1
|
Mazy D, Wang J, Dodin P, Lu D, Moldovan F, Nault ML. Emerging biologic augmentation strategies for meniscal repair: a systematic review. BMC Musculoskelet Disord 2024; 25:541. [PMID: 39003467 PMCID: PMC11245777 DOI: 10.1186/s12891-024-07644-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 07/01/2024] [Indexed: 07/15/2024] Open
Abstract
BACKGROUND Meniscal repair should be the gold standard. However, the meniscus is poorly vascularized and even an excellent meniscus repair may not heal. Therefore, numerous studies and systematic reviews have been carried out on platelet-rich plasma (PRP), mesenchymal stem cells (MSCs) and fibrin clots for meniscal augmentation, but the results remain controversial. This systematic review aimed to identify other emerging strategies for meniscal repair augmentation and to assess whether there are different avenues to explore in this field. METHODS A systematic literature review was conducted in August 2022. PubMed, Ovid MEDLINE(R) all, Ovid All EBM Reviews, Ovid Embase and ISI Web of Science databases were searched. In Vivo animal and human studies concerning the biological augmentation of meniscal lesions by factors other than PRP, MSCs or fibrin clots were included. Cartilage-only studies, previous systematic reviews and expert opinions were excluded. All data were analyzed by two independent reviewers. RESULTS Of 8965 studies only nineteen studies covering 12 different factors met the inclusion criteria. Eight studies investigated the use of growth factors for meniscal biologic augmentation, such as vascular endothelial growth factor or bone morphogenic protein 7. Five studies reported on cell therapy and six studies focused on other factors such as hyaluronic acid, simvastatin or atelocollagen. Most studies (n = 18) were performed on animal models with gross observation and histological evaluation as outcomes. Polymerase chain reaction and immunohistochemistry were also common. Biomechanical testing was the object of only two studies. CONCLUSIONS Although several augmentation strategies have been attempted, none has yielded conclusive results, testifying to a lack of understanding with regard to meniscal healing. More research is needed to better understand the pathways that regulate meniscus repair and how to act positively on them. LEVEL OF EVIDENCE Systematic review of case-control and animal laboratory studies.
Collapse
Affiliation(s)
- David Mazy
- CHU Sainte-Justine, Montréal, 7905-3175, Côte Ste-Catherine, QC, H3T 1C5, Canada
| | - Jessica Wang
- Faculty of Medicine, Université de Montréal, 2900 Boul. Edouard-Montpetit, Montreal, QC, H3T 1J4, Canada
| | - Philippe Dodin
- CHU Sainte-Justine, Montréal, 7905-3175, Côte Ste-Catherine, QC, H3T 1C5, Canada
| | - Daisy Lu
- Faculty of Medicine, Université de Montréal, 2900 Boul. Edouard-Montpetit, Montreal, QC, H3T 1J4, Canada
| | - Florina Moldovan
- CHU Sainte-Justine, Montréal, 7905-3175, Côte Ste-Catherine, QC, H3T 1C5, Canada
| | - Marie-Lyne Nault
- CHU Sainte-Justine, Montréal, 7905-3175, Côte Ste-Catherine, QC, H3T 1C5, Canada.
- Faculty of Medicine, Université de Montréal, 2900 Boul. Edouard-Montpetit, Montreal, QC, H3T 1J4, Canada.
- CHU Sainte-Justine Azrieli Research Center, Montréal, 7905-3175 Côte Ste-Catherine, H3T 1J4, QC, Canada.
- Department of Orthopedic Surgery, CIUSSS Hôpital du Sacré-Cœur de Montréal (HSCM), 5400, Boul. Gouin Ouest, Montreal, QC, H4J 1C5, Canada.
| |
Collapse
|
2
|
Bandyopadhyay A, Ghibhela B, Mandal BB. Current advances in engineering meniscal tissues: insights into 3D printing, injectable hydrogels and physical stimulation based strategies. Biofabrication 2024; 16:022006. [PMID: 38277686 DOI: 10.1088/1758-5090/ad22f0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 01/26/2024] [Indexed: 01/28/2024]
Abstract
The knee meniscus is the cushioning fibro-cartilage tissue present in between the femoral condyles and tibial plateau of the knee joint. It is largely avascular in nature and suffers from a wide range of tears and injuries caused by accidents, trauma, active lifestyle of the populace and old age of individuals. Healing of the meniscus is especially difficult due to its avascularity and hence requires invasive arthroscopic approaches such as surgical resection, suturing or implantation. Though various tissue engineering approaches are proposed for the treatment of meniscus tears, three-dimensional (3D) printing/bioprinting, injectable hydrogels and physical stimulation involving modalities are gaining forefront in the past decade. A plethora of new printing approaches such as direct light photopolymerization and volumetric printing, injectable biomaterials loaded with growth factors and physical stimulation such as low-intensity ultrasound approaches are being added to the treatment portfolio along with the contemporary tear mitigation measures. This review discusses on the necessary design considerations, approaches for 3D modeling and design practices for meniscal tear treatments within the scope of tissue engineering and regeneration. Also, the suitable materials, cell sources, growth factors, fixation and lubrication strategies, mechanical stimulation approaches, 3D printing strategies and injectable hydrogels for meniscal tear management have been elaborated. We have also summarized potential technologies and the potential framework that could be the herald of the future of meniscus tissue engineering and repair approaches.
Collapse
Affiliation(s)
- Ashutosh Bandyopadhyay
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Baishali Ghibhela
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Biman B Mandal
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| |
Collapse
|
3
|
Li X, Li D, Li J, Wang G, Yan L, Liu H, Jiu J, Li JJ, Wang B. Preclinical Studies and Clinical Trials on Cell-Based Treatments for Meniscus Regeneration. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:634-670. [PMID: 37212339 DOI: 10.1089/ten.teb.2023.0050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
This study aims at performing a thorough review of cell-based treatment strategies for meniscus regeneration in preclinical and clinical studies. The PubMed, Embase, and Web of Science databases were searched for relevant studies (both preclinical and clinical) published from the time of database construction to December 2022. Data related to cell-based therapies for in situ regeneration of the meniscus were extracted independently by two researchers. Assessment of risk of bias was performed according to the Cochrane Handbook for Systematic Reviews of Interventions. Statistical analyses based on the classification of different treatment strategies were performed. A total of 5730 articles were retrieved, of which 72 preclinical studies and 6 clinical studies were included in this review. Mesenchymal stem cells (MSCs), especially bone marrow MSCs (BMSCs), were the most commonly used cell type. Among preclinical studies, rabbit was the most commonly used animal species, partial meniscectomy was the most commonly adopted injury pattern, and 12 weeks was the most frequently chosen final time point for assessing repair outcomes. A range of natural and synthetic materials were used to aid cell delivery as scaffolds, hydrogels, or other morphologies. In clinical trials, there was large variation in the dose of cells, ranging from 16 × 106 to 150 × 106 cells with an average of 41.52 × 106 cells. The selection of treatment strategy for meniscus repair should be based on the nature of the injury. Cell-based therapies incorporating various "combination" strategies such as co-culture, composite materials, and extra stimulation may offer greater promise than single strategies for effective meniscal tissue regeneration, restoring natural meniscal anisotropy, and eventually achieving clinical translation. Impact Statement This review provides an up-to-date and comprehensive overview of preclinical and clinical studies that tested cell-based treatments for meniscus regeneration. It presents novel perspectives on studies published in the past 30 years, giving consideration to the cell sources and dose selection, delivery methods, extra stimulation, animal models and injury patterns, timing of outcome assessment, and histological and biomechanical outcomes, as well as a summary of findings for individual studies. These unique insights will help to shape future research on the repair of meniscus lesions and inform the clinical translation of new cell-based tissue engineering strategies.
Collapse
Affiliation(s)
- Xiaoke Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Dijun Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Jiarong Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, Australia
| | - Guishan Wang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Lei Yan
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Haifeng Liu
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Jingwei Jiu
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, Australia
| | - Bin Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
4
|
Veronesi F, Di Matteo B, Vitale N, Filardo G, Visani A, Kon E, Fini M. Biosynthetic scaffolds for partial meniscal loss: A systematic review from animal models to clinical practice. Bioact Mater 2021; 6:3782-3800. [PMID: 33898878 PMCID: PMC8044909 DOI: 10.1016/j.bioactmat.2021.03.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 03/21/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
Acute or degenerative meniscus tears are the most common knee lesions. Meniscectomy provides symptomatic relief and functional recovery only in the short- to mid-term follow-up but significantly increases the risk of osteoarthritis. For this reason, preserving the meniscus is key, although it remains a challenge. Allograft transplants present many disadvantages, so during the last 20 years preclinical and clinical research focused on developing and investigating meniscal scaffolds. The aim of this systematic review was to collect and evaluate all the available evidence on biosynthetic scaffolds for meniscus regeneration both in vivo and in clinical studies. Three databases were searched: 46 in vivo preclinical studies and 30 clinical ones were found. Sixteen natural, 15 synthetic, and 15 hybrid scaffolds were studied in vivo. Among them, only 2 were translated into clinic: the Collagen Meniscus Implant, used in 11 studies, and the polyurethane-based scaffold Actifit®, applied in 19 studies. Although positive outcomes were described in the short- to mid-term, the number of concurrent procedures and the lack of randomized trials are the major limitations of the available clinical literature. Few in vivo studies also combined the use of cells or growth factors, but these augmentation strategies have not been applied in the clinical practice yet. Current solutions offer a significant but incomplete clinical improvement, and the regeneration potential is still unsatisfactory. Building upon the overall positive results of these "old" technologies to address partial meniscal loss, further innovation is urgently needed in this field to provide patients better joint sparing treatment options.
Collapse
Affiliation(s)
- F. Veronesi
- Complex Structure of Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - B. Di Matteo
- Humanitas University, Department of Biomedical Sciences, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Milan, Italy
- First Moscow State Medical University - Sechenov University, Bol'shaya Pirogovskaya Ulitsa, 19c1, 119146, Moscow, Russia
| | - N.D. Vitale
- Humanitas University, Department of Biomedical Sciences, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - G. Filardo
- Applied and Translational Research Center, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Orthopaedic and Traumatology Unit, Ospedale Regionale di Lugano, EOC, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - A. Visani
- Complex Structure of Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - E. Kon
- Humanitas University, Department of Biomedical Sciences, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - M. Fini
- Complex Structure of Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| |
Collapse
|
5
|
Van Genechten W, Verdonk P, Krych AJ, Saris DB. Biologic Adjuvants in Meniscus Repair: A Review of Current Translational and Clinical Evidence. OPER TECHN SPORT MED 2020. [DOI: 10.1016/j.otsm.2020.150758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
6
|
Janarthanan G, Pillai MM, Kulasekaran SS, Rajendran S, Bhattacharyya A. Engineered knee meniscus construct: understanding the structure and impact of functionalization in 3D environment. Polym Bull (Berl) 2020. [DOI: 10.1007/s00289-019-02874-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
7
|
Li XG, Park IS, Choi BH, Kim UJ, Min BH. In Vivo Bioreactor Using Cellulose Membrane Benefit Engineering Cartilage by Improving the Chondrogenesis and Modulating the Immune Response. Tissue Eng Regen Med 2020; 17:165-181. [PMID: 32193874 PMCID: PMC7105552 DOI: 10.1007/s13770-019-00236-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND To regenerate tissue-engineered cartilage as a source of material for the restoration of cartilage defects, we used a human fetal cartilage progenitor cell pellet to improve chondrogenesis and modulation of the immune response in an in vivo bioreactor (IVB) system. METHODS IVB was buried subcutaneously in the host and then implanted into a cartilage defect. The IVB was composed of a silicone tube and a cellulose nano pore-sized membrane. First, fetal cartilage progenitor cell pellets were cultured in vitro for 3 days, then cultured in vitro, subcutaneously, and in an IVB for 3 weeks. First, the components and liquidity of IVB fluid were evaluated, then the chondrogenesis and immunogenicity of the pellets were evaluated using gross observation, cell viability assays, histology, biochemical analysis, RT-PCR, and Western blots. Finally, cartilage repair and synovial inflammation were evaluated histologically. RESULTS The fluid color and transparency of the IVB were similar to synovial fluid (SF) and the components were closer to SF than serum. The IVB system not only promoted the synthesis of cartilage matrix and maintained the cartilage phenotype, it also delayed calcification compared to the subcutaneously implanted pellets. CONCLUSION The IVB adopted to study cell differentiation was effective in preventing host immune rejection.
Collapse
Affiliation(s)
- Xue Guang Li
- Department of Orthopaedic Surgery, Ajou University School of Medicine, San 5, Wonchon-dong, Youngtong-gu, Suwon, 16499, Republic of Korea
- Cell Therapy Center, Ajou University School of Medicine, San 5, Wonchon-dong, Youngtong-gu, Suwon, 16499, Republic of Korea
| | - In-Su Park
- Cell Therapy Center, Ajou University School of Medicine, San 5, Wonchon-dong, Youngtong-gu, Suwon, 16499, Republic of Korea
| | - Byung Hyune Choi
- Department of Biomedical Sciences, Inha University College of Medicine, 100, Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea
| | - Ung-Jin Kim
- Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do, 17104, Republic of Korea
- Department of Plant and Environmental New Resources, College of Life Sciences, Kyung Hee University, 1 Seocheon-dong, Giheung-gu, Yongin-si, Gyeonggi-do, 17104, Republic of Korea
| | - Byoung-Hyun Min
- Department of Orthopaedic Surgery, Ajou University School of Medicine, San 5, Wonchon-dong, Youngtong-gu, Suwon, 16499, Republic of Korea.
- Cell Therapy Center, Ajou University School of Medicine, San 5, Wonchon-dong, Youngtong-gu, Suwon, 16499, Republic of Korea.
- Department of Molecular Science and Technology, Ajou University School of Medicine, San 5, Wonchon-dong, Youngtong-gu, Suwon, 16499, Republic of Korea.
| |
Collapse
|
8
|
Chen M, Feng Z, Guo W, Yang D, Gao S, Li Y, Shen S, Yuan Z, Huang B, Zhang Y, Wang M, Li X, Hao L, Peng J, Liu S, Zhou Y, Guo Q. PCL-MECM-Based Hydrogel Hybrid Scaffolds and Meniscal Fibrochondrocytes Promote Whole Meniscus Regeneration in a Rabbit Meniscectomy Model. ACS APPLIED MATERIALS & INTERFACES 2019; 11:41626-41639. [PMID: 31596568 DOI: 10.1021/acsami.9b13611] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Regeneration of an injured meniscus continues to be a scientific challenge due to its poor self-healing potential. Tissue engineering provides an avenue for regenerating a severely damaged meniscus. In this study, we first investigated the superiority of five concentrations (0%, 0.5%, 1%, 2%, and 4%) of meniscus extracellular matrix (MECM)-based hydrogel in promoting cell proliferation and the matrix-forming phenotype of meniscal fibrochondrocytes (MFCs). We found that the 2% group strongly enhanced chondrogenic marker mRNA expression and cell proliferation compared to the other groups. Moreover, the 2% group showed the highest glycosaminoglycan (GAG) and collagen production by day 14. We then constructed a hybrid scaffold by 3D printing a wedge-shaped poly(ε-caprolactone) (PCL) scaffold as a backbone, followed by injection with the optimized MECM-based hydrogel (2%), which served as a cell delivery system. The hybrid scaffold (PCL-hydrogel) clearly yielded favorable biomechanical properties close to those of the native meniscus. Finally, PCL scaffold, PCL-hydrogel, and MFCs-loaded hybrid scaffold (PCL-hydrogel-MFCs) were implanted into the knee joints of New Zealand rabbits that underwent total medial meniscectomy. Six months postimplantation we found that the PCL-hydrogel-MFCs group exhibited markedly better gross appearance and cartilage protection than the PCL scaffold and PCL-hydrogel groups. Moreover, the regenerated menisci in the PCL-hydrogel-MFCs group had similar histological structures, biochemical contents, and biomechanical properties as the native menisci in the sham operation group. In conclusion, PCL-MECM-based hydrogel hybrid scaffold seeded with MFCs can successfully promote whole meniscus regeneration, and cell-loaded PCL-MECM-based hydrogel hybrid scaffold may be a promising strategy for meniscus regeneration in the future.
Collapse
Affiliation(s)
- Mingxue Chen
- Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA , Institute of Orthopedics , No. 28 Fuxing Road, Haidian District , Beijing 100853 , People's Republic of China
- Department of Orthopedic Surgery, Beijing Jishuitan Hospital , Peking University Fourth School of Clinical Medicine , No. 31 Xinjiekou East Street, Xicheng District , Beijing 100035 , People's Republic of China
| | - Zhaoxuan Feng
- School of Material Science and Engineering , University of Science and Technology Beijing , No. 30 Xueyuan Road, Haidian District , Beijing 100083 , People's Republic of China
| | - Weimin Guo
- Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA , Institute of Orthopedics , No. 28 Fuxing Road, Haidian District , Beijing 100853 , People's Republic of China
- Department of Orthopaedic Surgery, First Affiliated Hospital , Sun Yat-sen University , No. 58 Zhongshan Second Road, Yuexiu District , Guangzhou , Guangdong 510080 , People's Republic of China
| | - Dejin Yang
- Department of Orthopedic Surgery, Beijing Jishuitan Hospital , Peking University Fourth School of Clinical Medicine , No. 31 Xinjiekou East Street, Xicheng District , Beijing 100035 , People's Republic of China
| | - Shuang Gao
- Academy for Advanced Interdisciplinary Studies , Peking University , No. 5 Yiheyuan Road, Haidian District , Beijing 100871 , People's Republic of China
| | - Yangyang Li
- Academy for Advanced Interdisciplinary Studies , Peking University , No. 5 Yiheyuan Road, Haidian District , Beijing 100871 , People's Republic of China
| | - Shi Shen
- Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA , Institute of Orthopedics , No. 28 Fuxing Road, Haidian District , Beijing 100853 , People's Republic of China
- Department of Bone and Joint Surgery , The Affiliated Hospital of Southwest Medical University , No. 25 Taiping Road , Luzhou 646000 , People's Republic of China
| | - Zhiguo Yuan
- Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA , Institute of Orthopedics , No. 28 Fuxing Road, Haidian District , Beijing 100853 , People's Republic of China
| | - Bo Huang
- Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA , Institute of Orthopedics , No. 28 Fuxing Road, Haidian District , Beijing 100853 , People's Republic of China
- Department of Bone and Joint Surgery , The Affiliated Hospital of Southwest Medical University , No. 25 Taiping Road , Luzhou 646000 , People's Republic of China
| | - Yu Zhang
- Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA , Institute of Orthopedics , No. 28 Fuxing Road, Haidian District , Beijing 100853 , People's Republic of China
| | - Mingjie Wang
- Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA , Institute of Orthopedics , No. 28 Fuxing Road, Haidian District , Beijing 100853 , People's Republic of China
| | - Xu Li
- Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA , Institute of Orthopedics , No. 28 Fuxing Road, Haidian District , Beijing 100853 , People's Republic of China
| | - Libo Hao
- Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA , Institute of Orthopedics , No. 28 Fuxing Road, Haidian District , Beijing 100853 , People's Republic of China
| | - Jiang Peng
- Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA , Institute of Orthopedics , No. 28 Fuxing Road, Haidian District , Beijing 100853 , People's Republic of China
| | - Shuyun Liu
- Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA , Institute of Orthopedics , No. 28 Fuxing Road, Haidian District , Beijing 100853 , People's Republic of China
| | - Yixin Zhou
- Department of Orthopedic Surgery, Beijing Jishuitan Hospital , Peking University Fourth School of Clinical Medicine , No. 31 Xinjiekou East Street, Xicheng District , Beijing 100035 , People's Republic of China
| | - Quanyi Guo
- Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA , Institute of Orthopedics , No. 28 Fuxing Road, Haidian District , Beijing 100853 , People's Republic of China
| |
Collapse
|
9
|
Pereira H, Fatih Cengiz I, Gomes S, Espregueira-Mendes J, Ripoll PL, Monllau JC, Reis RL, Oliveira JM. Meniscal allograft transplants and new scaffolding techniques. EFORT Open Rev 2019; 4:279-295. [PMID: 31210969 PMCID: PMC6549113 DOI: 10.1302/2058-5241.4.180103] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Clinical management of meniscal injuries has changed radically in recent years. We have moved from the model of systematic tissue removal (meniscectomy) to understanding the need to preserve the tissue.Based on the increased knowledge of the basic science of meniscal functions and their role in joint homeostasis, meniscus preservation and/or repair, whenever indicated and possible, are currently the guidelines for management.However, when repair is no longer possible or when facing the fact of the previous partial, subtotal or total loss of the meniscus, meniscus replacement has proved its clinical value. Nevertheless, meniscectomy remains amongst the most frequent orthopaedic procedures.Meniscus replacement is currently possible by means of meniscal allograft transplantation (MAT) which provides replacement of the whole meniscus with or without bone plugs/slots. Partial replacement has been achieved by means of meniscal scaffolds (mainly collagen or polyurethane-based). Despite the favourable clinical outcomes, it is still debatable whether MAT is capable of preventing progression to osteoarthritis. Moreover, current scaffolds have shown some fundamental limitations, such as the fact that the newly formed tissue may be different from the native fibrocartilage of the meniscus.Regenerative tissue engineering strategies have been used in an attempt to provide a new generation of meniscal implants, either for partial or total replacement. The goal is to provide biomaterials (acellular or cell-seeded constructs) which provide the biomechanical properties but also the biological features to replace the loss of native tissue. Moreover, these approaches include possibilities for patient-specific implants of correct size and shape, as well as advanced strategies combining cells, bioactive agents, hydrogels or gene therapy.Herein, the clinical evidence and tips concerning MAT, currently available meniscus scaffolds and future perspectives are discussed. Cite this article: EFORT Open Rev 2019;4 DOI: 10.1302/2058-5241.4.180103.
Collapse
Affiliation(s)
- Hélder Pereira
- Orthopedic Department of Póvoa de Varzim - Vila do Conde Hospital Centre, Vila do Conde, Portugal
- Ripoll y De Prado Sports Clinic, Murcia-Madrid, FIFA Medical Centre of Excellence, Madrid, Spain
- International Centre of Sports Traumatology of the Ave, Vila do Conde, Portugal
- 3Bs Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, Portugal
- ICVS/3Bs, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ibrahim Fatih Cengiz
- 3Bs Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, Portugal
- ICVS/3Bs, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sérgio Gomes
- International Centre of Sports Traumatology of the Ave, Vila do Conde, Portugal
| | - João Espregueira-Mendes
- 3Bs Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, Portugal
- ICVS/3Bs, PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Clínica do Dragão, Espregueira-Mendes Sports Centre, FIFA Medical Centre of Excellence, Porto, Portugal
- Orthopedic Department, University of Minho, Braga, Portugal
| | - Pedro L. Ripoll
- Ripoll y De Prado Sports Clinic, Murcia-Madrid, FIFA Medical Centre of Excellence, Madrid, Spain
| | - Joan C. Monllau
- Orthopaedic Department, Hospital del Mar, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Rui L. Reis
- 3Bs Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, Portugal
- ICVS/3Bs, PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, Barco, Guimarães, Portugal
| | - J. Miguel Oliveira
- 3Bs Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, Portugal
- ICVS/3Bs, PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Orthopaedic Department, Hospital del Mar, Universitat Autònoma de Barcelona, Barcelona, Spain
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, Barco, Guimarães, Portugal
| |
Collapse
|
10
|
Janmohammadi M, Nourbakhsh MS. Recent advances on 3D printing in hard and soft tissue engineering. INT J POLYM MATER PO 2019. [DOI: 10.1080/00914037.2019.1581196] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Mahsa Janmohammadi
- Biomaterial Group, Faculty of New Sciences and Technologies, Semnan University, Semnan, Iran
| | - Mohammad Sadegh Nourbakhsh
- Biomedical Engineering- Biomaterials, Faculty of Materials and Metallurgical Engineering, Semnan University, Semnan, Iran
| |
Collapse
|
11
|
Jiang X, Liu J, Liu Q, Lu Z, Zheng L, Zhao J, Zhang X. Therapy for cartilage defects: functional ectopic cartilage constructed by cartilage-simulating collagen, chondroitin sulfate and hyaluronic acid (CCH) hybrid hydrogel with allogeneic chondrocytes. Biomater Sci 2018; 6:1616-1626. [PMID: 29737330 DOI: 10.1039/c8bm00354h] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To regenerate functional cartilage-mimicking ectopic cartilage as a source for the restoration of cartilage defects, we used a previously synthesized three-phase collagen, chondroitin sulfate and hyaluronic acid (CCH) hydrogel for the encapsulation of allogeneic chondrocytes with a diffusion chamber system that was buried subcutaneously in the host for 4 weeks and then implanted into a cartilage defect. METHODS The CCH hydrogel was prepared and seeded with allogeneic chondrocytes from new-born rabbits, prior to being enveloped in a diffusion chamber that prevents cell ingrowth and vascular invasion of the host, as described previously. A collagen hydrogel (C) was used as the control. The diffusion chamber was embedded subcutaneously in an adult rabbit. 4 weeks later, the regenerated tissue was harvested from the diffusion chamber and then further used for cartilage repair in the same host. To evaluate the regenerated tissue, cell viability assay using calcein-acetoxymethyl (calcein-AM)/propidium iodide (PI) staining, biochemical analysis by examination of total DNA and GAG content, gene expression detection using RT-PCR for Col 1a1, Col 2a1, Acan, and Sox9, biomechanical detection and histological evaluation were implemented. RESULTS Analysis of the cell activity and biochemical evaluation in vitro showed that cell proliferation, GAG secretion and gene/protein expression of cartilage specific markers were much higher in the CCH group than those in the C group. The CCH constructed ectopic cartilage tissue in vivo showed the typical characteristics of hyaline cartilage with higher expression of cartilage matrix markers compared with the C groups, as evidenced by morphological and histological findings as well as RT-PCR analysis. Furthermore, ectopic cartilage from CCH successfully facilitated the cartilage restoration, with higher morphological and histological scores and greater mechanical strength than that from C. CONCLUSION The three-phase CCH hydrogel, which is closer to natural cartilage matrix and is stiffer than collagen, may replace collagen as the "gold standard" for cartilage tissue engineering. This study may provide a new insight for cartilage repair using ectopic cartilage reconstructed from functional materials and allogeneic cells.
Collapse
Affiliation(s)
- Xianfang Jiang
- The College of Stomatology of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | | | | | | | | | | | | |
Collapse
|
12
|
Grogan SP, Duffy SF, Pauli C, Lotz MK, D’Lima DD. Gene expression profiles of the meniscus avascular phenotype: A guide for meniscus tissue engineering. J Orthop Res 2018; 36:1947-1958. [PMID: 29411909 PMCID: PMC6326361 DOI: 10.1002/jor.23864] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 02/05/2018] [Indexed: 02/04/2023]
Abstract
Avascular (Avas) meniscus regeneration remains a challenge, which is partly a consequence of our limited knowledge of the cells that maintain this tissue region. In this study, we utilized microarrays to characterize gene expression profiles of intact human Avas meniscus tissue and of cells following culture expansion. Using these data, we examined various 3D culture conditions to redifferentiate Avas cells toward the tissue phenotype. RNA was isolated from either the tissue directly or following cell isolation and 2 weeks in monolayer culture. RNA was hybridized on human genome arrays. Differentially expressed (DE) genes were identified by ranking analysis. DAVID pathway analysis was performed and visualized using STRING analysis. Quantitative PCR (qPCR) on additional donor menisci (tissues and cells) were used to validate array data. Avas cells cultured in 3D were subjected to qPCR to compare with the array-generated data. A total of 387 genes were DE based on differentiation state (>3-fold change; p < 0.01). In Avas-cultured cells, the upregulated pathways included focal adhesion, ECM-receptor interaction, regulation of actin cytoskeleton, and PDGF Signaling. In 3D-cultured Avas cells, TGFβ1 or combinations of TGFβ1 and BMP6 were most effective to promote an Avas tissue phenotype. THBS2 and THBS4 expression levels were identified as a means to denote meniscus cell phenotype status. We identified the key gene expression profiles, new markers and pathways involved in characterizing the Avas meniscus phenotype in the native state and during in vitro dedifferentiation and redifferentiation. These data served to screen 3D conditions to generate meniscus-like neotissues. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1947-1958, 2018.
Collapse
Affiliation(s)
- Shawn P Grogan
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA
| | - Stuart F. Duffy
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Chantal Pauli
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Martin K Lotz
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Darryl D D’Lima
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA,Corresponding author: Darryl D D’Lima, MD, PhD, Shiley Center for Orthopaedic Research and Education at Scripps Clinic, 11025 North Torrey Pines Road, Suite 200, La Jolla, CA 92037, Tel 858 332 0166 Fax 858 332 0669,
| |
Collapse
|
13
|
Bilgen B, Jayasuriya CT, Owens BD. Current Concepts in Meniscus Tissue Engineering and Repair. Adv Healthc Mater 2018; 7:e1701407. [PMID: 29542287 PMCID: PMC6176857 DOI: 10.1002/adhm.201701407] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/22/2018] [Indexed: 12/13/2022]
Abstract
The meniscus is the most commonly injured structure in the human knee. Meniscus deficiency has been shown to lead to advanced osteoarthritis (OA) due to abnormal mechanical forces, and replacement strategies for this structure have lagged behind other tissue engineering endeavors. The challenges include the complex 3D structure with individualized size parameters, the significant compressive, tensile and shear loads encountered, and the poor blood supply. In this progress report, a review of the current clinical treatments for different types of meniscal injury is provided. The state-of-the-art research in cellular therapies and novel cell sources for these therapies is discussed. The clinically available cell-free biomaterial implants and the current progress on cell-free biomaterial implants are reviewed. Cell-based tissue engineering strategies for the repair and replacement of meniscus are presented, and the current challenges are identified. Tissue-engineered meniscal biocomposite implants may provide an alternative solution for the treatment of meniscal injury to prevent OA in the long run, because of the limitations of the existing therapies.
Collapse
Affiliation(s)
- Bahar Bilgen
- Department of Orthopaedics, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, 1 Hoppin St, Providence, RI, 02903, USA
- Providence VA Medical Center, Providence, RI, 02908, USA
| | - Chathuraka T Jayasuriya
- Department of Orthopaedics, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, 1 Hoppin St, Providence, RI, 02903, USA
| | - Brett D Owens
- Department of Orthopaedics, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, 1 Hoppin St, Providence, RI, 02903, USA
| |
Collapse
|
14
|
He A, Xia H, Xiao K, Wang T, Liu Y, Xue J, Li D, Tang S, Liu F, Wang X, Zhang W, Liu W, Cao Y, Zhou G. Cell yield, chondrogenic potential, and regenerated cartilage type of chondrocytes derived from ear, nasoseptal, and costal cartilage. J Tissue Eng Regen Med 2017; 12:1123-1132. [PMID: 29139602 DOI: 10.1002/term.2613] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 08/24/2017] [Accepted: 10/28/2017] [Indexed: 11/12/2022]
Abstract
Functional reconstruction of large cartilage defects in subcutaneous sites remains clinically challenging because of limited donor cartilage. Tissue engineering is a promising and widely accepted strategy for cartilage regeneration. To date, however, this strategy has not achieved a significant breakthrough in clinical translation owing to a lack of detailed preclinical data on cell yield and functionality of clinically applicable chondrocytes. To address this issue, the current study investigated the initial cell yield, proliferative potential, chondrogenic capacity, and regenerated cartilage type of human chondrocytes derived from auricular, nasoseptal, and costal cartilage using a scaffold-free cartilage regeneration model (cartilage sheet). Chondrocytes from all sources exhibited high sensitivity to basic fibroblast growth factor within 8 passages. Nasoseptal chondrocytes presented the strongest proliferation rate, whereas auricular chondrocytes obtained the highest total cell amount using comparable cartilage sample weights. Importantly, all chondrocytes at fifth passage showed strong chondrogenic capacity both in vitro and in the subcutaneous environment of nude mice. Although some significant differences in histological structure, cartilage matrix content and cartilage type specific proteins were observed between the in vitro engineered cartilage and original tissue; the in vivo regenerated cartilage showed mature cartilage features with high similarity to their original native tissue, except for minor matrix changes influenced by the in vivo environment. The current study provides detailed preclinical data for choice of chondrocyte source and thus promotes the clinical translation of cartilage regeneration approach.
Collapse
Affiliation(s)
- Aijuan He
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China.,National Tissue Engineering Center of China, Shanghai, China
| | - Huitang Xia
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China.,National Tissue Engineering Center of China, Shanghai, China.,Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, Shandong, China
| | - Kaiyan Xiao
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
| | - Tingting Wang
- National Tissue Engineering Center of China, Shanghai, China.,Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, Shandong, China
| | - Yu Liu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China.,National Tissue Engineering Center of China, Shanghai, China
| | - Jixin Xue
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China.,Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dan Li
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China.,National Tissue Engineering Center of China, Shanghai, China
| | - Shengjian Tang
- Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, Shandong, China
| | - Fangjun Liu
- Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, Shandong, China
| | - Xiaoyun Wang
- Department of General Surgery, Wu Jing Hospital, Minhang District, Shanghai, China
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China.,National Tissue Engineering Center of China, Shanghai, China
| | - Wei Liu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China.,National Tissue Engineering Center of China, Shanghai, China
| | - Yilin Cao
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China.,National Tissue Engineering Center of China, Shanghai, China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China.,National Tissue Engineering Center of China, Shanghai, China.,Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, Shandong, China
| |
Collapse
|
15
|
Chew E, Prakash R, Khan W. Mesenchymal stem cells in human meniscal regeneration: A systematic review. Ann Med Surg (Lond) 2017; 24:3-7. [PMID: 29062478 PMCID: PMC5644998 DOI: 10.1016/j.amsu.2017.09.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 09/25/2017] [Accepted: 09/26/2017] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Stem cell regeneration is the holy grail of meniscal tissue repair. Currently, the best treatment is to preserve the original meniscus but if it fails, a partial meniscectomy is indicated to delay the onset of osteoarthritis. MATERIALS AND METHODS The authors present a systematic review to determine the up-to-date evidence underlying the use of mesenchymal stem cells for meniscal regeneration in humans. A search was conducted using the electronic databases of MEDLINE/Pubmed, Google scholar, and the Cochrane Collaboration. Search keywords included human, meniscus, stem cells and regeneration. RESULTS After screening 10 non-duplicate studies, 5 were identified based on title and abstract. 4 were included in the analysis. There were marked differences in the method of stem cell harvest techniques. 3 studies administered stem cells through percutaneous injection into the knee and 1 study used a collagen scaffold. MRI analysis, functional scores and safety were assessed and the longest follow-up period was 2 years. The Visual Analogue Score (VAS) was most commonly used to assess function and patients generally showed an improvement. There were no reported adverse events. CONCLUSION Despite positive results from animal models, there is currently a lack of evidence in humans to conclude that stem cells can form durable neotissue similar to original human meniscus. There is a need for standardisation of protocol before further trials are considered. Initial outcomes from human studies are promising and mesenchymal stem cells may play an important role in meniscal repair in years to come.
Collapse
Affiliation(s)
- Ernest Chew
- Department of Trauma and Orthopaedics, St Mary's Hospital, London, W2 1NY, United Kingdom
| | - Rohan Prakash
- Department of Trauma and Orthopaedics, Royal Free Hospital, London, NW3 2QG, United Kingdom
| | - Wasim Khan
- Department of Trauma and Orthopaedics, Addenbrooke's Hospital, Cambridge, CB2 0QQ, United Kingdom
| |
Collapse
|
16
|
Baek J, Sovani S, Choi W, Jin S, Grogan SP, D'Lima DD. Meniscal Tissue Engineering Using Aligned Collagen Fibrous Scaffolds: Comparison of Different Human Cell Sources. Tissue Eng Part A 2017; 24:81-93. [PMID: 28463545 DOI: 10.1089/ten.tea.2016.0205] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hydrogel and electrospun scaffold materials support cell attachment and neotissue development and can be tuned to structurally and mechanically resemble native extracellular matrix by altering either electrospun fiber or hydrogel properties. In this study, we examined meniscus tissue generation from different human cell sources including meniscus cells derived from vascular and avascular regions, human bone marrow-derived mesenchymal stem cells, synovial cells, and cells from the infrapatellar fat pad (IPFP). All cells were seeded onto aligned electrospun collagen type I scaffolds and were optionally encapsulated in a tricomponent hydrogel. Single or multilayered constructs were generated and cultivated in defined medium with selected growth factors for 2 weeks. Cell viability, cell morphology, and gene-expression profiles were monitored using confocal microscopy, scanning electron microscopy, and quantitative polymerase chain reaction (qPCR), respectively. Multilayered constructs were examined with histology, immunohistochemistry, qPCR, and for tensile mechanical properties. For all cell types, TGFβ1 and TGFβ3 treatment increased COL1A1, COMP, Tenascin C (TNC), and Scleraxis (SCX) gene expression and deposition of collagen type I protein. IPFP cells generated meniscus-like tissues with higher meniscogenic gene expression, mechanical properties, and better cell distribution compared to other cell types studied. We show proof of concept that electrospun collagen scaffolds support neotissue formation and IPFP cells have potential for use in cell-based meniscus regeneration strategies.
Collapse
Affiliation(s)
- Jihye Baek
- 1 Shiley Center for Orthopaedic Research and Education at Scripps Clinic , La Jolla, California.,2 Department of Material Science and Engineering, University of California , San Diego, La Jolla, California
| | - Sujata Sovani
- 1 Shiley Center for Orthopaedic Research and Education at Scripps Clinic , La Jolla, California
| | - Wonchul Choi
- 1 Shiley Center for Orthopaedic Research and Education at Scripps Clinic , La Jolla, California
| | - Sungho Jin
- 2 Department of Material Science and Engineering, University of California , San Diego, La Jolla, California
| | - Shawn P Grogan
- 1 Shiley Center for Orthopaedic Research and Education at Scripps Clinic , La Jolla, California
| | - Darryl D D'Lima
- 1 Shiley Center for Orthopaedic Research and Education at Scripps Clinic , La Jolla, California
| |
Collapse
|
17
|
Korpershoek JV, de Windt TS, Hagmeijer MH, Vonk LA, Saris DBF. Cell-Based Meniscus Repair and Regeneration: At the Brink of Clinical Translation?: A Systematic Review of Preclinical Studies. Orthop J Sports Med 2017; 5:2325967117690131. [PMID: 28321424 PMCID: PMC5347439 DOI: 10.1177/2325967117690131] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background: Meniscus damage can be caused by trauma or degeneration and is therefore common among patients of all ages. Repair or regeneration of the menisci could be of great importance not only for pain relief or regaining function but also to prevent degenerative disease and osteoarthritis. Current treatment does not offer consistent long-term improvement. Although preclinical research focusing on augmentation of meniscal tear repair and regeneration after meniscectomy is encouraging, clinical translation remains difficult. Purpose: To systematically evaluate the literature on in vivo meniscus regeneration and explore the optimal cell sources and conditions for clinical translation. We aimed at thorough evaluation of current evidence as well as clarifying the challenges for future preclinical and clinical studies. Study Design: Systematic review. Methods: A search was conducted using the electronic databases of MEDLINE, Embase, and the Cochrane Collaboration. Search terms included meniscus, regeneration, and cell-based. Results: After screening 81 articles based on title and abstract, 51 articles on in vivo meniscus regeneration could be included; 2 additional articles were identified from the references. Repair and regeneration of the meniscus has been described by intra-articular injection of multipotent mesenchymal stromal (stem) cells from adipose tissue, bone marrow, synovium, or meniscus or the use of these cell types in combination with implantable or injectable scaffolds. The use of fibrochondrocytes, chondrocytes, and transfected myoblasts for meniscus repair and regeneration is limited to the combination with different scaffolds. The comparative in vitro and in vivo studies mentioned in this review indicate that the use of allogeneic cells is as successful as the use of autologous cells. In addition, the implantation or injection of cell-seeded scaffolds increased tissue regeneration and led to better structural organization compared with scaffold implantation or injection of a scaffold alone. None of the studies mentioned in this review compare the effectiveness of different (cell-seeded) scaffolds. Conclusion: There is heterogeneity in animal models, cell types, and scaffolds used, and limited comparative studies are available. The comparative in vivo research that is currently available is insufficient to draw strong conclusions as to which cell type is the most promising. However, there is a vast amount of in vivo research on the use of different types of multipotent mesenchymal stromal (stem) cells in different experimental settings, and good results are reported in terms of tissue formation. None of these studies compare the effectiveness of different cell-scaffold combinations, making it hard to conclude which scaffold has the greatest potential.
Collapse
Affiliation(s)
- Jasmijn V Korpershoek
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tommy S de Windt
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Michella H Hagmeijer
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lucienne A Vonk
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Daniel B F Saris
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands.; MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| |
Collapse
|
18
|
Treatments of Meniscus Lesions of the Knee: Current Concepts and Future Perspectives. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2017. [DOI: 10.1007/s40883-017-0025-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
19
|
Abstract
SUMMARY A recent revival of global interest for reconstruction of long-segment tracheal defects, which represents one of the most interesting and complex problems in head and neck and thoracic reconstructive surgery, has been witnessed. The trachea functions as a conduit for air, and its subunits including the epithelial layer, hyaline cartilage, and segmental blood supply make it particularly challenging to reconstruct. A myriad of attempts at replacing the trachea have been described. These along with the anatomy, indications, and approaches including microsurgical tracheal reconstruction will be reviewed. Novel techniques such as tissue-engineering approaches will also be discussed. Multiple attempts at replacing the trachea with synthetic scaffolds have been met with failure. The main lesson learned from such failures is that the trachea must not be treated as a "simple tube." Understanding the anatomy, developmental biology, physiology, and diseases affecting the trachea are required for solving this problem.
Collapse
|
20
|
Di Giancamillo A, Deponti D, Addis A, Domeneghini C, Peretti GM. Meniscus maturation in the swine model: changes occurring along with anterior to posterior and medial to lateral aspect during growth. J Cell Mol Med 2014; 18:1964-74. [PMID: 25216283 PMCID: PMC4244012 DOI: 10.1111/jcmm.12367] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 06/10/2014] [Indexed: 12/27/2022] Open
Abstract
The meniscus plays important roles in knee function and mechanics and is characterized by a heterogeneous matrix composition. The changes in meniscus vascularization observed during growth suggest that the tissue-specific composition may be the result of a maturation process. This study has the aim to characterize the structural and biochemical variations that occur in the swine meniscus with age. To this purpose, menisci were collected from young and adult pigs and divided into different zones. In study 1, both lateral and medial menisci were divided into the anterior horn, the body and the posterior horn for the evaluation of glycosaminoglycans (GAGs), collagen 1 and 2 content. In study 2, the menisci were sectioned into the inner, the intermediate and the outer zones to determine the variations in the cell phenotype along with the inner–outer direction, through gene expression analysis. According to the results, the swine meniscus is characterized by an increasing enrichment in the cartilaginous component with age, with an increasing deposition in the anterior horn (GAGs and collagen 2; P < 0.01 both); moreover, this cartilaginous matrix strongly increases in the inner avascular and intermediate zone, as a consequence of a specific differentiation of meniscal cells towards a cartilaginous phenotype (collagen 2, P < 0.01). The obtained data add new information on the changes that accompany meniscus maturation, suggesting a specific response of meniscal cells to the regional mechanical stimuli in the knee joint.
Collapse
|
21
|
Vinagre G. Are collagen meniscus implants a reality? Orthopedics 2014; 37:581-2. [PMID: 25198350 DOI: 10.3928/01477447-20140825-02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
22
|
Advances in tracheal reconstruction. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2014; 2:e178. [PMID: 25426361 PMCID: PMC4229282 DOI: 10.1097/gox.0000000000000097] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 03/24/2014] [Indexed: 12/26/2022]
Abstract
Summary: A recent revival of global interest for reconstruction of long-segment tracheal defects, which represents one of the most interesting and complex problems in head and neck and thoracic reconstructive surgery, has been witnessed. The trachea functions as a conduit for air, and its subunits including the epithelial layer, hyaline cartilage, and segmental blood supply make it particularly challenging to reconstruct. A myriad of attempts at replacing the trachea have been described. These along with the anatomy, indications, and approaches including microsurgical tracheal reconstruction will be reviewed. Novel techniques such as tissue-engineering approaches will also be discussed. Multiple attempts at replacing the trachea with synthetic scaffolds have been met with failure. The main lesson learned from such failures is that the trachea must not be treated as a “simple tube.” Understanding the anatomy, developmental biology, physiology, and diseases affecting the trachea are required for solving this problem.
Collapse
|
23
|
Grogan SP, Chung PH, Soman P, Chen P, Lotz MK, Chen S, D’Lima DD. Digital micromirror device projection printing system for meniscus tissue engineering. Acta Biomater 2013; 9:7218-26. [PMID: 23523536 DOI: 10.1016/j.actbio.2013.03.020] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 03/07/2013] [Accepted: 03/14/2013] [Indexed: 12/17/2022]
Abstract
Meniscus degeneration due to age or injury can lead to osteoarthritis. Although promising, current cell-based approaches show limited success. Here we present three-dimensional methacrylated gelatin (GelMA) scaffolds patterned via projection stereolithography to emulate the circumferential alignment of cells in native meniscus tissue. Cultured human avascular zone meniscus cells from normal meniscus were seeded on the scaffolds. Cell viability was monitored, and new tissue formation was assessed by gene expression analysis and histology after 2weeks in serum-free culture with transforming growth factor β1 (10ngml(-1)). Light, confocal and scanning electron microscopy were used to observe cell-GelMA interactions. Tensile mechanical testing was performed on unseeded, fresh scaffolds and 2-week-old cell-seeded and unseeded scaffolds. 2-week-old cell-GelMA constructs were implanted into surgically created meniscus defects in an explant organ culture model. No cytotoxic effects were observed 3weeks after implantation, and cells grew and aligned to the patterned GelMA strands. Gene expression profiles and histology indicated promotion of a fibrocartilage-like meniscus phenotype, and scaffold integration with repair tissue was observed in the explant model. We show that micropatterned GelMA scaffolds are non-toxic, produce organized cellular alignment, and promote meniscus-like tissue formation. Prefabrication of GelMA scaffolds with architectures mimicking the meniscus collagen bundle organization shows promise for meniscal repair. Furthermore, the technique presented may be scaled up to repair larger defects.
Collapse
|
24
|
Esposito AR, Moda M, Cattani SMDM, de Santana GM, Barbieri JA, Munhoz MM, Cardoso TP, Barbo MLP, Russo T, D'Amora U, Gloria A, Ambrosio L, Duek EADR. PLDLA/PCL-T Scaffold for Meniscus Tissue Engineering. Biores Open Access 2013; 2:138-47. [PMID: 23593566 PMCID: PMC3620496 DOI: 10.1089/biores.2012.0293] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The inability of the avascular region of the meniscus to regenerate has led to the use of tissue engineering to treat meniscal injuries. The aim of this study was to evaluate the ability of fibrochondrocytes preseeded on PLDLA/PCL-T [poly(L-co-D,L-lactic acid)/poly(caprolactone-triol)] scaffolds to stimulate regeneration of the whole meniscus. Porous PLDLA/PCL-T (90/10) scaffolds were obtained by solvent casting and particulate leaching. Compressive modulus of 9.5±1.0 MPa and maximum stress of 4.7±0.9 MPa were evaluated. Fibrochondrocytes from rabbit menisci were isolated, seeded directly on the scaffolds, and cultured for 21 days. New Zealand rabbits underwent total meniscectomy, after which implants consisting of cell-free scaffolds or cell-seeded scaffolds were introduced into the medial knee meniscus; the negative control group consisted of rabbits that received no implant. Macroscopic and histological evaluations of the neomeniscus were performed 12 and 24 weeks after implantation. The polymer scaffold implants adapted well to surrounding tissues, without apparent rejection, infection, or chronic inflammatory response. Fibrocartilaginous tissue with mature collagen fibers was observed predominantly in implants with seeded scaffolds compared to cell-free implants after 24 weeks. Similar results were not observed in the control group. Articular cartilage was preserved in the polymeric implants and showed higher chondrocyte cell number than the control group. These findings show that the PLDLA/PCL-T 90/10 scaffold has potential for orthopedic applications since this material allowed the formation of fibrocartilaginous tissue, a structure of crucial importance for repairing injuries to joints, including replacement of the meniscus and the protection of articular cartilage from degeneration.
Collapse
Affiliation(s)
- Andrea Rodrigues Esposito
- Department of Materials Engineering, Faculty of Mechanical Engineering, University of Campinas (UNICAMP) , Campinas, Brazil . ; Laboratory of Biomaterials, Faculty of Medicine and Health Sciences, Pontifical Catholic University of Sao Paulo (PUC-SP) , Sorocaba, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Murphy MK, Huey DJ, Reimer AJ, Hu JC, Athanasiou KA. Enhancing post-expansion chondrogenic potential of costochondral cells in self-assembled neocartilage. PLoS One 2013; 8:e56983. [PMID: 23437288 PMCID: PMC3578801 DOI: 10.1371/journal.pone.0056983] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 01/16/2013] [Indexed: 11/18/2022] Open
Abstract
The insufficient healing capacity of articular cartilage necessitates mechanically functional biologic tissue replacements. Using cells to form biomimetic cartilage implants is met with the challenges of cell scarcity and donor site morbidity, requiring expanded cells that possess the ability to generate robust neocartilage. To address this, this study assesses the effects of expansion medium supplementation (bFGF, TFP, FBS) and self-assembled construct seeding density (2, 3, 4 million cells/5 mm dia. construct) on the ability of costochondral cells to generate biochemically and biomechanically robust neocartilage. Results show TFP (1 ng/mL TGF-β1, 5 ng/mL bFGF, 10 ng/mL PDGF) supplementation of serum-free chondrogenic expansion medium enhances the post-expansion chondrogenic potential of costochondral cells, evidenced by increased glycosaminoglycan content, decreased type I/II collagen ratio, and enhanced compressive properties. Low density (2 million cells/construct) enhances matrix synthesis and tensile and compressive mechanical properties. Combined, TFP and Low density interact to further enhance construct properties. That is, with TFP, Low density increases type II collagen content by over 100%, tensile stiffness by over 300%, and compressive moduli by over 140%, compared with High density. In conclusion, the interaction of TFP and Low density seeding enhances construct material properties, allowing for a mechanically functional, biomimetic cartilage to be formed using clinically relevant costochondral cells.
Collapse
Affiliation(s)
- Meghan K. Murphy
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
| | - Daniel J. Huey
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
| | - Andrew J. Reimer
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
| | - Jerry C. Hu
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
| | - Kyriacos A. Athanasiou
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
26
|
Moriguchi Y, Tateishi K, Ando W, Shimomura K, Yonetani Y, Tanaka Y, Kita K, Hart DA, Gobbi A, Shino K, Yoshikawa H, Nakamura N. Repair of meniscal lesions using a scaffold-free tissue-engineered construct derived from allogenic synovial MSCs in a miniature swine model. Biomaterials 2012; 34:2185-93. [PMID: 23261221 DOI: 10.1016/j.biomaterials.2012.11.039] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 11/22/2012] [Indexed: 12/22/2022]
Abstract
The menisci of the knee are fibro-cartilaginous tissues and play important roles in the joint, and the loss of the meniscus predisposes the knee to degenerative changes. However, the menisci have limited healing potential due to the paucity of vascularity. The purpose of the present study was to test the feasibility of a scaffold-free tissue-engineered construct (TEC) derived from synovial mesenchymal stem cells (MSCs) to repair incurable meniscal lesions. Porcine synovial MSCs were cultured in monolayers at high density in the presence of ascorbic acid followed by the suspension culture to develop a three-dimensional cell/matrix construct (TEC). A 4-mm cylindrical defect was created bilaterally in the medial meniscus of skeletally mature miniature pigs. The defects were implanted with an allogenic TEC or were left empty. After 6 months, the TEC-treated defects were consistently repaired by a fibro-cartilaginous tissue with good tissue integration to the adjacent host meniscal tissue, while the untreated were either partially or not repaired. The ratio of Safranin O positive area within the central body of the meniscus adjacent to the original defect was significantly higher in the TEC-treated group than in the control group. Moreover, TEC treatment significantly reduced the size and severity of post-traumatic chondral lesions on the tibial plateau. These results suggest that the TEC could be a promising stem cell-based implant to repair meniscal lesions with preventive effects from meniscal body degeneration and the development of post-traumatic arthritis.
Collapse
Affiliation(s)
- Yu Moriguchi
- Department of Orthopaedics, Osaka University Graduate School of Medicine, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Hong HJ, Lee JS, Choi JW, Min BH, Lee HB, Kim CH. Transplantation of Autologous Chondrocytes Seeded on a Fibrin/Hyaluronan Composite Gel Into Tracheal Cartilage Defects in Rabbits: Preliminary Results. Artif Organs 2012; 36:998-1006. [DOI: 10.1111/j.1525-1594.2012.01486.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
28
|
Muzzarelli RAA, Greco F, Busilacchi A, Sollazzo V, Gigante A. Chitosan, hyaluronan and chondroitin sulfate in tissue engineering for cartilage regeneration: a review. Carbohydr Polym 2012; 89:723-39. [PMID: 24750856 DOI: 10.1016/j.carbpol.2012.04.057] [Citation(s) in RCA: 298] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 04/23/2012] [Accepted: 04/25/2012] [Indexed: 12/22/2022]
Abstract
Injection of hyaluronan into osteoarthritic joints restores the viscoelasticity, augments the flow of joint fluid, normalizes endogenous hyaluronan synthesis, and improves joint function. Chitosan easily forms polyelectrolyte complexes with hyaluronan and chondroitin sulfate. Synergy of chitosan with hyaluronan develops enhanced performances in regenerating hyaline cartilage, typical results being structural integrity of the hyaline-like neocartilage, and reconstitution of the subchondral bone, with positive cartilage staining for collagen-II and GAG in the treated sites. Chitosan qualifies for the preparation of scaffolds intended for the regeneration of cartilage: it yields mesoporous cryogels; it provides a friendly environment for chondrocytes to propagate, produce typical ECM, and assume the convenient phenotype; it is a good carrier for growth factors; it inactivates metalloproteinases thus preventing collagen degradation; it is suitable for the induction of the chondrogenic differentiation of mesenchymal stem cells; it is a potent means for hemostasis and platelet delivery.
Collapse
Affiliation(s)
- Riccardo A A Muzzarelli
- Clinical Orthopaedics, Department of Clinical and Experimental Sciences, Polytechnic University Delle Marche, Via Tronto 10-A, IT-60126 Ancona, Italy
| | - Francesco Greco
- Clinical Orthopaedics, Department of Clinical and Experimental Sciences, Polytechnic University Delle Marche, Via Tronto 10-A, IT-60126 Ancona, Italy
| | - Alberto Busilacchi
- Clinical Orthopaedics, Department of Clinical and Experimental Sciences, Polytechnic University Delle Marche, Via Tronto 10-A, IT-60126 Ancona, Italy
| | - Vincenzo Sollazzo
- Department of Orthopaedics, University of Ferrara, Corso Giovecca 203, IT-44100 Ferrara, Italy
| | - Antonio Gigante
- Clinical Orthopaedics, Department of Clinical and Experimental Sciences, Polytechnic University Delle Marche, Via Tronto 10-A, IT-60126 Ancona, Italy
| |
Collapse
|
29
|
Biological strategies to enhance healing of the avascular area of the meniscus. Stem Cells Int 2011; 2012:528359. [PMID: 22220179 PMCID: PMC3246301 DOI: 10.1155/2012/528359] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 11/01/2011] [Indexed: 12/15/2022] Open
Abstract
Meniscal injuries in the vascularized peripheral part of the meniscus have a better healing potential than tears in the central avascular zone because meniscal healing principally depends on its vascular supply. Several biological strategies have been proposed to enhance healing of the avascular area of the meniscus: abrasion therapy, fibrin clot, organ culture, cell therapy, and applications of growth factors. However, data are too heterogeneous to achieve definitive conclusions on the use of these techniques for routine management of meniscal lesions. Although most preclinical and clinical studies are very promising, they are still at an experimental stage. More prospective randomised controlled trials are needed to compare the different techniques for clinical results, applicability, and cost-effectiveness.
Collapse
|
30
|
Pereira H, Frias AM, Oliveira JM, Espregueira-Mendes J, Reis RL. Tissue engineering and regenerative medicine strategies in meniscus lesions. Arthroscopy 2011; 27:1706-19. [PMID: 22019234 DOI: 10.1016/j.arthro.2011.08.283] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 07/29/2011] [Accepted: 08/03/2011] [Indexed: 02/02/2023]
Abstract
PURPOSE The aim of this systematic review was to address tissue engineering and regenerative medicine (TERM) strategies applied to the meniscus, specifically (1) clinical applications, indications, results, and pitfalls and (2) the main trends in research assessed by evaluation of preclinical (in vivo) studies. METHODS Three independent reviewers performed a search on PubMed, from 2006 to March 31, 2011, using the term "meniscus" with all of the following terms: "scaffolds," "constructs," "cells," "growth factors," "implant," "tissue engineering," and "regenerative medicine." Inclusion criteria were English language-written, original clinical research (Level of Evidence I to IV) and preclinical studies of TERM application in knee meniscal lesions. Reference lists and related articles on journal Web sites of selected articles were checked until prepublication for potential studies that could not be identified eventually by our original search. The modified Coleman Methodology score was used for study quality analysis of clinical trials. RESULTS The PubMed search identified 286 articles (a similar search from 2000 to 2005 identified 161 articles). Non-English-language articles (n = 9), Level V publications (n = 19), in vitro studies (n = 118), and 102 studies not related to the topic were excluded. One reference was identified outside of PubMed. Thirty-eight references that met the inclusion criteria were identified from the original search. On the basis of our prepublication search, 2 other references were included. A total of 9 clinical and 31 preclinical studies were selected for further analysis. Of the clinical trials, 1 was classified as Level I, 2 as Level II, and 6 as Level IV. Eight referred to acellular scaffold implantation for partial meniscal replacement, and one comprised fibrin clot application. The mean modified Coleman Methodology score was 48.0 (SD, 15.7). Of the preclinical studies, 11 original works reported on studies using large animal models whereas 20 research studies used small animals. In these studies the experimental design favored cell-seeded scaffolds or scaffolds enhanced with growth factors (GFs) in attempts to improve tissue healing, as opposed to the plain acellular scaffolds that were predominant in clinical trials. Injection of mesenchymal stem cells and gene therapy are also presented as alternative strategies. CONCLUSIONS Partial meniscal substitution using acellular scaffolds in selected patients with irreparable loss of tissue may be a safe and promising procedure. However, there is only 1 randomized controlled study supporting its application, and globally, many methodologic issues of published trials limit further conclusions. We registered a different trend in preclinical trials, with most considering augmentation of scaffolds by cells and/or GFs, as opposed to the predominantly acellular approach in clinical trials. Different TERM approaches to enhance meniscal repair or regeneration are in preclinical analysis, such as the use of mesenchymal stem cells, gene therapy, and GFs alone or in combination, and thus could be considered in the design of subsequent trials. LEVEL OF EVIDENCE Level IV, systematic review of Level I to IV studies.
Collapse
Affiliation(s)
- Hélder Pereira
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, Minho University, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal.
| | | | | | | | | |
Collapse
|
31
|
Riera KM, Rothfusz NE, Wilusz RE, Weinberg JB, Guilak F, McNulty AL. Interleukin-1, tumor necrosis factor-alpha, and transforming growth factor-beta 1 and integrative meniscal repair: influences on meniscal cell proliferation and migration. Arthritis Res Ther 2011; 13:R187. [PMID: 22087734 PMCID: PMC3334636 DOI: 10.1186/ar3515] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 11/08/2011] [Accepted: 11/16/2011] [Indexed: 12/29/2022] Open
Abstract
Introduction Interleukin-1 (IL-1) and tumor necrosis factor-α (TNF-α) are up-regulated in injured and osteoarthritic knee joints. IL-1 and TNF-α inhibit integrative meniscal repair; however, the mechanisms by which this inhibition occurs are not fully understood. Transforming growth factor-β1 (TGF-β1) increases meniscal cell proliferation and accumulation, and enhances integrative meniscal repair. An improved understanding of the mechanisms modulating meniscal cell proliferation and migration will help to improve approaches for enhancing intrinsic or tissue-engineered repair of the meniscus. The goal of this study was to examine the hypothesis that IL-1 and TNF-α suppress, while TGF-β1 enhances, cellular proliferation and migration in cell and tissue models of meniscal repair. Methods A micro-wound assay was used to assess meniscal cell migration and proliferation in response to the following treatments for 0, 24, or 48 hours: 0 to 10 ng/mL IL-1, TNF-α, or TGF-β1, in the presence or absence of 10% serum. Proliferated and total cells were fluorescently labeled and imaged using confocal laser scanning microscopy and the number of proliferated, migrated, and total cells was determined in the micro-wound and edges of each image. Meniscal cell proliferation was also assessed throughout meniscal repair model explants treated with 0 or 10 ng/mL IL-1, TNF-α, or TGF-β1 for 14 days. At the end of the culture period, biomechanical testing and histological analyses were also performed. Statistical differences were assessed using an ANOVA and Newman-Keuls post hoc test. Results IL-1 and TNF-α decreased cell proliferation in both cell and tissue models of meniscal repair. In the presence of serum, TGF-β1 increased outer zone cell proliferation in the micro-wound and in the cross section of meniscal repair model explants. Both IL-1 and TNF-α decreased the integrative shear strength of repair and extracellular matrix deposition in the meniscal repair model system, while TGF-β1 had no effect on either measure. Conclusions Meniscal cell proliferation in vivo may be diminished following joint injury due to the up-regulation of inflammatory cytokines, thereby limiting native cellular repair of meniscal lesions. Therefore, therapies that can promote meniscal cell proliferation have promise to enhance meniscal repair and improve tissue engineering strategies.
Collapse
Affiliation(s)
- Katherine M Riera
- Department of Orthopaedic Surgery, Duke University Medical Center, DUMC Box 3093, Durham, NC 27710, USA
| | | | | | | | | | | |
Collapse
|
32
|
Advances in meniscal tissue engineering. Stem Cells Int 2011; 2012:420346. [PMID: 25098366 PMCID: PMC3205710 DOI: 10.1155/2012/420346] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 08/26/2011] [Indexed: 01/08/2023] Open
Abstract
Meniscal tears are the most common knee injuries and have a poor ability of healing. In the last few decades, several techniques have been increasingly used to optimize meniscal healing. Current research efforts of tissue engineering try to combine cell-based therapy, growth factors, gene therapy, and reabsorbable scaffolds to promote healing of meniscal defects. Preliminary studies did not allow to draw definitive conclusions on the use of these techniques for routine management of meniscal lesions. We performed a review of the available literature on current techniques of tissue engineering for the management of meniscal tears.
Collapse
|
33
|
The knee meniscus: structure-function, pathophysiology, current repair techniques, and prospects for regeneration. Biomaterials 2011; 32:7411-31. [PMID: 21764438 DOI: 10.1016/j.biomaterials.2011.06.037] [Citation(s) in RCA: 652] [Impact Index Per Article: 50.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 06/17/2011] [Indexed: 02/07/2023]
Abstract
Extensive scientific investigations in recent decades have established the anatomical, biomechanical, and functional importance that the meniscus holds within the knee joint. As a vital part of the joint, it acts to prevent the deterioration and degeneration of articular cartilage, and the onset and development of osteoarthritis. For this reason, research into meniscus repair has been the recipient of particular interest from the orthopedic and bioengineering communities. Current repair techniques are only effective in treating lesions located in the peripheral vascularized region of the meniscus. Healing lesions found in the inner avascular region, which functions under a highly demanding mechanical environment, is considered to be a significant challenge. An adequate treatment approach has yet to be established, though many attempts have been undertaken. The current primary method for treatment is partial meniscectomy, which commonly results in the progressive development of osteoarthritis. This drawback has shifted research interest toward the fields of biomaterials and bioengineering, where it is hoped that meniscal deterioration can be tackled with the help of tissue engineering. So far, different approaches and strategies have contributed to the in vitro generation of meniscus constructs, which are capable of restoring meniscal lesions to some extent, both functionally as well as anatomically. The selection of the appropriate cell source (autologous, allogeneic, or xenogeneic cells, or stem cells) is undoubtedly regarded as key to successful meniscal tissue engineering. Furthermore, a large variation of scaffolds for tissue engineering have been proposed and produced in experimental and clinical studies, although a few problems with these (e.g., byproducts of degradation, stress shielding) have shifted research interest toward new strategies (e.g., scaffoldless approaches, self-assembly). A large number of different chemical (e.g., TGF-β1, C-ABC) and mechanical stimuli (e.g., direct compression, hydrostatic pressure) have also been investigated, both in terms of encouraging functional tissue formation, as well as in differentiating stem cells. Even though the problems accompanying meniscus tissue engineering research are considerable, we are undoubtedly in the dawn of a new era, whereby recent advances in biology, engineering, and medicine are leading to the successful treatment of meniscal lesions.
Collapse
|
34
|
Sayed KE, Haisch A, John T, Marzahn U, Lohan A, Müller RD, Kohl B, Ertel W, Stoelzel K, Schulze-Tanzil G. Heterotopic Autologous Chondrocyte Transplantation—A Realistic Approach to Support Articular Cartilage Repair? TISSUE ENGINEERING PART B-REVIEWS 2010; 16:603-16. [DOI: 10.1089/ten.teb.2010.0167] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Karym El Sayed
- Department of Trauma and Reconstructive Surgery, Charité-Universitätsmedizin, Berlin, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-Universitätsmedizin, Berlin, Germany
| | - Andreas Haisch
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-Universitätsmedizin, Berlin, Germany
| | - Thilo John
- Department of Trauma and Reconstructive Surgery, Charité-Universitätsmedizin, Berlin, Germany
| | - Ulrike Marzahn
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-Universitätsmedizin, Berlin, Germany
| | - Anke Lohan
- Department of Trauma and Reconstructive Surgery, Charité-Universitätsmedizin, Berlin, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-Universitätsmedizin, Berlin, Germany
| | - Riccarda D. Müller
- Department of Trauma and Reconstructive Surgery, Charité-Universitätsmedizin, Berlin, Germany
| | - Benjamin Kohl
- Department of Trauma and Reconstructive Surgery, Charité-Universitätsmedizin, Berlin, Germany
| | - Wolfgang Ertel
- Department of Trauma and Reconstructive Surgery, Charité-Universitätsmedizin, Berlin, Germany
| | - Katharina Stoelzel
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-Universitätsmedizin, Berlin, Germany
| | - Gundula Schulze-Tanzil
- Department of Trauma and Reconstructive Surgery, Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|
35
|
Sun L, Reagan MR, Kaplan DL. Role of Cartilage Forming Cells in Regenerative Medicine for Cartilage Repair. Orthop Res Rev 2010; 2010:85-94. [PMID: 24049462 PMCID: PMC3773876 DOI: 10.2147/orr.s7194] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Currently, cartilage repair remains a major challenge for researchers and physicians due to its limited healing capacity. Cartilage regeneration requires suitable cells; these must be easily obtained and expanded, able to produce hyaline matrix with proper mechanical properties, and demonstrate sustained integration with native tissue. At present, there is a wide variety of possible cell sources for cartilage regeneration; this review explores the diversity of sources for cartilage forming cells and the distinctive characteristics, advantages, limitations, and potential applications of each cell source. We place emphasis on cell sources used for in vitro and clinical studies.
Collapse
Affiliation(s)
- Lin Sun
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby St, Medford MA., USA
| | - Michaela R. Reagan
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford MA., USA
| | - David L. Kaplan
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby St, Medford MA., USA
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford MA., USA
| |
Collapse
|
36
|
Gunja NJ, Athanasiou KA. Effects of co-cultures of meniscus cells and articular chondrocytes on PLLA scaffolds. Biotechnol Bioeng 2009; 103:808-16. [DOI: 10.1002/bit.22301] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
37
|
Healing of meniscal tissue by cellular fibrin glue: an in vivo study. Knee Surg Sports Traumatol Arthrosc 2009; 17:645-51. [PMID: 19296087 DOI: 10.1007/s00167-009-0745-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2008] [Accepted: 02/03/2009] [Indexed: 02/05/2023]
Abstract
Menisci represent fundamental structures for the maintenance of knee homeostasis, playing a key role in knee biomechanics. However, their intrinsic regenerative potential is poor. As a consequence, when a lesion occurs and the meniscus is partially removed by surgery, knee mechanics is subject to dramatic changes. These have been demonstrated to lead often to the development of early osteoarthritis. Therefore, menisci should be repaired whenever possible. In the last decades, tissue engineering approaches have been advocated to improve the reparative processes of joint tissues. In this study, the bonding capacity of an articular chondrocytes-fibrin glue hydrogel was tested as a biologic glue to improve the bonding between two swine meniscal slices in a nude mouse model. The composites were wrapped with acellular fibrin glue and implanted in subcutaneous pouches of nude mice for 4 weeks. Upon retrieval, a firm gross bonding was observed in the experimental samples while none of the control samples, prepared with acellular fibrin glue at the interface, presented any sign of bonding. This was consistent with the histological and scanning electron microscope findings. In particular, a fibrocartilaginous tissue was found at the interface between the meniscal slices, partially penetrating the native meniscus tissue. In order to overcome the lack of regenerative properties of the meniscus, the rationale of using cellular fibrin glue is that fibrin provides immediate stability while carrying cells in the site of lesion. Moreover, fibrin gel is recognized as an optimal scaffold for cell embedding and for promoting fibrocartilaginous differentiation of the cells which synthesize matrix having healing property. These results demonstrated the potential of this model for improving the meniscal bonding. However, further orthotopic studies in a large animal model are needed to evaluate its potential for clinical application.
Collapse
|
38
|
Peretti GM, Scotti C, Pozzi A, Mangiavini L, Vitari F, Domeneghini C, Fraschini G. Bonding of meniscal tissue: a nude mouse repair model. SPORT SCIENCES FOR HEALTH 2009. [DOI: 10.1007/s11332-008-0070-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
39
|
Hoben GM, Athanasiou KA. Creating a spectrum of fibrocartilages through different cell sources and biochemical stimuli. Biotechnol Bioeng 2008; 100:587-98. [PMID: 18078296 DOI: 10.1002/bit.21768] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In this study a scaffoldless approach was employed with two different cell sources and biochemical stimuli to engineer a spectrum of fibrocartilages representative of the different regions of the knee meniscus. Constructs composed of 100% fibrochondrocytes (FC) or a 50:50 co-culture of fibrochondrocytes and chondrocytes (CC) were cultured in 10% fetal bovine serum medium or serum-free "chondrogenic" medium, each +/-10 ng/mL TGF-beta1 (+T). Constructs from these two cell groups and four culture conditions were cultured for 6 weeks. By varying the cell type and presence of the growth factor, GAG per dry weight of the constructs spanned that of native tissue, ranging 16-45% and 1-7% in the CC and FC groups, respectively. Collagen density was most dependent on cell type and was significantly lower than tissue values. The collagen I/II ratio could be manipulated by cell type and serum presence to span the native range, from 3.5 in the serum-free CC group to over 1,000 in the FC groups treated with serum-containing medium. Using the CC cell group in the presence of serum-free medium dramatically increased the compressive stiffness to 128 +/- 34 kPa, similar to native tissue. Similarly, serum-free medium or TGF-beta1 treatment enhanced the tensile modulus by an order of magnitude, up to 3,000 kPa. Using two cell sources and manipulating biochemical stimuli, a range of fibrocartilaginous neotissues was engineered. Fibrocartilages such as the knee meniscus are characterized by heterogeneity in matrix and functional properties, and this work demonstrates a strategy for recreating these heterogeneous tissues.
Collapse
Affiliation(s)
- Gwendolyn M Hoben
- Department of Bioengineering, Rice University, 6100 Main St, Keck Hall Suite 116, Houston, Texas 77005, USA
| | | |
Collapse
|
40
|
Quintin A, Hirt-Burri N, Scaletta C, Schizas C, Pioletti DP, Applegate LA. Consistency and safety of cell banks for research and clinical use: preliminary analysis of fetal skin banks. Cell Transplant 2008; 16:675-84. [PMID: 18019357 DOI: 10.3727/000000007783465127] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Current restrictions for human cell-based therapies have been related to technological limitations with regards to cellular proliferation capacity, maintenance of differentiated phenotype for primary human cell culture, and transmission of communicable diseases. We have seen that cultured primary fetal cells from one organ donation could possibly meet the exigent and stringent technical aspects for development of therapeutic products. We could develop a master cell bank (MCB) of 50 homogenous ampoules of 4-5 million cells each from one fetal organ donation (skin) in short periods of time compared to other primary cell types. Safety tests were performed at all stages of the cell banking. MCB ampoules could create a working cell bank to be used for clinical or research use. Monolayer culture of fetal skin cells had a life span of 12-17 passages, and independent cultures obtained from the same organ donation were consistent for protein concentration (with 1.4-fold maximal difference between cultures) as well as gene expression of MMP-14, MMP-3, TIMP-3, and VEGF (1.4-, 1.9-, 2.1-, and 1.4-fold maximal difference between cultures, respectively). Cell cultures derived from four independent fetal skin donations were consistent for cell growth, protein concentration, and gene expression of MDK, PTN, TGF-beta1, and OPG. As it is the intention that banked primary fetal cells can profit from the potential treatment of hundreds of thousands of patients with only one organ donation, it is imperative to show consistency, tracability, and safety of the process, including donor tissue selection, cell banking, cell testing, and growth of cells in upscaling for the preparation of cell transplantation.
Collapse
Affiliation(s)
- Aurelie Quintin
- Orthopedic Cell Therapy Unit, University Hospital, Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
41
|
Judas F, Rosa S, Teixeira L, Lopes C, Ferreira Mendes A. Chondrocyte viability in fresh and frozen large human osteochondral allografts: effect of cryoprotective agents. Transplant Proc 2007; 39:2531-4. [PMID: 17954166 DOI: 10.1016/j.transproceed.2007.07.028] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Chondrocyte survival is a major goal for the effective storage and clinical performance of human osteochondral allografts. The majority of animal and human cryopreservation studies conducted so far have been performed in small osteochondral cylinders. Using human tibial plateaus as a model for large osteochondral pieces, this work sought to evaluate the cryoprotective efficiency of glycerol and dimethylsulfoxide (DMSO), and to identify cryopreservation conditions suitable for use in tissue banks. Human tibial plateaus harvested from 7 cadaveric tissue donors were incubated in the presence or absence of cryoprotective agents (CPA): 10% or 15% glycerol and 10% DMSO in a Ham F-12 nutrient mixture. Chondrocyte viability was assessed immediately after thawing, using the MTT reduction assay and a fluorescence microscopic method. The tibial plateaus frozen in the absence of CPA showed a significant decrease in chondrocyte viability. The use of CPA significantly increased chondrocyte viability compared with cartilage frozen without CPA (nearly 50% versus 80% living chondrocytes with 10% glycerol versus 10% DMSO, respectively) relative to that in fresh cartilage. In this regard, 10% DMSO was slightly more effective than either 10% or 15% glycerol, eliciting the recovery of approximately 15% relative to the living chondrocyte content in fresh cartilage. In all conditions, fluorescence microscopic studies showed that surviving chondrocytes were restricted to the superficial cartilage layer. Human tibial plateaus seemed to be a good experimental model to establish cryopreservation methods applicable to large human osteochondral pieces in tissue banks.
Collapse
Affiliation(s)
- F Judas
- Orthopaedics Department of Coimbra University Hospitals and Faculty of Medicine, University of Coimbra, 3000 Coimbra, Portugal.
| | | | | | | | | |
Collapse
|
42
|
Aufderheide AC, Athanasiou KA. Assessment of a bovine co-culture, scaffold-free method for growing meniscus-shaped constructs. ACTA ACUST UNITED AC 2007; 13:2195-205. [PMID: 17630876 DOI: 10.1089/ten.2006.0291] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Using a self-assembly (SA), scaffoldless method, five high-density co-cultures with varied ratios of meniscal fibrochondrocytes (MFCs) and articular chondrocytes (ACs) were seeded into novel meniscus-specific, ring-shaped agarose wells. The following ratios of MFCs to ACs were used: 0% MFC, 25% MFC, 50% MFC, 75% MFC, and 100% MFC. Over 4 weeks, all ratios of cells self-assembled into three-dimensional constructs with varying mechanobiological and morphological properties. All groups stained for collagen II (Col II), and all groups except the 0% MFC group stained for collagen I (Col I). It was found that the tensile modulus was proportional to the percentage of MFCs employed. The 100% MFC group yielded the greatest mechanical stiffness with 432.2 +/- 47 kPa tensile modulus and an ultimate tensile strength of 23.7 +/- 2.4 kPa. On gross inspection, the 50% MFC constructs were the most similar to our idealized meniscus shape, our primary criterion. A second experiment was performed to examine the anisotropy of constructs as well as to directly compare the scaffoldless, SA method with a poly-glycolic acid (PGA) scaffold-based construct. When compared to PGA constructs, the SA groups were 2-4 times stiffer and stronger in tension. Further, at 8 weeks, SA groups exhibited circumferential fiber bundles similar to native tissue. When pulled in the circumferential direction, the SA group had significantly higher tensile modulus (226 +/- 76 kPa) than when pulled in the radial direction (67 +/- 32 kPa). The PGA constructs had neither a directional collagen fiber orientation nor differences in mechanical properties in the radial or circumferential direction. It is suggested that the geometric constraint imposed by the ring-shaped, nonadhesive mold guides collagen fibril directionality and, thus, alters mechanical properties. Co-culturing ACs and MFCs in this manner appears to be a promising new method for tissue engineering fibrocartilaginous tissues exhibiting a spectrum of mechanical and biomechanical properties.
Collapse
Affiliation(s)
- Adam C Aufderheide
- Department of Bioengineering, Rice University, Houston, Texas 77251, USA
| | | |
Collapse
|