1
|
Peifer C, Oláh T, Venkatesan JK, Goebel L, Orth P, Schmitt G, Zurakowski D, Menger MD, Laschke MW, Cucchiarini M, Madry H. Locally Directed Recombinant Adeno- Associated Virus-Mediated IGF-1 Gene Therapy Enhances Osteochondral Repair and Counteracts Early Osteoarthritis In Vivo. Am J Sports Med 2024; 52:1336-1349. [PMID: 38482805 DOI: 10.1177/03635465241235149] [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] [Indexed: 04/04/2024]
Abstract
BACKGROUND Restoration of osteochondral defects is critical, because osteoarthritis (OA) can arise. HYPOTHESIS Overexpression of insulin-like growth factor 1 (IGF-1) via recombinant adeno-associated viral (rAAV) vectors (rAAV-IGF-1) would improve osteochondral repair and reduce parameters of early perifocal OA in sheep after 6 months in vivo. STUDY DESIGN Controlled laboratory study. METHODS Osteochondral defects were created in the femoral trochlea of adult sheep and treated with rAAV-IGF-1 or rAAV-lacZ (control) (24 defects in 6 knees per group). After 6 months in vivo, osteochondral repair and perifocal OA were assessed by well-established macroscopic, histological, and immunohistochemical scoring systems as well as biochemical and micro-computed tomography evaluations. RESULTS Application of rAAV-IGF-1 led to prolonged (6 months) IGF-1 overexpression without adverse effects, maintaining a significantly superior overall cartilage repair, together with significantly improved defect filling, extracellular matrix staining, cellular morphology, and surface architecture compared with rAAV-lacZ. Expression of type II collagen significantly increased and that of type I collagen significantly decreased. Subchondral bone repair and tidemark formation were significantly improved, and subchondral bone plate thickness and subarticular spongiosa mineral density returned to normal. The OA parameters of perifocal structure, cell cloning, and matrix staining were significantly better preserved upon rAAV-IGF-1 compared with rAAV-lacZ. Novel mechanistic associations between parameters of osteochondral repair and OA were identified. CONCLUSION Local rAAV-mediated IGF-1 overexpression enhanced osteochondral repair and ameliorated parameters of perifocal early OA. CLINICAL RELEVANCE IGF-1 gene therapy may be beneficial in repair of focal osteochondral defects and prevention of perifocal OA.
Collapse
Affiliation(s)
- Carolin Peifer
- Center of Experimental Orthopaedics, Saarland University, Homburg/Saar, Germany
| | - Tamás Oláh
- Center of Experimental Orthopaedics, Saarland University, Homburg/Saar, Germany
| | | | - Lars Goebel
- Center of Experimental Orthopaedics, Saarland University, Homburg/Saar, Germany
| | - Patrick Orth
- Center of Experimental Orthopaedics, Saarland University, Homburg/Saar, Germany
| | - Gertrud Schmitt
- Center of Experimental Orthopaedics, Saarland University, Homburg/Saar, Germany
| | - David Zurakowski
- Departments of Anesthesia and Surgery, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University, Homburg/Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University, Homburg/Saar, Germany
| |
Collapse
|
2
|
Shestovskaya MV, Bozhkova SA, Sopova JV, Khotin MG, Bozhokin MS. Methods of Modification of Mesenchymal Stem Cells and Conditions of Their Culturing for Hyaline Cartilage Tissue Engineering. Biomedicines 2021; 9:biomedicines9111666. [PMID: 34829895 PMCID: PMC8615732 DOI: 10.3390/biomedicines9111666] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 12/24/2022] Open
Abstract
The use of mesenchymal stromal cells (MSCs) for tissue engineering of hyaline cartilage is a topical area of regenerative medicine that has already entered clinical practice. The key stage of this procedure is to create conditions for chondrogenic differentiation of MSCs, increase the synthesis of hyaline cartilage extracellular matrix proteins by these cells and activate their proliferation. The first such works consisted in the indirect modification of cells, namely, in changing the conditions in which they are located, including microfracturing of the subchondral bone and the use of 3D biodegradable scaffolds. The most effective methods for modifying the cell culture of MSCs are protein and physical, which have already been partially introduced into clinical practice. Genetic methods for modifying MSCs, despite their effectiveness, have significant limitations. Techniques have not yet been developed that allow studying the effectiveness of their application even in limited groups of patients. The use of MSC modification methods allows precise regulation of cell culture proliferation, and in combination with the use of a 3D biodegradable scaffold, it allows obtaining a hyaline-like regenerate in the damaged area. This review is devoted to the consideration and comparison of various methods used to modify the cell culture of MSCs for their use in regenerative medicine of cartilage tissue.
Collapse
Affiliation(s)
- Maria V. Shestovskaya
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
| | - Svetlana A. Bozhkova
- Vreden National Medical Research Center of Traumatology and Orthopedics, Academica Baykova Str., 8, 195427 St. Petersburg, Russia;
| | - Julia V. Sopova
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
- Center of Transgenesis and Genome Editing, St. Petersburg State University, Universitetskaja Emb., 7/9, 199034 St. Petersburg, Russia
| | - Mikhail G. Khotin
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
| | - Mikhail S. Bozhokin
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
- Vreden National Medical Research Center of Traumatology and Orthopedics, Academica Baykova Str., 8, 195427 St. Petersburg, Russia;
- Correspondence:
| |
Collapse
|
3
|
Wen C, Xu L, Xu X, Wang D, Liang Y, Duan L. Insulin-like growth factor-1 in articular cartilage repair for osteoarthritis treatment. Arthritis Res Ther 2021; 23:277. [PMID: 34717735 PMCID: PMC8556920 DOI: 10.1186/s13075-021-02662-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 10/17/2021] [Indexed: 11/10/2022] Open
Abstract
Articular cartilage repair is a critical issue in osteoarthritis (OA) treatment. The insulin-like growth factor (IGF) signaling pathway has been implicated in articular cartilage repair. IGF-1 is a member of a family of growth factors that are structurally closely related to pro-insulin and can promote chondrocyte proliferation, enhance matrix production, and inhibit chondrocyte apoptosis. Here, we reviewed the role of IGF-1 in cartilage anabolism and catabolism. Moreover, we discussed the potential role of IGF-1 in OA treatment. Of note, we summarized the recent progress on IGF delivery systems. Optimization of IGF delivery systems will facilitate treatment application in cartilage repair and improve OA treatment efficacy.
Collapse
Affiliation(s)
- Caining Wen
- Department of Orthopedics, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Limei Xu
- Department of Orthopedics, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Xiao Xu
- Department of Orthopedics, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Daping Wang
- Department of Orthopedics, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.,Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yujie Liang
- Department of Orthopedics, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China. .,Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, 518003, China.
| | - Li Duan
- Department of Orthopedics, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.
| |
Collapse
|
4
|
Azami M, Beheshtizadeh N. Identification of regeneration-involved growth factors in cartilage engineering procedure promotes its reconstruction. Regen Med 2021; 16:719-731. [PMID: 34287065 DOI: 10.2217/rme-2021-0028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Aim: To fabricate mature cartilage for implantation, developmental biological processes and proteins should be understood and employed. Methods: A systems biology study of all protein-coding genes participating in cartilage regeneration resulted in a network graph with 11 nodes and 28 edges. Gene ontology and centrality analysis were performed based on the degree index. Results: The four most crucial biological processes along with the seven most interactive proteins involved in cartilage regeneration were identified. Some proteins, which are under serious discussion in cartilage developmental and disease processes, are included in regeneration. Conclusions: Findings positively correlate with the literature, supporting the use of the four most impressive proteins as growth factors applicable to cartilage tissue engineering, including COL2A1, SOX9, CTGF and TGFβ1.
Collapse
Affiliation(s)
- Mahmoud Azami
- Department of Tissue Engineering & Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Regenerative Medicine group (REMED), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Nima Beheshtizadeh
- Department of Tissue Engineering & Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Regenerative Medicine group (REMED), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| |
Collapse
|
5
|
Maihöfer J, Madry H, Rey‐Rico A, Venkatesan JK, Goebel L, Schmitt G, Speicher‐Mentges S, Cai X, Meng W, Zurakowski D, Menger MD, Laschke MW, Cucchiarini M. Hydrogel-Guided, rAAV-Mediated IGF-I Overexpression Enables Long-Term Cartilage Repair and Protection against Perifocal Osteoarthritis in a Large-Animal Full-Thickness Chondral Defect Model at One Year In Vivo. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2008451. [PMID: 33734514 PMCID: PMC11468525 DOI: 10.1002/adma.202008451] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/02/2021] [Indexed: 06/12/2023]
Abstract
The regeneration of focal articular cartilage defects is complicated by the reduced quality of the repair tissue and the potential development of perifocal osteoarthritis (OA). Biomaterial-guided gene therapy may enhance cartilage repair by controlling the release of therapeutic sequences in a spatiotemporal manner. Here, the benefits of delivering a recombinant adeno-associated virus (rAAV) vector coding for the human insulin-like growth factor I (IGF-I) via an alginate hydrogel (IGF-I/AlgPH155) to enhance repair of full-thickness chondral defects following microfracture surgery after one year in minipigs versus control (lacZ/AlgPH155) treatment are reported. Sustained IGF-I overexpression is significantly achieved in the repair tissue of defects treated with IGF-I/AlgPH155 versus those receiving lacZ/AlgPH155 for one year and in the cartilage surrounding the defects. Administration of IGF-I/AlgPH155 significantly improves parameters of cartilage repair at one year relative to lacZ/AlgPH155 (semiquantitative total histological score, cell densities, matrix deposition) without deleterious or immune reactions. Remarkably, delivery of IGF-I/AlgPH155 also significantly reduces perifocal OA and inflammation after one year versus lacZ/AlgPH155 treatment. Biomaterial-guided rAAV gene transfer represents a valuable clinical approach to promote cartilage repair and to protect against OA.
Collapse
Affiliation(s)
- Johanna Maihöfer
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterD‐66421Homburg/SaarGermany
| | - Henning Madry
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterD‐66421Homburg/SaarGermany
| | - Ana Rey‐Rico
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterD‐66421Homburg/SaarGermany
| | - Jagadeesh K. Venkatesan
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterD‐66421Homburg/SaarGermany
| | - Lars Goebel
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterD‐66421Homburg/SaarGermany
| | - Gertrud Schmitt
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterD‐66421Homburg/SaarGermany
| | - Susanne Speicher‐Mentges
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterD‐66421Homburg/SaarGermany
| | - Xiaoyu Cai
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterD‐66421Homburg/SaarGermany
| | - Weikun Meng
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterD‐66421Homburg/SaarGermany
| | - David Zurakowski
- Departments of Anesthesia and SurgeryChildren's Hospital BostonHarvard Medical SchoolBostonMA02115USA
| | - Michael D. Menger
- Institute for Clinical and Experimental SurgerySaarland UniversityD‐66421Homburg/SaarGermany
| | - Matthias W. Laschke
- Institute for Clinical and Experimental SurgerySaarland UniversityD‐66421Homburg/SaarGermany
| | - Magali Cucchiarini
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterD‐66421Homburg/SaarGermany
| |
Collapse
|
6
|
Oláh T, Michaelis JC, Cai X, Cucchiarini M, Madry H. Comparative anatomy and morphology of the knee in translational models for articular cartilage disorders. Part II: Small animals. Ann Anat 2020; 234:151630. [PMID: 33129976 DOI: 10.1016/j.aanat.2020.151630] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 10/14/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND Small animal models are critical to model the complex disease mechanisms affecting a functional joint leading to articular cartilage disorders. They are advantageous for several reasons and significantly contributed to the understanding of the mechanisms of cartilage diseases among which osteoarthritis. METHODS Literature search in Pubmed. RESULTS AND DISCUSSION This narrative review summarizes the most relevant anatomical structural and functional characteristics of the knee (stifle) joints of the major small animal species, including mice, rats, guinea pigs, and rabbits compared with humans. Specific characteristics of each species, including kinematical gait parameters are provided and compared with the human situation. When placed in a proper context respecting their challenges and limitations, small animal models are important and appropriate models for articular cartilage disorders.
Collapse
Affiliation(s)
- Tamás Oláh
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany
| | | | - Xiaoyu Cai
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany; Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg, Germany.
| |
Collapse
|
7
|
Dual delivery of stem cells and insulin-like growth factor-1 in coacervate-embedded composite hydrogels for enhanced cartilage regeneration in osteochondral defects. J Control Release 2020; 327:284-295. [PMID: 32763434 DOI: 10.1016/j.jconrel.2020.08.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/07/2020] [Accepted: 08/02/2020] [Indexed: 12/31/2022]
Abstract
Exogenous dual delivery of progenitor cell population and therapeutic growth factors (GFs) is one of alternative tissue engineering strategies for osteochondral tissue regeneration. In the present study, an implantable dual delivery platform was developed using coacervates (Coa) (i.e., a tertiary complex of poly(ethylene argininylaspartate diglyceride) (PEAD) polycation, heparin, and cargo insulin-like growth factor-1 (IGF-1), in thiolated gelatin (gelatin-SH)/ poly(ethylene glycol) diacrylate (PEGDA) interpenetrating network (IPN) hydrogels. Since Coa is able to protect cargo GF and maintain its long-term bioactivity, it is speculated that Coa-mediated delivery of chondrogenic factor IGF-1 with the aid of adipose-derived stem cells (ADSCs) would synergistically facilitate osteochondral tissue repair during physiological regeneration process. Our results indicate that gelatin-SH/PEGDA IPN hydrogels demonstrated biocompatibility and mechanical properties for a possible long-term transplantation, and PEAD-base Coa exhibited a sustained release of bioactive IGF-1 over 3 weeks. Subsequently, released IGF-1 from Coa could effectively induce chondrogenic differentiation of embedded ADSCs in the hydrogel, by showing enhanced glycosaminoglycan deposition and expression of chondrogenesis-associated genes. More importantly, at 12 weeks post-implantation in a rabbit full thickness osteochondral defect model, the quality of regenerative tissues in both chondral and subchondral layers was significantly improved in dual delivery of ADSC and IGF-1 in Coa encapsulated in gelatin-SH/PEGDA IPN hydrogels, as compared with a single delivery of ADSC only and a dual delivery without Coa. Therefore, we conclude that our Coa-embedded composite hydrogel platform could effectively augment osteochondral tissue regeneration holds promise for a feasible osteoarthritis therapeutic application.
Collapse
|
8
|
Wang S, Liu R, Fu Y, Kao WJ. Release mechanisms and applications of drug delivery systems for extended-release. Expert Opin Drug Deliv 2020; 17:1289-1304. [PMID: 32619149 DOI: 10.1080/17425247.2020.1788541] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Drug delivery systems with extended-release profiles are ideal in improving patient compliance with enhanced efficacy. To develop devices capable of a prolonged delivery kinetics, it is crucial to understand the various underlying mechanisms contributing to extended drug release and the impact thereof on modulating the long-term performance of such systems in a practical application environment. AREAS COVERED This review article intends to provide a comprehensive summary of release mechanisms in extended-release drug delivery systems, particularly polymer-based systems; however, other material types will also be mentioned. Selected current research in the delivery of small molecule drugs and macromolecules is highlighted. Emphasis is placed on the combined impact of different release mechanisms and drug properties on the long-term release kinetics in vitro and in vivo. EXPERT OPINION The development of drug delivery systems over an extended duration is promising but also challenging when considering the numerous interrelated delivery-related parameters. Achieving a well-controlled extended drug release requires advanced techniques to minimize burst release and lag phase, a better understanding of the dynamic interrelationship between drug properties and release profiles over time, and a thorough elucidation of the impact of multiple in vivo conditions to methodically evaluate the eventual clinical efficacy.
Collapse
Affiliation(s)
- Shuying Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University , Chengdu, China
| | - Renhe Liu
- Global Health Drug Discovery Institute , Beijing, China
| | - Yao Fu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University , Chengdu, China
| | - W John Kao
- Department of Industrial and Manufacturing Systems Engineering, Biomedical Engineering Programme, Chemical Biology Centre, and Li Ka Shing Faculty of Medicine, The University of Hong Kong , Pokfulam, China
| |
Collapse
|
9
|
Muir SM, Reisbig N, Baria M, Kaeding C, Bertone AL. The Concentration of Plasma Provides Additional Bioactive Proteins in Platelet and Autologous Protein Solutions. Am J Sports Med 2019; 47:1955-1963. [PMID: 31125271 DOI: 10.1177/0363546519849671] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Currently, platelet-poor plasma (PPP) is a discarded waste product of platelet-rich plasma (PRP) and may contain valuable proteins. PURPOSE/HYPOTHESIS The study's goal was to evaluate the concentration of plasma as a potential additive biotherapy for the treatment of osteoarthritis. We hypothesized that a novel polyacrylamide concentration device would efficiently concentrate insulin-like growth factor-1 (IGF-1) from PPP and be additive to PRP or autologous protein solution (APS). STUDY DESIGN Descriptive laboratory study. METHODS A laboratory study was conducted with human and equine whole blood from healthy volunteers/donors. Fresh samples of blood and plasma were processed and characterized for platelet, white blood cell, and growth factor/cytokine content and then quantified by enzyme-linked immunosorbent assays specific for IGF-1, transforming growth factor-β, interleukin-1β, and interleukin-1 receptor antagonist as representatives of cartilage anabolic and inflammatory mediators. RESULTS A potent cartilage anabolic protein, IGF-1, was significantly concentrated by the polyacrylamide concentration device in both human and equine PPP. The polyacrylamide device also substantially increased plasma proteins over whole blood, most dramatically key proteins relevant to the treatment of osteoarthritis, including transforming growth factor-β (29-fold over blood) and interleukin-1 receptor antagonist (70-fold over plasma). CONCLUSION Concentrated PPP is a unique source for biologically relevant concentrations of IGF-1. PRP and APS can produce greater concentrations of other anabolic and anti-inflammatory proteins not found in plasma. CLINICAL RELEVANCE The polyacrylamide device efficiently concentrated PPP to create a unique source of IGF-1 that may supplement orthopaedic biologic therapies.
Collapse
Affiliation(s)
- Sean M Muir
- The Ohio State University, Columbus, Ohio, USA
| | | | | | | | | |
Collapse
|
10
|
Rey-Rico A, Cucchiarini M. Supramolecular Cyclodextrin-Based Hydrogels for Controlled Gene Delivery. Polymers (Basel) 2019; 11:polym11030514. [PMID: 30960498 PMCID: PMC6473339 DOI: 10.3390/polym11030514] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 03/15/2019] [Accepted: 03/17/2019] [Indexed: 12/27/2022] Open
Abstract
Controlled delivery of gene transfer vectors is a powerful strategy to enhance the temporal and spatial presentation of therapeutic agents in a defined target. Hydrogels are adapted biomaterials for gene delivery capable of acting as a localized depot of genes while maintaining the long term local availability of DNA vectors at a specific location. Supramolecular hydrogels based on cyclodextrins (CDs) have attracted considerable attention as potential biomaterials in a broad range of drug delivery applications. Their unique characteristics of thixotropicity and low cytotoxicity due to their production under mild conditions make them potential candidates to form injectable delivery systems. This work aims to provide an overview of the use of CD-based polypseudorotaxane hydrogels as controlled gene delivery systems for different applications in regenerative medicine.
Collapse
Affiliation(s)
- Ana Rey-Rico
- Cell Therapy and Regenerative Medicine Unit, Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña, Campus de A Coruña, 15071 A Coruña, Spain.
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, D-66421 Homburg/Saar, Germany.
| |
Collapse
|
11
|
Gonzalez-Fernandez T, Rathan S, Hobbs C, Pitacco P, Freeman FE, Cunniffe GM, Dunne NJ, McCarthy HO, Nicolosi V, O'Brien FJ, Kelly DJ. Pore-forming bioinks to enable spatio-temporally defined gene delivery in bioprinted tissues. J Control Release 2019; 301:13-27. [PMID: 30853527 DOI: 10.1016/j.jconrel.2019.03.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/02/2019] [Accepted: 03/06/2019] [Indexed: 12/17/2022]
Abstract
The regeneration of complex tissues and organs remains a major clinical challenge. With a view towards bioprinting such tissues, we developed a new class of pore-forming bioink to spatially and temporally control the presentation of therapeutic genes within bioprinted tissues. By blending sacrificial and stable hydrogels, we were able to produce bioinks whose porosity increased with time following printing. When combined with amphipathic peptide-based plasmid DNA delivery, these bioinks supported enhanced non-viral gene transfer to stem cells in vitro. By modulating the porosity of these bioinks, it was possible to direct either rapid and transient (pore-forming bioinks), or slower and more sustained (solid bioinks) transfection of host or transplanted cells in vivo. To demonstrate the utility of these bioinks for the bioprinting of spatially complex tissues, they were next used to zonally position stem cells and plasmids encoding for either osteogenic (BMP2) or chondrogenic (combination of TGF-β3, BMP2 and SOX9) genes within networks of 3D printed thermoplastic fibers to produce mechanically reinforced, gene activated constructs. In vivo, these bioprinted tissues supported the development of a vascularised, bony tissue overlaid by a layer of stable cartilage. When combined with multiple-tool biofabrication strategies, these gene activated bioinks can enable the bioprinting of a wide range of spatially complex tissues.
Collapse
Affiliation(s)
- T Gonzalez-Fernandez
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons, Ireland
| | - S Rathan
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - C Hobbs
- Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons, Ireland; School of Physics, Trinity College Dublin, Ireland; Centre for Research of Adaptive Nanostructures and Nanodevices, Trinity College Dublin, Ireland
| | - P Pitacco
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - F E Freeman
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - G M Cunniffe
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - N J Dunne
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons, Ireland; Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Ireland; School of Mechanical and Manufacturing Engineering, Dublin City University, Ireland; School of Pharmacy, Queen's University Belfast, UK
| | - H O McCarthy
- School of Pharmacy, Queen's University Belfast, UK
| | - V Nicolosi
- Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons, Ireland; School of Physics, Trinity College Dublin, Ireland; Centre for Research of Adaptive Nanostructures and Nanodevices, Trinity College Dublin, Ireland
| | - F J O'Brien
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons, Ireland; Tissue Engineering Research Group, Dept. of Anatomy, Royal College of Surgeons in, Ireland
| | - D J Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons, Ireland; Tissue Engineering Research Group, Dept. of Anatomy, Royal College of Surgeons in, Ireland.
| |
Collapse
|
12
|
Biomaterial-guided delivery of gene vectors for targeted articular cartilage repair. Nat Rev Rheumatol 2018; 15:18-29. [DOI: 10.1038/s41584-018-0125-2] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
13
|
Graceffa V, Vinatier C, Guicheux J, Evans CH, Stoddart M, Alini M, Zeugolis DI. State of art and limitations in genetic engineering to induce stable chondrogenic phenotype. Biotechnol Adv 2018; 36:1855-1869. [DOI: 10.1016/j.biotechadv.2018.07.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/16/2018] [Accepted: 07/12/2018] [Indexed: 12/18/2022]
|
14
|
Improved Chondrogenic Differentiation of rAAV SOX9-Modified Human MSCs Seeded in Fibrin-Polyurethane Scaffolds in a Hydrodynamic Environment. Int J Mol Sci 2018; 19:ijms19092635. [PMID: 30189664 PMCID: PMC6163252 DOI: 10.3390/ijms19092635] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 08/30/2018] [Accepted: 09/03/2018] [Indexed: 12/15/2022] Open
Abstract
The repair of focal articular cartilage defects remains a problem. Combining gene therapy with tissue engineering approaches using bone marrow-derived mesenchymal stem cells (MSCs) may allow the development of improved options for cartilage repair. Here, we examined whether a three-dimensional fibrin-polyurethane scaffold provides a favorable environment for the effective chondrogenic differentiation of human MSCs (hMSCs) overexpressing the cartilage-specific SOX9 transcription factor via recombinant adeno-associated virus (rAAV) -mediated gene transfer cultured in a hydrodynamic environment in vitro. Sustained SOX9 expression was noted in the constructs for at least 21 days, the longest time point evaluated. Such spatially defined SOX9 overexpression enhanced proliferative, metabolic, and chondrogenic activities compared with control (reporter lacZ gene transfer) treatment. Of further note, administration of the SOX9 vector was also capable of delaying premature hypertrophic and osteogenic differentiation in the constructs. This enhancement of chondrogenesis by spatially defined overexpression of human SOX9 demonstrate the potential benefits of using rAAV-modified hMSCs seeded in fibrin-polyurethane scaffolds as a promising approach for implantation in focal cartilage lesions to improve cartilage repair.
Collapse
|
15
|
Controlled Non-Viral Gene Delivery in Cartilage and Bone Repair: Current Strategies and Future Directions. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800038] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
16
|
da Silva Morais A, Oliveira JM, Reis RL. Small Animal Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1059:423-439. [DOI: 10.1007/978-3-319-76735-2_19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
17
|
Wu P, Chen H, Jin R, Weng T, Ho JK, You C, Zhang L, Wang X, Han C. Non-viral gene delivery systems for tissue repair and regeneration. J Transl Med 2018; 16:29. [PMID: 29448962 PMCID: PMC5815227 DOI: 10.1186/s12967-018-1402-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 02/07/2018] [Indexed: 12/11/2022] Open
Abstract
Critical tissue defects frequently result from trauma, burns, chronic wounds and/or surgery. The ideal treatment for such tissue loss is autografting, but donor sites are often limited. Tissue engineering (TE) is an inspiring alternative for tissue repair and regeneration (TRR). One of the current state-of-the-art methods for TRR is gene therapy. Non-viral gene delivery systems (nVGDS) have great potential for TE and have several advantages over viral delivery including lower immunogenicity and toxicity, better cell specificity, better modifiability, and higher productivity. However, there is no ideal nVGDS for TRR, hence, there is widespread research to improve their properties. This review introduces the basic principles and key aspects of commonly-used nVGDSs. We focus on recent advances in their applications, current challenges, and future directions.
Collapse
Affiliation(s)
- Pan Wu
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, 310009, China
| | - Haojiao Chen
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, 310009, China
| | - Ronghua Jin
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, 310009, China
| | - Tingting Weng
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, 310009, China
| | - Jon Kee Ho
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, 310009, China
| | - Chuangang You
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, 310009, China
| | - Liping Zhang
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, 310009, China
| | - Xingang Wang
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, 310009, China.
| | - Chunmao Han
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, 310009, China.
| |
Collapse
|
18
|
Liu R, Chen Y, Liu L, Gong Y, Wang M, Li S, Chen C, Yu B. Long-term delivery of rhIGF-1 from biodegradable poly(lactic acid)/hydroxyapatite@Eudragit double-layer microspheres for prevention of bone loss and articular degeneration in C57BL/6 mice. J Mater Chem B 2018; 6:3085-3095. [PMID: 32254343 DOI: 10.1039/c8tb00324f] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Insulin-like growth factor (IGF-1) has encouraged researchers to investigate its various potential therapeutic uses such as in the treatment of osteoporosis and repair of articular cartilage.
Collapse
Affiliation(s)
- Rui Liu
- Department of Orthopedics
- Zhujiang Hospital
- Southern Medical University
- Guangzhou 510282
- China
| | - Yan Chen
- Department of Ultrasonic Diagnosis
- Zhujiang Hospital
- Southern Medical University
- Guangzhou 510282
- China
| | - Lanlan Liu
- Key Laboratory of Biomedical Materials and Implant Devices
- Research Institute of Tsinghua University in Shenzhen
- Shenzhen 518057
- P. R. China
| | - Yong Gong
- Department of Orthopedics
- Zhujiang Hospital
- Southern Medical University
- Guangzhou 510282
- China
| | - Mingbo Wang
- Key Laboratory of Biomedical Materials and Implant Devices
- Research Institute of Tsinghua University in Shenzhen
- Shenzhen 518057
- P. R. China
| | - Songjian Li
- Department of Orthopedics
- Zhujiang Hospital
- Southern Medical University
- Guangzhou 510282
- China
| | - Changsheng Chen
- Key Laboratory of Biomedical Materials and Implant Devices
- Research Institute of Tsinghua University in Shenzhen
- Shenzhen 518057
- P. R. China
| | - Bo Yu
- Department of Orthopedics
- Zhujiang Hospital
- Southern Medical University
- Guangzhou 510282
- China
| |
Collapse
|
19
|
Generating Chondromimetic Mesenchymal Stem Cell Spheroids by Regulating Media Composition and Surface Coating. Cell Mol Bioeng 2017; 11:99-115. [PMID: 29623134 DOI: 10.1007/s12195-017-0517-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Spheroids of mesenchymal stem cells (MSCs) in cartilage tissue engineering have been shown to enhance regenerative potential owing to their 3D structure. In this study, we explored the possibility of priming spheroids under different media to replace the use of inductive surface coatings for chondrogenic differentiation. METHODS Rat bone marrow-derived MSCs were organized into cell spheroids by the hanging drop technique and subsequently cultured on hyaluronic acid (HA) coated or non-coated well plates under different cell media conditions. Endpoint analysis included cell viability, DNA and Glycosaminoglycan (GAG) and collagen content, gene expression and immunohistochemistry. RESULTS For chondrogenic applications, MSC spheroids derived on non-coated surfaces outperformed the spheroids derived from HA-coated surfaces in matrix synthesis and collagen II gene expression. Spheroids on non-coated surfaces gave rise to the highest collagen and GAG when primed with medium containing insulin-like growth factor (IGF) for 1 week during spheroid formation. Spheroids that were grown in chondroinductive raw material-inclusive media such as aggrecan or chondroitin sulfate exhibited the highest Collagen II gene expression in the non-coated surface at 1 week. CONCLUSION Media priming by growth factors and raw materials might be a more predictive influencer of chondrogenesis compared to inductive-surfaces. Such tailored bioactivity of the stem cell spheroids in the stage of the spheroid formation may give rise to a platform technology that may eventually produce spheroids capable of chondrogenesis achieved by mere media manipulation, skipping the need for additional culture on a modified surface, that paves the way for cost-effective technologies.
Collapse
|
20
|
Betz VM, Keller A, Foehr P, Thirion C, Salomon M, Rammelt S, Zwipp H, Burgkart R, Jansson V, Müller PE, Betz OB. BMP-2 gene activated muscle tissue fragments for osteochondral defect regeneration in the rabbit knee. J Gene Med 2017; 19. [PMID: 28744947 DOI: 10.1002/jgm.2972] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 07/19/2017] [Accepted: 07/19/2017] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Previously published data indicate that BMP-2 gene activated muscle tissue grafts can repair large bone defects in rats. This innovative abbreviated ex vivo gene therapy is appealing because it does not require elaborative and time-consuming extraction and expansion of cells. Hence, in the present study, we evaluated the potential of this expedited tissue engineering approach for regenerating osteochondral defects in rabbits. METHODS Autologous muscle tissue grafts from female White New Zealand rabbits were directly transduced with an adenoviral BMP-2 vector or remained unmodified. Osteochondral defects in the medial condyle of rabbit knees were treated with either BMP-2 activated muscle tissue implants or unmodified muscle tissue or remained empty. After 13 weeks, repair of osteochondral defects was examined by biomechanical indentation testing and by histology/imunohistochemistry applying an extended O'Driscoll scoring system and histomorphometry. RESULTS Biomechanical investigations revealed a trend towards slightly improved mechanical properties of the group receiving BMP-2 activated muscle tissue compared to unmodified muscle treatment and empty defect controls. However, a statistically significant difference was noted only between BMP-2 muscle and unmodified muscle treatment. Also, histological evaluation resulted in slightly higher histological scores and improved collagen I/II ratio without statistical significance in the BMP-2 treatment group. Histomorphometry indicated enhanced repair of subchondral bone after treatment with BMP-2 muscle, with a significantly larger bone area compared to untreated defects. CONCLUSIONS Gene activated muscle tissue grafts showed potential for osteochondral defect repair. There is room for improvement via the use of appropriate growth factor combinations.
Collapse
Affiliation(s)
- Volker M Betz
- University Center of Orthopaedics and Traumatology and Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus Dresden, Technical University Dresden, Dresden, Germany
| | - Alexander Keller
- Department of Orthopaedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Peter Foehr
- Department of Orthopaedics and Sportsorthopaedics, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | | | | | - Stefan Rammelt
- University Center of Orthopaedics and Traumatology and Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus Dresden, Technical University Dresden, Dresden, Germany.,DFG-Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Hans Zwipp
- University Center of Orthopaedics and Traumatology and Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus Dresden, Technical University Dresden, Dresden, Germany.,DFG-Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Rainer Burgkart
- Department of Orthopaedics and Sportsorthopaedics, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Volkmar Jansson
- Department of Orthopaedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Peter E Müller
- Department of Orthopaedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Oliver B Betz
- Department of Orthopaedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
21
|
IGF-1 Gene Transfer to Human Synovial MSCs Promotes Their Chondrogenic Differentiation Potential without Induction of the Hypertrophic Phenotype. Stem Cells Int 2017; 2017:5804147. [PMID: 28740513 PMCID: PMC5504993 DOI: 10.1155/2017/5804147] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 05/07/2017] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem cell- (MSC-) based therapy is a promising treatment for cartilage. However, repair tissue in general fails to regenerate an original hyaline-like tissue. In this study, we focused on increasing the expression levels for insulin-like growth factor-1 (IGF-1) to improve repair tissue quality. The IGF-1 gene was introduced into human synovial MSCs with a lentiviral vector and examined the levels of gene expression and morphological status of MSCs under chondrogenic differentiation condition using pellet cultures. The size of the pellets derived from IGF-1-MSCs were significantly larger than those of the control group. The abundance of glycosaminoglycan (GAG) was also significantly higher in the IGF-1-MSC group. The histology of the IGF-1-induced pellets demonstrated similarities to hyaline cartilage without exhibiting features of a hypertrophic chondrocyte phenotype. Expression levels for the Col2A1 gene and protein were significantly higher in the IGF-1 pellets than in the control pellets, but expression levels for Col10, MMP-13, ALP, and Osterix were not higher. Thus, IGF-1 gene transfer to human synovial MSCs led to an improved chondrogenic differentiation capacity without the detectable induction of a hypertrophic or osteogenic phenotype.
Collapse
|
22
|
Venkatesan JK, Frisch J, Rey-Rico A, Schmitt G, Madry H, Cucchiarini M. Impact of mechanical stimulation on the chondrogenic processes in human bone marrow aspirates modified to overexpress sox9 via rAAV vectors. J Exp Orthop 2017. [PMID: 28634835 PMCID: PMC5478551 DOI: 10.1186/s40634-017-0097-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Evaluation of gene-based approaches to target human bone marrow aspirates in conditions of mechanical stimulation that aim at reproducing the natural joint environment may allow to develop improved treatments for articular cartilage injuries. In the present study, we investigated the potential of rAAV-mediated sox9 gene transfer to enhance the chondrogenic differentiation processes in human bone marrow aspirates under established hydrodynamic conditions compared with the more commonly employed static culture conditions. Methods Fresh human bone marrow aspirates were transduced with rAAV-FLAG-hsox9 (40 μl) and maintained for up to 28 days in chondrogenic medium under mechanically-induced conditions in dynamic flow rotating bioreactors that permit tissue growth and matrix deposition relative to static culture conditions. The samples were then processed to examine the potential effects of sox9 overexpression on the cellular activities (matrix synthesis, proliferation) and on the chondrogenic differentiation potency compared with control treatments (absence of rAAV vector; reporter rAAV-lacZ, rAAV-RFP, and rAAV-luc gene transfer). Results Prolonged, significant sox9 overexpression via rAAV was achieved in the aspirates for at least 28 days when applying the rAAV-FLAG-hsox9 construct, leading to higher, prolonged levels of matrix biosynthesis and to enhanced chondrogenic activities relative to control treatments especially when maintaining the samples under mechanical stimulation. Administration of sox9 however did not impact the indices of proliferation in the aspirates. Remarkably, sox9 gene transfer also durably delayed hypertrophic and osteogenic differentiation in the samples regardless of the conditions of culture applied versus control treatments. Conclusions The current observations show the value of genetically modifying human bone marrow aspirates upon mechanical stimulation by rAAV sox9 as a promising strategy for future treatments to improve cartilage repair by implantation in lesions where the tissue is submitted to natural mechanical forces.
Collapse
Affiliation(s)
- Jagadeesh K Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Janina Frisch
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Ana Rey-Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Gertrud Schmitt
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany.,Department of Orthopaedic Surgery, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany.
| |
Collapse
|
23
|
Li K, Zhang C, Qiu L, Gao L, Zhang X. Advances in Application of Mechanical Stimuli in Bioreactors for Cartilage Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2017; 23:399-411. [PMID: 28463576 DOI: 10.1089/ten.teb.2016.0427] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Articular cartilage (AC) is the weight-bearing tissue in diarthroses. It lacks the capacity for self-healing once there are injuries or diseases due to its avascularity. With the development of tissue engineering, repairing cartilage defects through transplantation of engineered cartilage that closely matches properties of native cartilage has become a new option for curing cartilage diseases. The main hurdle for clinical application of engineered cartilage is how to develop functional cartilage constructs for mass production in a credible way. Recently, impressive hyaline cartilage that may have the potential to provide capabilities for treating large cartilage lesions in the future has been produced in laboratories. The key to functional cartilage construction in vitro is to identify appropriate mechanical stimuli. First, they should ensure the function of metabolism because mechanical stimuli play the role of blood vessels in the metabolism of AC, for example, acquiring nutrition and removing wastes. Second, they should mimic the movement of synovial joints and produce phenotypically correct tissues to achieve the adaptive development between the micro- and macrostructure and function. In this article, we divide mechanical stimuli into three types according to forces transmitted by different media in bioreactors, namely forces transmitted through the liquid medium, solid medium, or other media, then we review and summarize the research status of bioreactors for cartilage tissue engineering (CTE), mainly focusing on the effects of diverse mechanical stimuli on engineered cartilage. Based on current researches, there are several motion patterns in knee joints; but compression, tension, shear, fluid shear, or hydrostatic pressure each only partially reflects the mechanical condition in vivo. In this study, we propose that rolling-sliding-compression load consists of various stimuli that will represent better mechanical environment in CTE. In addition, engineers often ignore the importance of biochemical factors to the growth and development of engineered cartilage. In our point of view, only by fully considering synergistic effects of mechanical and biochemical factors can we find appropriate culture conditions for functional cartilage constructs. Once again, rolling-sliding-compression load under appropriate biochemical conditions may be conductive to realize the adaptive development between the structure and function of engineered cartilage in vitro.
Collapse
Affiliation(s)
- Ke Li
- Tianjin Key Laboratory of Design and Intelligent Control of the Advanced Mechatronical System, School of Mechanical Engineering, Tianjin University of Technology , Tianjin, China
| | - Chunqiu Zhang
- Tianjin Key Laboratory of Design and Intelligent Control of the Advanced Mechatronical System, School of Mechanical Engineering, Tianjin University of Technology , Tianjin, China
| | - Lulu Qiu
- Tianjin Key Laboratory of Design and Intelligent Control of the Advanced Mechatronical System, School of Mechanical Engineering, Tianjin University of Technology , Tianjin, China
| | - Lilan Gao
- Tianjin Key Laboratory of Design and Intelligent Control of the Advanced Mechatronical System, School of Mechanical Engineering, Tianjin University of Technology , Tianjin, China
| | - Xizheng Zhang
- Tianjin Key Laboratory of Design and Intelligent Control of the Advanced Mechatronical System, School of Mechanical Engineering, Tianjin University of Technology , Tianjin, China
| |
Collapse
|
24
|
Zhang Z, Li L, Yang W, Cao Y, Shi Y, Li X, Zhang Q. The effects of different doses of IGF-1 on cartilage and subchondral bone during the repair of full-thickness articular cartilage defects in rabbits. Osteoarthritis Cartilage 2017; 25:309-320. [PMID: 27662821 DOI: 10.1016/j.joca.2016.09.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 09/08/2016] [Accepted: 09/13/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To investigate the effects of different doses of insulin-like growth factor 1 (IGF-1) on the cartilage layer and subchondral bone (SB) during repair of full-thickness articular cartilage (AC) defects. DESIGN IGF-1-loaded collagen membrane was implanted into full-thickness AC defects in rabbits. The effects of two different doses of IGF-1 on cartilage layer and SB adjacent to the defect, the cartilage structure, formation and integration, and the new SB formation were evaluated at the 1st, 4th and 8th week postoperation. Meanwhile, after 1 week treatment, the relative mRNA expressions in tissues adjacent to the defect, including cartilage and SB were determined by quantitative real-time RT-PCR (qRT-PCR), respectively. RESULTS Different doses of IGF-1 induced different gene expression profiles in tissues adjacent to the defect and resulted in different repair outcomes. Particularly, at high dose IGF-1 aided cell survival, regulated the gene expressions in cartilage layer adjacent defect and altered ECM composition more effectively, improved the formation and integrity of neo-cartilage. While, at low dose IGF-1 regulated the gene expressions in SB more efficaciously and subsequently promoted the SB remodeling and reconstruction. CONCLUSION Different doses of IGF-1 induced different responses of cartilage or SB during the repair of full-thickness AC defects. Particularly, high dose of IGF-1 was more beneficial to the neo-cartilage formation and integration, while low dose of it was more effective for the SB formation.
Collapse
Affiliation(s)
- Z Zhang
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences, Peking Union Medical College, The Key Laboratory of Biomedical Material of Tianjin, Tianjin 300192, PR China.
| | - L Li
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences, Peking Union Medical College, The Key Laboratory of Biomedical Material of Tianjin, Tianjin 300192, PR China.
| | - W Yang
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences, Peking Union Medical College, The Key Laboratory of Biomedical Material of Tianjin, Tianjin 300192, PR China.
| | - Y Cao
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences, Peking Union Medical College, The Key Laboratory of Biomedical Material of Tianjin, Tianjin 300192, PR China.
| | - Y Shi
- School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin 300384, PR China.
| | - X Li
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences, Peking Union Medical College, The Key Laboratory of Biomedical Material of Tianjin, Tianjin 300192, PR China.
| | - Q Zhang
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences, Peking Union Medical College, The Key Laboratory of Biomedical Material of Tianjin, Tianjin 300192, PR China.
| |
Collapse
|
25
|
Osteochondral Tissue Engineering and Regenerative Strategies. REGENERATIVE STRATEGIES FOR THE TREATMENT OF KNEE JOINT DISABILITIES 2017. [DOI: 10.1007/978-3-319-44785-8_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
26
|
Chen J, Yuan Z, Liu Y, Zheng R, Dai Y, Tao R, Xia H, Liu H, Zhang Z, Zhang W, Liu W, Cao Y, Zhou G. Improvement of In Vitro Three-Dimensional Cartilage Regeneration by a Novel Hydrostatic Pressure Bioreactor. Stem Cells Transl Med 2016; 6:982-991. [PMID: 28297584 PMCID: PMC5442788 DOI: 10.5966/sctm.2016-0118] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 08/10/2016] [Indexed: 12/21/2022] Open
Abstract
In vitro three‐dimensional (3D) cartilage regeneration is a promising strategy for repair of cartilage defects. However, inferior mechanical strength and tissue homogeneity greatly restricted its clinical translation. Simulation of mechanical stress through a bioreactor is an important approach for improving in vitro cartilage regeneration. The current study developed a hydrostatic pressure (HP) bioreactor based on a novel pressure‐transmitting mode achieved by slight deformation of a flexible membrane in a completely sealed stainless steel device. The newly developed bioreactor efficiently avoided the potential risks of previously reported pressure‐transmitting modes and simultaneously addressed a series of important issues, such as pressure scopes, culture chamber sizes, sealability, contamination control, and CO2 balance. The whole bioreactor system realized stable long‐term (8 weeks) culture under high HP (5–10 MPa) without the problems of medium leakage and contamination. Furthermore, the results of in vitro 3D tissue culture based on a cartilage regeneration model revealed that HP provided by the newly developed bioreactor efficiently promoted in vitro 3D cartilage formation by improving its mechanical strength, thickness, and homogeneity. Detailed analysis in cell proliferation, cartilage matrix production, and cross‐linking level of collagen macromolecules, as well as density and alignment of collagen fibers, further revealed the possible mechanisms that HP regulated in vitro cartilage regeneration. The current study provided a highly efficient and stable bioreactor system for improving in vitro 3D cartilage regeneration and thus will help to accelerate its clinical translation. Stem Cells Translational Medicine2017;6:982–991
Collapse
Affiliation(s)
- Jie Chen
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- National Tissue Engineering Center of China, Shanghai, People's Republic of China
- Department of Anesthesiology, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Zhaoyuan Yuan
- Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, Shandong, People's Republic of China
| | - Yu Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- National Tissue Engineering Center of China, Shanghai, People's Republic of China
| | - Rui Zheng
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- National Tissue Engineering Center of China, Shanghai, People's Republic of China
| | - Yao Dai
- College of Materials Science and Engineering, Hunan University, Changsha, Hunan, People's Republic of China
| | - Ran Tao
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- National Tissue Engineering Center of China, Shanghai, People's Republic of China
| | - Huitang Xia
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- National Tissue Engineering Center of China, Shanghai, People's Republic of China
- Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, Shandong, People's Republic of China
| | - Hairong Liu
- College of Materials Science and Engineering, Hunan University, Changsha, Hunan, People's Republic of China
| | - Zhiyong Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- National Tissue Engineering Center of China, Shanghai, People's Republic of China
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- National Tissue Engineering Center of China, Shanghai, People's Republic of China
| | - Wei Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- National Tissue Engineering Center of China, Shanghai, People's Republic of China
| | - Yilin Cao
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- National Tissue Engineering Center of China, Shanghai, People's Republic of China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- National Tissue Engineering Center of China, Shanghai, People's Republic of China
- Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, Shandong, People's Republic of China
| |
Collapse
|
27
|
Bardsley K, Kwarciak A, Freeman C, Brook I, Hatton P, Crawford A. Repair of bone defects in vivo using tissue engineered hypertrophic cartilage grafts produced from nasal chondrocytes. Biomaterials 2016; 112:313-323. [PMID: 27770634 DOI: 10.1016/j.biomaterials.2016.10.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 10/03/2016] [Accepted: 10/11/2016] [Indexed: 10/20/2022]
Abstract
The regeneration of large bone defects remains clinically challenging. The aim of our study was to use a rat model to use nasal chondrocytes to engineer a hypertrophic cartilage tissue which could be remodelled into bone in vivo by endochondral ossification. Primary adult rat nasal chondrocytes were isolated from the nasal septum, the cell numbers expanded in monolayer culture and the cells cultured in vitro on polyglycolic acid scaffolds in chondrogenic medium for culture periods of 5-10 weeks. Hypertrophic differentiation was assessed by determining the temporal expression of key marker genes and proteins involved in hypertrophic cartilage formation. The temporal changes in the genes measured reflected the temporal changes observed in the growth plate. Collagen II gene expression increased 6 fold by day 7 and was then significantly downregulated from day 14 onwards. Conversely, collagen X gene expression was detectable by day 14 and increased 100-fold by day 35. The temporal increase in collagen X expression was mirrored by increases in alkaline phosphatase gene expression which also was detectable by day 14 with a 30-fold increase in gene expression by day 35. Histological and immunohistochemical analysis of the engineered constructs showed increased chondrocyte cell volume (31-45 μm), deposition of collagen X in the extracellular matrix and expression of alkaline phosphatase activity. However, no cartilage mineralisation was observed in in vitro culture of up to 10 weeks. On subcutaneous implantation of the hypertrophic engineered constructs, the grafts became vascularised, cartilage mineralisation occurred and loss of the proteoglycan in the matrix was observed. Implantation of the hypertrophic engineered constructs into a rat cranial defect resulted in angiogenesis, mineralisation and remodelling of the cartilage tissue into bone. Micro-CT analysis indicated that defects which received the engineered hypertrophic constructs showed 38.48% in bone volume compared to 7.01% in the control defects. Development of tissue engineered hypertrophic cartilage to use as a bone graft substitute is an exciting development in regenerative medicine. This is a proof of principal study demonstrating the potential of nasal chondrocytes to engineer hypertrophic cartilage which will remodel into bone on in vivo transplantation. This approach to making engineered hypertrophic cartilage grafts could form the basis of a new potential future clinical treatment for maxillofacial reconstruction.
Collapse
Affiliation(s)
- Katie Bardsley
- School of Clinical Dentistry, University of Sheffield, 19 Claremont Crescent, Sheffield, South Yorkshire, S10 2TA, UK; Guy Hilton Research Centre, University of Keele, Staffordshire, ST4 7QB, UK
| | - Agnieska Kwarciak
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials & Methodology, Aviation House, 125 Kingsway, London, WC2B 6NH, UK
| | - Christine Freeman
- School of Clinical Dentistry, University of Sheffield, 19 Claremont Crescent, Sheffield, South Yorkshire, S10 2TA, UK
| | - Ian Brook
- School of Clinical Dentistry, University of Sheffield, 19 Claremont Crescent, Sheffield, South Yorkshire, S10 2TA, UK
| | - Paul Hatton
- School of Clinical Dentistry, University of Sheffield, 19 Claremont Crescent, Sheffield, South Yorkshire, S10 2TA, UK
| | - Aileen Crawford
- School of Clinical Dentistry, University of Sheffield, 19 Claremont Crescent, Sheffield, South Yorkshire, S10 2TA, UK.
| |
Collapse
|
28
|
Hydrogel-Based Controlled Delivery Systems for Articular Cartilage Repair. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1215263. [PMID: 27642587 PMCID: PMC5011507 DOI: 10.1155/2016/1215263] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 07/12/2016] [Indexed: 12/19/2022]
Abstract
Delivery of bioactive factors is a very valuable strategy for articular cartilage repair. Nevertheless, the direct supply of such biomolecules is limited by several factors including rapid degradation, the need for supraphysiological doses, the occurrence of immune and inflammatory responses, and the possibility of dissemination to nontarget sites that may impair their therapeutic action and raise undesired effects. The use of controlled delivery systems has the potential of overcoming these hurdles by promoting the temporal and spatial presentation of such factors in a defined target. Hydrogels are promising materials to develop delivery systems for cartilage repair as they can be easily loaded with bioactive molecules controlling their release only where required. This review exposes the most recent technologies on the design of hydrogels as controlled delivery platforms of bioactive molecules for cartilage repair.
Collapse
|
29
|
Zhou Q, Li B, Zhao J, Pan W, Xu J, Chen S. IGF-I induces adipose derived mesenchymal cell chondrogenic differentiation in vitro and enhances chondrogenesis in vivo. In Vitro Cell Dev Biol Anim 2016; 52:356-364. [PMID: 26822434 DOI: 10.1007/s11626-015-9969-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 10/16/2015] [Indexed: 12/29/2022]
Abstract
Recent studies have demonstrated that insulin-like growth factor-1 (IGF-I) modulates bone mesenchymal stem cell chondrogenic differentiation independent of transforming growth factor beta (TGF-β) signaling in vitro. However, it is unclear whether IGF-I can solely modulate human adipose-derived mesenchymal cell (hAMC) chondrogenic differentiation, or whether it has additive effects with TGF-β1 to induce chondrogenic differentiation in vitro and development of mature cartilage in vivo. We investigated the effect of IGF-I on the induction of hAMC chondrogenic differentiation in the presence or absence of transforming growth factor beta 1 (TGF-β1) in vitro, and chondrogenesis of the induced hAMC in vivo. The results showed that IGF-I alone induced collagen type II, aggrecan, and Sox9 mRNA expression and collagen type II and aggrecan proteins expressions in hAMCs. Notably, there was greater mRNA expression of collagen type II, aggrecan and Sox9, and greater protein expression of collagen type II and aggrecan following TGF-β1 + IGF-I treatment, compared to either TGF-β1 or IGF-I-treated hAMCs. These results were confirmed in cartilage tissues derived from induced hAMCs. These findings indicate that IGF-I alone has the ability to induce chondrogenic differentiation and has additive effects with TGF-β1 to induce chondrogenic differentiation in vitro and in vivo.
Collapse
Affiliation(s)
- Quan Zhou
- Department of Orthopaedics, Huai'an Hospital Affiliated of Xuzhou Medical College and Huai'an Second Hospital, No. 62 Huaihai Road South, Huai'an, 223002, China
| | - Baojun Li
- Department of Joint Surgery, Second People's Hospital of Hunan Province, Changsha, 410007, China
| | - Jiali Zhao
- Department of Orthopaedics, Huai'an Hospital Affiliated of Xuzhou Medical College and Huai'an Second Hospital, No. 62 Huaihai Road South, Huai'an, 223002, China
| | - Wei Pan
- Department of Orthopaedics, Huai'an Hospital Affiliated of Xuzhou Medical College and Huai'an Second Hospital, No. 62 Huaihai Road South, Huai'an, 223002, China
| | - Jin Xu
- Department of Orthopaedics, Huai'an Hospital Affiliated of Xuzhou Medical College and Huai'an Second Hospital, No. 62 Huaihai Road South, Huai'an, 223002, China
| | - Sumei Chen
- Department of Orthopaedics, Huai'an Hospital Affiliated of Xuzhou Medical College and Huai'an Second Hospital, No. 62 Huaihai Road South, Huai'an, 223002, China.
| |
Collapse
|
30
|
Madry H, Ochi M, Cucchiarini M, Pape D, Seil R. Large animal models in experimental knee sports surgery: focus on clinical translation. J Exp Orthop 2015; 2:9. [PMID: 26914877 PMCID: PMC4545948 DOI: 10.1186/s40634-015-0025-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/25/2015] [Indexed: 02/06/2023] Open
Abstract
Large animal models play a crucial role in sports surgery of the knee, as they are critical for the exploration of new experimental strategies and the clinical translation of novel techniques. The purpose of this contribution is to provide critical aspects of relevant animal models in this field, with a focus on paediatric anterior cruciate ligament (ACL) reconstruction, high tibial osteotomy, and articular cartilage repair. Although there is no single large animal model strictly replicating the human knee joint, the sheep stifle joint shares strong similarities. Studies in large animal models of paediatric ACL reconstruction identified specific risk factors associated with the different surgical techniques. The sheep model of high tibial osteotomy is a powerful new tool to advance the understanding of the effect of axial alignment on the lower extremity on specific issues of the knee joint. Large animal models of both focal chondral and osteochondral defects and of osteoarthritis have brought new findings about the mechanisms of cartilage repair and treatment options. The clinical application of a magnetic device for targeted cell delivery serves as a suitable example of how data from such animal models are directly translated into in clinical cartilage repair. As novel insights from studies in these translational models will advance the basic science, close cooperation in this important field of clinical translation will improve current reconstructive surgical options and open novel avenues for regenerative therapies of musculoskeletal disorders.
Collapse
Affiliation(s)
- Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University, Bldg 37, Kirrbergerstr. 1, D-66421, Homburg, Germany.
- Cartilage Net of the Greater Region, Homburg, Germany.
- Department of Orthopaedic Surgery, Saarland University Medical Center and Saarland University, D-66421, Homburg/Saar, Germany.
| | - Mitsuo Ochi
- Department of Orthopaedic Surgery, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan.
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University, Bldg 37, Kirrbergerstr. 1, D-66421, Homburg, Germany.
- Cartilage Net of the Greater Region, Homburg, Germany.
| | - Dietrich Pape
- Cartilage Net of the Greater Region, Homburg, Germany.
- Department of Orthopaedic Surgery, Centre Hospitalier du Luxembourg, L-1460, Luxembourg, Luxembourg.
- Sports Medicine Research Laboratory, Public Research Centre for Health, Luxembourg, Centre Médical de la Fondation Norbert Metz, 76 rue d'Eich, L-1460, Luxembourg, Luxembourg.
| | - Romain Seil
- Cartilage Net of the Greater Region, Homburg, Germany.
- Department of Orthopaedic Surgery, Centre Hospitalier du Luxembourg, L-1460, Luxembourg, Luxembourg.
- Sports Medicine Research Laboratory, Public Research Centre for Health, Luxembourg, Centre Médical de la Fondation Norbert Metz, 76 rue d'Eich, L-1460, Luxembourg, Luxembourg.
| |
Collapse
|
31
|
Steinmetz NJ, Aisenbrey EA, Westbrook KK, Qi HJ, Bryant SJ. Mechanical loading regulates human MSC differentiation in a multi-layer hydrogel for osteochondral tissue engineering. Acta Biomater 2015; 21:142-53. [PMID: 25900444 DOI: 10.1016/j.actbio.2015.04.015] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Revised: 04/11/2015] [Accepted: 04/13/2015] [Indexed: 12/15/2022]
Abstract
A bioinspired multi-layer hydrogel was developed for the encapsulation of human mesenchymal stem cells (hMSCs) as a platform for osteochondral tissue engineering. The spatial presentation of biochemical cues, via incorporation of extracellular matrix analogs, and mechanical cues, via both hydrogel crosslink density and externally applied mechanical loads, were characterized in each layer. A simple sequential photopolymerization method was employed to form stable poly(ethylene glycol)-based hydrogels with a soft cartilage-like layer of chondroitin sulfate and low RGD concentrations, a stiff bone-like layer with high RGD concentrations, and an intermediate interfacial layer. Under a compressive load, the variation in hydrogel stiffness within each layer produced high strains in the soft cartilage-like layer, low strains in the stiff bone-like layer, and moderate strains in the interfacial layer. When hMSC-laden hydrogels were cultured statically in osteochondral differentiation media, the local biochemical and matrix stiffness cues were not sufficient to spatially guide hMSC differentiation after 21 days. However dynamic mechanical stimulation led to differentially high expression of collagens with collagen II in the cartilage-like layer, collagen X in the interfacial layer and collagen I in the bone-like layer and mineral deposits localized to the bone layer. Overall, these findings point to external mechanical stimulation as a potent regulator of hMSC differentiation toward osteochondral cellular phenotypes.
Collapse
|
32
|
Madry H, Cucchiarini M. Tissue-engineering strategies to repair joint tissue in osteoarthritis: nonviral gene-transfer approaches. Curr Rheumatol Rep 2015; 16:450. [PMID: 25182678 DOI: 10.1007/s11926-014-0450-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Loss of articular cartilage is a common clinical consequence of osteoarthritis (OA). In the past decade, substantial progress in tissue engineering, nonviral gene transfer, and cell transplantation have provided the scientific foundation for generating cartilaginous constructs from genetically modified cells. Combining tissue engineering with overexpression of therapeutic genes enables immediate filling of a cartilage defect with an engineered construct that actively supports chondrogenesis. Several pioneering studies have proved that spatially defined nonviral overexpression of growth-factor genes in constructs of solid biomaterials or hydrogels is advantageous compared with gene transfer or scaffold alone, both in vitro and in vivo. Notably, these investigations were performed in models of focal cartilage defects, because advanced cartilage-repair strategies based on the principles of tissue engineering have not advanced sufficiently to enable resurfacing of extensively degraded cartilage as therapy for OA. These studies serve as prototypes for future technological developments, because they raise the possibility that cartilage constructs engineered from genetically modified chondrocytes providing autocrine and paracrine stimuli could similarly compensate for the loss of articular cartilage in OA. Because cartilage-tissue-engineering strategies are already used in the clinic, combining tissue engineering and nonviral gene transfer could prove a powerful approach to treat OA.
Collapse
Affiliation(s)
- Henning Madry
- Center of Experimental Orthopaedics and Department of Orthopaedic Surgery, Saarland University, 66421, Homburg, Germany,
| | | |
Collapse
|
33
|
Li KC, Hu YC. Cartilage tissue engineering: recent advances and perspectives from gene regulation/therapy. Adv Healthc Mater 2015; 4:948-68. [PMID: 25656682 DOI: 10.1002/adhm.201400773] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 01/10/2015] [Indexed: 12/16/2022]
Abstract
Diseases in articular cartilages affect millions of people. Despite the relatively simple biochemical and cellular composition of articular cartilages, the self-repair ability of cartilage is limited. Successful cartilage tissue engineering requires intricately coordinated interactions between matrerials, cells, biological factors, and phycial/mechanical factors, and still faces a multitude of challenges. This article presents an overview of the cartilage biology, current treatments, recent advances in the materials, biological factors, and cells used in cartilage tissue engineering/regeneration, with strong emphasis on the perspectives of gene regulation (e.g., microRNA) and gene therapy.
Collapse
Affiliation(s)
- Kuei-Chang Li
- Department of Chemical Engineering; National Tsing Hua University; Hsinchu Taiwan 300
| | - Yu-Chen Hu
- Department of Chemical Engineering; National Tsing Hua University; Hsinchu Taiwan 300
| |
Collapse
|
34
|
Cucchiarini M, Henrionnet C, Mainard D, Pinzano A, Madry H. New trends in articular cartilage repair. J Exp Orthop 2015; 2:8. [PMID: 26914876 PMCID: PMC4544617 DOI: 10.1186/s40634-015-0026-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/25/2015] [Indexed: 12/31/2022] Open
Abstract
Damage to the articular cartilage is an important, prevalent, and unsolved clinical issue for the orthopaedic surgeon. This review summarizes innovative basic research approaches that may improve the current understanding of cartilage repair processes and lead to novel therapeutic options. In this regard, new aspects of cartilage tissue engineering with a focus on the choice of the best-suited cell source are presented. The importance of non-destructive cartilage imaging is highlighted with the recent availability of adapted experimental tools such as Second Harmonic Generation (SHG) imaging. Novel insights into cartilage pathophysiology based on the involvement of the infrapatellar fat pad in osteoarthritis are also described. Also, recombinant adeno-associated viral vectors are discussed as clinically adapted, efficient tools for potential gene-based medicines in a variety of articular cartilage disorders. Taken as a whole, such advances in basic research in diverse fields of articular cartilage repair may lead to the development of improved therapies in the clinics for an improved, effective treatment of cartilage lesions in a close future.
Collapse
Affiliation(s)
- Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University, Bldg 37, Kirrbergerstr. 1, D-66421, Homburg, Germany. .,Cartilage Net of the Greater Region, Homburg, Germany.
| | - Christel Henrionnet
- Cartilage Net of the Greater Region, Homburg, Germany. .,Ingénierie Moléculaire et Physiopathologie Articulaire, Unité Mixte de Recherches 7365 Centre National de la Recherche Scientifique, Université de Lorraine, F-54505, Vandoeuvre Lès Nancy, France.
| | - Didier Mainard
- Cartilage Net of the Greater Region, Homburg, Germany. .,Ingénierie Moléculaire et Physiopathologie Articulaire, Unité Mixte de Recherches 7365 Centre National de la Recherche Scientifique, Université de Lorraine, F-54505, Vandoeuvre Lès Nancy, France.
| | - Astrid Pinzano
- Cartilage Net of the Greater Region, Homburg, Germany. .,Ingénierie Moléculaire et Physiopathologie Articulaire, Unité Mixte de Recherches 7365 Centre National de la Recherche Scientifique, Université de Lorraine, F-54505, Vandoeuvre Lès Nancy, France.
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University, Bldg 37, Kirrbergerstr. 1, D-66421, Homburg, Germany. .,Cartilage Net of the Greater Region, Homburg, Germany. .,Department of Orthopaedic Surgery, Saarland University Medical Center and Saarland University, D-66421, Homburg/Saar, Germany.
| |
Collapse
|
35
|
Yan LP, Oliveira JM, Oliveira AL, Reis RL. Current Concepts and Challenges in Osteochondral Tissue Engineering and Regenerative Medicine. ACS Biomater Sci Eng 2015; 1:183-200. [DOI: 10.1021/ab500038y] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Le-Ping Yan
- 3B’s
Research Group−Biomaterials, Biodegradables and Biomimetics,
Headquarters of the European Institute of Excellence on Tissue Engineering
and Regenerative Medicine, University of Minho, AvePark, S. Cláudio
de Barco, 4806-909 Taipas, Guimarães, Portugal
- ICVS/3B’s−PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joaquim M. Oliveira
- 3B’s
Research Group−Biomaterials, Biodegradables and Biomimetics,
Headquarters of the European Institute of Excellence on Tissue Engineering
and Regenerative Medicine, University of Minho, AvePark, S. Cláudio
de Barco, 4806-909 Taipas, Guimarães, Portugal
- ICVS/3B’s−PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana L. Oliveira
- 3B’s
Research Group−Biomaterials, Biodegradables and Biomimetics,
Headquarters of the European Institute of Excellence on Tissue Engineering
and Regenerative Medicine, University of Minho, AvePark, S. Cláudio
de Barco, 4806-909 Taipas, Guimarães, Portugal
- ICVS/3B’s−PT Government Associate Laboratory, Braga/Guimarães, Portugal
- CBQF−Center
for Biotechnology and Fine Chemistry, School of Biotechnology, Portuguese Catholic University, Porto 4200−072, Portugal
| | - Rui L. Reis
- 3B’s
Research Group−Biomaterials, Biodegradables and Biomimetics,
Headquarters of the European Institute of Excellence on Tissue Engineering
and Regenerative Medicine, University of Minho, AvePark, S. Cláudio
de Barco, 4806-909 Taipas, Guimarães, Portugal
- ICVS/3B’s−PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
36
|
Mullen LM, Best SM, Ghose S, Wardale J, Rushton N, Cameron RE. Bioactive IGF-1 release from collagen-GAG scaffold to enhance cartilage repair in vitro. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2015; 26:5325. [PMID: 25577208 PMCID: PMC4289525 DOI: 10.1007/s10856-014-5325-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 07/20/2014] [Indexed: 05/28/2023]
Abstract
Tissue engineering is a promising technique for cartilage repair. Toward this goal, a porous collagen-glycosaminoglycan (CG) scaffold was loaded with different concentrations of insulin-like growth factor-1 (IGF-1) and evaluated as a growth factor delivery device. The biological response was assessed by monitoring the amount of type II collagen and proteoglycan synthesised by the chondrocytes seeded within the scaffolds. IGF-1 release was dependent on the IGF-1 loading concentration used to adsorb IGF-1 onto the CG scaffolds and the amount of IGF-1 released into the media was highest at day 4. This initial IGF-1 release could be modelled using linear regression analysis. Osteoarthritic (OA) chondrocytes seeded within scaffolds containing adsorbed IGF-1 deposited decorin and type II collagen in a dose dependent manner and the highest type II collagen deposition was achieved via loading the scaffold with 50 μg/ml IGF-1. Cells seeded within the IGF-1 loaded scaffolds also deposited more extracellular matrix than the no growth factor control group thus the IGF-1 released from the scaffold remained bioactive and exerted an anabolic effect on OA chondrocytes. The effectiveness of adsorbing IGF-1 onto the scaffold may be due to protection of the molecule from proteolytic digestion allowing a more sustained release of IGF-1 over time compared to adding multiple doses of exogenous growth factor. Incorporating IGF-1 into the CG scaffold provided an initial therapeutic burst release of IGF-1 which is beneficial in initiating ECM deposition and repair in this in vitro model and shows potential for developing this delivery device in vivo.
Collapse
Affiliation(s)
- Leanne M Mullen
- Department of Materials Science and Metallurgy, University of Cambridge, Pembroke Street, Cambridge, CB2 3QZ, UK,
| | | | | | | | | | | |
Collapse
|
37
|
Madeira C, Santhagunam A, Salgueiro JB, Cabral JM. Advanced cell therapies for articular cartilage regeneration. Trends Biotechnol 2015; 33:35-42. [DOI: 10.1016/j.tibtech.2014.11.003] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 09/29/2014] [Accepted: 11/07/2014] [Indexed: 01/25/2023]
|
38
|
Liu Y, Wang DA. Viral vector-mediated transgenic cell therapy in regenerative medicine: safety of the process. Expert Opin Biol Ther 2014; 15:559-67. [DOI: 10.1517/14712598.2015.995086] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
39
|
Zhao R, Peng X, Li Q, Song W. Effects of phosphorylatable short peptide-conjugated chitosan-mediated IL-1Ra and igf-1 gene transfer on articular cartilage defects in rabbits. PLoS One 2014; 9:e112284. [PMID: 25390659 PMCID: PMC4229204 DOI: 10.1371/journal.pone.0112284] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 10/10/2014] [Indexed: 11/25/2022] Open
Abstract
Previously, we reported an improvement in the transfection efficiency of the plasmid DNA-chitosan (pDNA/CS) complex by the utilization of phosphorylatable short peptide-conjugated chitosan (pSP-CS). In this study, we investigated the effects of pSP-CS-mediated gene transfection of interleukin-1 receptor antagonist protein (IL-1Ra) combined with insulin-like growth factor-1 (IGF-1) in rabbit chondrocytes and in a rabbit model of cartilage defects. pBudCE4.1-IL-1Ra+igf-1, pBudCE4.1-IL-1Ra and pBudCE4.1-igf-1 were constructed and combined with pSP-CS to form pDNA/pSP-CS complexes. These complexes were transfected into rabbit primary chondrocytes or injected into the joint cavity. Seven weeks after treatment, all rabbits were sacrificed and analyzed. High levels of IL-1Ra and igf-1 expression were detected both in the cell culture supernatant and in the synovial fluid. In vitro, the transgenic complexes caused significant proliferation of chondrocytes, promotion of glycosaminoglycan (GAG) and collagen II synthesis, and inhibition of chondrocyte apoptosis and nitric oxide (NO) synthesis. In vivo, the exogenous genes resulted in increased collagen II synthesis and reduced NO and GAG concentrations in the synovial fluid; histological studies revealed that pDNA/pSP-CS treatment resulted in varying degrees of hyaline-like cartilage repair and Mankin score decrease. The co-expression of both genes produced greater effects than each single gene alone both in vitro and in vivo. The results suggest that pSP-CS is a good candidate for use in gene therapy for the treatment of cartilage defects and that igf-1 and IL-1Ra co-expression produces promising biologic effects on cartilage defects.
Collapse
Affiliation(s)
- Ronglan Zhao
- Department of Medical Laboratory, Shandong Provincial Key Laboratory of Clinical Laboratory Diagnostics, Weifang Medical University, Weifang, Shandong, China
| | - Xiaoxiang Peng
- Department of Medical Laboratory, Shandong Provincial Key Laboratory of Clinical Laboratory Diagnostics, Weifang Medical University, Weifang, Shandong, China
- * E-mail:
| | - Qian Li
- Department of Medical Laboratory, Shandong Provincial Key Laboratory of Clinical Laboratory Diagnostics, Weifang Medical University, Weifang, Shandong, China
| | - Wei Song
- Department of Medical Laboratory, Shandong Provincial Key Laboratory of Clinical Laboratory Diagnostics, Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
40
|
Evans C. Using genes to facilitate the endogenous repair and regeneration of orthopaedic tissues. INTERNATIONAL ORTHOPAEDICS 2014; 38:1761-9. [PMID: 25038968 DOI: 10.1007/s00264-014-2423-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Accepted: 06/09/2014] [Indexed: 10/25/2022]
Abstract
Traditional tissue engineering approaches to the restoration of orthopaedic tissues promise to be expensive and not well suited to treating large numbers of patients. Advances in gene transfer technology offer the prospect of developing expedited techniques in which all manipulations can be performed percutaneously or in a single operation. This rests on the ability of gene delivery to provoke the sustained synthesis of relevant gene products in situ without further intervention. Regulated gene expression is also possible, but its urgency is reduced by our ignorance of exactly what levels and periods of expression are needed for specific gene products. This review describes various strategies by which gene therapy can be used to expedite the repair and regeneration of orthopaedic tissues. Strategies include the direct injection of vectors into sites of injury, the use of genetically modified, allogeneic cell lines and the intra-operative harvest of autologous tissues that are quickly transduced and returned to the body, either intact or following rapid cell isolation. Data obtained from pre-clinical experiments in animal models have provided much encouragement that such approaches may eventually find clinical application in human and veterinary medicine.
Collapse
Affiliation(s)
- Christopher Evans
- Rehabilitation Medicine Research Center, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA,
| |
Collapse
|
41
|
Overexpression of human IGF-I via direct rAAV-mediated gene transfer improves the early repair of articular cartilage defects in vivo. Gene Ther 2014; 21:811-9. [PMID: 24989812 DOI: 10.1038/gt.2014.58] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 05/06/2014] [Accepted: 05/28/2014] [Indexed: 11/09/2022]
Abstract
Direct therapeutic gene transfer is a promising tool to treat articular cartilage defects. Here, we tested the ability of an recombinant adeno-associated virus (rAAV) insulin-like growth factor I (IGF-I) vector to improve the early repair of cartilage lesions in vivo. The vector was administered for 3 weeks in osteochondral defects created in the knee joints of rabbits compared with control (lacZ) treatment and in cells that participate in the repair processes (mesenchymal stem cells, chondrocytes). Efficient IGF-I expression was observed in the treated lesions and in isolated cells in vitro. rAAV-mediated IGF-I overexpression was capable of stimulating the biologic activities (proliferation, matrix synthesis) both in vitro and in vivo. IGF-I treatment in vivo was well tolerated, revealing significant improvements of the repair capabilities of the entire osteochondral unit. IGF-I overexpression delayed terminal differentiation and hypertrophy in the newly formed cartilage, possibly due to contrasting effects upon the osteogenic expression of RUNX2 and β-catenin and to stimulating effects of this factor on the parathyroid hormone/parathyroid hormone-related protein pathway in this area. Production of IGF-I improved the reconstitution of the subchondral bone layer in the defects, showing increased RUNX2 expression levels in this zone. These findings show the potential of directly applying therapeutic rAAVs to treat cartilage lesions.
Collapse
|
42
|
Lohan A, Marzahn U, El Sayed K, Haisch A, Müller RD, Kohl B, Stölzel K, Ertel W, John T, Schulze-Tanzil G. Osteochondral articular defect repair using auricle-derived autologous chondrocytes in a rabbit model. Ann Anat 2014; 196:317-26. [PMID: 24812031 DOI: 10.1016/j.aanat.2014.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 03/07/2014] [Accepted: 03/08/2014] [Indexed: 12/29/2022]
Abstract
Hypothesizing that the implantation of non-articular (heterotopic) chondrocytes might be an alternative approach to support articular cartilage repair, we analyzed joint cartilage defect healing in the rabbit model after implantation of autologous auricle-derived (auricular) chondrocytes. Autologous lapine articular and auricular chondrocytes were cultured for 3 weeks in polyglycolic acid (PGA) scaffolds before being implanted into critical sized osteochondral defects of the rabbit knee femoropatellar groove. Cell-free PGA scaffolds and empty defects served as controls. Construct quality was determined before implantation and defect healing was monitored after 6 and 12 weeks using vitality assays, macroscopical and histological score systems. Neo-cartilage was formed in the PGA constructs seeded with both articular and auricular chondrocytes in vitro and in vivo. At the histological level, cartilage repair was slightly improved when using autologous articular chondrocyte seeded constructs compared to empty defects and was significantly superior compared to defects treated with auricular chondrocytes 6 weeks after implantation. Although only the immunohistological differences were significant, auricular chondrocyte implantation induced an inferior healing response compared with the empty defects. Elastic auricular chondrocytes might maintain some tissue-specific characteristics when implanted into joint cartilage defects which limit its repair capacity.
Collapse
Affiliation(s)
- Anke Lohan
- Department of Orthopaedic, Trauma and Reconstructive Surgery, Charité-University of Medicine, Campus Benjamin Franklin, Berlin, Germany.
| | - Ulrike Marzahn
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-University of Medicine, Campus Benjamin Franklin, Berlin, Germany
| | - Karym El Sayed
- Department of Orthopaedic, Trauma and Reconstructive Surgery, Charité-University of Medicine, Campus Benjamin Franklin, Berlin, Germany
| | - Andreas Haisch
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-University of Medicine, Campus Benjamin Franklin, Berlin, Germany
| | - Riccarda Dolores Müller
- Department of Orthopaedic, Trauma and Reconstructive Surgery, Charité-University of Medicine, Campus Benjamin Franklin, Berlin, Germany
| | - Benjamin Kohl
- Department of Orthopaedic, Trauma and Reconstructive Surgery, Charité-University of Medicine, Campus Benjamin Franklin, Berlin, Germany
| | - Katharina Stölzel
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-University of Medicine, Campus Benjamin Franklin, Berlin, Germany
| | - Wolfgang Ertel
- Department of Orthopaedic, Trauma and Reconstructive Surgery, Charité-University of Medicine, Campus Benjamin Franklin, Berlin, Germany
| | - Thilo John
- Department of Orthopaedic, Trauma and Reconstructive Surgery, Charité-University of Medicine, Campus Benjamin Franklin, Berlin, Germany
| | - Gundula Schulze-Tanzil
- Department of Orthopaedic, Trauma and Reconstructive Surgery, Charité-University of Medicine, Campus Benjamin Franklin, Berlin, Germany
| |
Collapse
|
43
|
Scaffold-mediated lentiviral transduction for functional tissue engineering of cartilage. Proc Natl Acad Sci U S A 2014; 111:E798-806. [PMID: 24550481 DOI: 10.1073/pnas.1321744111] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The ability to develop tissue constructs with matrix composition and biomechanical properties that promote rapid tissue repair or regeneration remains an enduring challenge in musculoskeletal engineering. Current approaches require extensive cell manipulation ex vivo, using exogenous growth factors to drive tissue-specific differentiation, matrix accumulation, and mechanical properties, thus limiting their potential clinical utility. The ability to induce and maintain differentiation of stem cells in situ could bypass these steps and enhance the success of engineering approaches for tissue regeneration. The goal of this study was to generate a self-contained bioactive scaffold capable of mediating stem cell differentiation and formation of a cartilaginous extracellular matrix (ECM) using a lentivirus-based method. We first showed that poly-L-lysine could immobilize lentivirus to poly(ε-caprolactone) films and facilitate human mesenchymal stem cell (hMSC) transduction. We then demonstrated that scaffold-mediated gene delivery of transforming growth factor β3 (TGF-β3), using a 3D woven poly(ε-caprolactone) scaffold, induced robust cartilaginous ECM formation by hMSCs. Chondrogenesis induced by scaffold-mediated gene delivery was as effective as traditional differentiation protocols involving medium supplementation with TGF-β3, as assessed by gene expression, biochemical, and biomechanical analyses. Using lentiviral vectors immobilized on a biomechanically functional scaffold, we have developed a system to achieve sustained transgene expression and ECM formation by hMSCs. This method opens new avenues in the development of bioactive implants that circumvent the need for ex vivo tissue generation by enabling the long-term goal of in situ tissue engineering.
Collapse
|