1
|
Rajankunte Mahadeshwara M, Al-Jawad M, Hall RM, Pandit H, El-Gendy R, Bryant M. How Do Cartilage Lubrication Mechanisms Fail in Osteoarthritis? A Comprehensive Review. Bioengineering (Basel) 2024; 11:541. [PMID: 38927777 PMCID: PMC11200606 DOI: 10.3390/bioengineering11060541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/02/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Cartilage degeneration is a characteristic of osteoarthritis (OA), which is often observed in aging populations. This degeneration is due to the breakdown of articular cartilage (AC) mechanical and tribological properties primarily attributed to lubrication failure. Understanding the reasons behind these failures and identifying potential solutions could have significant economic and societal implications, ultimately enhancing quality of life. This review provides an overview of developments in the field of AC, focusing on its mechanical and tribological properties. The emphasis is on the role of lubrication in degraded AC, offering insights into its structure and function relationship. Further, it explores the fundamental connection between AC mechano-tribological properties and the advancement of its degradation and puts forth recommendations for strategies to boost its lubrication efficiency.
Collapse
Affiliation(s)
- Manoj Rajankunte Mahadeshwara
- Institute of Functional Surfaces, Mechanical Engineering, University of Leeds, Leeds LS2 9JT, UK
- Department of Oral Biology, Faculty of Dentistry, University of Leeds, Leeds LS2 9JT, UK; (M.A.-J.); (R.E.-G.)
| | - Maisoon Al-Jawad
- Department of Oral Biology, Faculty of Dentistry, University of Leeds, Leeds LS2 9JT, UK; (M.A.-J.); (R.E.-G.)
| | - Richard M. Hall
- School of Engineering, College of Engineering and Physical Sciences, University of Birmingham, Birmingham B15 2TT, UK;
| | - Hemant Pandit
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Chapel Allerton Hospital, Leeds LS7 4SA, UK;
| | - Reem El-Gendy
- Department of Oral Biology, Faculty of Dentistry, University of Leeds, Leeds LS2 9JT, UK; (M.A.-J.); (R.E.-G.)
- Department of Oral Pathology, Faculty of Dentistry, Suez Canal University, Ismailia 3, Ismailia Governorate 8366004, Egypt
| | - Michael Bryant
- Institute of Functional Surfaces, Mechanical Engineering, University of Leeds, Leeds LS2 9JT, UK
- School of Engineering, College of Engineering and Physical Sciences, University of Birmingham, Birmingham B15 2TT, UK;
| |
Collapse
|
2
|
Nasiri N, Hosseini S, Alini M, Khademhosseini A, Baghaban Eslaminejad M. Targeted cell delivery for articular cartilage regeneration and osteoarthritis treatment. Drug Discov Today 2019; 24:2212-2224. [DOI: 10.1016/j.drudis.2019.07.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/31/2019] [Accepted: 07/31/2019] [Indexed: 12/17/2022]
|
3
|
Khorraminejad-Shirazi M, Dorvash M, Estedlal A, Hoveidaei AH, Mazloomrezaei M, Mosaddeghi P. Aging: A cell source limiting factor in tissue engineering. World J Stem Cells 2019; 11:787-802. [PMID: 31692986 PMCID: PMC6828594 DOI: 10.4252/wjsc.v11.i10.787] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/03/2019] [Accepted: 09/05/2019] [Indexed: 02/06/2023] Open
Abstract
Tissue engineering has yet to reach its ideal goal, i.e. creating profitable off-the-shelf tissues and organs, designing scaffolds and three-dimensional tissue architectures that can maintain the blood supply, proper biomaterial selection, and identifying the most efficient cell source for use in cell therapy and tissue engineering. These are still the major challenges in this field. Regarding the identification of the most appropriate cell source, aging as a factor that affects both somatic and stem cells and limits their function and applications is a preventable and, at least to some extents, a reversible phenomenon. Here, we reviewed different stem cell types, namely embryonic stem cells, adult stem cells, induced pluripotent stem cells, and genetically modified stem cells, as well as their sources, i.e. autologous, allogeneic, and xenogeneic sources. Afterward, we approached aging by discussing the functional decline of aged stem cells and different intrinsic and extrinsic factors that are involved in stem cell aging including replicative senescence and Hayflick limit, autophagy, epigenetic changes, miRNAs, mTOR and AMPK pathways, and the role of mitochondria in stem cell senescence. Finally, various interventions for rejuvenation and geroprotection of stem cells are discussed. These interventions can be applied in cell therapy and tissue engineering methods to conquer aging as a limiting factor, both in original cell source and in the in vitro proliferated cells.
Collapse
Affiliation(s)
- Mohammadhossein Khorraminejad-Shirazi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Cell and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Mohammadreza Dorvash
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Cell and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
| | - Alireza Estedlal
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Cell and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Amir Human Hoveidaei
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Mohsen Mazloomrezaei
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Cell and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Pouria Mosaddeghi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Cell and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
| |
Collapse
|
4
|
Abstract
The study objectives include, enhancing the proliferations of aged bone marrow stem cells (BMSCs) and adipose stem cells (ADSCs); and evaluating the shelf lives of clinical grade chondrogenically induced cells from both samples. ADSCs and BMSCs from 56 patients (76 ± 8 yrs) were proliferated using basal medium (FD) and at (5, 10, 15, 20 and 25) ng/ml of basal fibroblast growth factor (bFGF). They were induced to chondrogenic lineage and stored for more than 120 hrs in FD, serum, Dulbecco’s phosphate buffered saline (DPBS) and saline at 4 °C. In FD, cells stagnated and BMSCs’ population doubling time (PDT) was 137 ± 30 hrs, while ADSCs’ was 129.7 ± 40 hrs. bFGF caused PDT’s decrease to 24.5 ± 5.8 hrs in BMSCs and 22.0 ± 6.5 hrs in ADSCs (p = 0.0001). Both cells were positive to stem cell markers before inductions and thereafter, expressed significantly high chondrogenic genes (p = 0.0001). On shelf life, both cells maintained viabilities and counts above 70% in FD and serum after 120 hrs. BMSCs’ viabilities in DPBS fell below 70% after 96 hrs and saline after 72 hrs. ADSCs’ viability fell below 70% in DPBS after 24 hrs and saline within 24 hrs. Concentrations between 20 ng/ml bFGF is ideal for aged adult cells’ proliferation and delivery time of induced BMSCs and ADSCs can be 120 hrs in 4 °C serum.
Collapse
|
5
|
Gabner S, Hlavaty J, Velde K, Renner M, Jenner F, Egerbacher M. Inflammation-induced transgene expression in genetically engineered equine mesenchymal stem cells. J Gene Med 2018; 18:154-64. [PMID: 27272202 DOI: 10.1002/jgm.2888] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 05/31/2016] [Accepted: 05/31/2016] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Osteoarthritis, a chronic and progressive degenerative joint disorder, ranks amongst the top five causes of disability. Given the high incidence, associated socioeconomic costs and the absence of effective disease-modifying therapies of osteoarthritis, cell-based treatments offer a promising new approach. Owing to their paracrine, differentiation and self-renewal abilities, mesenchymal stem cells (MSCs) have great potential for regenerative medicine, which might be further enhanced by targeted gene therapy. Hence, the development of systems allowing transgene expression, particularly when regulated by natural disease-dependent occuring substances, is of high interest. METHODS Bone marrow-isolated equine MSCs were stably transduced with an HIV-1 based lentiviral vector expressing the luciferase gene under control of an inducible nuclear factor κB (NFκB)-responsive promoter. Marker gene expression was analysed by determining luciferase activity in transduced cells stimulated with different concentrations of interleukin (IL)-1β or tumour necrosis factor (TNF)α. RESULTS A dose-dependent increase in luciferase expression was observed in transduced MSCs upon cytokine stimulation. The induction effect was more potent in cells treated with TNFα compared to those treated with IL-1β. Maximum transgene expression was obtained after 48 h of stimulation and the same time was necessary to return to baseline luciferase expression levels after withdrawal of the stimulus. Repeated cycles of induction allowed on-off modulation of transgene expression without becoming refractory to induction. The NFκB-responsive promoter retained its inducibility also in chondrogenically differentiated MSC/Luc cells. CONCLUSIONS The results of the present study demonstrate that on demand transgene expression from the NFκB-responsive promoter using naturally occurring inflammatory cytokines can be induced in undifferentiated and chondrogenically differentiated equine MSCs. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Simone Gabner
- Institute of Anatomy, Histology and Embryology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Juraj Hlavaty
- Institute of Anatomy, Histology and Embryology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Karsten Velde
- Equine University Hospital, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Matthias Renner
- Division of Medical Biotechnology, Paul-Ehrlich-Institut, Langen, Germany
| | - Florien Jenner
- Equine University Hospital, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Monika Egerbacher
- Institute of Anatomy, Histology and Embryology, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
6
|
Wang W, Guo H, Li H, Yan Y, Wu C, Wang X, He X, Zhao N. Interleukin-35 Gene-Modified Mesenchymal Stem Cells Protect Concanavalin A-Induced Fulminant Hepatitis by Decreasing the Interferon Gamma Level. Hum Gene Ther 2017; 29:234-241. [PMID: 29054137 DOI: 10.1089/hum.2017.171] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Interleukin 35 (IL-35) is a relatively newly identified cytokine required for the regulatory and suppressive functions of regulatory T cells (Treg), playing an important role in the prevention of autoimmune diseases. This study used mesenchymal stem cells (MSCs) as the gene-delivery vehicles for IL-35 gene therapy and investigated their protective effects in Concanavalin A (Con A)-induced autoimmune hepatitis. Results showed that IL-35 gene modified MSCs (IL-35-MSCs) can specifically migrate to the injured liver tissues and significantly narrow the necrosis areas of injured livers. IL-35-MSCs prevented hepatocyte apoptosis by reducing the FASL expression by mononuclear cells. Although MSC treatment can alleviate liver injury to some extent, IL-35-MSCs showed a stronger protective effect, which means some novel mechanisms exist. The results show that IL-35-MSCs could decrease the level of interferon gamma secreted by liver mononuclear cells through the JAK1-STAT1/STAT4 signal pathway. In summary, this study thus demonstrates a novel and efficient treatment for Con A-induced fulminant hepatitis through negatively regulating the secretion of interferon gamma, thus providing a novel therapeutic approach for this devastating liver disease.
Collapse
Affiliation(s)
- Wei Wang
- Department of General Surgery, Tianjin Medical University General Hospital , Tianjin, China
| | - Hao Guo
- Department of General Surgery, Tianjin Medical University General Hospital , Tianjin, China
| | - Hongyue Li
- Department of General Surgery, Tianjin Medical University General Hospital , Tianjin, China
| | - Yongjia Yan
- Department of General Surgery, Tianjin Medical University General Hospital , Tianjin, China
| | - Chao Wu
- Department of General Surgery, Tianjin Medical University General Hospital , Tianjin, China
| | - Xiaodong Wang
- Department of General Surgery, Tianjin Medical University General Hospital , Tianjin, China
| | - Xianghui He
- Department of General Surgery, Tianjin Medical University General Hospital , Tianjin, China
| | - Na Zhao
- Department of General Surgery, Tianjin Medical University General Hospital , Tianjin, China
| |
Collapse
|
7
|
Lolli A, Penolazzi L, Narcisi R, van Osch GJVM, Piva R. Emerging potential of gene silencing approaches targeting anti-chondrogenic factors for cell-based cartilage repair. Cell Mol Life Sci 2017; 74:3451-3465. [PMID: 28434038 PMCID: PMC11107620 DOI: 10.1007/s00018-017-2531-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/14/2017] [Accepted: 04/19/2017] [Indexed: 12/18/2022]
Abstract
The field of cartilage repair has exponentially been growing over the past decade. Here, we discuss the possibility to achieve satisfactory regeneration of articular cartilage by means of human mesenchymal stem cells (hMSCs) depleted of anti-chondrogenic factors and implanted in the site of injury. Different types of molecules including transcription factors, transcriptional co-regulators, secreted proteins, and microRNAs have recently been identified as negative modulators of chondroprogenitor differentiation and chondrocyte function. We review the current knowledge about these molecules as potential targets for gene knockdown strategies using RNA interference (RNAi) tools that allow the specific suppression of gene function. The critical issues regarding the optimization of the gene silencing approach as well as the delivery strategies are discussed. We anticipate that further development of these techniques will lead to the generation of implantable hMSCs with enhanced potential to regenerate articular cartilage damaged by injury, disease, or aging.
Collapse
Affiliation(s)
- Andrea Lolli
- Department of Orthopaedics, Erasmus MC, University Medical Center, 3015 CN, Rotterdam, The Netherlands.
| | - Letizia Penolazzi
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Roberto Narcisi
- Department of Orthopaedics, Erasmus MC, University Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Gerjo J V M van Osch
- Department of Orthopaedics, Erasmus MC, University Medical Center, 3015 CN, Rotterdam, The Netherlands
- Department of Otorhinolaryngology, Erasmus MC, University Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Roberta Piva
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
8
|
Minicircle Mediated Gene Delivery to Canine and Equine Mesenchymal Stem Cells. Int J Mol Sci 2017; 18:ijms18040819. [PMID: 28417917 PMCID: PMC5412403 DOI: 10.3390/ijms18040819] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 04/03/2017] [Accepted: 04/10/2017] [Indexed: 12/31/2022] Open
Abstract
Gene-directed tissue repair offers the clinician, human or veterinary, the chance to enhance cartilage regeneration and repair at a molecular level. Non-viral plasmid vectors have key biosafety advantages over viral vector systems for regenerative therapies due to their episomal integration however, conventional non-viral vectors can suffer from low transfection efficiency. Our objective was to identify and validate in vitro a novel non-viral gene expression vector that could be utilized for ex vivo and in vivo delivery to stromal-derived mesenchymal stem cells (MSCs). Minicircle plasmid DNA vector containing green fluorescent protein (GFP) was generated and transfected into adipose-derived MSCs from three species: canine, equine and rodent and transfection efficiency was determined. Both canine and rat cells showed transfection efficiencies of approximately 40% using minicircle vectors with equine cells exhibiting lower transfection efficiency. A Sox9-expressing minicircle vector was generated and transfected into canine MSCs. Successful transfection of the minicircle-Sox9 vector was confirmed in canine cells by Sox9 immunostaining. This study demonstrate the application and efficacy of a novel non-viral expression vector in canine and equine MSCs. Minicircle vectors have potential use in gene-directed regenerative therapies in non-rodent animal models for treatment of cartilage injury and repair.
Collapse
|
9
|
Tan AR, Hung CT. Concise Review: Mesenchymal Stem Cells for Functional Cartilage Tissue Engineering: Taking Cues from Chondrocyte-Based Constructs. Stem Cells Transl Med 2017; 6:1295-1303. [PMID: 28177194 PMCID: PMC5442836 DOI: 10.1002/sctm.16-0271] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 12/21/2016] [Indexed: 01/01/2023] Open
Abstract
Osteoarthritis, the most prevalent form of joint disease, afflicts 9% of the U.S. population over the age of 30 and costs the economy nearly $100 billion annually in healthcare and socioeconomic costs. It is characterized by joint pain and dysfunction, though the pathophysiology remains largely unknown. Due to its avascular nature and limited cellularity, articular cartilage exhibits a poor intrinsic healing response following injury. As such, significant research efforts are aimed at producing engineered cartilage as a cell-based approach for articular cartilage repair. However, the knee joint is mechanically demanding, and during injury, also a milieu of harsh inflammatory agents. The unforgiving mechano-chemical environment requires tissue replacements that are capable of bearing such burdens. The use of mesenchymal stem cells (MSCs) for cartilage tissue engineering has emerged as a promising cell source due to their ease of isolation, capacity to readily expand in culture, and ability to undergo lineage-specific differentiation into chondrocytes. However, to date, very few studies utilizing MSCs have successfully recapitulated the structural and functional properties of native cartilage, exposing the difficult process of uniformly differentiating stem cells into desired cell fates and maintaining the phenotype during in vitro culture and after in vivo implantation. To address these shortcomings, here, we present a concise review on modulating stem cell behavior, tissue development and function using well-developed techniques from chondrocyte-based cartilage tissue engineering. Stem Cells Translational Medicine 2017;6:1295-1303.
Collapse
|
10
|
The superior regenerative potential of muscle-derived stem cells for articular cartilage repair is attributed to high cell survival and chondrogenic potential. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16065. [PMID: 27990446 PMCID: PMC5129874 DOI: 10.1038/mtm.2016.65] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 06/22/2016] [Indexed: 11/08/2022]
Abstract
Three populations of muscle-derived cells (PP1, PP3, and PP6) were isolated from mouse skeletal muscle using modified preplate technique and retrovirally transduced with BMP4/GFP. In vitro, the PP1 cells (fibroblasts) proliferated significantly slower than the PP3 (myoblasts) and PP6 cells (muscle-derived stem cells); the PP1 and PP6 cells showed a superior rate of survival compared with PP3 cells under oxidative stress; and the PP6 cells showed significantly superior chondrogenic capabilities than PP1 and PP3 cells. In vivo, the PP6 cells promoted superior cartilage regeneration compared with the other muscle-derived cell populations. The cartilage defects in the PP6 group had significantly higher histological scores than those of the other muscle-derived cell groups, and GFP detection revealed that the transplanted PP6 cells showed superior in vivo cell survival and chondrogenic capabilities compared with the PP1 and PP3 cells. PP6 cells (muscle-derived stem cells) are superior to other primary muscle-derived cells for use as a cellular vehicle for BMP4-based ex vivo gene therapy to heal full-thickness osteo-chondral defects. The superiority of the PP6/muscle-derived stem cells appears to be attributable to a combination of increased rate of in vivo survival and superior chondrogenic differentiation capacity.
Collapse
|
11
|
Im GI. Gene Transfer Strategies to Promote Chondrogenesis and Cartilage Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2016; 22:136-48. [DOI: 10.1089/ten.teb.2015.0347] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Gun-Il Im
- Department of Orthopedics, Dongguk University Ilsan Hospital, Goyang, Republic of Korea
| |
Collapse
|
12
|
Madry H, Cucchiarini M. Gene therapy for human osteoarthritis: principles and clinical translation. Expert Opin Biol Ther 2015; 16:331-46. [PMID: 26593049 DOI: 10.1517/14712598.2016.1124084] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Osteoarthritis (OA) is the most prevalent chronic joint disease. Its key feature is a progressive articular cartilage loss. Gene therapy for OA aims at delivering gene-based therapeutic agents to the osteoarthritic cartilage, resulting in a controlled, site-specific, long-term presence to rebuild the damaged cartilage. AREAS COVERED An overview is provided of the principles of gene therapy for OA based on a PubMed literature search. Gene transfer to normal and osteoarthritic cartilage in vitro and in animal models in vivo is reviewed. Results from recent clinical gene therapy trials for OA are discussed and placed into perspective. EXPERT OPINION Recombinant adeno-associated viral (rAAV) vectors enable to directly transfer candidate sequences in human articular chondrocytes in situ, providing a potent tool to modulate the structure of osteoarthritic cartilage. However, few preclinical animal studies in OA models have been performed thus far. Noteworthy, several gene therapy clinical trials have been carried out in patients with end-stage knee OA based on the intraarticular injection of human juvenile allogeneic chondrocytes overexpressing a cDNA encoding transforming growth factor-beta-1 via retroviral vectors. In a recent placebo-controlled randomized trial, clinical scores were improved compared with placebo. These translational results provide sufficient reason to proceed with further clinical testing of gene transfer protocols for the treatment of OA.
Collapse
Affiliation(s)
- Henning Madry
- a Center of Experimental Orthopaedics , Saarland University , Homburg/Saar , Germany
| | - Magali Cucchiarini
- a Center of Experimental Orthopaedics , Saarland University , Homburg/Saar , Germany
| |
Collapse
|
13
|
D'souza N, Rossignoli F, Golinelli G, Grisendi G, Spano C, Candini O, Osturu S, Catani F, Paolucci P, Horwitz EM, Dominici M. Mesenchymal stem/stromal cells as a delivery platform in cell and gene therapies. BMC Med 2015; 13:186. [PMID: 26265166 PMCID: PMC4534031 DOI: 10.1186/s12916-015-0426-0] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 07/17/2015] [Indexed: 02/07/2023] Open
Abstract
Regenerative medicine relying on cell and gene therapies is one of the most promising approaches to repair tissues. Multipotent mesenchymal stem/stromal cells (MSC), a population of progenitors committing into mesoderm lineages, are progressively demonstrating therapeutic capabilities far beyond their differentiation capacities. The mechanisms by which MSC exert these actions include the release of biomolecules with anti-inflammatory, immunomodulating, anti-fibrogenic, and trophic functions. While we expect the spectra of these molecules with a therapeutic profile to progressively expand, several human pathological conditions have begun to benefit from these biomolecule-delivering properties. In addition, MSC have also been proposed to vehicle genes capable of further empowering these functions. This review deals with the therapeutic properties of MSC, focusing on their ability to secrete naturally produced or gene-induced factors that can be used in the treatment of kidney, lung, heart, liver, pancreas, nervous system, and skeletal diseases. We specifically focus on the different modalities by which MSC can exert these functions. We aim to provide an updated understanding of these paracrine mechanisms as a prerequisite to broadening the therapeutic potential and clinical impact of MSC.
Collapse
Affiliation(s)
- Naomi D'souza
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Filippo Rossignoli
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Giulia Golinelli
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Giulia Grisendi
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Carlotta Spano
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Olivia Candini
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Satoru Osturu
- The Division of Hematology/Oncology/BMT, Nationwide Children's Hospital, Departments of Pediatrics and Medicine, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Fabio Catani
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Paolo Paolucci
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Edwin M Horwitz
- The Division of Hematology/Oncology/BMT, Nationwide Children's Hospital, Departments of Pediatrics and Medicine, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy.
| |
Collapse
|
14
|
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
|
15
|
Qu F, Zhao Z, Yuan B, Qi W, Li C, Shen X, Liu C, Li H, Zhao G, Wang J, Guo Q, Liu Y. CaMKII plays a part in the chondrogenesis of bone marrow-derived mesenchymal stem cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:5981-5987. [PMID: 26191331 PMCID: PMC4503202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/16/2015] [Indexed: 06/04/2023]
Abstract
AIMS The purpose of the study is to observe the functions of calcium/calmodulin dependent protein kinase II (CaMKII) in the induced chondrogenic differentiation of bone marrow derived mesenchymal stem cells (BMSCs). METHODS BMSCs was in vitro isolated and cultured for induced chondrogenesis. Western blot was used to ascertain the expression of CaMKII and phosphorylated CaMKII (PCaMKII, activatory CaMKII) in chondrogenic induced BMSCs. MTT method was utilized to observe the impact of CaMKII on the proliferation of BMSCs. The generation of cartilage matrix in BMSCs cells was detected by toluidine blue staining. The levels of cartilage marker genes COL2A1, Aggrecan and SOX9 in BMSCs were gained by real-time fluorescence quantitative polymerase chain reaction (RT-QPCR). Finally, BMSCs proliferation, cartilage matrix generation and the changes of COL2A1, Aggrecan and SOX9 were surveyed after CaMKII being blocked by CaMKII inhibitor KN93. RESULTS Expression of CaMKII and PCaMKII could be found in chondrogenic induced BMSCs. CaMKII had no significant influence on BMSCs proliferation, but the toluidine blue staining was obviously lighter, indicating a significant decline in the expression of COL2A1, Aggrecan and SOX9. CONCLUSION As one of the factors influencing the chondrogenic capacity of BMSCs, CaMKII does not impact on BMSCs proliferation, but it can inhibit the chondrogenic ability of BMSCs by influencing its differentiation.
Collapse
Affiliation(s)
- Feng Qu
- Department of Orthopedics, Chinese PLA General Hospital28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Zhikun Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chinese PLA General Hospital51 Fucheng Road, Haidian District, Beijing 100048, China
| | - Bangtuo Yuan
- Department of Orthopedics, Chinese PLA General Hospital28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Wei Qi
- Department of Orthopedics, Chinese PLA General Hospital28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Chunbao Li
- Department of Orthopedics, Chinese PLA General Hospital28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Xuezhen Shen
- Department of Orthopedics, Chinese PLA General Hospital28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Chang Liu
- Department of Orthopedics, Chinese PLA General Hospital28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Hongliang Li
- Department of Orthopedics, Chinese PLA General Hospital28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Gang Zhao
- Department of Orthopedics, Chinese PLA General Hospital28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Jiangtao Wang
- Department of Orthopedics, Chinese PLA General Hospital28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Qi Guo
- Department of Orthopedics, Chinese PLA General Hospital28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Yujie Liu
- Department of Orthopedics, Chinese PLA General Hospital28 Fuxing Road, Haidian District, Beijing 100853, China
| |
Collapse
|
16
|
Zhang H, Leng P, He T, Wang Y. Comparison of hIGF-1 gene transfection to the hBMSCs and human meniscal fibrochondrocytes. Med Sci Monit 2015; 21:681-8. [PMID: 25738974 PMCID: PMC4357485 DOI: 10.12659/msm.891410] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background Treatment strategies for meniscal injury are shifting from meniscectomy to repair, especially cell-based therapy. Delivering selected genes to donor cells can modify differentiation and proliferation. Efficiency of gene transfection and expression may relate to cell type. Material/Methods Full-length hIGF-1 cDNA was cloned into eukaryotic expression vector by PCR. Human BMSCs and meniscal fibrochondrocytes were isolated and cultured in vitro and hIGF-1 gene was transfected by FuGene 6. Expression of EGFP and hIGF-1 were determined. Biological activity of the hIGF-1 in medium was assessed by MTT chromatometry. Real-time quantitative PCR and Western blot were used to assess the expression of exogenous genes. Efficacy of gene transfection was detected by immunohistochemistry staining and flow cytometry. Results Sequences of hIGF-1 were verified by sequence analysis. Expression of EGFP increased gradually and reached peak intensity 48 h after transfection. Transfection efficiency of BMSCs was higher than meniscal fibrochondrocytes. The population doubling time was decreased in both cell types. Peak concentration of hIGF-1 in the medium of BMSCs and meniscal cells was 32.5±4.8 ng/ml and 24.5±4.6 ng/ml, respectively. Secreted hIGF-1 possessed the ability to enhance proliferation of the cell line. Results of qPCR and Western blot confirmed the expression of hIGF-1. Type II collagen appeared within the cells, and percentage of cells in S stage was increased in both cell types after transfection. Conclusions hIGF-1 cDNA can be transfected into BMSCs and meniscal fibrochondrocytes, resulting in gene expression. Expression efficiency in BMSCs was higher than that in fibrochondrocytes.
Collapse
Affiliation(s)
- Haining Zhang
- Department of Joint Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Ping Leng
- Department of Pharmacy, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Tian He
- Department of Trauma Surgery, Affiliated Yantai Yuhuangding Hospital of Qingdao University Medical College, Yantai, Shandong, China (mainland)
| | - Yingzhen Wang
- Department of Joint Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| |
Collapse
|
17
|
Rey-Rico A, Frisch J, Venkatesan JK, Schmitt G, Madry H, Cucchiarini M. Determination of effective rAAV-mediated gene transfer conditions to support chondrogenic differentiation processes in human primary bone marrow aspirates. Gene Ther 2015; 22:50-7. [PMID: 25338919 DOI: 10.1038/gt.2014.90] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/23/2014] [Accepted: 08/26/2014] [Indexed: 11/09/2022]
Abstract
The genetic modification of freshly aspirated bone marrow may provide convenient tools to enhance the regenerative capacities of cartilage defects compared with the complex manipulation of isolated progenitor cells. In the present study, we examined the ability and safety of recombinant adeno-associated virus (rAAV) serotype 2 vectors to deliver various reporter gene sequences in primary human bone marrow aspirates over time without altering the chondrogenic processes in the samples. The results demonstrate that successful rAAV-mediated gene transfer and expression of the lacZ and red fluorescent protein marker genes were achieved in transduced aspirates at very high efficiencies (90-94%) and over extended periods of time (up to 125 days) upon treatment with hirudin, an alternative anticoagulant that does not prevent the adsorption of the rAAV-2 particles at the surface of their targets compared with heparin. Application of rAAV was safe, displaying neither cytotoxic nor detrimental effects on the cellular and proliferative activities or on the chondrogenic processes in the aspirates especially using an optimal dose of 0.5 mg ml(-1) hirudin, and application of the potent SOX9 transcription factor even enhanced these processes while counteracting hypertrophic differentiation. The current findings demonstrate the clinical value of this class of vector to durably and safely modify bone marrow aspirates as a means to further develop convenient therapeutic approaches to improve the healing of cartilage defects.
Collapse
Affiliation(s)
- A Rey-Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - J Frisch
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - J K Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - G Schmitt
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - H Madry
- 1] Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany [2] Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| | - M Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| |
Collapse
|
18
|
Bone marrow derived stem cells in joint and bone diseases: a concise review. INTERNATIONAL ORTHOPAEDICS 2014; 38:1787-801. [PMID: 25005462 DOI: 10.1007/s00264-014-2445-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 06/21/2014] [Indexed: 12/11/2022]
Abstract
Stem cells have huge applications in the field of tissue engineering and regenerative medicine. Their use is currently not restricted to the life-threatening diseases but also extended to disorders involving the structural tissues, which may not jeopardize the patients' life, but certainly influence their quality of life. In fact, a particularly popular line of research is represented by the regeneration of bone and cartilage tissues to treat various orthopaedic disorders. Most of these pioneering research lines that aim to create new treatments for diseases that currently have limited therapies are still in the bench of the researchers. However, in recent years, several clinical trials have been started with satisfactory and encouraging results. This article aims to review the concept of stem cells and their characterization in terms of site of residence, differentiation potential and therapeutic prospective. In fact, while only the bone marrow was initially considered as a "reservoir" of this cell population, later, adipose tissue and muscle tissue have provided a considerable amount of cells available for multiple differentiation. In reality, recently, the so-called "stem cell niche" was identified as the perivascular space, recognizing these cells as almost ubiquitous. In the field of bone and joint diseases, their potential to differentiate into multiple cell lines makes their application ideally immediate through three main modalities: (1) cells selected by withdrawal from bone marrow, subsequent culture in the laboratory, and ultimately transplant at the site of injury; (2) bone marrow aspirate, concentrated and directly implanted into the injury site; (3) systemic mobilization of stem cells and other bone marrow precursors by the use of growth factors. The use of this cell population in joint and bone disease will be addressed and discussed, analysing both the clinical outcomes but also the basic research background, which has justified their use for the treatment of bone, cartilage and meniscus tissues.
Collapse
|
19
|
Nanduri V, Tattikota SM, T AR, Sriramagiri VRR, Kantipudi S, Pande G. Reconstruction of Hyaline Cartilage Deep Layer Properties in 3-Dimensional Cultures of Human Articular Chondrocytes. Orthop J Sports Med 2014; 2:2325967114539122. [PMID: 26535340 PMCID: PMC4555603 DOI: 10.1177/2325967114539122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Background: Articular cartilage (AC) injuries and malformations are commonly noticed because of trauma or age-related degeneration. Many methods have been adopted for replacing or repairing the damaged tissue. Currently available AC repair methods, in several cases, fail to yield good-quality long-lasting results, perhaps because the reconstructed tissue lacks the cellular and matrix properties seen in hyaline cartilage (HC). Purpose: To reconstruct HC tissue from 2-dimensional (2D) and 3-dimensional (3D) cultures of AC-derived human chondrocytes that would specifically exhibit the cellular and biochemical properties of the deep layer of HC. Study Design: Descriptive laboratory study. Methods: Two-dimensional cultures of human AC–derived chondrocytes were established in classical medium (CM) and newly defined medium (NDM) and maintained for a period of 6 weeks. These cells were suspended in 2 mm–thick collagen I gels, placed in 24-well culture inserts, and further cultured up to 30 days. Properties of chondrocytes, grown in 2D cultures and the reconstructed 3D cartilage tissue, were studied by optical and scanning electron microscopic techniques, immunohistochemistry, and cartilage-specific gene expression profiling by reverse transcription polymerase chain reaction and were compared with those of the deep layer of native human AC. Results: Two-dimensional chondrocyte cultures grown in NDM, in comparison with those grown in CM, showed more chondrocyte-specific gene activity and matrix properties. The NDM-grown chondrocytes in 3D cultures also showed better reproduction of deep layer properties of HC, as confirmed by microscopic and gene expression analysis. The method used in this study can yield cartilage tissue up to approximately 1.6 cm in diameter and 2 mm in thickness that satisfies the very low cell density and matrix composition properties present in the deep layer of normal HC. Conclusion: This study presents a novel and reproducible method for long-term culture of AC-derived chondrocytes and reconstruction of cartilage tissue with properties similar to the deep layer of HC in vitro. Clinical Relevance: The HC tissue obtained by the method described can be used to develop an implantable product for the replacement of damaged or malformed AC, especially in younger patients where the lesions are caused by trauma or mechanical stress.
Collapse
Affiliation(s)
- Vibudha Nanduri
- Council of Scientific and Industrial Research-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Surendra Mohan Tattikota
- Council of Scientific and Industrial Research-Centre for Cellular and Molecular Biology, Hyderabad, India. ; Center for Thrombosis & Hemostasis, Universitaetsmedizin Mainz, Mainz, Germany
| | - Avinash Raj T
- Council of Scientific and Industrial Research-Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | - Suma Kantipudi
- Sridevi Maternity & Nursing Home, Warasiguda, Hyderabad, India
| | - Gopal Pande
- Council of Scientific and Industrial Research-Centre for Cellular and Molecular Biology, Hyderabad, India
| |
Collapse
|
20
|
Gene modification of mesenchymal stem cells and articular chondrocytes to enhance chondrogenesis. BIOMED RESEARCH INTERNATIONAL 2014; 2014:369528. [PMID: 24963479 PMCID: PMC4052490 DOI: 10.1155/2014/369528] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 03/27/2014] [Indexed: 01/14/2023]
Abstract
Current cell based treatment for articular cartilage and osteochondral defects are hampered by issues such as cellular dedifferentiation and hypertrophy of the resident or transplanted cells. The reduced expression of chondrogenic signalling molecules and transcription factors is a major contributing factor to changes in cell phenotype. Gene modification of chondrocytes may be one approach to redirect cells to their primary phenotype and recent advances in nonviral and viral gene delivery technologies have enabled the expression of these lost factors at high efficiency and specificity to regain chondrocyte function. This review focuses on the various candidate genes that encode signalling molecules and transcription factors that are specific for the enhancement of the chondrogenic phenotype and also how epigenetic regulators of chondrogenesis in the form of microRNA may also play an important role.
Collapse
|
21
|
Madry H, Cucchiarini M. Advances and challenges in gene-based approaches for osteoarthritis. J Gene Med 2014; 15:343-55. [PMID: 24006099 DOI: 10.1002/jgm.2741] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 08/06/2013] [Accepted: 08/30/2013] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA), a paramount cause of physical disability for which there is no definitive cure, is mainly characterized by the gradual loss of the articular cartilage. Current nonsurgical and reconstructive surgical therapies have not met success in reversing the OA phenotype so far. Gene transfer approaches allow for a long-term and site-specific presence of a therapeutic agent to re-equilibrate the metabolic balance in OA cartilage and may consequently be suited to treat this slow and irreversible disorder. The distinct stages of OA need to be respected in individual gene therapy strategies. In this context, molecular therapy appears to be most effective for early OA. A critical step forward has been made by directly transferring candidate sequences into human articular chondrocytes embedded within their native extracellular matrix via recombinant adeno-associated viral vectors. Although extensive studies in vitro attest to a growing interest in this approach, data from animal models of OA are sparse. A phase I dose-escalating trial was recently performed in patients with advanced knee OA to examine the safety and activity of chondrocytes modified to produce the transforming growth factor β1 via intra-articular injection, showing a dose-dependent trend toward efficacy. Proof-of-concept studies in patients prior to undergoing total knee replacement may be privileged in the future to identify the best mode of translating this approach to clinical application, followed by randomized controlled trials.
Collapse
Affiliation(s)
- Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Saarland University, Homburg, Saar, Germany
| | | |
Collapse
|
22
|
Bidarra SJ, Barrias CC, Granja PL. Injectable alginate hydrogels for cell delivery in tissue engineering. Acta Biomater 2014; 10:1646-62. [PMID: 24334143 DOI: 10.1016/j.actbio.2013.12.006] [Citation(s) in RCA: 343] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 11/28/2013] [Accepted: 12/05/2013] [Indexed: 12/16/2022]
Abstract
Alginate hydrogels are extremely versatile and adaptable biomaterials, with great potential for use in biomedical applications. Their extracellular matrix-like features have been key factors for their choice as vehicles for cell delivery strategies aimed at tissue regeneration. A variety of strategies to decorate them with biofunctional moieties and to modulate their biophysical properties have been developed recently, which further allow their tailoring to the desired application. Additionally, their potential use as injectable materials offers several advantages over preformed scaffold-based approaches, namely: easy incorporation of therapeutic agents, such as cells, under mild conditions; minimally invasive local delivery; and high contourability, which is essential for filling in irregular defects. Alginate hydrogels have already been explored as cell delivery systems to enhance regeneration in different tissues and organs. Here, the in vitro and in vivo potential of injectable alginate hydrogels to deliver cells in a targeted fashion is reviewed. In each example, the selected crosslinking approach, the cell type, the target tissue and the main findings of the study are highlighted.
Collapse
Affiliation(s)
- Sílvia J Bidarra
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre, 823, 4150-180 Porto, Portugal.
| | - Cristina C Barrias
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre, 823, 4150-180 Porto, Portugal.
| | - Pedro L Granja
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre, 823, 4150-180 Porto, Portugal; FEUP - Faculdade de Engenharia da Universidade do Porto, Departamento de Engenharia Metalúrgica e de Materiais, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
23
|
Li B, Li F, Ma L, Yang J, Wang C, Wang D, Gao C. Poly(lactide-co-glycolide)/Fibrin Gel Construct as a 3D Model to Evaluate Gene Therapy of Cartilage in Vivo. Mol Pharm 2014; 11:2062-70. [DOI: 10.1021/mp5000136] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Bo Li
- MOE
Key Laboratory of Macromolecular Synthesis and Functionalization,
Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Feifei Li
- MOE
Key Laboratory of Macromolecular Synthesis and Functionalization,
Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Lie Ma
- MOE
Key Laboratory of Macromolecular Synthesis and Functionalization,
Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Junzhou Yang
- MOE
Key Laboratory of Macromolecular Synthesis and Functionalization,
Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Chunfen Wang
- MOE
Key Laboratory of Macromolecular Synthesis and Functionalization,
Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Dongan Wang
- Division of Bioengineering, School of Chemical & Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, N1.3-B2-13, 637457 Singapore
| | - Changyou Gao
- MOE
Key Laboratory of Macromolecular Synthesis and Functionalization,
Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
24
|
Smith Callahan LA, Childers EP, Bernard SL, Weiner SD, Becker ML. Maximizing phenotype constraint and extracellular matrix production in primary human chondrocytes using arginine-glycine-aspartate concentration gradient hydrogels. Acta Biomater 2013; 9:7420-8. [PMID: 23567942 DOI: 10.1016/j.actbio.2013.04.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 03/01/2013] [Accepted: 04/01/2013] [Indexed: 01/30/2023]
Abstract
New systematic approaches are necessary to determine and optimize the chemical and mechanical scaffold properties for hyaline cartilage generation using the limited cell numbers obtained from primary human sources. Peptide functionalized hydrogels possessing continuous variations in physico-chemical properties are an efficient three-dimensional platform for studying several properties simultaneously. Herein, we describe a polyethylene glycol dimethacrylate (PEGDM) hydrogel system possessing a gradient of arginine-glycine-aspartic acid peptide (RGD) concentrations from 0mM to 10mM. The system is used to correlate primary human osteoarthritic chondrocyte proliferation, phenotype maintenance and extracellular matrix (ECM) production to the gradient hydrogel properties. Cell number and chondrogenic phenotype (CD14:CD90 ratios) were found to decline in regions with higher RGD concentrations, while regions with lower RGD concentrations maintained cell number and phenotype. Over three weeks of culture, hydrogel regions containing lower RGD concentrations experience an increase in ECM content compared to regions with higher RGD concentrations. Variations in actin amounts and vinculin organization were observed within the RGD concentration gradients that contribute to the differences in chondrogenic phenotype maintenance and ECM expression.
Collapse
|
25
|
Madry H, Kaul G, Zurakowski D, Vunjak-Novakovic G, Cucchiarini M. Cartilage constructs engineered from chondrocytes overexpressing IGF-I improve the repair of osteochondral defects in a rabbit model. Eur Cell Mater 2013; 25:229-47. [PMID: 23588785 PMCID: PMC4476264 DOI: 10.22203/ecm.v025a17] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Tissue engineering combined with gene therapy is a promising approach for promoting articular cartilage repair. Here, we tested the hypothesis that engineered cartilage with chondrocytes overexpressing a human insulin-like growth factor I (IGF-I) gene can enhance the repair of osteochondral defects, in a manner dependent on the duration of cultivation. Genetically modified chondrocytes were cultured on biodegradable polyglycolic acid scaffolds in dynamic flow rotating bioreactors for either 10 or 28 d. The resulting cartilaginous constructs were implanted into osteochondral defects in rabbit knee joints. After 28 weeks of in vivo implantation, immunoreactivity to ß-gal was detectable in the repair tissue of defects that received lacZ constructs. Engineered cartilaginous constructs based on IGF-I-overexpressing chondrocytes markedly improved osteochondral repair compared with control (lacZ) constructs. Moreover, IGF-I constructs cultivated for 28 d in vitro significantly promoted osteochondral repair vis-à-vis similar constructs cultivated for 10 d, leading to significantly decreased osteoarthritic changes in the cartilage adjacent to the defects. Hence, the combination of spatially defined overexpression of human IGF-I within a tissue-engineered construct and prolonged bioreactor cultivation resulted in most enhanced articular cartilage repair and reduction of osteoarthritic changes in the cartilage adjacent to the defect. Such genetically enhanced tissue engineering provides a versatile tool to evaluate potential therapeutic genes in vivo and to improve our comprehension of the development of the repair tissue within articular cartilage defects. Insights gained with additional exploration using this model may lead to more effective treatment options for acute cartilage defects.
Collapse
Affiliation(s)
- Henning Madry
- Centre of Experimental Orthopaedics, Saarland University, Homburg, Germany,Department of Orthopaedic Surgery, Saarland University, Homburg, Germany,Address for correspondence: Henning Madry Centre of Experimental Orthopaedics Medical Faculty Building 37 Saarland University D-66421 Homburg, Germany Telephone Number: +49-6841-1624515 FAX Number: +49-6841-1624988
| | - Gunter Kaul
- Centre of Experimental Orthopaedics, Saarland University, Homburg, Germany
| | - David Zurakowski
- Departments of Anaesthesia and Surgery, Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Magali Cucchiarini
- Centre of Experimental Orthopaedics, Saarland University, Homburg, Germany
| |
Collapse
|
26
|
Odabas S, Feichtinger GA, Korkusuz P, Inci I, Bilgic E, Yar AS, Cavusoglu T, Menevse S, Vargel I, Piskin E. Auricular cartilage repair using cryogel scaffolds loaded with BMP-7-expressing primary chondrocytes. J Tissue Eng Regen Med 2012; 7:831-40. [PMID: 23281155 DOI: 10.1002/term.1634] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 08/08/2012] [Accepted: 09/25/2012] [Indexed: 12/13/2022]
Abstract
The loss of cartilage tissue due to trauma, tumour surgery or congenital defects, such as microtia and anotia, is one of the major concerns in head and neck surgery. Recently tissue-engineering approaches, including gene delivery, have been proposed for the regeneration of cartilage tissue. In this study, primary chondrocytes were genetically modified with plasmid-encoding bone morphogenetic protein-7 (BMP-7) via the commercially available non-viral Turbofect vector, with the aim of bringing ex vivo transfected chondrocytes to resynthesize BMP-7 in vitro as they would in vivo. Genetically modified cells were implanted into gelatin-oxidized dextran scaffolds and cartilage tissue formation was investigated in 15 × 15 mm auricular cartilage defects in vivo in 48 New Zealand (NZ) white rabbits for 4 months. The results were evaluated via histology and early gene expression. Early gene expression results indicated a strong effect of exogenous BMP-7 on matrix synthesis and chondrocyte growth. In addition, histological analysis results exhibited significantly better cartilage healing with BMP-7-modified (transfected) cells than in the non-modified (non-transfected) group and as well as the control.
Collapse
Affiliation(s)
- S Odabas
- Chemical Engineering Department and Bioengineering Division and Centre for Bioengineering (Biyomedtek), Hacettepe University, Beytepe, Ankara, Turkey
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Current world literature. Curr Opin Organ Transplant 2012; 17:688-99. [PMID: 23147911 DOI: 10.1097/mot.0b013e32835af316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
28
|
Al Faqeh H, Nor Hamdan BMY, Chen HC, Aminuddin BS, Ruszymah BHI. The potential of intra-articular injection of chondrogenic-induced bone marrow stem cells to retard the progression of osteoarthritis in a sheep model. Exp Gerontol 2012; 47:458-64. [PMID: 22759409 DOI: 10.1016/j.exger.2012.03.018] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 03/08/2012] [Accepted: 03/29/2012] [Indexed: 12/13/2022]
Abstract
In recent years, the use of bone marrow mesenchymal stem cell (BMSC) implantation has provided an alternative treatment for osteoarthritis. The objective of this study is to determine whether or not an intra-articular injection of a single dose of autologous chondrogenic induced BMSC could retard the progressive destruction of cartilage in a surgically induced osteoarthritis in sheep. Sheep BMSCs were isolated and divided into two groups. One group was cultured in chondrogenic media containing (Ham's F12:DMEM, 1:1) FD+1% FBS+5 ng/ml TGFβ3+50 ng/ml IGF-1 (CM), and the other group was cultured in the basal media, FD+10% FBS (BM). The procedure for surgically induced osteoarthritis was performed on the donor sheep 6 weeks prior to intra-articular injection into the knee joint of a single dose of BMSC from either group, suspended in 5 ml FD at density of 2 million cells/ml. The control groups were injected with basal media, without cells. Six weeks after injection, gross evidence of retardation of cartilage destruction was seen in the osteoarthritic knee joints treated with CM as well as BM. No significant ICRS (International Cartilage Repair Society) scoring was detected between the two groups with cells. However macroscopically, meniscus repair was observed in the knee joint treated with CM. Severe osteoarthritis and meniscal injury was observed in the control group. Interestingly, histologically the CM group demonstrated good cartilage histoarchitecture, thickness and quality, comparable to normal knee joint cartilage. As a conclusion, intra-articular injection of a single dose of BMSC either chondrogenically induced or not, could retard the progression of osteoarthritis (OA) in a sheep model, but the induced cells indicated better results especially in meniscus regeneration.
Collapse
Affiliation(s)
- Hamoud Al Faqeh
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia.
| | | | | | | | | |
Collapse
|
29
|
General principles for the regeneration of bone and cartilage. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2012; 130:69-88. [PMID: 22805879 DOI: 10.1007/10_2012_145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
For the regeneration of bone and cartilage, mesenchymal stem cells are currently used invitro and in-vivo. For bone, the existence of viable cells, scaffolds, mechanical environment, growth factors and vascularization are of paramount importance. Mesenchymal stem cells can be harvested from the bone marrow using minimally invasive techniques. Centrifugation can increase the number of transplanted cells per volume. The use of cell therapy is under current clinical investigation and the benefit from these systems has to be proven in level I studies. For cartilage, current techniques recruiting stem cells from the subchondral bone have been demonstrated to be nearly as effective as autologous chondrocyte transplantation, requiring less invasive surgery. The efficacy of mesenchymal stem cell concentrates remains to be proven. There is high potential for tissue engineered joint surfaces to become an option for joint surface defects and degeneration.
Collapse
|