood ChemistryUniversity of GoettingenBüsgenweg 4Göttingen37077Germany
| | - Thomas Heinze
- Institute of Organic Chemistry and Macromolecular ChemistryFriedrich Schiller University of JenaCentre of Excellence for Polysaccharide ResearchHumboldt Straße 10JenaD‐07743Germany
| | - Thomas Groth
- Department Biomedical MaterialsInstitute of PharmacyMartin Luther University Halle‐WittenbergHeinrich‐Damerow‐Strasse 4Halle (Saale)06120Germany
- Interdisciplinary Center of Materials ScienceMartin Luther University Halle‐WittenbergHalle (Saale)06120Germany
- Laboratory of Biomedical NanotechnologiesInstitute of Bionic Technologies and EngineeringI. M. Sechenov First Moscow State UniversityTrubetskaya Street 8119991MoscowRussian Federation
| | - Kai Zhang
- Wood Technology and Wood ChemistryUniversity of GoettingenBüsgenweg 4Göttingen37077Germany
| |
Collapse
|
15
|
Chiesa-Estomba CM, Aiastui A, González-Fernández I, Hernáez-Moya R, Rodiño C, Delgado A, Garces JP, Paredes-Puente J, Aldazabal J, Altuna X, Izeta A. Three-Dimensional Bioprinting Scaffolding for Nasal Cartilage Defects: A Systematic Review. Tissue Eng Regen Med 2021; 18:343-353. [PMID: 33864626 PMCID: PMC8169726 DOI: 10.1007/s13770-021-00331-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/11/2021] [Accepted: 01/22/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND In recent years, three-dimensional (3D)-printing of tissue-engineered cartilaginous scaffolds is intended to close the surgical gap and provide bio-printed tissue designed to fit the specific geometric and functional requirements of each cartilage defect, avoiding donor site morbidity and offering a personalizing therapy. METHODS To investigate the role of 3D-bioprinting scaffolding for nasal cartilage defects repair a systematic review of the electronic databases for 3D-Bioprinting articles pertaining to nasal cartilage bio-modelling was performed. The primary focus was to investigate cellular source, type of scaffold utilization, biochemical evaluation, histological analysis, in-vitro study, in-vivo study, animal model used, length of research, and placement of experimental construct and translational investigation. RESULTS From 1011 publications, 16 studies were kept for analysis. About cellular sources described, most studies used primary chondrocyte cultures. The cartilage used for cell isolation was mostly nasal septum. The most common biomaterial used for scaffold creation was polycaprolactone alone or in combination. About mechanical evaluation, we found a high heterogeneity, making it difficult to extract any solid conclusion. Regarding biological and histological characteristics of each scaffold, we found that the expression of collagen type I, collagen Type II and other ECM components were the most common patterns evaluated through immunohistochemistry on in-vitro and in-vivo studies. Only two studies made an orthotopic placement of the scaffolds. However, in none of the studies analyzed, the scaffold was placed in a subperichondrial pocket to rigorously simulate the cartilage environment. In contrast, scaffolds were implanted in a subcutaneous plane in almost all of the studies included. CONCLUSION The role of 3D-bioprinting scaffolding for nasal cartilage defects repair is growing field. Despite the amount of information collected in the last years and the first surgical applications described recently in humans. Further investigations are needed due to the heterogeneity on mechanical evaluation parameters, the high level of heterotopic scaffold implantation and the need for quantitative histological data.
Collapse
Affiliation(s)
- Carlos M Chiesa-Estomba
- Otorhinolaryngology - Head and Neck surgery Department, Osakidetza Basque Health Service, Donostia University Hospital, 20014, San Sebastian, Spain.
- Multidisciplinary 3D Printing Platform (3DPP), Biodonostia Health Research Institute, 20014, San Sebastian, Spain.
| | - Ana Aiastui
- Multidisciplinary 3D Printing Platform (3DPP), Biodonostia Health Research Institute, 20014, San Sebastian, Spain
- Biodonostia Health Research Institute, Histology Platform, 20014, San Sebastian, Spain
| | | | - Raquel Hernáez-Moya
- Multidisciplinary 3D Printing Platform (3DPP), Biodonostia Health Research Institute, 20014, San Sebastian, Spain
| | - Claudia Rodiño
- Biodonostia Health Research Institute, Histology Platform, 20014, San Sebastian, Spain
| | - Alba Delgado
- Biodonostia Health Research Institute, Histology Platform, 20014, San Sebastian, Spain
| | - Juan P Garces
- Multidisciplinary 3D Printing Platform (3DPP), Biodonostia Health Research Institute, 20014, San Sebastian, Spain
- Department of Pathology, Osakidetza Basque Health Service, Donostia University Hospital, 20014, San Sebastian, Spain
| | - Jacobo Paredes-Puente
- Multidisciplinary 3D Printing Platform (3DPP), Biodonostia Health Research Institute, 20014, San Sebastian, Spain
- Tecnun-University of Navarra, Pso. Mikeletegi 48, 20009, San Sebastian, Spain
| | - Javier Aldazabal
- Multidisciplinary 3D Printing Platform (3DPP), Biodonostia Health Research Institute, 20014, San Sebastian, Spain
- Tecnun-University of Navarra, Pso. Mikeletegi 48, 20009, San Sebastian, Spain
| | - Xabier Altuna
- Otorhinolaryngology - Head and Neck surgery Department, Osakidetza Basque Health Service, Donostia University Hospital, 20014, San Sebastian, Spain
| | - Ander Izeta
- Multidisciplinary 3D Printing Platform (3DPP), Biodonostia Health Research Institute, 20014, San Sebastian, Spain
- Tecnun-University of Navarra, Pso. Mikeletegi 48, 20009, San Sebastian, Spain
- Tissue Engineering Group, Biodonostia Health Research Institute, 20014, San Sebastian, Spain
| |
Collapse
|
16
|
Samadi A, Premaratne ID, Wright MA, Bernstein JL, Lara DO, Kim J, Zhao R, Bonassar LJ, Spector JA. Nipple Engineering: Maintaining Nipple Geometry with Externally Scaffolded Processed Autologous Costal Cartilage. J Plast Reconstr Aesthet Surg 2021; 74:2596-2603. [PMID: 33863678 DOI: 10.1016/j.bjps.2021.03.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 12/28/2020] [Accepted: 03/13/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Nipple reconstruction is the essential last step of breast reconstruction after total mastectomy, resulting in improved general and aesthetic satisfaction. However, most techniques are limited by secondary scar contracture and loss of neo-nipple projection leading to patient dissatisfaction. Approximately, 16,000 patients undergo autologous flap breast reconstruction annually, during which the excised costal cartilage (CC) is discarded. We propose utilizing processed CC placed within biocompatible 3D-printed external scaffolds to generate tissue cylinders that mimic the shape, size and biomechanical properties of native human nipple tissue while mitigating contracture and projection loss. METHODS External scaffolds were designed and then 3D-printed using polylactic acid (PLA). Patient-derived CC was processed by mincing or zesting, then packed into the scaffolds, implanted into nude rats and explanted after 3 months for volumetric, histologic and biomechanical analyses. Similar analyses were performed on native human nipple tissue and unprocessed CC. RESULTS After 3 months in vivo, gross analysis demonstrated significantly greater preservation of contour, projection and volume of the scaffolded nipples. Mechanical analysis demonstrated that processing of the cartilage resulted in implant equilibrium modulus values closer to that of the human nipple. Histologic analysis showed the presence of healthy and viable cartilage after 3 months in vivo, invested with fibrovascular tissue. CONCLUSIONS Autologous CC can be processed intraoperatively and placed within biocompatible external scaffolds to mimic the shape and biomechanical properties of the native human nipple. This allows for custom design and fabrication of individualized engineered autologous implants tailored to patient desire, without the loss of projection seen with traditional approaches.
Collapse
Affiliation(s)
- Arash Samadi
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medicine, New York, NY, United States of America
| | - Ishani D Premaratne
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medicine, New York, NY, United States of America
| | - Matthew A Wright
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medicine, New York, NY, United States of America
| | - Jaime L Bernstein
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medicine, New York, NY, United States of America
| | - Daniel O Lara
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medicine, New York, NY, United States of America
| | - Jongkil Kim
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Runlei Zhao
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medicine, New York, NY, United States of America
| | - Lawrence J Bonassar
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Jason A Spector
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medicine, New York, NY, United States of America; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America.
| |
Collapse
|
17
|
Otto IA, Capendale PE, Garcia JP, de Ruijter M, van Doremalen RFM, Castilho M, Lawson T, Grinstaff MW, Breugem CC, Kon M, Levato R, Malda J. Biofabrication of a shape-stable auricular structure for the reconstruction of ear deformities. Mater Today Bio 2021; 9:100094. [PMID: 33665603 PMCID: PMC7903133 DOI: 10.1016/j.mtbio.2021.100094] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 01/04/2021] [Accepted: 01/08/2021] [Indexed: 11/04/2022] Open
Abstract
Bioengineering of the human auricle remains a significant challenge, where the complex and unique shape, the generation of high-quality neocartilage, and shape preservation are key factors. Future regenerative medicine–based approaches for auricular cartilage reconstruction will benefit from a smart combination of various strategies. Our approach to fabrication of an ear-shaped construct uses hybrid bioprinting techniques, a recently identified progenitor cell population, previously validated biomaterials, and a smart scaffold design. Specifically, we generated a 3D-printed polycaprolactone (PCL) scaffold via fused deposition modeling, photocrosslinked a human auricular cartilage progenitor cell–laden gelatin methacryloyl (gelMA) hydrogel within the scaffold, and cultured the bioengineered structure in vitro in chondrogenic media for 30 days. Our results show that the fabrication process maintains the viability and chondrogenic phenotype of the cells, that the compressive properties of the combined PCL and gelMA hybrid auricular constructs are similar to native auricular cartilage, and that biofabricated hybrid auricular structures exhibit excellent shape fidelity compared with the 3D digital model along with deposition of cartilage-like matrix in both peripheral and central areas of the auricular structure. Our strategy affords an anatomically enhanced auricular structure with appropriate mechanical properties, ensures adequate preservation of the auricular shape during a dynamic in vitro culture period, and enables chondrogenically potent progenitor cells to produce abundant cartilage-like matrix throughout the auricular construct. The combination of smart scaffold design with 3D bioprinting and cartilage progenitor cells holds promise for the development of clinically translatable regenerative medicine strategies for auricular reconstruction. First application of human auricular cartilage progenitor cells for bioprinting. Dual-printing of hybrid ear-shaped constructs with excellent shape fidelity over time. Strategy and design ensured adequate deposition of cartilage-like matrix throughout large auricular constructs.
Collapse
Affiliation(s)
- I A Otto
- Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands.,Department of Plastic, Reconstructive and Hand Surgery, University Medical Center Utrecht, Utrecht, the Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - P E Capendale
- Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - J P Garcia
- Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - M de Ruijter
- Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - R F M van Doremalen
- Robotics and Mechatronics, Faculty of Electrical Engineering, Mathematics & Computer Science, University of Twente, Enschede, the Netherlands.,Bureau Science & Innovation, Deventer Hospital, Deventer, the Netherlands
| | - M Castilho
- Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - T Lawson
- Departments of Chemistry and Biomedical Engineering, Boston University, Boston, USA
| | - M W Grinstaff
- Departments of Chemistry and Biomedical Engineering, Boston University, Boston, USA
| | - C C Breugem
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam University Medical Center, Emma Children's Hospital, Amsterdam, the Netherlands
| | - M Kon
- Department of Plastic, Reconstructive and Hand Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - R Levato
- Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - J Malda
- Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, the Netherlands.,Department of Clinical Sciences, Faculty of Veterinary Science, Utrecht University, the Netherlands
| |
Collapse
|
18
|
Jang CH, Koo Y, Kim G. ASC/chondrocyte-laden alginate hydrogel/PCL hybrid scaffold fabricated using 3D printing for auricle regeneration. Carbohydr Polym 2020; 248:116776. [DOI: 10.1016/j.carbpol.2020.116776] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/12/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022]
|
19
|
Abstract
The field of Tissue Engineering and Regenerative Medicine has evolved rapidly over the past thirty years. This review will summarize its history, current status and direction through the lens of clinical need, its progress through science in the laboratory and application back into patients. We can take pride in the fact that much effort and progress began with the surgical problems of children and that many surgeons in the pediatric surgical specialties have become pioneers and investigators in this new field of science, engineering, and medicine. Although the field has yet to fulfill its great promise, there have been several examples where a therapy has progressed from the first idea to human application within a short span of time and, in many cases, it has been applied in the surgical care of children.
Collapse
|
20
|
Wu Y, Heikal L, Ferns G, Ghezzi P, Nokhodchi A, Maniruzzaman M. 3D Bioprinting of Novel Biocompatible Scaffolds for Endothelial Cell Repair. Polymers (Basel) 2019; 11:E1924. [PMID: 31766610 PMCID: PMC6960937 DOI: 10.3390/polym11121924] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 12/16/2022] Open
Abstract
The aim of this study was to develop and evaluate an optimized 3D bioprinting technology in order to fabricate novel scaffolds for the application of endothelial cell repair. Various biocompatible and biodegradable macroporous scaffolds (D = 10 mm) with interconnected pores (D = ~500 µm) were fabricated using a commercially available 3D bioprinter (r3bEL mini, SE3D, USA). The resolution of the printing layers was set at ~100 µm for all scaffolds. Various compositions of polylactic acid (PLA), polyethylene glycol (PEG) and pluronic F127 (F127) formulations were prepared and optimized to develop semi-solid viscous bioinks. Either dimethyloxalylglycine (DMOG) or erythroprotein (EPO) was used as a model drug and loaded in the viscous biocompatible ink formulations with a final concentration of 30% (w/w). The surface analysis of the bioinks via a spectroscopic analysis revealed a homogenous distribution of the forming materials throughout the surface, whereas SEM imaging of the scaffolds showed a smooth surface with homogenous macro-porous texture and precise pore size. The rheological and mechanical analyses showed optimum rheological and mechanical properties of each scaffold. As the drug, DMOG, is a HIF-1 inducer, its release from the scaffolds into PBS solution was measured indirectly using a bioassay for HIF-1α. This showed that the release of DMOG was sustained over 48 h. The release of DMOG was enough to cause a significant increase in HIF-1α levels in the bioassay, and when incubated with rat aortic endothelial cells (RAECs) for 2 h resulted in transcriptional activation of a HIF-1α target gene (VEGF). The optimum time for the increased expression of VEGF gene was approximately 30 min and was a 3-4-fold increase above baseline. This study provides a proof of concept, that a novel bioprinting platform can be exploited to develop biodegradable composite scaffolds for potential clinical applications in endothelial cell repair in cardiovascular disease (CVD), or in other conditions in which endothelial damage occurs.
Collapse
Affiliation(s)
- Yan Wu
- Department of Pharmacy (Chemistry), School of Life Sciences, University of Sussex, Brighton BN1 9QJ, UK;
| | - Lamia Heikal
- Brighton and Sussex Medical School, Brighton BN1 9RH, UK or (G.F.); (P.G.)
- Faculty of Pharmacy, Department of Pharmaceutics, Alexandria University, El Sultan Hussein St AZARITA-Qesm Al Attarin, Alexandria Governorate 21521, Egypt
| | - Gordon Ferns
- Brighton and Sussex Medical School, Brighton BN1 9RH, UK or (G.F.); (P.G.)
| | - Pietro Ghezzi
- Brighton and Sussex Medical School, Brighton BN1 9RH, UK or (G.F.); (P.G.)
| | - Ali Nokhodchi
- Department of Pharmacy (Chemistry), School of Life Sciences, University of Sussex, Brighton BN1 9QJ, UK;
- Drug Applied Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 51664, Iran
| | - Mohammed Maniruzzaman
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas, Austin, TX 78712, USA
| |
Collapse
|
21
|
Shen S, Chen M, Guo W, Li H, Li X, Huang S, Luo X, Wang Z, Wen Y, Yuan Z, Zhang B, Peng L, Gao C, Guo Q, Liu S, Zhuo N. Three Dimensional Printing-Based Strategies for Functional Cartilage Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:187-201. [PMID: 30608012 DOI: 10.1089/ten.teb.2018.0248] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Shi Shen
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Mingxue Chen
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Weimin Guo
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Haojiang Li
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xu Li
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Suqiong Huang
- Department of Liver and Gallbladder Disease, The Affiliated Chinese Traditional Medicine Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Xujiang Luo
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Zhenyong Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
- First Department of Orthopedics, First Affiliated Hospital of Jiamusi University, Jiamusi, People's Republic of China
| | - Yang Wen
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Zhiguo Yuan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Bin Zhang
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Liqing Peng
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Chao Gao
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Quanyi Guo
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Shuyun Liu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Naiqiang Zhuo
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| |
Collapse
|
22
|
Lehmann J, Nürnberger S, Narcisi R, Stok KS, van der Eerden BCJ, Koevoet WJLM, Kops N, Ten Berge D, van Osch GJ. Recellularization of auricular cartilage via elastase-generated channels. Biofabrication 2019; 11:035012. [PMID: 30921774 DOI: 10.1088/1758-5090/ab1436] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Decellularized tissue matrices are promising substrates for tissue generation by stem cells to replace poorly regenerating tissues such as cartilage. However, the dense matrix of decellularized cartilage impedes colonisation by stem cells. Here, we show that digestion of elastin fibre bundles traversing auricular cartilage creates channels through which cells can migrate into the matrix. Human chondrocytes and bone marrow-derived mesenchymal stromal cells efficiently colonise elastin-treated scaffolds through these channels, restoring a glycosaminoglycan-rich matrix and improving mechanical properties while maintaining size and shape of the restored tissue. The scaffolds are also rapidly colonised by endogenous cartilage-forming cells in a subcutaneously implanted osteochondral biopsy model. Creating channels for cells in tissue matrices may be a broadly applicable strategy for recellularization and restoration of tissue function.
Collapse
Affiliation(s)
- J Lehmann
- Department of Otorhinolaryngology and Head and Neck Surgery Erasmus MC, Rotterdam, The Netherlands. Department of Cell Biology Erasmus MC, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Liao J, Chen Y, Chen J, He B, Qian L, Xu J, Wang A, Li Q, Xie H, Zhou J. Auricle shaping using 3D printing and autologous diced cartilage. Laryngoscope 2019; 129:2467-2474. [PMID: 30843613 PMCID: PMC6850318 DOI: 10.1002/lary.27752] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/06/2018] [Accepted: 11/19/2018] [Indexed: 11/05/2022]
Abstract
OBJECTIVE To reconstruct the auricle using a porous, hollow, three-dimensional (3D)-printed mold and autologous diced cartilage mixed with platelet-rich plasma (PRP). METHODS Materialise Magics v20.03 was used to design a 3D, porous, hollow auricle mold. Ten molds were printed by selective laser sintering with polyamide. Cartilage grafts were harvested from one ear of a New Zealand rabbit, and PRP was prepared using 10 mL of auricular blood from the same animal. Ear cartilage was diced into 0.5- to 2.0-mm pieces, weighed, mixed with PRP, and then placed inside the hollow mold. Composite grafts were then implanted into the backs of respective rabbits (n = 10) for 4 months. The shape and composition of the diced cartilage were assessed histologically, and biomechanical testing was used to determine stiffness. RESULTS The 3D-printed auricle molds were 0.6-mm thick and showed connectivity between the internal and external surfaces, with round pores of 0.1 to 0.3 cm. After 4 months, the diced cartilage pieces had fused into an auricular shape with high fidelity to the anthropotomy. The weight of the diced cartilage was 5.157 ± 0.230 g (P > 0.05, compared with preoperative). Histological staining showed high chondrocyte viability and the production of collagen II, glycosaminoglycans, and other cartilaginous matrix components. In unrestricted compression tests, auricle stiffness was 0.158 ± 0.187 N/mm, similar to that in humans. CONCLUSION Auricle grafts were constructed successfully through packing a 3D-printed, porous, hollow auricle mold with diced cartilage mixed with PRP. The auricle cartilage contained viable chondrocytes, appropriate extracellular matrix components, and good mechanical properties. LEVELS OF EVIDENCE NA. Laryngoscope, 129:2467-2474, 2019.
Collapse
Affiliation(s)
- Junlin Liao
- Departments of Medical Cosmetology, The First Affiliated Hospital, University of South China, Hengyang.,Departments of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha
| | - Yong Chen
- Departments of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha.,Emergency Department, The First Hospital of Changsha, Changsha
| | - Jia Chen
- Departments of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha
| | - Bin He
- Departments of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha.,Departments of Burn and Plastic Surgery, Ningxiang People's Hospital, Ningxiang, Hunan
| | - Li Qian
- Departments of Burn and Plastic Surgery, The Second Xiangya Hospital, Central South University, Changsha
| | - Jiaqin Xu
- Departments of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha.,Departments of Burn and Plastic Surgery, Hainan People's Hospital, Haikou, Hainan
| | - Aijun Wang
- Department of Surgery, Davis Health System, University of California, Sacramento, California, U.S.A
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Jiaotong University Medical School, Ninth People's Hospital, Shanghai, People's Republic of China
| | - Hongju Xie
- Departments of Medical Cosmetology, The First Affiliated Hospital, University of South China, Hengyang
| | - Jianda Zhou
- Departments of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha
| |
Collapse
|
24
|
Chiu LLY, Weber JF, Waldman SD. Engineering of scaffold-free tri-layered auricular tissues for external ear reconstruction. Laryngoscope 2019; 129:E272-E283. [DOI: 10.1002/lary.27823] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/15/2018] [Accepted: 12/31/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Loraine L. Y. Chiu
- Department of Chemical Engineering; Ryerson University; Toronto Ontario Canada
- Li Ka Shing Knowledge Institute; St. Michael's Hospital; Toronto Ontario Canada
| | - Joanna F. Weber
- Department of Chemical Engineering; Ryerson University; Toronto Ontario Canada
- Li Ka Shing Knowledge Institute; St. Michael's Hospital; Toronto Ontario Canada
| | - Stephen D. Waldman
- Department of Chemical Engineering; Ryerson University; Toronto Ontario Canada
- Li Ka Shing Knowledge Institute; St. Michael's Hospital; Toronto Ontario Canada
| |
Collapse
|
25
|
Cohen BP, Bernstein JL, Morrison KA, Spector JA, Bonassar LJ. Tissue engineering the human auricle by auricular chondrocyte-mesenchymal stem cell co-implantation. PLoS One 2018; 13:e0202356. [PMID: 30356228 PMCID: PMC6200177 DOI: 10.1371/journal.pone.0202356] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 08/01/2018] [Indexed: 01/21/2023] Open
Abstract
Children suffering from microtia have few options for auricular reconstruction. Tissue engineering approaches attempt to replicate the complex anatomy and structure of the ear with autologous cartilage but have been limited by access to clinically accessible cell sources. Here we present a full-scale, patient-based human ear generated by implantation of human auricular chondrocytes and human mesenchymal stem cells in a 1:1 ratio. Additional disc construct surrogates were generated with 1:0, 1:1, and 0:1 combinations of auricular chondrocytes and mesenchymal stem cells. After 3 months in vivo, monocellular auricular chondrocyte discs and 1:1 disc and ear constructs displayed bundled collagen fibers in a perichondrial layer, rich proteoglycan deposition, and elastin fiber network formation similar to native human auricular cartilage, with the protein composition and mechanical stiffness of native tissue. Full ear constructs with a 1:1 cell combination maintained gross ear structure and developed a cartilaginous appearance following implantation. These studies demonstrate the successful engineering of a patient-specific human auricle using exclusively human cell sources without extensive in vitro tissue culture prior to implantation, a critical step towards the clinical application of tissue engineering for auricular reconstruction.
Collapse
Affiliation(s)
- Benjamin P Cohen
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
| | - Jaime L Bernstein
- Division of Plastic Surgery, Weill Cornell Medical College, New York, New York, United States of America
| | - Kerry A Morrison
- Division of Plastic Surgery, Weill Cornell Medical College, New York, New York, United States of America
| | - Jason A Spector
- Division of Plastic Surgery, Weill Cornell Medical College, New York, New York, United States of America
| | - Lawrence J Bonassar
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, United States of America.,Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
26
|
Morrison RJ, Nasser HB, Kashlan KN, Zopf DA, Milner DJ, Flanangan CL, Wheeler MB, Green GE, Hollister SJ. Co-culture of adipose-derived stem cells and chondrocytes on three-dimensionally printed bioscaffolds for craniofacial cartilage engineering. Laryngoscope 2018; 128:E251-E257. [PMID: 29668079 PMCID: PMC6105552 DOI: 10.1002/lary.27200] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 02/16/2018] [Accepted: 03/01/2018] [Indexed: 11/11/2022]
Abstract
OBJECTIVES/HYPOTHESIS Reconstruction of craniofacial cartilagenous defects are among the most challenging surgical procedures in facial plastic surgery. Bioengineered craniofacial cartilage holds immense potential to surpass current reconstructive options, but limitations to clinical translation exist. We endeavored to determine the viability of utilizing adipose-derived stem cell-chondrocyte co-culture and three-dimensional (3D) printing to produce 3D bioscaffolds for cartilage tissue engineering. We describe a feasibility study revealing a novel approach for cartilage tissue engineering with in vitro and in vivo animal data. METHODS Porcine adipose-derived stem cells and chondrocytes were isolated and co-seeded at 1:1, 2:1, 5:1, 10:1, and 0:1 experimental ratios in a hyaluronic acid/collagen hydrogel in the pores of 3D-printed polycaprolactone scaffolds to form 3D bioscaffolds for cartilage tissue engineering. Bioscaffolds were cultured in vitro without growth factors for 4 weeks and then implanted into the subcutaneous tissue of athymic rats for an additional 4 weeks before sacrifice. Bioscaffolds were subjected to histologic, immunohistochemical, and biochemical analysis. RESULTS Successful production of cartilage was achieved using a co-culture model of adipose-derived stem cells and chondrocytes without the use of exogenous growth factors. Histology demonstrated cartilage growth for all experimental ratios at the post-in vivo time point confirmed with type II collagen immunohistochemistry. There was no difference in sulfated-glycosaminoglycan production between experimental groups. CONCLUSION Tissue-engineered cartilage was successfully produced on 3D-printed bioresorbable scaffolds using an adipose-derived stem cell and chondrocyte co-culture technique. This potentiates co-culture as a solution for several key barriers to a clinically translatable cartilage tissue engineering process. LEVEL OF EVIDENCE NA. Laryngoscope, 128:E251-E257, 2018.
Collapse
Affiliation(s)
- Robert J. Morrison
- Department of Otolaryngology-Head & Neck Surgery, Vanderbilt University, Nashville, TN, USA
| | - Hassan B. Nasser
- Department of Otolaryngology-Head & Neck Surgery, University of California Los Angeles, Los Angeles, CA, USA
| | - Khaled N. Kashlan
- Department of Otolaryngology-Head & Neck Surgery, Henry Ford Hospital, Detroit, MI, USA
| | - David A. Zopf
- Department of Otolaryngology-Head & Neck Surgery, Division of Pediatric Otolaryngology, University of Michigan, Ann Arbor, MI, USA
| | - Derek J. Milner
- Carel R. Woese Institute for Genomic Biology, Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana-Champaign, IL, USA
| | - Colleen L. Flanangan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Matthew B. Wheeler
- Carel R. Woese Institute for Genomic Biology, Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana-Champaign, IL, USA
| | - Glenn E. Green
- Department of Otolaryngology-Head & Neck Surgery, Division of Pediatric Otolaryngology, University of Michigan, Ann Arbor, MI, USA
| | - Scott J. Hollister
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Wallace A. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
27
|
Rotter N, Zenobi-Wong M. [Regeneration - A New Therapeutic Dimension in Otorhinolaryngology]. Laryngorhinootologie 2018; 97:S185-S213. [PMID: 29905357 PMCID: PMC6290928 DOI: 10.1055/s-0043-122309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Regeneration as a therapeutic priniciple and regenerative medicine in general are promising new strategies to add new therapeutic dimensions to our current treatment options. Today, reconstructive surgery, drugs and implants such as the cochlear implant can replace the functions of damaged tissues. In contrast, regenerative therapies aim at the replacement of the damaged tissues themselves while at the same time replacing their lost tissue function. In this review article new technologies such as 3D-bioprinting and the application of decellularised tissues as biomaterials are introduced and explained. A summary of current preclinical and clinical regenerative studies in otorhinolaryngology is complementing these basic aspects.
Collapse
Affiliation(s)
- Nicole Rotter
- Klinik für Hals-Nasen-Ohrenheilkunde, Universitätsmedizin Mannheim, Universitätsklinikum Mannheim
| | | |
Collapse
|
28
|
Abstract
Simulating natural characteristics and aesthetics in reconstructed ears has provided a complex 3-dimensional puzzle for those treating patients with microtia. Costochondral grafts remain the gold standard for autologous reconstruction. However, other options such as Medpor and prosthetics are indicated depending on patient circumstances and personal choice. Research into tissue engineering offers an alternative method to a traditional surgical approach that may reduce donor-site morbidity. However, tissue engineering for microtia reconstruction brings new challenges such as cell sourcing, promotion of chondrogenesis, scaffold vascularization, and prevention of scaffold contraction. Advancements in 3D printing, nanofiber utilization, stem cell technologies, and decellularization techniques have played significant roles in overcoming these challenges. These recent advancements and reports of a successful clinical-scale study in an immunocompetent animal suggest a promising outlook for future clinical application of tissue engineering for auricular reconstruction.
Collapse
|
29
|
Reighard CL, Hollister SJ, Zopf DA. Auricular reconstruction from rib to 3D printing. JOURNAL OF 3D PRINTING IN MEDICINE 2018; 2:35-41. [PMID: 29607095 PMCID: PMC5824712 DOI: 10.2217/3dp-2017-0017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/16/2017] [Indexed: 12/19/2022]
Abstract
The human ear imparts critical form and function and remains one of the most challenging facial features to reconstruct. Over the past century, surgeons have developed numerous techniques and materials for total auricular reconstruction. Refined costal cartilage techniques have remained the gold standard for the past half-century. Recent advancements with novel materials, tissue engineering and 3D printing provide immense potential; however, prohibitive costs and regulatory steps remain as barriers to clinical translation.
Collapse
Affiliation(s)
| | - Scott J Hollister
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - David A Zopf
- Otolaryngology – Head & Neck Surgery, Pediatric Division, University of Michigan Health Systems, CS Mott Children's Hospital, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
30
|
Auricular Tissue Engineering Using Osteogenic Differentiation of Adipose Stem Cells with Small Intestine Submucosa. Plast Reconstr Surg 2017; 140:297-305. [DOI: 10.1097/prs.0000000000003522] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
31
|
Gu Y, Kang N, Dong P, Liu X, Wang Q, Fu X, Yan L, Jiang H, Cao Y, Xiao R. Chondrocytes from congenital microtia possess an inferior capacity for in vivo cartilage regeneration to healthy ear chondrocytes. J Tissue Eng Regen Med 2017; 12:e1737-e1746. [PMID: 27860439 DOI: 10.1002/term.2359] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 09/06/2016] [Accepted: 11/09/2016] [Indexed: 01/01/2023]
Abstract
The remnant auricular cartilage from microtia has become a valuable cell source for ear regeneration. It is important to clarify the issue of whether the genetically defective microtia chondrocytes could engineer cartilage tissue comparable to healthy ear chondrocytes. In the current study, the histology and cell yield of native microtia and normal ear cartilage were investigated, and the biological characteristics of derived chondrocytes examined, including proliferation, chondrogenic phenotype and cell migration. Furthermore, the in vivo cartilage-forming capacity of passaged microtia and normal auricular chondrocytes were systematically compared by seeding them onto polyglycolic acid/polylactic acid scaffold to generate tissue engineered cartilage in nude mice. Through histological examinations and quantitative analysis of glycosaminoglycan, Young's modulus, and the expression of cartilage-related genes, it was found that microtia chondrocytes had a slower dedifferentiation rate with the decreased expression of stemness-related genes, and weaker migration ability than normal ear chondrocytes, and the microtia chondrocytes-engineered cartilage was biochemically and biomechanically inferior to that constructed using normal ear chondrocytes. This study provides valuable information for the clinical application of the chondrocytes derived from congenital microtia to engineer cartilage. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Yunpeng Gu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Ning Kang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Ping Dong
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Xia Liu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Qian Wang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Xin Fu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Li Yan
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Haiyue Jiang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Yilin Cao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Ran Xiao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| |
Collapse
|
32
|
Otto IA, van Doremalen RFM, Melchels FPW, Kolodzynski MN, Pouran B, Malda J, Kon M, Breugem CC. Accurate Measurements of the Skin Surface Area of the Healthy Auricle and Skin Deficiency in Microtia Patients. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2016; 4:e1146. [PMID: 28293505 PMCID: PMC5222650 DOI: 10.1097/gox.0000000000001146] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 10/06/2016] [Indexed: 12/11/2022]
Abstract
Background: The limited cranial skin covering auricular implants is an important yet underrated factor in auricular reconstruction for both reconstruction surgery and tissue engineering strategies. We report exact measurements on skin deficiency in microtia patients and propose an accessible preoperative method for these measurements. Methods: Plaster ear models (n = 11; male:female = 2:1) of lobular-type microtia patients admitted to the University Medical Center Utrecht in The Netherlands were scanned using a micro-computed tomographic scanner or a cone-beam computed tomographic scanner. The resulting images were converted into mesh models from which the surface area could be calculated. Results: The mean total skin area of an adult-size healthy ear was 47.3 cm2, with 49.0 cm2 in men and 44.3 cm2 in women. Microtia ears averaged 14.5 cm2, with 15.6 cm2 in men and 12.6 cm2 in women. The amount of skin deficiency was 25.4 cm2, with 26.7 cm2 in men and 23.1 cm2 in women. Conclusions: This study proposes a novel method to provide quantitative data on the skin surface area of the healthy adult auricle and the amount of skin deficiency in microtia patients. We demonstrate that the microtia ear has less than 50% of skin available compared with healthy ears. Limited skin availability in microtia patients can lead to healing problems after auricular reconstruction and poses a significant challenge in the development of tissue-engineered cartilage implants. The results of this study could be used to evaluate outcomes and investigate new techniques with regard to tissue-engineered auricular constructs.
Collapse
Affiliation(s)
- Iris A Otto
- Departments of Plastic, Reconstructive and Hand Surgery and Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Delft, The Netherlands; Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands; and Department of Plastic Surgery, Meander Medical Centre, Amersfoort, The Netherlands
| | - Rob F M van Doremalen
- Departments of Plastic, Reconstructive and Hand Surgery and Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Delft, The Netherlands; Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands; and Department of Plastic Surgery, Meander Medical Centre, Amersfoort, The Netherlands
| | - Ferry P W Melchels
- Departments of Plastic, Reconstructive and Hand Surgery and Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Delft, The Netherlands; Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands; and Department of Plastic Surgery, Meander Medical Centre, Amersfoort, The Netherlands
| | - Michail N Kolodzynski
- Departments of Plastic, Reconstructive and Hand Surgery and Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Delft, The Netherlands; Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands; and Department of Plastic Surgery, Meander Medical Centre, Amersfoort, The Netherlands
| | - Behdad Pouran
- Departments of Plastic, Reconstructive and Hand Surgery and Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Delft, The Netherlands; Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands; and Department of Plastic Surgery, Meander Medical Centre, Amersfoort, The Netherlands
| | - Jos Malda
- Departments of Plastic, Reconstructive and Hand Surgery and Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Delft, The Netherlands; Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands; and Department of Plastic Surgery, Meander Medical Centre, Amersfoort, The Netherlands
| | - Moshe Kon
- Departments of Plastic, Reconstructive and Hand Surgery and Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Delft, The Netherlands; Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands; and Department of Plastic Surgery, Meander Medical Centre, Amersfoort, The Netherlands
| | - Corstiaan C Breugem
- Departments of Plastic, Reconstructive and Hand Surgery and Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Delft, The Netherlands; Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands; and Department of Plastic Surgery, Meander Medical Centre, Amersfoort, The Netherlands
| |
Collapse
|
33
|
Morrison KA, Cohen BP, Asanbe O, Dong X, Harper A, Bonassar LJ, Spector JA. Optimizing cell sourcing for clinical translation of tissue engineered ears. Biofabrication 2016; 9:015004. [PMID: 27917821 DOI: 10.1088/1758-5090/9/1/015004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Background . Currently, the major impediment to clinical translation of our previously described platform for the fabrication of high fidelity, patient-specific tissue engineered ears is the development of a clinically optimal cell sourcing strategy. A limited autologous auricular chondrocyte (AuC) supply in conjunction with rapid chondrocyte de-differentiation during in vitro expansion currently makes clinical translation more challenging. Mesenchymal stem cells (MSCs) offer significant promise due to their inherent chondrogenic potential, and large availability through minimally invasive procedures. Herein, we demonstrate the promise of AuC/MSC co-culture to fabricate elastic cartilage using 50% fewer AuC than standard approaches. METHODS Bovine auricular chondrocytes (bAuC) and bovine MSC (bMSC) were encapsulated within 10 mg ml-1 type I collagen hydrogels in ratios of bAuC:bMSC 100:0, 50:50, and 0:100 at a density of 25 million cells ml-1 hydrogel. One mm thick collagen sheet gels were fabricated, and thereafter, 8 mm diameter discs were extracted using a biopsy punch. Discs were implanted subcutaneously in the dorsa of nude mice (NU/NU) and harvested after 1 and 3 months. RESULTS Gross analysis of explanted discs revealed bAuC:bMSC co-culture discs maintained their size and shape, and exhibited native auricular cartilage-like elasticity after 1 and 3 months of implantation. Co-culture discs developed into auricular cartilage, with viable chondrocytes within lacunae, copious proteoglycan and elastic fiber deposition, and a distinct perichondrial layer. Biochemical analysis confirmed that co-culture discs deposited critical cartilage molecular components more readily than did both bAuC and bMSC discs after 1 and 3 months, and proteoglycan content significantly increased between 1 and 3 months. CONCLUSION We have successfully demonstrated an innovative cell sourcing strategy that facilitates our efforts to achieve clinical translation of our high fidelity, patient-specific ears for auricular reconstruction utilizing only half of the requisite auricular chondrocytes to fabricate mature elastic cartilage.
Collapse
Affiliation(s)
- Kerry A Morrison
- Laboratory for Bioregenerative Medicine and Surgery, Department of Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, NY, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Bos E, Doerga P, Breugem C, van Zuijlen P. The burned ear; possibilities and challenges in framework reconstruction and coverage. Burns 2016; 42:1387-1395. [DOI: 10.1016/j.burns.2016.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 01/20/2016] [Accepted: 02/09/2016] [Indexed: 10/21/2022]
|
35
|
Modulevsky DJ, Cuerrier CM, Pelling AE. Biocompatibility of Subcutaneously Implanted Plant-Derived Cellulose Biomaterials. PLoS One 2016; 11:e0157894. [PMID: 27328066 PMCID: PMC4915699 DOI: 10.1371/journal.pone.0157894] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 06/07/2016] [Indexed: 12/22/2022] Open
Abstract
There is intense interest in developing novel biomaterials which support the invasion and proliferation of living cells for potential applications in tissue engineering and regenerative medicine. Decellularization of existing tissues have formed the basis of one major approach to producing 3D scaffolds for such purposes. In this study, we utilize the native hypanthium tissue of apples and a simple preparation methodology to create implantable cellulose scaffolds. To examine biocompatibility, scaffolds were subcutaneously implanted in wild-type, immunocompetent mice (males and females; 6-9 weeks old). Following the implantation, the scaffolds were resected at 1, 4 and 8 weeks and processed for histological analysis (H&E, Masson's Trichrome, anti-CD31 and anti-CD45 antibodies). Histological analysis revealed a characteristic foreign body response to the scaffold 1 week post-implantation. However, the immune response was observed to gradually disappear by 8 weeks post-implantation. By 8 weeks, there was no immune response in the surrounding dermis tissue and active fibroblast migration within the cellulose scaffold was observed. This was concomitant with the deposition of a new collagen extracellular matrix. Furthermore, active blood vessel formation within the scaffold was observed throughout the period of study indicating the pro-angiogenic properties of the native scaffolds. Finally, while the scaffolds retain much of their original shape they do undergo a slow deformation over the 8-week length of the study. Taken together, our results demonstrate that native cellulose scaffolds are biocompatible and exhibit promising potential as a surgical biomaterial.
Collapse
Affiliation(s)
- Daniel J. Modulevsky
- Centre for Interdisciplinary NanoPhysics, University of Ottawa, Ottawa, Ontario, Canada
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Charles M. Cuerrier
- Centre for Interdisciplinary NanoPhysics, University of Ottawa, Ottawa, Ontario, Canada
- Department of Physics, University of Ottawa, Ottawa, Ontario, Canada
| | - Andrew E. Pelling
- Centre for Interdisciplinary NanoPhysics, University of Ottawa, Ottawa, Ontario, Canada
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
- Department of Physics, University of Ottawa, Ottawa, Ontario, Canada
- Institute for Science, Society and Policy, University of Ottawa, Ottawa, Ontario, Canada
- SymbioticA, School of Anatomy, Physiology and Human Biology, University of Western Australia, Perth WA 6009, Australia
| |
Collapse
|
36
|
Chondrogenesis by bone marrow‐derived mesenchymal stem cells grown in chondrocyte‐conditioned medium for auricular reconstruction. J Tissue Eng Regen Med 2016; 11:2763-2773. [DOI: 10.1002/term.2171] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 01/15/2016] [Accepted: 02/10/2016] [Indexed: 01/10/2023]
|
37
|
Visscher DO, Bos EJ, Peeters M, Kuzmin NV, Groot ML, Helder MN, van Zuijlen PPM. Cartilage Tissue Engineering: Preventing Tissue Scaffold Contraction Using a 3D-Printed Polymeric Cage. Tissue Eng Part C Methods 2016; 22:573-84. [PMID: 27089896 DOI: 10.1089/ten.tec.2016.0073] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Scaffold contraction is a common but underestimated problem in the field of tissue engineering. It becomes particularly problematic when creating anatomically complex shapes such as the ear. The aim of this study was to develop a contraction-free biocompatible scaffold construct for ear cartilage tissue engineering. To address this aim, we used three constructs: (i) a fibrin/hyaluronic acid (FB/HA) hydrogel, (ii) a FB/HA hydrogel combined with a collagen I/III scaffold, and (iii) a cage construct containing (ii) surrounded by a 3D-printed poly-ɛ-caprolactone mold. A wide range of different cell types were tested within these constructs, including chondrocytes, perichondrocytes, adipose-derived mesenchymal stem cells, and their combinations. After in vitro culturing for 1, 14, and 28 days, all constructs were analyzed. Macroscopic observation showed severe contraction of the cell-seeded hydrogel (i). This could be prevented, in part, by combining the hydrogel with the collagen scaffold (ii) and prevented in total using the 3D-printed cage construct (iii). (Immuno)histological analysis, multiphoton laser scanning microscopy, and biomechanical analysis showed extracellular matrix deposition and increased Young's modulus and thereby the feasibility of ear cartilage engineering. These results demonstrated that the 3D-printed cage construct is an adequate model for contraction-free ear cartilage engineering using a range of cell combinations.
Collapse
Affiliation(s)
- Dafydd O Visscher
- 1 Department of Plastic, Reconstructive & Hand Surgery, VU Medical Center , Amsterdam, Netherlands
- 2 CTRM/MOVE Research Institute , Amsterdam, Netherlands
| | - Ernst J Bos
- 1 Department of Plastic, Reconstructive & Hand Surgery, VU Medical Center , Amsterdam, Netherlands
- 2 CTRM/MOVE Research Institute , Amsterdam, Netherlands
| | - Mirte Peeters
- 2 CTRM/MOVE Research Institute , Amsterdam, Netherlands
- 3 Department of Orthopedic Surgery, VU Medical Center , Amsterdam, Netherlands
| | - Nikolay V Kuzmin
- 4 LaserLaB Amsterdam, Department of Physics, Vrije Universiteit , Amsterdam, Netherlands
| | - Marie Louise Groot
- 4 LaserLaB Amsterdam, Department of Physics, Vrije Universiteit , Amsterdam, Netherlands
| | - Marco N Helder
- 2 CTRM/MOVE Research Institute , Amsterdam, Netherlands
- 3 Department of Orthopedic Surgery, VU Medical Center , Amsterdam, Netherlands
| | - Paul P M van Zuijlen
- 1 Department of Plastic, Reconstructive & Hand Surgery, VU Medical Center , Amsterdam, Netherlands
- 2 CTRM/MOVE Research Institute , Amsterdam, Netherlands
- 5 Red Cross Hospital Beverwijk , Beverwijk, Netherlands
| |
Collapse
|
38
|
Cohen BP, Hooper RC, Puetzer JL, Nordberg R, Asanbe O, Hernandez KA, Spector JA, Bonassar LJ. Long-Term Morphological and Microarchitectural Stability of Tissue-Engineered, Patient-Specific Auricles In Vivo. Tissue Eng Part A 2016; 22:461-8. [PMID: 26847742 DOI: 10.1089/ten.tea.2015.0323] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Current techniques for autologous auricular reconstruction produce substandard ear morphologies with high levels of donor-site morbidity, whereas alloplastic implants demonstrate poor biocompatibility. Tissue engineering, in combination with noninvasive digital photogrammetry and computer-assisted design/computer-aided manufacturing technology, offers an alternative method of auricular reconstruction. Using this method, patient-specific ears composed of collagen scaffolds and auricular chondrocytes have generated auricular cartilage with great fidelity following 3 months of subcutaneous implantation, however, this short time frame may not portend long-term tissue stability. We hypothesized that constructs developed using this technique would undergo continued auricular cartilage maturation without degradation during long-term (6 month) implantation. Full-sized, juvenile human ear constructs were injection molded from high-density collagen hydrogels encapsulating juvenile bovine auricular chondrocytes and implanted subcutaneously on the backs of nude rats for 6 months. Upon explantation, constructs retained overall patient morphology and displayed no evidence of tissue necrosis. Limited contraction occurred in vivo, however, no significant change in size was observed beyond 1 month. Constructs at 6 months showed distinct auricular cartilage microstructure, featuring a self-assembled perichondrial layer, a proteoglycan-rich bulk, and rounded cellular lacunae. Verhoeff's staining also revealed a developing elastin network comparable to native tissue. Biochemical measurements for DNA, glycosaminoglycan, and hydroxyproline content and mechanical properties of aggregate modulus and hydraulic permeability showed engineered tissue to be similar to native cartilage at 6 months. Patient-specific auricular constructs demonstrated long-term stability and increased cartilage tissue development during extended implantation, and offer a potential tissue-engineered solution for the future of auricular reconstructions.
Collapse
Affiliation(s)
- Benjamin Peter Cohen
- 1 Meinig School of Biomedical Engineering, Cornell University , Ithaca, New York
| | - Rachel C Hooper
- 2 Laboratory for Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Weill Cornell Medical College , New York, New York
| | - Jennifer L Puetzer
- 1 Meinig School of Biomedical Engineering, Cornell University , Ithaca, New York
| | - Rachel Nordberg
- 1 Meinig School of Biomedical Engineering, Cornell University , Ithaca, New York
| | - Ope Asanbe
- 2 Laboratory for Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Weill Cornell Medical College , New York, New York
| | - Karina A Hernandez
- 2 Laboratory for Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Weill Cornell Medical College , New York, New York
| | - Jason A Spector
- 1 Meinig School of Biomedical Engineering, Cornell University , Ithaca, New York.,2 Laboratory for Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Weill Cornell Medical College , New York, New York
| | - Lawrence J Bonassar
- 1 Meinig School of Biomedical Engineering, Cornell University , Ithaca, New York.,3 Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York
| |
Collapse
|
39
|
Martínez Ávila H, Schwarz S, Rotter N, Gatenholm P. 3D bioprinting of human chondrocyte-laden nanocellulose hydrogels for patient-specific auricular cartilage regeneration. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.bprint.2016.08.003] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
40
|
Park JY, Choi YJ, Shim JH, Park JH, Cho DW. Development of a 3D cell printed structure as an alternative to autologs cartilage for auricular reconstruction. J Biomed Mater Res B Appl Biomater 2016; 105:1016-1028. [PMID: 26922876 DOI: 10.1002/jbm.b.33639] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 01/04/2016] [Accepted: 02/03/2016] [Indexed: 12/12/2022]
Abstract
Surgical technique using autologs cartilage is considered as the best treatment for cartilage tissue reconstruction, although the burdens of donor site morbidity and surgical complications still remain. The purpose of this study is to apply three-dimensional (3D) cell printing to fabricate a tissue-engineered graft, and evaluate its effects on cartilage reconstruction. A multihead tissue/organ building system is used to print cell-printed scaffold (CPS), then assessed the effect of the CPS on cartilage regeneration in a rabbit ear. The cell viability and functionality of chondrocytes were significantly higher in CPS than in cell-seeded scaffold (CSS) and cell-seeded hybrid scaffold (CSHS) in vitro. CPS was then implanted into a rabbit ear that had an 8 mm-diameter cartilage defect; at 3 months after implantation the CPS had fostered complete cartilage regeneration whereas CSS and autologs cartilage (AC) fostered only incomplete healing. This result demonstrates that cell printing technology can provide an appropriate environment in which encapsulated chondrocytes can survive and differentiate into cartilage tissue in vivo. Moreover, the effects of CPS on cartilage regeneration were even better than those of AC. Therefore, we confirmed the feasibility of CPS as an alternative to AC for auricular reconstruction. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 1016-1028, 2017.
Collapse
Affiliation(s)
- Ju Young Park
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Korea
| | - Yeong-Jin Choi
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Korea
| | - Jin-Hyung Shim
- Department of Mechanical Engineering, Korea Polytechnic University, Siheung, Korea
| | - Jeong Hun Park
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Korea
| |
Collapse
|
41
|
Tissue composition regulates distinct viscoelastic responses in auricular and articular cartilage. J Biomech 2016; 49:344-52. [DOI: 10.1016/j.jbiomech.2015.12.032] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 11/24/2015] [Accepted: 12/16/2015] [Indexed: 01/24/2023]
|
42
|
Jessop ZM, Javed M, Otto IA, Combellack EJ, Morgan S, Breugem CC, Archer CW, Khan IM, Lineaweaver WC, Kon M, Malda J, Whitaker IS. Combining regenerative medicine strategies to provide durable reconstructive options: auricular cartilage tissue engineering. Stem Cell Res Ther 2016; 7:19. [PMID: 26822227 PMCID: PMC4730656 DOI: 10.1186/s13287-015-0273-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Recent advances in regenerative medicine place us in a unique position to improve the quality of engineered tissue. We use auricular cartilage as an exemplar to illustrate how the use of tissue-specific adult stem cells, assembly through additive manufacturing and improved understanding of postnatal tissue maturation will allow us to more accurately replicate native tissue anisotropy. This review highlights the limitations of autologous auricular reconstruction, including donor site morbidity, technical considerations and long-term complications. Current tissue-engineered auricular constructs implanted into immune-competent animal models have been observed to undergo inflammation, fibrosis, foreign body reaction, calcification and degradation. Combining biomimetic regenerative medicine strategies will allow us to improve tissue-engineered auricular cartilage with respect to biochemical composition and functionality, as well as microstructural organization and overall shape. Creating functional and durable tissue has the potential to shift the paradigm in reconstructive surgery by obviating the need for donor sites.
Collapse
Affiliation(s)
- Zita M Jessop
- Reconstructive Surgery & Regenerative Medicine Research Group, Swansea University Medical School, Room 509, ILS2, Swansea, SA2 8SS, UK.
- The Welsh Centre for Burns and Plastic Surgery, Morriston Hospital, Swansea, SA6 6NL, UK.
| | - Muhammad Javed
- Reconstructive Surgery & Regenerative Medicine Research Group, Swansea University Medical School, Room 509, ILS2, Swansea, SA2 8SS, UK.
- The Welsh Centre for Burns and Plastic Surgery, Morriston Hospital, Swansea, SA6 6NL, UK.
| | - Iris A Otto
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, 3584 CX, The Netherlands.
- Department of Plastic and Reconstructive Surgery, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Emman J Combellack
- Reconstructive Surgery & Regenerative Medicine Research Group, Swansea University Medical School, Room 509, ILS2, Swansea, SA2 8SS, UK.
- The Welsh Centre for Burns and Plastic Surgery, Morriston Hospital, Swansea, SA6 6NL, UK.
| | - Siân Morgan
- Reconstructive Surgery & Regenerative Medicine Research Group, Swansea University Medical School, Room 509, ILS2, Swansea, SA2 8SS, UK.
- The Welsh Centre for Burns and Plastic Surgery, Morriston Hospital, Swansea, SA6 6NL, UK.
| | - Corstiaan C Breugem
- Department of Plastic and Reconstructive Surgery, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Charles W Archer
- Reconstructive Surgery & Regenerative Medicine Research Group, Swansea University Medical School, Room 509, ILS2, Swansea, SA2 8SS, UK.
| | - Ilyas M Khan
- KhanLab, Swansea University, ILS2, Swansea, SA2 8SS, UK.
| | - William C Lineaweaver
- Division of Plastic Surgery, University of Mississippi Medical Center, Jackson, Mississippi, 39216, USA.
| | - Moshe Kon
- Department of Plastic and Reconstructive Surgery, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, 3584 CX, The Netherlands.
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Domplein 29, 3512 JE, Utrecht, The Netherlands.
| | - Iain S Whitaker
- Reconstructive Surgery & Regenerative Medicine Research Group, Swansea University Medical School, Room 509, ILS2, Swansea, SA2 8SS, UK.
- The Welsh Centre for Burns and Plastic Surgery, Morriston Hospital, Swansea, SA6 6NL, UK.
| |
Collapse
|
43
|
Pomerantseva I, Bichara DA, Tseng A, Cronce MJ, Cervantes TM, Kimura AM, Neville CM, Roscioli N, Vacanti JP, Randolph MA, Sundback CA. Ear-Shaped Stable Auricular Cartilage Engineered from Extensively Expanded Chondrocytes in an Immunocompetent Experimental Animal Model. Tissue Eng Part A 2015; 22:197-207. [PMID: 26529401 DOI: 10.1089/ten.tea.2015.0173] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Advancement of engineered ear in clinical practice is limited by several challenges. The complex, largely unsupported, three-dimensional auricular neocartilage structure is difficult to maintain. Neocartilage formation is challenging in an immunocompetent host due to active inflammatory and immunological responses. The large number of autologous chondrogenic cells required for engineering an adult human-sized ear presents an additional challenge because primary chondrocytes rapidly dedifferentiate during in vitro culture. The objective of this study was to engineer a stable, human ear-shaped cartilage in an immunocompetent animal model using expanded chondrocytes. The impact of basic fibroblast growth factor (bFGF) supplementation on achieving clinically relevant expansion of primary sheep chondrocytes by in vitro culture was determined. Chondrocytes expanded in standard medium were either combined with cryopreserved, primary passage 0 chondrocytes at the time of scaffold seeding or used alone as control. Disk and human ear-shaped scaffolds were made from porous collagen; ear scaffolds had an embedded, supporting titanium wire framework. Autologous chondrocyte-seeded scaffolds were implanted subcutaneously in sheep after 2 weeks of in vitro incubation. The quality of the resulting neocartilage and its stability and retention of the original ear size and shape were evaluated at 6, 12, and 20 weeks postimplantation. Neocartilage produced from chondrocytes that were expanded in the presence of bFGF was superior, and its quality improved with increased implantation time. In addition to characteristic morphological cartilage features, its glycosaminoglycan content was high and marked elastin fiber formation was present. The overall shape of engineered ears was preserved at 20 weeks postimplantation, and the dimensional changes did not exceed 10%. The wire frame within the engineered ear was able to withstand mechanical forces during wound healing and neocartilage maturation and prevented shrinkage and distortion. This is the first demonstration of a stable, ear-shaped elastic cartilage engineered from auricular chondrocytes that underwent clinical-scale expansion in an immunocompetent animal over an extended period of time.
Collapse
Affiliation(s)
- Irina Pomerantseva
- 1 Department of Surgery, Massachusetts General Hospital , Boston, Massachusetts.,2 Harvard Medical School , Boston, Massachusetts
| | - David A Bichara
- 2 Harvard Medical School , Boston, Massachusetts.,3 Plastic Surgery Research Laboratory, Massachusetts General Hospital , Boston, Massachusetts
| | - Alan Tseng
- 1 Department of Surgery, Massachusetts General Hospital , Boston, Massachusetts
| | - Michael J Cronce
- 1 Department of Surgery, Massachusetts General Hospital , Boston, Massachusetts
| | - Thomas M Cervantes
- 1 Department of Surgery, Massachusetts General Hospital , Boston, Massachusetts
| | - Anya M Kimura
- 1 Department of Surgery, Massachusetts General Hospital , Boston, Massachusetts
| | - Craig M Neville
- 1 Department of Surgery, Massachusetts General Hospital , Boston, Massachusetts.,2 Harvard Medical School , Boston, Massachusetts
| | | | - Joseph P Vacanti
- 1 Department of Surgery, Massachusetts General Hospital , Boston, Massachusetts.,2 Harvard Medical School , Boston, Massachusetts
| | - Mark A Randolph
- 2 Harvard Medical School , Boston, Massachusetts.,3 Plastic Surgery Research Laboratory, Massachusetts General Hospital , Boston, Massachusetts
| | - Cathryn A Sundback
- 1 Department of Surgery, Massachusetts General Hospital , Boston, Massachusetts.,2 Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
44
|
Otto IA, Melchels FPW, Zhao X, Randolph MA, Kon M, Breugem CC, Malda J. Auricular reconstruction using biofabrication-based tissue engineering strategies. Biofabrication 2015. [PMID: 26200941 DOI: 10.1088/1758-5090/7/3/032001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Auricular malformations, which impose a significant social and psychological burden, are currently treated using ear prostheses, synthetic implants or autologous implants derived from rib cartilage. Advances in the field of regenerative medicine and biofabrication provide the possibility to engineer functional cartilage with intricate architectures and complex shapes using patient-derived or donor cells. However, the development of a successful auricular cartilage implant still faces a number of challenges. These challenges include the generation of a functional biochemical matrix, the fabrication of a customized anatomical shape, and maintenance of that shape. Biofabrication technologies may have the potential to overcome these challenges due to their ability to reproducibly deposit multiple materials in complex geometries in a highly controllable manner. This topical review summarizes this potential of biofabrication technologies for the generation of implants for auricular reconstruction. In particular, it aims to discuss how biofabrication technologies, although still in pre-clinical phase, could overcome the challenges of generating and maintaining the desired auricular shapes. Finally, remaining bottlenecks and future directions are discussed.
Collapse
Affiliation(s)
- I A Otto
- Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA Utrecht, The Netherlands. Department of Plastic, Reconstructive and Hand Surgery, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
45
|
Prefabricated, ear-shaped cartilage tissue engineering by scaffold-free porcine chondrocyte membrane. Plast Reconstr Surg 2015; 135:313e-321e. [PMID: 25626816 DOI: 10.1097/prs.0000000000001105] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Ear defects caused by traumatic injury, tumor ablation, and congenital deficiency are still challenging problems for the plastic and reconstructive surgeon. The authors developed a scaffold-free, ear-shaped cartilage by tailoring a multilayered chondrocyte membrane on an ear-shaped titanium alloy model and investigated the possibility of long-term ear-shaped maintenance in nude mice. METHODS High-density chondrocytes (approximately 30 × 10 cells) were seeded to produce chondrocyte membranes after cultivation under chondrogenic medium for 2 weeks. Then, three-layer chondrocyte membranes were tailored on the ear-shaped titanium mold and fixed by 6-0 nylon. The constructs were implanted onto the dorsal pockets of nude mice for 8 and 24 weeks. The chondrocyte membrane, 8- and 24-week implants were analyzed by safranin O, toluidine blue, elastica van Gieson, and collagen type II immunohistochemistry stains and quantitative measurement of glycosaminoglycan and total collagen compared with native cartilage. Mechanical strength was compared by compressive Young's modulus. RESULTS Results showed that the chondrocyte membrane was durable and nonfragile and easily manipulated by forceps. The composite of chondrocyte membrane and titanium alloy maintained the stable ear-like shape after 8 and 24 weeks of subcutaneous implantation. Histologic examination verified that the newly formed tissue at the implant construct was elastic cartilage at both 8 and 24 weeks by safranin O, toluidine blue, elastica van Gieson, and collagen type II immunohistochemistry stains. The Young's modulus was only half of and similar to normal cartilage in 8- and 24-week implants, respectively. CONCLUSION This study demonstrated that an ear-shaped elastic cartilage could be regenerated by a scaffold-free chondrocyte membrane shaped by a prefabricated, three-dimensional, ear-shaped titanium mold.
Collapse
|
46
|
Conditions for seeding and promoting neo-auricular cartilage formation in a fibrous collagen scaffold. J Craniomaxillofac Surg 2015; 43:382-9. [DOI: 10.1016/j.jcms.2014.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 12/07/2014] [Accepted: 12/12/2014] [Indexed: 01/25/2023] Open
|
47
|
Martínez Ávila H, Feldmann EM, Pleumeekers MM, Nimeskern L, Kuo W, de Jong WC, Schwarz S, Müller R, Hendriks J, Rotter N, van Osch GJ, Stok KS, Gatenholm P. Novel bilayer bacterial nanocellulose scaffold supports neocartilage formation in vitro and in vivo. Biomaterials 2015; 44:122-33. [DOI: 10.1016/j.biomaterials.2014.12.025] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/01/2014] [Accepted: 12/20/2014] [Indexed: 10/24/2022]
|
48
|
Tseng A, Pomerantseva I, Cronce MJ, Kimura AM, Neville CM, Randolph MA, Vacanti JP, Sundback CA. Extensively Expanded Auricular Chondrocytes Form Neocartilage In Vivo. Cartilage 2014; 5:241-51. [PMID: 26069703 PMCID: PMC4335768 DOI: 10.1177/1947603514546740] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVE Our goal was to engineer cartilage in vivo using auricular chondrocytes that underwent clinically relevant expansion and using methodologies that could be easily translated into health care practice. DESIGN Sheep and human chondrocytes were isolated from auricular cartilage biopsies and expanded in vitro. To reverse dedifferentiation, expanded cells were either mixed with cryopreserved P0 chondrocytes at the time of seeding onto porous collagen scaffolds or proliferated with basic fibroblast growth factor (bFGF). After 2-week in vitro incubation, seeded scaffolds were implanted subcutaneously in nude mice for 6 weeks. The neocartilage quality was evaluated histologically; DNA and glycosaminoglycans were quantified. Cell proliferation rates and collagen gene expression profiles were assessed. RESULTS Clinically sufficient over 500-fold chondrocyte expansion was achieved at passage 3 (P3); cell dedifferentiation was confirmed by the simultaneous COL1A1/3A1 gene upregulation and COL2A1 downregulation. The chondrogenic phenotype of sheep but not human P3 cells was rescued by addition of cryopreserved P0 chondrocytes. With bFGF supplementation, chondrocytes achieved clinically sufficient expansion at P2; COL2A1 expression was not rescued but COL1A1/3A1genes were downregulated. Although bFGF failed to rescue COL2A1 expression during chondrocyte expansion in vitro, elastic neocartilage with obvious collagen II expression was observed on porous collagen scaffolds after implantation in mice for 6 weeks. CONCLUSIONS Both animal and human auricular chondrocytes expanded with low-concentration bFGF supplementation formed high-quality elastic neocartilage on porous collagen scaffolds in vivo.
Collapse
Affiliation(s)
- Alan Tseng
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA,Center for Regenerative Medicine, Boston, MA, USA
| | - Irina Pomerantseva
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA,Center for Regenerative Medicine, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Michael J. Cronce
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA,Center for Regenerative Medicine, Boston, MA, USA
| | - Anya M. Kimura
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA,Center for Regenerative Medicine, Boston, MA, USA
| | - Craig M. Neville
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA,Center for Regenerative Medicine, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Mark A. Randolph
- Harvard Medical School, Boston, MA, USA,Plastic Surgery Research Laboratory, Massachusetts General Hospital, Boston, MA, USA
| | - Joseph P. Vacanti
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA,Center for Regenerative Medicine, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Cathryn A. Sundback
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA,Center for Regenerative Medicine, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| |
Collapse
|
49
|
|
50
|
Magnetic resonance imaging of the ear for patient-specific reconstructive surgery. PLoS One 2014; 9:e104975. [PMID: 25144306 PMCID: PMC4140740 DOI: 10.1371/journal.pone.0104975] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 07/06/2014] [Indexed: 11/22/2022] Open
Abstract
Introduction Like a fingerprint, ear shape is a unique personal feature that should be reconstructed with a high fidelity during reconstructive surgery. Ear cartilage tissue engineering (TE) advantageously offers the possibility to use novel 3D manufacturing techniques to reconstruct the ear, thus allowing for a detailed auricular shape. However it also requires detailed patient-specific images of the 3D cartilage structures of the patient’s intact contralateral ear (if available). Therefore the aim of this study was to develop and evaluate an imaging strategy for acquiring patient-specific ear cartilage shape, with sufficient precision and accuracy for use in a clinical setting. Methods and Materials Magnetic resonance imaging (MRI) was performed on 14 volunteer and six cadaveric auricles and manually segmented. Reproducibility of cartilage volume (Cg.V), surface (Cg.S) and thickness (Cg.Th) was assessed, to determine whether raters could repeatedly define the same volume of interest. Additionally, six cadaveric auricles were harvested, scanned and segmented using the same procedure, then dissected and scanned using high resolution micro-CT. Correlation between MR and micro-CT measurements was assessed to determine accuracy. Results Good inter- and intra-rater reproducibility was observed (precision errors <4% for Cg.S and <9% for Cg.V and Cg.Th). Intraclass correlations were good for Cg.V and Cg.S (>0.82), but low for Cg.Th (<0.23) due to similar average Cg.Th between patients. However Pearson’s coefficients showed that the ability to detect local cartilage shape variations is unaffected. Good correlation between clinical MRI and micro-CT (r>0.95) demonstrated high accuracy. Discussion and Conclusion This study demonstrated that precision and accuracy of the proposed method was high enough to detect patient-specific variation in ear cartilage geometry. The present study provides a clinical strategy to access the necessary information required for the production of 3D ear scaffolds for TE purposes, including detailed patient-specific shape. Furthermore, the protocol is applicable in daily clinical practice with existing infrastructure.
Collapse
|