1
|
Lara-Arias J, Peña-Martínez VM, Rodriguez-Corpus LA, Romero-Díaz VJ, Álvarez-Lozano E, Martínez-Rodríguez HG. The Effect of the Overexpression or Addition of IGF1 and FGF2 in Human Chondrocytes Included in a Fibrin Matrix and Cultivated in a Dynamic Environment. Polymers (Basel) 2024; 16:1968. [PMID: 39065286 PMCID: PMC11281257 DOI: 10.3390/polym16141968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/24/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Hyaline cartilage is a highly specialized tissue. When injured, its repair capacity is low, which results in the massive destruction of the articular surface. Using tissue engineering and genetic engineering techniques, it is possible to provide a suitable microenvironment providing chondrocyte growth factors involved in the development of hyaline cartilage proteins, as well as cell proliferation and differentiation. Our aim was to stimulate the synthesis of an extracellular matrix via the chondrocytes included in a fibrin matrix through the addition or overexpression of IGF1 and/or FGF2, while maintaining a constant agitation of the culture medium. Collagen type II and glycosaminoglycans increased during the entire incubation time. In contrast, collagen type I decreased its expression under the same culture conditions, transfecting or supplementing growth factors to chondrocytes. However, chondrocytes that were not transfected or supplemented showed a general increase in the proteins analyzed in this study. The presence of IGF1 and FGF2 increased the protein synthesis of the hyaline cartilage, regardless of which one was the source of growth factors. Continuous agitation using the spinner flask allows for the adequate nutrition of chondrocytes included in the fibrin matrix. However, they require growth factors to up-regulate or down-regulate collagenous proteins.
Collapse
Affiliation(s)
- Jorge Lara-Arias
- Orthopedics and Traumatology Service, Hospital Dr. José E. González, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Mitras Centro, Monterrey 64460, NL, Mexico; (J.L.-A.); (V.M.P.-M.); (L.A.R.-C.)
| | - Victor Manuel Peña-Martínez
- Orthopedics and Traumatology Service, Hospital Dr. José E. González, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Mitras Centro, Monterrey 64460, NL, Mexico; (J.L.-A.); (V.M.P.-M.); (L.A.R.-C.)
| | - Luis Alejandro Rodriguez-Corpus
- Orthopedics and Traumatology Service, Hospital Dr. José E. González, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Mitras Centro, Monterrey 64460, NL, Mexico; (J.L.-A.); (V.M.P.-M.); (L.A.R.-C.)
| | - Viktor J. Romero-Díaz
- Histology Department, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Mitras Centro, Monterrey 64460, NL, Mexico;
| | - Eduardo Álvarez-Lozano
- Orthopedics and Traumatology Service, Hospital Dr. José E. González, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Mitras Centro, Monterrey 64460, NL, Mexico; (J.L.-A.); (V.M.P.-M.); (L.A.R.-C.)
| | - Herminia G. Martínez-Rodríguez
- Biochemistry and Molecular Medicine Department, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Mitras Centro, Monterrey 64460, NL, Mexico
| |
Collapse
|
2
|
Ciamillo SA, Pownder SL, Potter HG, Stefanovski D, Nixon AJ, Ortved KF. Correlation of Arthroscopic Grading and Optical Coherence Tomography as Markers of Early Repair and Predictors of Later Healing Evident on MRI and Histomorphometric Assessment of Cartilage Defects Implanted with Chondrocytes Overexpressing IGF-I. Cartilage 2023; 14:210-219. [PMID: 36864720 PMCID: PMC10416204 DOI: 10.1177/19476035231154508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 03/04/2023] Open
Abstract
OBJECTIVE Injury of articular cartilage is common, and due to the poor intrinsic capabilities of chondrocytes, it can precipitate joint degradation and osteoarthritis (OA). Implantation of autologous chondrocytes into cartilaginous defects has been used to bolster repair. Accurate assessment of the quality of repair tissue remains challenging. This study aimed to investigate the utility of noninvasive imaging modalities, including arthroscopic grading and optical coherence tomography (OCT) for assessment of early cartilage repair (8 weeks), and MRI to determine long-term healing (8 months). DESIGN Large (15 mm diameter), full-thickness chondral defects were created on both lateral trochlear ridges of the femur in 24 horses. Defects were implanted with autologous chondrocytes transduced with rAAV5-IGF-I, autologous chondrocytes transduced with rAAV5-GFP, naïve autologous chondrocytes, or autologous fibrin. Healing was evaluated at 8 weeks post-implantation using arthroscopy and OCT, and at 8 months post-implantation using MRI, gross pathology, and histopathology. RESULTS OCT and arthroscopic scoring of short-term repair tissue were significantly correlated. Arthroscopy was also correlated with later gross pathology and histopathology of repair tissue at 8 months post-implantation, while OCT was not correlated. MRI was not correlated with any other assessment variable. CONCLUSIONS This study indicated that arthroscopic inspection and manual probing to develop an early repair score may be a better predictor of long-term cartilage repair quality following autologous chondrocyte implantation. Furthermore, qualitative MRI may not provide additional discriminatory information when assessing mature repair tissue, at least in this equine model of cartilage repair.
Collapse
Affiliation(s)
- Sarah A. Ciamillo
- New Bolton Center, Department of Clinical Studies, University of Pennsylvania, Kennett Square, PA, USA
| | | | | | - Darko Stefanovski
- New Bolton Center, Department of Clinical Studies, University of Pennsylvania, Kennett Square, PA, USA
| | - Alan J. Nixon
- Department of Clinical Sciences, Cornell University, Ithaca, NY, USA
| | - Kyla F. Ortved
- New Bolton Center, Department of Clinical Studies, University of Pennsylvania, Kennett Square, PA, USA
| |
Collapse
|
3
|
Bi R, Luo X, Li Q, Li P, Li H, Fan Y, Ying B, Zhu S. Igf1 Regulates Fibrocartilage Stem Cells, Cartilage Growth, and Homeostasis in the Temporomandibular Joint of Mice. J Bone Miner Res 2023; 38:556-567. [PMID: 36722289 DOI: 10.1002/jbmr.4782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 01/19/2023] [Accepted: 01/27/2023] [Indexed: 02/02/2023]
Abstract
Temporomandibular joint (TMJ) growth requires orchestrated interactions between various cell types. Recent studies revealed that fibrocartilage stem cells (FCSCs) in the TMJ cartilage play critical roles as cell resources for joint development and repair. However, the detailed molecular network that influences FCSC fate during TMJ cartilage development remains to be elucidated. Here, we investigate the functional role of Igf1 in FCSCs for TMJ cartilage growth and homeostasis by lineage tracing using Gli1-CreER+ ; Tmflfl mice and conditional Igf1 deletion using Gli1-/Col2-CreER+ ; Igf1fl/fl mice. In Gli1-CreER+ ; Tmflfl mice, red fluorescence+ (RFP+ ) FCSCs show a favorable proliferative capacity. Igf1 deletion in Gli1+ /Col2+ cell lineages leads to distinct pathological changes in TMJ cartilage. More serious cartilage thickness and cell density reductions are found in the superficial layers in Gli1-CreER+ ; Igf1fl/fl mice. After long-term Igf1 deletion, a severe disordered cell arrangement is found in both groups. When Igf1 is conditionally deleted in vivo, the red fluorescent protein-labeled Gli1+ FCSC shows a significant disruption of chondrogenic differentiation, cell proliferation, and apoptosis leading to TMJ cartilage disarrangement and subchondral bone loss. Immunostaining shows that pAkt signaling is blocked in all cartilage layers after the Gli1+ -specific deletion of Igf1. In vitro, Igf1 deletion disrupts FCSC capacities, including proliferation and chondrogenesis. Moreover, the deletion of Igf1 in FCSCs significantly aggravates the joint osteoarthritis phenotype in the unilateral anterior crossbite mouse model, characterized by decreased cartilage thickness and cell numbers as well as a loss of extracellular matrix secretions. These findings uncover Igf1 as a regulator of TMJ cartilage growth and repair. The deletion of Igf1 disrupts the progenitor capacity of FCSCs, leading to a disordered cell distribution and exaggerating TMJ cartilage dysfunction. © 2023 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Ruiye Bi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xueting Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qianli Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Peiran Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Haohan Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Fan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Binbin Ying
- Department of Stomatology, Ningbo First Hospital, Ningbo, China
| | - Songsong Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Thampi P, Samulski RJ, Grieger JC, Phillips JN, McIlwraith CW, Goodrich LR. Gene therapy approaches for equine osteoarthritis. Front Vet Sci 2022; 9:962898. [PMID: 36246316 PMCID: PMC9558289 DOI: 10.3389/fvets.2022.962898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/08/2022] [Indexed: 01/24/2023] Open
Abstract
With an intrinsically low ability for self-repair, articular cartilage injuries often progress to cartilage loss and joint degeneration resulting in osteoarthritis (OA). Osteoarthritis and the associated articular cartilage changes can be debilitating, resulting in lameness and functional disability both in human and equine patients. While articular cartilage damage plays a central role in the pathogenesis of OA, the contribution of other joint tissues to the pathogenesis of OA has increasingly been recognized thus prompting a whole organ approach for therapeutic strategies. Gene therapy methods have generated significant interest in OA therapy in recent years. These utilize viral or non-viral vectors to deliver therapeutic molecules directly into the joint space with the goal of reprogramming the cells' machinery to secrete high levels of the target protein at the site of injection. Several viral vector-based approaches have demonstrated successful gene transfer with persistent therapeutic levels of transgene expression in the equine joint. As an experimental model, horses represent the pathology of human OA more accurately compared to other animal models. The anatomical and biomechanical similarities between equine and human joints also allow for the use of similar imaging and diagnostic methods as used in humans. In addition, horses experience naturally occurring OA and undergo similar therapies as human patients and, therefore, are a clinically relevant patient population. Thus, further studies utilizing this equine model would not only help advance the field of human OA therapy but also benefit the clinical equine patients with naturally occurring joint disease. In this review, we discuss the advancements in gene therapeutic approaches for the treatment of OA with the horse as a relevant patient population as well as an effective and commonly utilized species as a translational model.
Collapse
Affiliation(s)
- Parvathy Thampi
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States
| | - R. Jude Samulski
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, United States
| | - Joshua C. Grieger
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, United States
| | - Jennifer N. Phillips
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States
| | - C. Wayne McIlwraith
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States
| | - Laurie R. Goodrich
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States,*Correspondence: Laurie R. Goodrich
| |
Collapse
|
5
|
Predicting Transport of Intra-Articularly Injected Growth Factor Fusion Proteins into Human Knee Joint Cartilage. Acta Biomater 2022; 153:243-259. [DOI: 10.1016/j.actbio.2022.09.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/23/2022]
|
6
|
Rao C, Shi S. Development of Nanomaterials to Target Articular Cartilage for Osteoarthritis Therapy. Front Mol Biosci 2022; 9:900344. [PMID: 36032667 PMCID: PMC9402910 DOI: 10.3389/fmolb.2022.900344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/09/2022] [Indexed: 01/10/2023] Open
Abstract
Osteoarthritis (OA) is an obstinate, degradative, and complicated disease that has drawn much attention worldwide. Characterized by its stubborn symptoms and various sequela, OA causes much financial burden on both patients and the health system. What’s more, conventional systematic therapy is not effective enough and causes multiple side effects. There’s much evidence that nanoparticles have unique properties such as high penetration, biostability, and large specific surface area. Thus, it is urgent to exploit novel medications for OA. Nanomaterials have been sufficiently studied, exploiting diverse nano-drug delivery systems (DDSs) and targeted nano therapeutical molecules. The nanomaterials are primarily intra-articular injected under the advantages of high topical concentration and low dosage. After administration, the DDS and targeted nano therapeutical molecules can specifically react with the components, including cartilage and synovium of a joint in OA, furthermore attenuate the chondrocyte apoptosis, matrix degradation, and macrophage recruitment. Thus, arthritis would be alleviated. The DDSs could load with conventional anti-inflammatory drugs, antibodies, RNA, and so on, targeting chondrocytes, synovium, or extracellular matrix (ECM) and releasing the molecules sequentially. The targeted nano therapeutical molecules could directly get to the targeted tissue, alleviating the inflammation and promoting tissue healing. This review will comprehensively collect and evaluate the targeted nanomaterials to articular cartilage in OA.
Collapse
|
7
|
Vedadghavami A, Hakim B, He T, Bajpayee AG. Cationic peptide carriers enable long-term delivery of insulin-like growth factor-1 to suppress osteoarthritis-induced matrix degradation. Arthritis Res Ther 2022; 24:172. [PMID: 35858920 PMCID: PMC9297664 DOI: 10.1186/s13075-022-02855-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/27/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Insulin-like growth factor-1 (IGF-1) has the potential to be used for osteoarthritis (OA) treatment but has not been evaluated in clinics yet owing to toxicity concerns. It suffers from short intra-joint residence time and a lack of cartilage targeting following its intra-articular administration. Here, we synthesize an electrically charged cationic formulation of IGF-1 by using a short-length arginine-rich, hydrophilic cationic peptide carrier (CPC) with a net charge of +14, designed for rapid and high uptake and retention in both healthy and arthritic cartilage. METHODS IGF-1 was conjugated to CPC by using a site-specific sulfhydryl reaction via a bifunctional linker. Intra-cartilage depth of penetration and retention of CPC-IGF-1 was compared with the unmodified IGF-1. The therapeutic effectiveness of a single dose of CPC-IGF-1 was compared with free IGF-1 in an IL-1α-challenged cartilage explant culture post-traumatic OA model. RESULTS CPC-IGF-1 rapidly penetrated through the full thickness of cartilage creating a drug depot owing to electrostatic interactions with negatively charged aggrecan-glycosaminoglycans (GAGs). CPC-IGF-1 remained bound within the tissue while unmodified IGF-1 cleared out. Treatment with a single dose of CPC-IGF-1 effectively suppressed IL-1α-induced GAG loss and nitrite release and rescued cell metabolism and viability throughout the 16-day culture period, while free IGF at the equivalent dose was not effective. CONCLUSIONS CPC-mediated depot delivery of IGF-1 protected cartilage by suppressing cytokine-induced catabolism with only a single dose. CPC is a versatile cationic motif that can be used for intra-cartilage delivery of other similar-sized drugs.
Collapse
Affiliation(s)
| | - Bill Hakim
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Tengfei He
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Ambika G Bajpayee
- Department of Bioengineering, Northeastern University, Boston, MA, USA.
- Departments of Mechanical Engineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
8
|
Trompeter N, Gardinier JD, DeBarros V, Boggs M, Gangadharan V, Cain WJ, Hurd L, Duncan RL. Insulin-like growth factor-1 regulates the mechanosensitivity of chondrocytes by modulating TRPV4. Cell Calcium 2021; 99:102467. [PMID: 34530313 PMCID: PMC8541913 DOI: 10.1016/j.ceca.2021.102467] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 08/12/2021] [Accepted: 08/26/2021] [Indexed: 10/20/2022]
Abstract
Both mechanical and biochemical stimulation are required for maintaining the integrity of articular cartilage. However, chondrocytes respond differently to mechanical stimuli in osteoarthritic cartilage when biochemical signaling pathways, such as Insulin-like Growth Factor-1 (IGF-1), are altered. The Transient Receptor Potential Vanilloid 4 (TRPV4) channel is central to chondrocyte mechanotransduction and regulation of cartilage homeostasis. Here, we propose that changes in IGF-1 can modulate TRPV4 channel activity. We demonstrate that physiologic levels of IGF-1 suppress hypotonic-induced TRPV4 currents and intracellular calcium flux by increasing apparent cell stiffness that correlates with actin stress fiber formation. Disruption of F-actin following IGF-1 treatment results in the return of the intracellular calcium response to hypotonic swelling. Using point mutations of the TRPV4 channel at the microtubule-associated protein 7 (MAP-7) site shows that regulation of TRPV4 by actin is mediated via the interaction of actin with the MAP-7 domain of TRPV4. We further highlight that ATP release, a down-stream response to mechanical stimulation in chondrocytes, is mediated by TRPV4 during hypotonic challenge. This response is significantly abrogated with IGF-1 treatment. As chondrocyte mechanosensitivity is greatly altered during osteoarthritis progression, IGF-1 presents as a promising candidate for prevention and treatment of articular cartilage damage.
Collapse
Affiliation(s)
- Nicholas Trompeter
- Biomedical Engineering, University of Delaware, Newark, DE, United States
| | - Joseph D Gardinier
- Biomechanics and Movement Science Program, University of Delaware, Newark, DE, United States; Bone and Joint Center, Henry Ford Hospital, Detroit, MI, United States
| | - Victor DeBarros
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Mary Boggs
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Vimal Gangadharan
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - William J Cain
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Lauren Hurd
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Randall L Duncan
- Biomedical Engineering, University of Delaware, Newark, DE, United States; Biomechanics and Movement Science Program, University of Delaware, Newark, DE, United States; Department of Biological Sciences, University of Delaware, Newark, DE, United States; Department of Biology, University of Michigan-Flint, Flint, MI, United States.
| |
Collapse
|
9
|
Li X, Dai B, Guo J, Zheng L, Guo Q, Peng J, Xu J, Qin L. Nanoparticle-Cartilage Interaction: Pathology-Based Intra-articular Drug Delivery for Osteoarthritis Therapy. NANO-MICRO LETTERS 2021; 13:149. [PMID: 34160733 PMCID: PMC8222488 DOI: 10.1007/s40820-021-00670-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/19/2021] [Indexed: 05/03/2023]
Abstract
Osteoarthritis is the most prevalent chronic and debilitating joint disease, resulting in huge medical and socioeconomic burdens. Intra-articular administration of agents is clinically used for pain management. However, the effectiveness is inapparent caused by the rapid clearance of agents. To overcome this issue, nanoparticles as delivery systems hold considerable promise for local control of the pharmacokinetics of therapeutic agents. Given the therapeutic programs are inseparable from pathological progress of osteoarthritis, an ideal delivery system should allow the release of therapeutic agents upon specific features of disorders. In this review, we firstly introduce the pathological features of osteoarthritis and the design concept for accurate localization within cartilage for sustained drug release. Then, we review the interactions of nanoparticles with cartilage microenvironment and the rational design. Furthermore, we highlight advances in the therapeutic schemes according to the pathology signals. Finally, armed with an updated understanding of the pathological mechanisms, we place an emphasis on the development of "smart" bioresponsive and multiple modality nanoparticles on the near horizon to interact with the pathological signals. We anticipate that the exploration of nanoparticles by balancing the efficacy, safety, and complexity will lay down a solid foundation tangible for clinical translation.
Collapse
Affiliation(s)
- Xu Li
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
| | - Bingyang Dai
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
| | - Jiaxin Guo
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
| | - Lizhen Zheng
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
| | - Quanyi Guo
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jiang Peng
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China.
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China.
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China.
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China.
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China.
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China.
| |
Collapse
|
10
|
Aoki E, Asawa Y, Hikita A, Hoshi K. Establishment of a new technique for the fabrication of regenerative cartilage with a microslicer device to prepare three dimensional diced cartilage. Biomed Res 2021; 41:67-80. [PMID: 32307400 DOI: 10.2220/biomedres.41.67] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Chondrocytes are utilized to cartilage regeneration by being harvested through enzymatic digestion and expanded by monolayer culture. However, these procedures will cause deterioration and dedifferentiation of the chondrocytes. In addition, scaffolds are often needed to provide the cartilage with mechanical strength and three-dimensional structures. We tried to use diced cartilage prepared using a micro-slicer without digestion, monolayer culture or scaffolds. In this study, an appropriate culture condition to induce the fusion of diced cartilage in vitro and cartilage regeneration in vitro and in vivo was determined to realize a scaffold-free cartilage regeneration. As a result, diced cartilages aggregated when they were cultured more than 5 weeks in the media containing 10% fetal bovine serum (FBS). Diced cartilage cultured for 7 weeks with the media containing 10%, followed by the culture with the media containing insulin-like growth factor-1 for 5 weeks in the ultralow attachment plate showed most prominent cartilage formation both in vitro and in vivo. The volume of regenerated cartilage was 2.14 times larger than that of the original cartilage. These results indicated that large regenerative cartilage from a small amount of cartilage was achieved without deterioration or dedifferentiation.
Collapse
Affiliation(s)
- Erika Aoki
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo
| | - Yukiyo Asawa
- Division of Tissue Engineering, The University of Tokyo Hospital
| | - Atsuhiko Hikita
- Division of Tissue Engineering, The University of Tokyo Hospital
| | - Kazuto Hoshi
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo.,Division of Tissue Engineering, The University of Tokyo Hospital.,Department of Oral-maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Hospital
| |
Collapse
|
11
|
Watanabe T, Asawa Y, Watanabe M, Okubo R, Nio M, Takato T, Hoshi K, Hikita A. The usefulness of the decellularized matrix from three-dimensional regenerative cartilage as a scaffold material. Regen Ther 2020; 15:312-322. [PMID: 33426234 PMCID: PMC7770429 DOI: 10.1016/j.reth.2020.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/30/2020] [Accepted: 11/18/2020] [Indexed: 11/30/2022] Open
Abstract
In cartilage tissue engineering, research on materials for three-dimensional (3D) scaffold has attracted attention. Decellularized matrix can be one of the candidates for the scaffold material. In this study, decellularization of regenerated cartilage was carried out and its effectiveness as a scaffold material was examined. Three-dimensionally-cultured cartilage constructs in the differentiation medium containing IGF-1 produced more cartilage matrix than those in the proliferation medium. Detergent-enzymatic method (DEM) could decellularize 3D-cultured cartilage constructs only by 1 cycle without breaking down the structure of the constructs. In vitro, newly-seeded chondrocytes were infiltrated and engrafted into decellularized constructs in the proliferation medium, and newly formed fibers were observed around the surface where newly-seeded cells were attached. Recellularized constructs could mature similarly as those without decellularization in vivo. The decellularized 3D-cultured matrix from regenerative cartilage is expected to be used as a scaffold material in the future.
Collapse
Affiliation(s)
- Tomohiko Watanabe
- Department of Pediatric Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryomachi, Aoba-ku, Sendai 980-8574, Japan
| | - Yukiyo Asawa
- Department of Cell & Tissue Engineering (Fujisoft), Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan.,Department of Tissue Engineering, The University of Tokyo Hospital, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Makoto Watanabe
- Department of Tissue Engineering, The University of Tokyo Hospital, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Ryuji Okubo
- Department of Pediatric Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryomachi, Aoba-ku, Sendai 980-8574, Japan
| | - Masaki Nio
- Department of Pediatric Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryomachi, Aoba-ku, Sendai 980-8574, Japan
| | - Tsuyoshi Takato
- JR Tokyo General Hospital, Yoyogi 2-1-3, Shibuya-ku, Tokyo 151-8528, Japan
| | - Kazuto Hoshi
- Department of Tissue Engineering, The University of Tokyo Hospital, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan.,Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Atsuhiko Hikita
- Department of Cell & Tissue Engineering (Fujisoft), Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan.,Department of Tissue Engineering, The University of Tokyo Hospital, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
12
|
Hyaluronic acid-binding insulin-like growth factor-1: Creation of a gene encoding a bifunctional fusion protein. Mol Biol Rep 2020; 47:9749-9756. [PMID: 33263932 DOI: 10.1007/s11033-020-06034-w] [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] [Received: 08/10/2020] [Accepted: 11/24/2020] [Indexed: 10/22/2022]
Abstract
Chondrogenic growth factors are promising therapeutic agents for articular cartilage repair. A persistent impediment to fulfilling this promise is a limited ability to apply and retain the growth factors within the region of cartilage damage that is in need of repair. Current therapies successfully deliver cells and/or matrices, but growth factors are subject to diffusion into the joint space and then loss from the joint. To address this problem, we created a novel gene that encodes a bifunctional fusion protein comprised by a matrix binding domain and a growth factor. The gene encodes the hyaluronic acid binding region of the cartilage matrix molecule, versican, and the chondrogenic growth factor, insulin-like growth factor-1 (IGF-1). We delivered the gene in an adeno-associated virus-based plasmid vector to articular chondrocytes. The cells synthesized and secreted the fusion protein gene product. The fusion protein bound to hyaluronic acid and retained the anabolic and mitogenic actions of IGF-1 on the chondrocytes. This proof-of-concept study suggests that the bifunctional fusion protein, in concert with chondrocytes and a hyaluronic acid-based delivery vehicle, may serve as an intra-articular therapy to help achieve articular cartilage repair.
Collapse
|
13
|
Mobasheri A, Choi H, Martín-Vasallo P. Over-Production of Therapeutic Growth Factors for Articular Cartilage Regeneration by Protein Production Platforms and Protein Packaging Cell Lines. BIOLOGY 2020; 9:biology9100330. [PMID: 33050357 PMCID: PMC7599991 DOI: 10.3390/biology9100330] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/28/2022]
Abstract
Simple Summary Osteoarthritis (OA) is the most common form of arthritis across the world. Most of the existing drugs for OA treat the symptoms of pain and inflammation. There are no drugs that can dure the disease. There are a number of new treatments for OA including cell therapy and gene therapy. This articles outlines the concept behind TissueGene-C, a new biological drug for OA. This new treatment includes cartilage cells mixed with a genetically modified cell line called GP2-293, which is effectively a “drug factory”, over-producing the growth factors that are important for cartilage regeneration and changing the environment inside joints. The mixture is injected into the affected knee joint. These cells are designed to be short-lived and cannot reproduce. Therefore, after they have done their job, they die and are cleared by immune cells. This is a new and modern approach to treating OA and TissueGene-C is the prototype cell therapy for OA. In the future, it is entirely possible to combine different clones of genetically engineered cells like GP2-293 that have been designed to over-produce a growth factor or biological drug with cells from the cartilage endplate of the intervertebral disc to treat degeneration in the spine. Abstract This review article focuses on the current state-of-the-art cellular and molecular biotechnology for the over-production of clinically relevant therapeutic and anabolic growth factors. We discuss how the currently available tools and emerging technologies can be used for the regenerative treatment of osteoarthritis (OA). Transfected protein packaging cell lines such as GP-293 cells may be used as “cellular factories” for large-scale production of therapeutic proteins and pro-anabolic growth factors, particularly in the context of cartilage regeneration. However, when irradiated with gamma or x-rays, these cells lose their capacity for replication, which makes them safe for use as a live cell component of intra-articular injections. This innovation is already here, in the form of TissueGene-C, a new biological drug that consists of normal allogeneic primary chondrocytes combined with transduced GP2-293 cells that overexpress the growth factor transforming growth factor β1 (TGF-β1). TissueGene-C has revolutionized the concept of cell therapy, allowing drug companies to develop live cells as biological drug delivery systems for direct intra-articular injection of growth factors whose half-lives are in the order of minutes. Therefore, in this paper, we discuss the potential for new innovations in regenerative medicine for degenerative diseases of synovial joints using mammalian protein production platforms, specifically protein packaging cell lines, for over-producing growth factors for cartilage tissue regeneration and give recent examples. Mammalian protein production platforms that incorporate protein packaging eukaryotic cell lines are superior to prokaryotic bacterial expression systems and are likely to have a significant impact on the development of new humanized biological growth factor therapies for treating focal cartilage defects and more generally for the treatment of degenerative joint diseases such as OA, especially when injected directly into the joint.
Collapse
Affiliation(s)
- Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, FI-90014 Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
- Departments of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
- Versus Arthritis Centre for Sport, Exercise and Osteoarthritis Research, Queen’s Medical Centre, Nottingham NG7 2UH, UK
- Correspondence: or
| | - Heonsik Choi
- Kolon TissueGene, Inc., Rockville, MD 20850, USA;
- Healthcare Research Institute, Kolon Advanced Research Center, Kolon Industries, Inc., Magok-dong, Gangseo-gu, Seoul 07793, Korea
| | - Pablo Martín-Vasallo
- UD of Biochemistry and Molecular Biology, Instituto de Tecnologías Biomédicas de Canarias, Universidad de La Laguna, San Cristóbal de La Laguna, 38071 Tenerife, Spain;
| |
Collapse
|
14
|
Schivo S, Khurana S, Govindaraj K, Scholma J, Kerkhofs J, Zhong L, Huang X, van de Pol J, Langerak R, van Wijnen AJ, Geris L, Karperien M, Post JN. ECHO, the executable CHOndrocyte: A computational model to study articular chondrocytes in health and disease. Cell Signal 2020; 68:109471. [DOI: 10.1016/j.cellsig.2019.109471] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 11/01/2019] [Accepted: 11/12/2019] [Indexed: 12/18/2022]
|
15
|
Griffiths R, Woods S, Cheng A, Wang P, Griffiths-Jones S, Ronshaugen M, Kimber SJ. The Transcription Factor-microRNA Regulatory Network during hESC-chondrogenesis. Sci Rep 2020; 10:4744. [PMID: 32179818 PMCID: PMC7075910 DOI: 10.1038/s41598-020-61734-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 02/19/2020] [Indexed: 12/21/2022] Open
Abstract
Human embryonic stem cells (ESCs) offer a promising therapeutic approach for osteoarthritis (OA). The unlimited source of cells capable of differentiating to chondrocytes has potential for repairing damaged cartilage or to generate disease models via gene editing. However their use is limited by the efficiency of chondrogenic differentiation. An improved understanding of the transcriptional and post-transcriptional regulation of chondrogenesis will enable us to improve hESC chondrogenic differentiation protocols. Small RNA-seq and whole transcriptome sequencing was performed on distinct stages of hESC-directed chondrogenesis. This revealed significant changes in the expression of several microRNAs including upregulation of known cartilage associated microRNAs and those transcribed from the Hox complexes, and the downregulation of pluripotency associated microRNAs. Integration of miRomes and transcriptomes generated during hESC-directed chondrogenesis identified key functionally related clusters of co-expressed microRNAs and protein coding genes, associated with pluripotency, primitive streak, limb development and extracellular matrix. Analysis identified regulators of hESC-directed chondrogenesis such as miR-29c-3p with 10 of its established targets identified as co-regulated 'ECM organisation' genes and miR-22-3p which is highly co-expressed with ECM genes and may regulate these genes indirectly by targeting the chondrogenic regulators SP1 and HDAC4. We identified several upregulated transcription factors including HOXA9/A10/D13 involved in limb patterning and RELA, JUN and NFAT5, which have targets enriched with ECM associated genes. We have developed an unbiased approach for integrating transcriptome and miRome using protein-protein interactions, transcription factor regulation and miRNA target interactions and identified key regulatory networks prominent in hESC chondrogenesis.
Collapse
Affiliation(s)
- Rosie Griffiths
- Divisions of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology Medicine and Health, Michael Smith Building, Oxford Road, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, United Kingdom
| | - Steven Woods
- Divisions of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology Medicine and Health, Michael Smith Building, Oxford Road, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Aixin Cheng
- Divisions of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology Medicine and Health, Michael Smith Building, Oxford Road, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
- Salford Royal NHS Foundation Trust, Department of Trauma and Orthopaedic, Stott Lane, Salford, M6 8HD, United Kingdom
| | - Ping Wang
- Evolution and Genomic Science, Faculty of Biology Medicine and Health, Michael Smith Building, Oxford Road, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Sam Griffiths-Jones
- Evolution and Genomic Science, Faculty of Biology Medicine and Health, Michael Smith Building, Oxford Road, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Matthew Ronshaugen
- Developmental Biology and Medicine, Faculty of Biology Medicine and Health, Michael Smith Building, Oxford Road, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Susan J Kimber
- Divisions of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology Medicine and Health, Michael Smith Building, Oxford Road, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK.
| |
Collapse
|
16
|
Menarim BC, Gillis KH, Oliver A, Mason C, Werre SR, Luo X, Byron CR, Kalbfleisch TS, MacLeod JN, Dahlgren LA. Inflamed synovial fluid induces a homeostatic response in bone marrow mononuclear cells in vitro: Implications for joint therapy. FASEB J 2020; 34:4430-4444. [PMID: 32030831 DOI: 10.1096/fj.201902698r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/13/2020] [Accepted: 01/13/2020] [Indexed: 02/06/2023]
Abstract
Synovial inflammation is a central feature of osteoarthritis (OA), elicited when local regulatory macrophages (M2-like) become overwhelmed, activating an inflammatory response (M1-like). Bone marrow mononuclear cells (BMNC) are a source of naïve macrophages capable of reducing joint inflammation and producing molecules essential for cartilage metabolism. This study investigated the response of BMNC to normal (SF) and inflamed synovial fluid (ISF). Equine BMNC cultured in autologous SF or ISF (n = 8 horses) developed into macrophage-rich cultures with phenotypes similar to cells native to normal SF and became more confluent in ISF (~100%) than SF (~25%). BMNC cultured in SF or ISF were neither M1- nor M2-like, but exhibited aspects of both phenotypes and a regulatory immune response, characterized by increasing counts of IL-10+ macrophages, decreasing IL-1β concentrations and progressively increasing IL-10 and IGF-1 concentrations. Changes were more marked in ISF and suggest that homeostatic mechanisms were preserved over time and were potentially favored by progressive cell proliferation. Collectively, our data suggest that intra-articular BMNC could increase synovial macrophage counts, potentiating the macrophage- and IL-10-associated mechanisms of joint homeostasis lost during the progression of OA, preserving the production of cytokines involved in tissue repair (PGE2 , IL-10) generally impaired by frequently used corticosteroids.
Collapse
Affiliation(s)
- Bruno C Menarim
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Kiersten H Gillis
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Andrea Oliver
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Caitlin Mason
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Stephen R Werre
- Laboratory for Study Design and Statistical Analysis, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Xin Luo
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Christopher R Byron
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Theodore S Kalbfleisch
- Maxwell Gluck Equine Research Center, College of Agricultural and Veterinary Sciences, University of Kentucky, Lexington, KY, USA
| | - James N MacLeod
- Maxwell Gluck Equine Research Center, College of Agricultural and Veterinary Sciences, University of Kentucky, Lexington, KY, USA
| | - Linda A Dahlgren
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| |
Collapse
|
17
|
Okubo R, Asawa Y, Watanabe M, Nagata S, Nio M, Takato T, Hikita A, Hoshi K. Proliferation medium in three-dimensional culture of auricular chondrocytes promotes effective cartilage regeneration in vivo. Regen Ther 2019; 11:306-315. [PMID: 31687424 PMCID: PMC6818372 DOI: 10.1016/j.reth.2019.10.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/11/2019] [Accepted: 10/01/2019] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Cartilage regeneration have been attracted attentions because of the poor ability of cartilage tissues to regenerate. Three-dimensional (3D) culture of chondrocytes is considered to be advantageous for cartilage regeneration. Although it is plausible that maturation of the constructs before transplantation positively affects the chondrogenesis, matured constructs after cultures for longer periods do not necessarily result in effective cartilage regeneration. In this study, we compared different types of culture media including growth factors which are clinically available. We prepared differentiation medium containing insulin-like growth factor-1 (IGF-1), proliferation medium containing fibroblast growth factor-2 (FGF-2) and insulin, and combination of them, and compared their efficacies on chondrogenesis when used in 3D culture of engineered cartilage constructs. METHODS Cartilage constructs were fabricated by auricular chondrocytes and atelocollagen, and they were 3D-cultured with four types of media: control medium, differentiation medium, proliferation medium, and combination medium. After 3 weeks of culture, the constructs were analyzed for cell number, gene and protein expressions and mechanical properties. The constructs were also transplanted into nude mice. After 8 weeks, the degree of cartilage regeneration was evaluated. Constructs manufactured with canine auricular chondrocytes were subjected to autologous transplantation into beagles and examined for cartilage regeneration. RESULTS During 3D culture, remarkably high gene expression of type II collagen was detected in the construct cultured with the differentiation medium whereas cell apoptosis were suppressed in the proliferation medium. When transplanted into nude mice, the constructs 3D-cultured in the proliferation medium produced abundant cartilage matrices. In autologous implantation model, the construct cultured in the proliferation medium again showed better chondrogenesis than those in other media. CONCLUSIONS The present study indicates that 3D culture with the proliferation medium maintains the cell viability to potentiate the subsequent cartilage regeneration. Here, we propose that not only differentiation but also high cell viability accompanied by proliferation factors should be taken into account to improve cartilage regeneration.
Collapse
Affiliation(s)
- Ryuji Okubo
- Department of Pediatric Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryomachi, Aoba-ku, Sendai 980-8574, Japan
| | - Yukiyo Asawa
- Department of Cell & Tissue Engineering (Fujisoft), Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Makoto Watanabe
- Department of Cell & Tissue Engineering (Fujisoft), Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Satoru Nagata
- Nagata Microtia and Reconstructive Plastic Surgery Clinic, Sasameminamicho 22-1, Toda, Saitama 335-0035, Japan
| | - Masaki Nio
- Department of Pediatric Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryomachi, Aoba-ku, Sendai 980-8574, Japan
| | - Tsuyoshi Takato
- JR Tokyo General Hospital, Yoyogi 2-1-3, Shibuya-ku, Tokyo 151-8528, Japan
| | - Atsuhiko Hikita
- Department of Cell & Tissue Engineering (Fujisoft), Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kazuto Hoshi
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
18
|
Törnqvist AE, Sophocleous A, Ralston SH, Ohlsson C, Svensson J. Liver-derived IGF-I is not required for protection against osteoarthritis in male mice. Am J Physiol Endocrinol Metab 2019; 317:E1150-E1157. [PMID: 31638855 DOI: 10.1152/ajpendo.00330.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Insulin-like growth factor-I (IGF-I) is anabolic for cartilage and important for cartilage integrity, which might suggest a connection between IGF-I and osteoarthritis (OA) development. However, the results of studies performed so far are conflicting, and we aimed to clarify the role of endocrine IGF-I in rodent OA. Male mice with inducible inactivation of circulating, liver-derived IGF-I (LI-IGF-I-/- mice, serum IGF-I reduced by ~80%) were used. Experimental OA was induced in young adult LI-IGF-I-/- and control mice by destabilization of the medial meniscus (DMM); age-related OA was also evaluated in 1-yr-old mice. DMM-operated LI-IGF-I-/- mice had thinner lateral subchondral bone plate in tibia compared with control mice, whereas osteophyte volume and articular cartilage damage were unaffected at the medial side of the DMM knee. However, the control mice but not the LI-IGF-I-/- mice also developed mild OA on the lateral side of the DMM knee compared with the unoperated knee. One-year-old LI-IGF-I-/- mice had lower mid-diaphyseal cortical bone area than the 1-yr-old control mice, whereas analyses of joint tissues displayed smaller osteophyte volume and thicker calcified cartilage than the control mice. There was no difference in OA severity in the articular cartilage between old LI-IGF-I-/- and control mice. Our study is the first to investigate whether there is an association between circulating IGF-I and OA in mice. We conclude that, although there is an ~80% reduction of circulating IGF-I and a decrease in cortical bone in male LI-IGF-I-/- mice, cartilage damage is clearly not intensified and may instead be slightly reduced.
Collapse
Affiliation(s)
- Anna E Törnqvist
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
- Rheumatology and Bone Diseases Unit, Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, United Kingdom
| | - Antonia Sophocleous
- Rheumatology and Bone Diseases Unit, Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, United Kingdom
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Stuart H Ralston
- Rheumatology and Bone Diseases Unit, Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, United Kingdom
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Johan Svensson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| |
Collapse
|
19
|
Geiger BC, Wang S, Padera RF, Grodzinsky AJ, Hammond PT. Cartilage-penetrating nanocarriers improve delivery and efficacy of growth factor treatment of osteoarthritis. Sci Transl Med 2019; 10:10/469/eaat8800. [PMID: 30487252 DOI: 10.1126/scitranslmed.aat8800] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 11/07/2018] [Indexed: 11/02/2022]
Abstract
Osteoarthritis is a debilitating joint disease affecting nearly 30 million people for which there are no disease-modifying therapies. Several drugs that have failed clinical trials have shown inefficient and inadequate delivery to target cells. Anabolic growth factors are one class of such drugs that could be disease-modifying if delivered directly to chondrocytes, which reside deep within dense, anionic cartilage tissue. To overcome this biological barrier, we conjugated a growth factor to a cationic nanocarrier for targeted delivery to chondrocytes and retention within joint cartilage after direct intra-articular injection. The nanocarrier uses reversible electrostatic interactions with anionic cartilage tissue to improve tissue binding, penetration, and residence time. Amine terminal polyamidoamine (PAMAM) dendrimers were end functionalized with variable molar ratios of poly(ethylene glycol) (PEG) to control surface charge. From this small family of variably PEGylated dendrimers, an optimal formulation showing 70% uptake into cartilage tissue and 100% cell viability was selected. When conjugated to insulin-like growth factor 1 (IGF-1), the dendrimer penetrated bovine cartilage of human thickness within 2 days and enhanced therapeutic IGF-1 joint residence time in rat knees by 10-fold for up to 30 days. In a surgical model of rat osteoarthritis, a single injection of dendrimer-IGF-1 rescued cartilage and bone more effectively than free IGF-1. Dendrimer-IGF-1 reduced width of cartilage degeneration by 60% and volumetric osteophyte burden by 80% relative to untreated rats at 4 weeks after surgery. These results suggest that PEGylated PAMAM dendrimer nanocarriers could improve pharmacokinetics and efficacy of disease-modifying osteoarthritis drugs in the clinic.
Collapse
Affiliation(s)
- Brett C Geiger
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.,Koch Institute for Integrative Cancer Research, 500 Main Street, Cambridge, MA 02142, USA
| | - Sheryl Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.,Koch Institute for Integrative Cancer Research, 500 Main Street, Cambridge, MA 02142, USA
| | - Robert F Padera
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 45 Francis Street, Boston, MA 02115, USA
| | - Alan J Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.,Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.,Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Paula T Hammond
- Koch Institute for Integrative Cancer Research, 500 Main Street, Cambridge, MA 02142, USA. .,Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| |
Collapse
|
20
|
Abstract
Nanoparticulate materials displaying enzyme-like properties, so-called nanozymes, are explored as substitutes for natural enzymes in several industrial, energy-related, and biomedical applications. Outstanding high stability, enhanced catalytic activities, low cost, and availability at industrial scale are some of the fascinating features of nanozymes. Furthermore, nanozymes can also be equipped with the unique attributes of nanomaterials such as magnetic or optical properties. Due to the impressive development of nanozymes during the last decade, their potential in the context of tissue engineering and regenerative medicine also started to be explored. To highlight the progress, in this review, we discuss the two most representative nanozymes, namely, cerium- and iron-oxide nanomaterials, since they are the most widely studied. Special focus is placed on their applications ranging from cardioprotection to therapeutic angiogenesis, bone tissue engineering, and wound healing. Finally, current challenges and future directions are discussed.
Collapse
|
21
|
Shi S, Mercer S, Eckert GJ, Trippel SB. Regulation of articular chondrocyte catabolic genes by growth factor interaction. J Cell Biochem 2019; 120:11127-11139. [PMID: 30809855 PMCID: PMC6716380 DOI: 10.1002/jcb.28389] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/26/2018] [Accepted: 01/09/2019] [Indexed: 01/25/2023]
Abstract
Osteoarthritis is characterized by a loss of articular cartilage homeostasis in which degradation exceeds formation. Several growth factors have been shown to promote cartilage formation by augmenting articular chondrocyte anabolic activity. This study tests the hypothesis that such growth factors also play an anticatabolic role. We transferred individual or combinations of the genes encoding insulin-like growth factor-I, bone morphogenetic protein-2, bone morphogenetic protein-7, transforming growth factor-β1, and fibroblast growth factor-2, into adult bovine articular chondrocytes and measured the expression of catabolic marker genes encoding A disintegrin and metalloproteinase with thrombospondin motifs-4 and -5, matrix metalloproteinases-3 and -13, and interleukin-6. When delivered individually, or in combination, these growth factor transgenes differentially regulated the direction, magnitude, and time course of expression of the catabolic marker genes. In concert, the growth factor transgenes regulated the marker genes in an interactive fashion that ranged from synergistic inhibition to synergistic stimulation. Synergistic stimulation prevailed over synergistic inhibition, reaching maxima of 15.2- and 2.7-fold, respectively. Neither the magnitude nor the time course of the effect of the transgene combinations could be predicted on the basis of the individual transgene effects. With few exceptions, the data contradict our hypothesis. The results demonstrate that growth factors that are traditionally viewed as chondrogenic tend also to promote catabolic gene expression. The competing actions of these potential therapeutic agents add an additional level of complexity to the selection of regulatory factors for restoring articular cartilage homeostasis or promoting repair.
Collapse
Affiliation(s)
- Shuiliang Shi
- Department of Orthopaedic Surgery, Indiana University
School of Medicine
| | - Scott Mercer
- Department of Orthopaedic Surgery, Indiana University
School of Medicine
| | - George J. Eckert
- Department of Biostatistics, Indiana University School of
Medicine
| | - Stephen B. Trippel
- Department of Orthopaedic Surgery, Indiana University
School of Medicine.,Department of Anatomy and Cell Biology, Indiana University
School of Medicine.,Department of Biomedical Engineering, Indiana University
Purdue University Indianapolis,To whom correspondence should be addressed:
Stephen B. Trippel: Department of Orthopaedics, Indiana University School of
Medicine, Indianapolis, IN 46202; ; Tel. (317)
278-0085; Fax. (317) 274-3702
| |
Collapse
|
22
|
Stefani RM, Halder SS, Estell EG, Lee AJ, Silverstein AM, Sobczak E, Chahine NO, Ateshian GA, Shah RP, Hung CT. A Functional Tissue-Engineered Synovium Model to Study Osteoarthritis Progression and Treatment. Tissue Eng Part A 2019; 25:538-553. [PMID: 30203722 PMCID: PMC6482911 DOI: 10.1089/ten.tea.2018.0142] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 08/31/2018] [Indexed: 01/15/2023] Open
Abstract
IMPACT STATEMENT The synovium envelops the diarthrodial joint and plays a key regulatory role in defining the composition of the synovial fluid through filtration and biosynthesis of critical boundary lubricants. Synovium changes often precede cartilage damage in osteoarthritis. We describe a novel in vitro tissue engineered model, validated against native synovium explants, to investigate the structure-function of synovium through quantitative solute transport measures. Synovium was evaluated in the presence of a proinflammatory cytokine, interleukin-1, or the clinically relevant corticosteroid, dexamethasone. We anticipate that a better understanding of synovium transport would support efforts to develop more effective strategies aimed at restoring joint health.
Collapse
Affiliation(s)
- Robert M. Stefani
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Saiti S. Halder
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Eben G. Estell
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Andy J. Lee
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Amy M. Silverstein
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Evie Sobczak
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Nadeen O. Chahine
- Department of Biomedical Engineering, Columbia University, New York, New York
- Department of Orthopedic Surgery, Columbia University, New York, New York
| | - Gerard A. Ateshian
- Department of Biomedical Engineering, Columbia University, New York, New York
- Department of Mechanical Engineering, Columbia University, New York, New York
| | - Roshan P. Shah
- Department of Orthopedic Surgery, Columbia University, New York, New York
| | - Clark T. Hung
- Department of Biomedical Engineering, Columbia University, New York, New York
| |
Collapse
|
23
|
Lasarzik de Ascurra J, Ehrle A, Einspanier R, Lischer C. Influence of Incubation Time and Incubation Tube on the Cytokine and Growth Factor Concentrations of Autologous Conditioned Serum in Horses. J Equine Vet Sci 2019; 75:30-34. [DOI: 10.1016/j.jevs.2018.12.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/16/2022]
|
24
|
Sato M, Tsutsui T, Moroi A, Yoshizawa K, Aikawa Y, Sakamoto H, Ueki K. Adaptive change in temporomandibular joint tissue and mandibular morphology following surgically induced anterior disc displacement by bFGF injection in a rabbit model. J Craniomaxillofac Surg 2018; 47:320-327. [PMID: 30579745 DOI: 10.1016/j.jcms.2018.11.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/27/2018] [Accepted: 11/29/2018] [Indexed: 10/27/2022] Open
Abstract
PURPOSE The purpose of this study was to examine the effect of injecting basic fibroblast growth factor following surgical induced anterior disc displacement in temporomandibular joints (TMJ). MATERIALS AND METHODS Adult male Japanese white rabbits (n = 16; 2.0-2.5 kg; 10 weeks old) were assigned to experimental and control groups. In the experimental group, anterior disc displacement was induced in the bilateral TMJ. Recombinant human basic fibroblast growth factor (rh bFGF) 0.1 μg/1 μL aqueous solution was injected into the left retro-discal connective tissue close to the disc (ADL group), and saline alone was injected into the same site on the right (ADR group). In the control group, a sham operation without disc position change was performed in the bilateral TMJ (CR group and CL group). Four animals from the experimental (ADR and ADL) and control (CR and CL) groups were sacrificed at 1 and 12 weeks postoperatively to evaluate the mandibular morphology and computed tomographic (CT) value of the condylar head, using 3 dimensional computed tomography. Furthermore, cartilage layers and disc tissue were examined histologically. RESULTS Regarding CT value at the 0° site of the condylar surface, ADR showed the lowest value after 1 week (P = 0.0325). However, there were no significant differences among the 4 groups regarding CT values at the other degree sites after 1 and 12 weeks. Regarding mandibular length, ADR showed the lowest value after 12 weeks (P = 0.0079). In condylar width, ADR showed the lowest value after 1 week (P = 0.0097). CONCLUSION This study suggested that surgically induced anterior disc displacement could affect condylar morphology in the early stage, and could decrease mandibular length in the late stage. However, bFGF injection into the TMJ might prevent the degenerative change derived from anterior disc displacement and inhibition of sequential mandibular growth.
Collapse
Affiliation(s)
- Momoko Sato
- Department of Oral and Maxillofacial Surgery (Head: Prof. Dr. K Ueki), Division of Medicine, Interdisciplinary Graduate School, University of Yamanashi, 1110 Shimokato, Chuo-shi, Yamanashi, 409-3898, Japan
| | - Takamitsu Tsutsui
- Department of Oral and Maxillofacial Surgery (Head: Prof. Dr. K Ueki), Division of Medicine, Interdisciplinary Graduate School, University of Yamanashi, 1110 Shimokato, Chuo-shi, Yamanashi, 409-3898, Japan
| | - Akinori Moroi
- Department of Oral and Maxillofacial Surgery (Head: Prof. Dr. K Ueki), Division of Medicine, Interdisciplinary Graduate School, University of Yamanashi, 1110 Shimokato, Chuo-shi, Yamanashi, 409-3898, Japan
| | - Kunio Yoshizawa
- Department of Oral and Maxillofacial Surgery (Head: Prof. Dr. K Ueki), Division of Medicine, Interdisciplinary Graduate School, University of Yamanashi, 1110 Shimokato, Chuo-shi, Yamanashi, 409-3898, Japan
| | - Yoshihito Aikawa
- Radiology Unit (Head: Radiology Technician. H Sakamoto), University of Yamanashi Hospital, 1110 Shimokato, Chuo-shi, Yamanashi, 409-3898, Japan
| | - Hajime Sakamoto
- Radiology Unit (Head: Radiology Technician. H Sakamoto), University of Yamanashi Hospital, 1110 Shimokato, Chuo-shi, Yamanashi, 409-3898, Japan
| | - Koichiro Ueki
- Department of Oral and Maxillofacial Surgery (Head: Prof. Dr. K Ueki), Division of Medicine, Interdisciplinary Graduate School, University of Yamanashi, 1110 Shimokato, Chuo-shi, Yamanashi, 409-3898, Japan.
| |
Collapse
|
25
|
Soul J, Dunn SL, Anand S, Serracino-Inglott F, Schwartz JM, Boot-Handford RP, Hardingham TE. Stratification of knee osteoarthritis: two major patient subgroups identified by genome-wide expression analysis of articular cartilage. Ann Rheum Dis 2017; 77:423. [PMID: 29273645 PMCID: PMC5867416 DOI: 10.1136/annrheumdis-2017-212603] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 11/21/2017] [Accepted: 11/28/2017] [Indexed: 12/13/2022]
Abstract
Introduction Osteoarthritis (OA) is a heterogeneous and complex disease. We have used a network biology approach based on genome-wide analysis of gene expression in OA knee cartilage to seek evidence for pathogenic mechanisms that may distinguish different patient subgroups. Methods Results from RNA-Sequencing (RNA-Seq) were collected from intact knee cartilage at total knee replacement from 44 patients with OA, from 16 additional patients with OA and 10 control patients with non-OA. Results were analysed to identify patient subsets and compare major active pathways. Results The RNA-Seq results showed 2692 differentially expressed genes between OA and non-OA. Analysis by unsupervised clustering identified two distinct OA groups: Group A with 24 patients (55%) and Group B with 18 patients (41%). A 10 gene subgroup classifier was validated by RT-qPCR in 16 further patients with OA. Pathway analysis showed increased protein expression in both groups. PhenomeExpress analysis revealed group differences in complement activation, innate immune responses and altered Wnt and TGFβ signalling, but no activation of inflammatory cytokine expression. Both groups showed suppressed circadian regulators and whereas matrix changes in Group A were chondrogenic, in Group B they were non-chondrogenic with changes in mechanoreceptors, calcium signalling, ion channels and in cytoskeletal organisers. The gene expression changes predicted 478 potential biomarkers for detection in synovial fluid to distinguish patients from the two groups. Conclusions Two subgroups of knee OA were identified by network analysis of RNA-Seq data with evidence for the presence of two major pathogenic pathways. This has potential importance as a new basis for the stratification of patients with OA for drug trials and for the development of new targeted treatments.
Collapse
Affiliation(s)
- Jamie Soul
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Sara L Dunn
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Sanjay Anand
- Department of Orthopaedic Surgery, Stockport NHS Foundation Trust, Stockport, UK
| | | | - Jean-Marc Schwartz
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Ray P Boot-Handford
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Tim E Hardingham
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| |
Collapse
|
26
|
Shi S, Kelly BJ, Wang C, Klingler K, Chan A, Eckert GJ, Trippel SB. Human IGF-I propeptide A promotes articular chondrocyte biosynthesis and employs glycosylation-dependent heparin binding. Biochim Biophys Acta Gen Subj 2017; 1862:567-575. [PMID: 29174671 DOI: 10.1016/j.bbagen.2017.11.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Insulin-like growth factor I (IGF-I) is a key regulator of chondrogenesis, but its therapeutic application to articular cartilage damage is limited by rapid elimination from the repair site. The human IGF-I gene gives rise to three IGF-I propeptides (proIGF-IA, proIGF-IB and proIGF-IC) that are cleaved to create mature IGF-I. In this study, we elucidate the processing of IGF-I precursors by articular chondrocytes, and test the hypotheses that proIGF-I isoforms bind to heparin and regulate articular chondrocyte biosynthesis. METHODS Human IGF-I propeptides and mutants were overexpressed in bovine articular chondrocytes. IGF-I products were characterized by ELISA, western blot and FPLC using a heparin column. The biosynthetic activity of IGF-I products on articular chondrocytes was assayed for DNA and glycosaminoglycan that the cells produced. RESULTS Secreted IGF-I propeptides stimulated articular chondrocyte biosynthetic activity to the same degree as mature IGF-I. Of the three IGF-I propeptides, only one, proIGF-IA, strongly bound to heparin. Interestingly, heparin binding of proIGF-IA depended on N-glycosylation at Asn92 in the EA peptide. To our knowledge, this is the first demonstration that N-glycosylation determines the binding of a heparin-binding protein to heparin. CONCLUSION The biosynthetic and heparin binding abilities of proIGF-IA, coupled with its generation of IGF-I, suggest that proIGF-IA may have therapeutic value for articular cartilage repair. GENERAL SIGNIFICANCE These data identify human pro-insulin-like growth factor IA as a bifunctional protein. Its combined ability to bind heparin and augment chondrocyte biosynthesis makes it a promising therapeutic agent for cartilage damage due to trauma and osteoarthritis.
Collapse
Affiliation(s)
- Shuiliang Shi
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States.
| | - Brian J Kelly
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Congrong Wang
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Ken Klingler
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Albert Chan
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - George J Eckert
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Stephen B Trippel
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Orthopaedic Surgery Service, Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, United States
| |
Collapse
|
27
|
Sermer C, Kandel R, Anderson J, Hurtig M, Theodoropoulos J. Platelet‐rich plasma enhances the integration of bioengineered cartilage with native tissue in an
in vitro
model. J Tissue Eng Regen Med 2017; 12:427-436. [DOI: 10.1002/term.2468] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 03/16/2017] [Accepted: 05/04/2017] [Indexed: 01/06/2023]
Affiliation(s)
- Corey Sermer
- Department of Surgery, Division of OrthopaedicsMt. Sinai and Women's College Hospital Toronto Ontario Canada
- Department of Pathology and Laboratory Medicine, Lunenfeld‐Tannenbaum Research InstituteMt. Sinai Hospital Toronto Ontario Canada
| | - Rita Kandel
- Department of Pathology and Laboratory Medicine, Lunenfeld‐Tannenbaum Research InstituteMt. Sinai Hospital Toronto Ontario Canada
| | - Jesse Anderson
- Department of Surgery, Division of OrthopaedicsMt. Sinai and Women's College Hospital Toronto Ontario Canada
| | - Mark Hurtig
- Department of Clinical Studies, Ontario Veterinary CollegeUniversity of Guelph Guelph Canada
| | - John Theodoropoulos
- Department of Surgery, Division of OrthopaedicsMt. Sinai and Women's College Hospital Toronto Ontario Canada
| |
Collapse
|
28
|
van Dalen SCM, Blom AB, Slöetjes AW, Helsen MMA, Roth J, Vogl T, van de Loo FAJ, Koenders MI, van der Kraan PM, van den Berg WB, van den Bosch MHJ, van Lent PLEM. Interleukin-1 is not involved in synovial inflammation and cartilage destruction in collagenase-induced osteoarthritis. Osteoarthritis Cartilage 2017; 25:385-396. [PMID: 27654963 DOI: 10.1016/j.joca.2016.09.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 09/05/2016] [Accepted: 09/12/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Interleukin-1 (IL-1) is an alleged important cytokine in osteoarthritis (OA), although the exact contribution of IL-1 to joint destruction remains unclear. Here we investigated the involvement of IL-1α and IL-1β in joint pathology during collagenase-induced OA (CiOA). METHODS CiOA was induced in wild type (WT) and IL-1αβ-/- mice. Additionally, IL-1 signaling was inhibited in WT mice with CiOA using osmotic pumps containing IL-1RA. Joint pathology was assessed using histology. Activity of cartilage-degrading enzymes was determined using antibodies against aggrecan neo-epitopes VDIPEN and NITEGE. Synovial gene expression was analyzed using quantitative real-time polymerase chain reaction (qRT-PCR). Serum protein levels were measured with Luminex or enzyme-linked immunosorbent assay (ELISA). RESULTS Synovial IL-1β expression was strongly elevated 7 days after induction of CiOA in WT mice but decreased afterwards, whereas S100A8/A9, previously described to aggravate OA, remained elevated for 21 days. Remarkably, synovial inflammation was comparable between WT and IL-1αβ-/- mice on day 7 of CiOA. In line, synovial mRNA expression of genes involved in IL-1 signaling and inflammatory mediators was comparable between WT and IL-1αβ-/- mice, and serum levels for Keratinocyte Chemoattractant (KC)/IL-6/S100A8/S100A9/IL-10 were equal. Synovial matrix metalloproteinase (MMP)/aggrecanase expression and activity in cartilage was not different in WT and IL-1αβ-/- mice on day 7 of CiOA. Cartilage destruction on day 42 was not different between WT and IL-1αβ-/- mice, which was supported by our finding that IL-1RA treatment in WT mice with CiOA did not alter joint destruction. CONCLUSIONS IL-1α and IL-1β are not involved in synovial inflammation and cartilage destruction during CiOA, implicating that other mediators are responsible for the joint damage.
Collapse
Affiliation(s)
- S C M van Dalen
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - A B Blom
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - A W Slöetjes
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - M M A Helsen
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - J Roth
- Institute of Immunology, University of Münster, Münster, Germany.
| | - T Vogl
- Institute of Immunology, University of Münster, Münster, Germany.
| | - F A J van de Loo
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - M I Koenders
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - P M van der Kraan
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - W B van den Berg
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - M H J van den Bosch
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - P L E M van Lent
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
29
|
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
|
30
|
Synovial chondromatosis of the temporomandibular joint: Immunohistochemical examinations regarding the role of insulin-like growth factors and their binding proteins in the etiology of this disease. J Craniomaxillofac Surg 2017; 45:198-202. [DOI: 10.1016/j.jcms.2016.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 11/28/2016] [Accepted: 12/07/2016] [Indexed: 11/21/2022] Open
|
31
|
Wang J, Cao W, Niu F. Adenoviral vector expressing IGF-1 protects murine chondrogenic ATDC5 cells against hydrogen peroxide-induced mitochondrial dysfunction and apoptosis. J Toxicol Sci 2016; 40:585-95. [PMID: 26354375 DOI: 10.2131/jts.40.585] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Insulin-like growth factor-1 (IGF-1), with an age-related decline, regulates the proliferation and survival of multiple cell types, particularly stimulates cartilage matrix synthesis, and inhibits matrix degradation. The present study was to investigate the regulatory role of IGF-1 against hydrogen peroxide(H2O2)-induced mitochondrial dysfunction and apoptosis in murine chondrocytic ATDC5 cells. We firstly determined mitochondrial dysfunction and apoptosis in ATDC5 cells which were exposed to H2O2. We then constructed an IGF-1-overexpressed adenovirus (IGF-1-Ad) harboring the IGF-1 coding sequence, and investigated the regulatory role of the overexpressed IGF-1 against the H2O2-induced mitochondrial dysfunction and apoptosis in ATDC5 cells. It was demonstrated that H2O2 treatment promoted the mitochondrial dysfunction, and further reduced the viability and induced apoptosis of ATDC5 cells. However, the IGF-1 overexpression by adenovirus inhibited the H2O2-induced mitochondrial dysfunction and further inhibited the H2O2-promoted apoptosis in ATDC5 cells. In conclusion, the present study found that oxidative stress promoted mitochondrial dysfunction and induced apoptosis in the murine chondrocytic ATDC5 cells, and the adenoviral vector-expressed IGF-1 protected the murine chondrocytic ATDC5 cells against such mitochondrial dysfunction and apoptosis. This study implies the protective role of IGF-1 against the oxidative stress in murine chondrocytic ATDC5 cells and demonstrates the promising anti-oxidative stress effect of the recombinant IGF-1-Ad against oxidative stress in chondrocytic cells.
Collapse
Affiliation(s)
- Jinfeng Wang
- Department of Orthopaedics, Tianjin 4th Centre Hospital, China
| | | | | |
Collapse
|
32
|
Kostereva NV, Wang Y, Fletcher DR, Unadkat JV, Schnider JT, Komatsu C, Yang Y, Stolz DB, Davis MR, Plock JA, Gorantla VS. IGF-1 and Chondroitinase ABC Augment Nerve Regeneration after Vascularized Composite Limb Allotransplantation. PLoS One 2016; 11:e0156149. [PMID: 27272754 PMCID: PMC4896437 DOI: 10.1371/journal.pone.0156149] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 05/10/2016] [Indexed: 12/04/2022] Open
Abstract
Impaired nerve regeneration and inadequate recovery of motor and sensory function following peripheral nerve repair remain the most significant hurdles to optimal functional and quality of life outcomes in vascularized tissue allotransplantation (VCA). Neurotherapeutics such as Insulin-like Growth Factor-1 (IGF-1) and chondroitinase ABC (CH) have shown promise in augmenting or accelerating nerve regeneration in experimental models and may have potential in VCA. The aim of this study was to evaluate the efficacy of low dose IGF-1, CH or their combination (IGF-1+CH) on nerve regeneration following VCA. We used an allogeneic rat hind limb VCA model maintained on low-dose FK506 (tacrolimus) therapy to prevent rejection. Experimental animals received neurotherapeutics administered intra-operatively as multiple intraneural injections. The IGF-1 and IGF-1+CH groups received daily IGF-1 (intramuscular and intraneural injections). Histomorphometry and immunohistochemistry were used to evaluate outcomes at five weeks. Overall, compared to controls, all experimental groups showed improvements in nerve and muscle (gastrocnemius) histomorphometry. The IGF-1 group demonstrated superior distal regeneration as confirmed by Schwann cell (SC) immunohistochemistry as well as some degree of extrafascicular regeneration. IGF-1 and CH effectively promote nerve regeneration after VCA as confirmed by histomorphometric and immunohistochemical outcomes.
Collapse
Affiliation(s)
- Nataliya V. Kostereva
- Department of Plastic Surgery, Thomas E. Starzl Transplant Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Yong Wang
- Department of Plastic Surgery, Thomas E. Starzl Transplant Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Trauma Surgery, East Hospital of Shanghai, Shanghai, China
| | - Derek R. Fletcher
- Department of Plastic Surgery, Thomas E. Starzl Transplant Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jignesh V. Unadkat
- Department of Plastic Surgery, Thomas E. Starzl Transplant Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jonas T. Schnider
- Department of Plastic Surgery, Thomas E. Starzl Transplant Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Chiaki Komatsu
- Department of Plastic Surgery, Thomas E. Starzl Transplant Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Yang Yang
- Department of Plastic Surgery, Thomas E. Starzl Transplant Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Donna B. Stolz
- Center for Biological Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Michael R. Davis
- United States Army Institute for Surgical Research, San Antonio Military Medical Center, 3698 Chambers Road, San Antonio, Texas, United States of America
| | - Jan A. Plock
- Department of Plastic Surgery, Thomas E. Starzl Transplant Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Vijay S. Gorantla
- Department of Plastic Surgery, Thomas E. Starzl Transplant Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
33
|
Bottini M, Bhattacharya K, Fadeel B, Magrini A, Bottini N, Rosato N. Nanodrugs to target articular cartilage: An emerging platform for osteoarthritis therapy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:255-68. [DOI: 10.1016/j.nano.2015.09.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 09/16/2015] [Indexed: 01/12/2023]
|
34
|
Tan AR, VandenBerg CD, Attur M, Abramson SB, Knight MM, Bulinski JC, Ateshian GA, Cook JL, Hung CT. Cytokine preconditioning of engineered cartilage provides protection against interleukin-1 insult. Arthritis Res Ther 2015; 17:361. [PMID: 26667364 PMCID: PMC4704536 DOI: 10.1186/s13075-015-0876-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 11/26/2015] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND During osteoarthritis and following surgical procedures, the environment of the knee is rich in proinflammatory cytokines such as IL-1. Introduction of tissue-engineered cartilage constructs to a chemically harsh milieu may limit the functionality of the implanted tissue over long periods. A chemical preconditioning scheme (application of low doses of IL-1) was tested as a method to prepare developing engineered tissue to withstand exposure to a higher concentration of the cytokine, known to elicit proteolysis as well as rapid degeneration of cartilage. METHODS Using an established juvenile bovine model system, engineered cartilage was preconditioned with low doses of IL-1α (0.1 ng/mL, 0.5 ng/mL, and 1.0 ng/mL) for 7 days before exposure to an insult dose (10 ng/mL). The time frame over which this protection is afforded was investigated by altering the amount of time between preconditioning and insult as well as the time following insult. To explore a potential mechanism for this protection, one set of constructs was preconditioned with CoCl2, a chemical inducer of hypoxia, before exposure to the IL-1α insult. Finally, we examined the translation of this preconditioning method to extend to clinically relevant adult, passaged chondrocytes from a preclinical canine model. RESULTS Low doses of IL-1α (0.1 ng/mL and 0.5 ng/mL) protected against subsequent catabolic degradation by cytokine insult, preserving mechanical stiffness and biochemical composition. Regardless of amount of time between preconditioning scheme and insult, protection was afforded. In a similar manner, preconditioning with CoCl2 similarly allowed for mediation of catabolic damage by IL-1α. The effects of preconditioning on clinically relevant adult, passaged chondrocytes from a preclinical canine model followed the same trends with low-dose IL-1β offering variable protection against insult. CONCLUSIONS Chemical preconditioning schemes have the ability to protect engineered cartilage constructs from IL-1-induced catabolic degradation, offering potential modalities for therapeutic treatments.
Collapse
Affiliation(s)
- Andrea R Tan
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, NY, USA.
| | - Curtis D VandenBerg
- Department of Orthopedic Surgery, St. Luke's-Roosevelt Hospital Center, 1000 10th Avenue, New York, NY, USA.
| | - Mukundan Attur
- New York University Hospital for Joint Disease, 301 E. 17th Street, New York, NY, USA.
| | - Steven B Abramson
- New York University Hospital for Joint Disease, 301 E. 17th Street, New York, NY, USA.
| | - Martin M Knight
- Institute of Bioengineering and School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, London, UK.
| | - J Chloe Bulinski
- Department of Biological Sciences, Columbia University, 1212 Amsterdam Avenue, New York, NY, USA.
| | - Gerard A Ateshian
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, NY, USA.
- Department of Mechanical Engineering, Columbia University, 500 W. 120th Street, New York, NY, USA.
| | - James L Cook
- Comparative Orthopaedic Laboratory, University of Missouri, 1100 Virginia Avenue, Columbia, MO, USA.
| | - Clark T Hung
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, NY, USA.
| |
Collapse
|
35
|
Wardale J, Mullen L, Howard D, Ghose S, Rushton N. An ex vivo model using human osteoarthritic cartilage demonstrates the release of bioactive insulin-like growth factor-1 from a collagen-glycosaminoglycan scaffold. Cell Biochem Funct 2015; 33:277-84. [PMID: 26059711 PMCID: PMC4528234 DOI: 10.1002/cbf.3112] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/01/2015] [Accepted: 04/07/2015] [Indexed: 01/04/2023]
Abstract
Biomimetic scaffolds hold great promise for therapeutic repair of cartilage, but although most scaffolds are tested with cells in vitro, there are very few ex vivo models (EVMs) where adult cartilage and scaffolds are co-cultured to optimize their interaction prior to in vivo studies. This study describes a simple, non-compressive method that is applicable to mammalian or human cartilage and provides a reasonable throughput of samples. Rings of full-depth articular cartilage slices were derived from human donors undergoing knee replacement for osteoarthritis and a 3 mm core of a collagen/glycosaminoglycan biomimetic scaffold (Tigenix, UK) inserted to create the EVM. Adult osteoarthritis chondrocytes were seeded into the scaffold and cultures maintained for up to 30 days. Ex vivo models were stable throughout experiments, and cells remained viable. Chondrocytes seeded into the EVM attached throughout the scaffold and in contact with the cartilage explants. Cell migration and deposition of extracellular matrix proteins in the scaffold was enhanced by growth factors particularly if the scaffold was preloaded with growth factors. This study demonstrates that the EVM represents a suitable model that has potential for testing a range of therapeutic parameters such as numbers/types of cell, growth factors or therapeutic drugs before progressing to costly pre-clinical trials. © 2015 The Authors. Cell Biochemistry and Function Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- J Wardale
- Orthopaedic Research Unit, University of Cambridge, Cambridge, UK
| | | | - D Howard
- Orthopaedic Research Unit, University of Cambridge, Cambridge, UK
| | | | - N Rushton
- Orthopaedic Research Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
36
|
Pasold J, Zander K, Heskamp B, Grüttner C, Lüthen F, Tischer T, Jonitz-Heincke A, Bader R. Positive impact of IGF-1-coupled nanoparticles on the differentiation potential of human chondrocytes cultured on collagen scaffolds. Int J Nanomedicine 2015; 10:1131-43. [PMID: 25709437 PMCID: PMC4327566 DOI: 10.2147/ijn.s72872] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE In the present study, silica nanoparticles (sNP) coupled with insulin-like growth factor 1 (IGF-1) were loaded on a collagen-based scaffold intended for cartilage repair, and the influence on the viability, proliferation, and differentiation potential of human primary articular chondrocytes was examined. METHODS Human chondrocytes were isolated from the hyaline cartilage of patients (n=4, female, mean age: 73±5.1 years) undergoing primary total knee joint replacement. Cells were dedifferentiated and then cultivated on a bioresorbable collagen matrix supplemented with fluorescent sNP coupled with IGF-1 (sNP-IGF-1). After 3, 7, and 14 days of cultivation, cell viability and integrity into the collagen scaffold as well as metabolic cell activity and synthesis rate of matrix proteins (collagen type I and II) were analyzed. RESULTS The number of vital cells increased over 14 days of cultivation, and the cells were able to infiltrate the collagen matrix (up to 120 μm by day 7). Chondrocytes cultured on the collagen scaffold supplemented with sNP-IGF-1 showed an increase in metabolic activity (5.98-fold), and reduced collagen type I (1.58-fold), but significantly increased collagen type II expression levels (1.53-fold; P=0.02) after 7 days of cultivation compared to 3 days. In contrast, chondrocytes grown in a monolayer on plastic supplemented with sNP-IGF-1 had significantly lower metabolic activity (1.32-fold; P=0.007), a consistent amount of collagen type I, and significantly reduced collagen type II protein expression (1.86-fold; P=0.001) after 7 days compared to 3 days. CONCLUSION Collagen-based scaffolds enriched with growth factors, such as IGF-1 coupled to nanoparticles, represent an improved therapeutic intervention for the targeted and controlled treatment of articular cartilage lesions.
Collapse
Affiliation(s)
- Juliane Pasold
- Department of Orthopaedics, Biomechanics and Implant Technology Laboratory, University Medicine Rostock, Rostock, Germany
| | - Kathleen Zander
- Department of Orthopaedics, Biomechanics and Implant Technology Laboratory, University Medicine Rostock, Rostock, Germany
| | - Benjamin Heskamp
- Department of Orthopaedics, Biomechanics and Implant Technology Laboratory, University Medicine Rostock, Rostock, Germany
| | | | - Frank Lüthen
- Institute of Cell Biology, University Medicine Rostock, Rostock, Germany
| | - Thomas Tischer
- Department of Orthopaedics, Biomechanics and Implant Technology Laboratory, University Medicine Rostock, Rostock, Germany
| | - Anika Jonitz-Heincke
- Department of Orthopaedics, Biomechanics and Implant Technology Laboratory, University Medicine Rostock, Rostock, Germany
| | - Rainer Bader
- Department of Orthopaedics, Biomechanics and Implant Technology Laboratory, University Medicine Rostock, Rostock, Germany
| |
Collapse
|
37
|
Effects of insulin-like growth factor-1 and dexamethasone on cytokine-challenged cartilage: relevance to post-traumatic osteoarthritis. Osteoarthritis Cartilage 2015; 23:266-74. [PMID: 25450855 PMCID: PMC4304966 DOI: 10.1016/j.joca.2014.11.006] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 10/27/2014] [Accepted: 11/02/2014] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Interleukin-1 is one of the inflammatory cytokines elevated after traumatic joint injury that plays a critical role in mediating cartilage tissue degradation, suppressing matrix biosynthesis, and inducing chondrocyte apoptosis, events associated with progression to post-traumatic osteoarthritis (PTOA). We studied the combined use of insulin-like growth factor-1 (IGF-1) and dexamethasone (Dex) to block these multiple degradative effects of cytokine challenge to articular cartilage. METHODS Young bovine and adult human articular cartilage explants were treated with IL-1α in the presence or absence of IGF-1, Dex, or their combination. Loss of sulfated glycosaminoglycans (sGAG) and collagen were evaluated by the DMMB and hydroxyproline assays, respectively. Matrix biosynthesis was measured via radiolabel incorporation, chondrocyte gene expression by qRT-PCR, and cell viability by fluorescence staining. RESULTS In young bovine cartilage, the combination of IGF-1 and Dex significantly inhibited the loss of sGAG and collagen, rescued the suppression of matrix biosynthesis, and inhibited the loss of chondrocyte viability caused by IL-1α treatment. In adult human cartilage, only IGF-1 rescued matrix biosynthesis and only Dex inhibited sGAG loss and improved cell viability. Thus, the combination of IGF-1 + Dex together showed combined beneficial effects in human cartilage. CONCLUSIONS Our findings suggest that the combination of IGF-1 and Dex has greater beneficial effects than either molecule alone in preventing cytokine-mediated cartilage degradation in adult human and young bovine cartilage. Our results support the use of such a combined approach as a potential treatment relevant to early cartilage degradative changes associated with joint injury.
Collapse
|
38
|
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
|
39
|
Ponnurangam S, O'Connell GD, Chernyshova IV, Wood K, Hung CTH, Somasundaran P. Beneficial effects of cerium oxide nanoparticles in development of chondrocyte-seeded hydrogel constructs and cellular response to interleukin insults. Tissue Eng Part A 2014; 20:2908-19. [PMID: 24762195 PMCID: PMC4229709 DOI: 10.1089/ten.tea.2013.0592] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 04/24/2014] [Indexed: 12/18/2022] Open
Abstract
The harsh inflammatory environment associated with injured and arthritic joints represents a major challenge to articular cartilage repair. In this study, we report the effect of cerium oxide nanoparticles, or nanoceria, in modulating development of engineered cartilage and in combating the deleterious effects of interleukin-1α. Nanoceria was found to be biocompatible with bovine chondrocytes up to a concentration of 1000 μg/mL (60,000 cells/μg of nanoceria), and its presence significantly improved compressive mechanical properties and biochemical composition (i.e., glycosaminoglycans) of engineered cartilage. Raman microspectroscopy revealed that individual chondrocytes with internalized nanoceria have increased concentrations of proline, procollagen, and glycogen as compared with cells without the nanoparticles in their vicinity. The inflammatory response due to physiologically relevant quantities of interluekin-1α (0.5 ng/mL) is partially inhibited by nanoceria. To the best of the authors' knowledge, these results are the first to demonstrate a high potential for nanoceria to improve articular cartilage tissue properties and for their long-term treatment against an inflammatory reaction.
Collapse
Affiliation(s)
- Sathish Ponnurangam
- Department of Earth and Environmental Engineering, Columbia University, New York, New York
| | - Grace D. O'Connell
- Department of Mechanical Engineering, University of California, Berkeley, California
| | - Irina V. Chernyshova
- Department of Earth and Environmental Engineering, Columbia University, New York, New York
| | - Katherine Wood
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Clark Tung-Hui Hung
- Department of Biomedical Engineering, Columbia University, New York, New York
| | | |
Collapse
|
40
|
Florine EM, Miller RE, Liebesny PH, Mroszczyk KA, Lee RT, Patwari P, Grodzinsky AJ. Delivering heparin-binding insulin-like growth factor 1 with self-assembling peptide hydrogels. Tissue Eng Part A 2014; 21:637-46. [PMID: 25231349 DOI: 10.1089/ten.tea.2013.0679] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Heparin-binding insulin-like growth factor 1 (HB-IGF-1) is a fusion protein of IGF-1 with the HB domain of heparin-binding epidermal growth factor-like growth factor. A single dose of HB-IGF-1 has been shown to bind specifically to cartilage and to promote sustained upregulation of proteoglycan synthesis in cartilage explants. Achieving strong integration between native cartilage and tissue-engineered cartilage remains challenging. We hypothesize that if a growth factor delivered by the tissue engineering scaffold could stimulate enhanced matrix synthesis by both the cells within the scaffold and the adjacent native cartilage, integration could be enhanced. In this work, we investigated methods for adsorbing HB-IGF-1 to self-assembling peptide hydrogels to deliver the growth factor to encapsulated chondrocytes and cartilage explants cultured with growth factor-loaded hydrogels. We tested multiple methods for adsorbing HB-IGF-1 in self-assembling peptide hydrogels, including adsorption prior to peptide assembly, following peptide assembly, and with/without heparan sulfate (HS, a potential linker between peptide molecules and HB-IGF-1). We found that HB-IGF-1 and HS were retained in the peptide for all tested conditions. A subset of these conditions was then studied for their ability to stimulate increased matrix production by gel-encapsulated chondrocytes and by chondrocytes within adjacent native cartilage. Adsorbing HB-IGF-1 or IGF-1 prior to peptide assembly was found to stimulate increased sulfated glycosaminoglycan per DNA and hydroxyproline content of chondrocyte-seeded hydrogels compared with basal controls at day 10. Cartilage explants cultured adjacent to functionalized hydrogels had increased proteoglycan synthesis at day 10 when HB-IGF-1 was adsorbed, but not IGF-1. We conclude that delivery of HB-IGF-1 to focal defects in cartilage using self-assembling peptide hydrogels is a promising technique that could aid cartilage repair via enhanced matrix production and integration with native tissue.
Collapse
Affiliation(s)
- Emily M Florine
- 1 Biological Engineering, Massachusetts Institute of Technology , Cambridge, Massachusetts
| | | | | | | | | | | | | |
Collapse
|
41
|
Ortved KF, Begum L, Mohammed HO, Nixon AJ. Implantation of rAAV5-IGF-I transduced autologous chondrocytes improves cartilage repair in full-thickness defects in the equine model. Mol Ther 2014; 23:363-73. [PMID: 25311491 DOI: 10.1038/mt.2014.198] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 10/02/2014] [Indexed: 01/18/2023] Open
Abstract
Cartilage injury often precipitates osteoarthritis which has driven research to bolster repair in cartilage impact damage. Autologous chondrocytes transduced with rAAV5-IGF-I were evaluated in chondral defects in a well-established large animal model. Cartilage was harvested from the talus of 24 horses; chondrocytes were isolated and stored frozen. Twenty million cells were cultured and transduced with 10(5) AAV vg/cell prior to implantation. Chondrocytes from eight horses were transduced with rAAV5-IGF-I, chondrocytes from eight horses with rAAV5-GFP, and chondrocytes from eight horses were not transduced. A 15 mm full-thickness chondral defect was created arthroscopically in the lateral trochlear ridge of the femur in both femoropatellar joints. Treated defects were filled with naive or gene-enhanced chondrocytes, in fibrin vehicle. Control defects in the opposite limb received fibrin alone. rAAV5-IGF-I transduced chondrocytes resulted in significantly better healing at 8 week arthroscopy and 8 month necropsy examination when compared to controls. At 8 months, defects implanted with cells expressing IGF-I had better histological scores compared to control defects and defects repaired with naive chondrocytes. This included increased chondrocyte predominance and collagen type II, both features of hyaline-like repair tissue. The equine model closely approximates human cartilage healing, indicating AAV-mediated genetic modification of chondrocytes may be clinically beneficial to humans.
Collapse
Affiliation(s)
- Kyla F Ortved
- Comparative Orthopaedics Laboratory, Cornell University, Ithaca, New York, USA
| | - Laila Begum
- Comparative Orthopaedics Laboratory, Cornell University, Ithaca, New York, USA
| | - Hussni O Mohammed
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Alan J Nixon
- Comparative Orthopaedics Laboratory, Cornell University, Ithaca, New York, USA
| |
Collapse
|
42
|
Loffredo FS, Pancoast JR, Cai L, Vannelli T, Dong JZ, Lee RT, Patwari P. Targeted delivery to cartilage is critical for in vivo efficacy of insulin-like growth factor 1 in a rat model of osteoarthritis. Arthritis Rheumatol 2014; 66:1247-55. [PMID: 24470361 DOI: 10.1002/art.38357] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 01/07/2014] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Acute articular injuries lead to an increased risk of progressive joint damage and osteoarthritis (OA), and no therapies are currently available to repair or protect the injured joint tissue. Intraarticular delivery of therapeutic proteins has been limited by their rapid clearance from the joint space and lack of retention within cartilage. The aim of this study was to test whether targeted delivery to cartilage by fusion with a heparin-binding domain would be sufficient to prolong the in vivo function of the insulin-like growth factor 1 (IGF-1). METHODS We produced a humanized and optimized recombinant HB-IGF-1 fusion protein. By injecting HB-IGF-1, IGF-1, or saline alone into the knee joints of adult Lewis rats, we tested whether fusion with a heparin-binding domain 1) altered the kinetics of retention in joint tissues, 2) prolonged functional stimulation as measured by radiolabel incorporation, and 3) enhanced efficacy in a rat model of surgically induced OA, using weekly injections. RESULTS Fusion of heparin-binding domain with IGF-1 prolonged retention in articular and meniscal cartilage from <1 day to 8 days after injection. Unmodified IGF-1 had no functional effect 2 days after injection, whereas HB-IGF-1 stimulated meniscal cartilage at least 4 days after injection. HB-IGF-1, but not IGF-1, significantly slowed cartilage damage in a rat model of OA. CONCLUSION Heparin-binding domain fusions can transform rapidly cleared proteins into potential intraarticular therapies by targeting them to cartilage.
Collapse
Affiliation(s)
- Francesco S Loffredo
- Brigham and Women's Hospital and Harvard Medical School, Cambridge, Massachusetts
| | | | | | | | | | | | | |
Collapse
|
43
|
Evans DS, Cailotto F, Parimi N, Valdes AM, Castaño-Betancourt MC, Liu Y, Kaplan RC, Bidlingmaier M, Vasan RS, Teumer A, Tranah GJ, Nevitt MC, Cummings SR, Orwoll ES, Barrett-Connor E, Renner JB, Jordan JM, Doherty M, Doherty SA, Uitterlinden AG, van Meurs JBJ, Spector TD, Lories RJ, Lane NE. Genome-wide association and functional studies identify a role for IGFBP3 in hip osteoarthritis. Ann Rheum Dis 2014; 74:1861-7. [PMID: 24928840 DOI: 10.1136/annrheumdis-2013-205020] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 05/22/2014] [Indexed: 01/10/2023]
Abstract
OBJECTIVES To identify genetic associations with hip osteoarthritis (HOA), we performed a meta-analysis of genome-wide association studies (GWAS) of HOA. METHODS The GWAS meta-analysis included approximately 2.5 million imputed HapMap single nucleotide polymorphisms (SNPs). HOA cases and controls defined radiographically and by total hip replacement were selected from the Osteoporotic Fractures in Men (MrOS) Study and the Study of Osteoporotic Fractures (SOF) (654 cases and 4697 controls, combined). Replication of genome-wide significant SNP associations (p ≤5×10(-8)) was examined in five studies (3243 cases and 6891 controls, combined). Functional studies were performed using in vitro models of chondrogenesis and osteogenesis. RESULTS The A allele of rs788748, located 65 kb upstream of the IGFBP3 gene, was associated with lower HOA odds at the genome-wide significance level in the discovery stage (OR 0.71, p=2×10(-8)). The association replicated in five studies (OR 0.92, p=0.020), but the joint analysis of discovery and replication results was not genome-wide significant (p=1×10(-6)). In separate study populations, the rs788748 A allele was also associated with lower circulating IGFBP3 protein levels (p=4×10(-13)), suggesting that this SNP or a variant in linkage disequilibrium could be an IGFBP3 regulatory variant. Results from functional studies were consistent with association results. Chondrocyte hypertrophy, a deleterious event in OA pathogenesis, was largely prevented upon IGFBP3 knockdown in chondrocytes. Furthermore, IGFBP3 overexpression induced cartilage catabolism and osteogenic differentiation. CONCLUSIONS Results from GWAS and functional studies provided suggestive links between IGFBP3 and HOA.
Collapse
Affiliation(s)
- Daniel S Evans
- California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Frederic Cailotto
- Laboratory of Tissue Homeostasis and Disease, Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Neeta Parimi
- California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Ana M Valdes
- Department of Academic Rheumatology, University of Nottingham, Nottingham City Hospital, Nottingham, UK
| | - Martha C Castaño-Betancourt
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands The Netherlands Genomics Initiative-sponsored Netherlands Consortium for Healthy Aging (NGI-NCHA), Rotterdam/Leiden, The Netherlands
| | - Youfang Liu
- Departments of Medicine and Orthopedics, Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Martin Bidlingmaier
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ramachandran S Vasan
- California Pacific Medical Center Research Institute, San Francisco, California, USA Laboratory of Tissue Homeostasis and Disease, Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium Department of Academic Rheumatology, University of Nottingham, Nottingham City Hospital, Nottingham, UK Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands The Netherlands Genomics Initiative-sponsored Netherlands Consortium for Healthy Aging (NGI-NCHA), Rotterdam/Leiden, The Netherlands Departments of Medicine and Orthopedics, Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA Albert Einstein College of Medicine, Bronx, New York, USA Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, Munich, Germany Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, Massachusetts, USA Institute of Functional Genomics, Ernst Moritz Arndt University, University of Greifswald, Greifswald, Germany Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA School of Medicine, Oregon Health & Science University, Portland, Oregon, USA Division of Epidemiology, Departments of Family and Preventive Medicine and Medicine, University of California San Diego, La Jolla, California, USA Departments of Medicine and Radiology, Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands Department of Twin Research and Genetic Epidemiology Unit, King's College London, London, UK Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium University of California at Davis, Sacramento, California, USA
| | - Alexander Teumer
- Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, Massachusetts, USA Institute of Functional Genomics, Ernst Moritz Arndt University, University of Greifswald, Greifswald, Germany
| | - Gregory J Tranah
- California Pacific Medical Center Research Institute, San Francisco, California, USA Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| | - Michael C Nevitt
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| | - Steven R Cummings
- California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Eric S Orwoll
- School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Elizabeth Barrett-Connor
- Division of Epidemiology, Departments of Family and Preventive Medicine and Medicine, University of California San Diego, La Jolla, California, USA
| | - Jordan B Renner
- Departments of Medicine and Radiology, Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joanne M Jordan
- Departments of Medicine and Orthopedics, Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michael Doherty
- Department of Academic Rheumatology, University of Nottingham, Nottingham City Hospital, Nottingham, UK
| | - Sally A Doherty
- Department of Academic Rheumatology, University of Nottingham, Nottingham City Hospital, Nottingham, UK
| | - Andre G Uitterlinden
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands The Netherlands Genomics Initiative-sponsored Netherlands Consortium for Healthy Aging (NGI-NCHA), Rotterdam/Leiden, The Netherlands Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Joyce B J van Meurs
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology Unit, King's College London, London, UK
| | - Rik J Lories
- Laboratory of Tissue Homeostasis and Disease, Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Nancy E Lane
- University of California at Davis, Sacramento, California, USA
| |
Collapse
|
44
|
Development and retranslational validation of an in vitro model to characterize acute infections in large human joints. BIOMED RESEARCH INTERNATIONAL 2014; 2014:848604. [PMID: 24877141 PMCID: PMC4021994 DOI: 10.1155/2014/848604] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 04/09/2014] [Indexed: 11/17/2022]
Abstract
Bacterial infections can destroy cartilage integrity, resulting in osteoarthritis. Goal was to develop an in vitro model with in vivo validation of acute joint inflammation. Inflammation in cocultivated human synovial fibroblasts (SFB), chondrocytes (CHDR), and mononuclear cells (MNC) was successively relieved for 10 days. Articular effusions from patients with (n = 7) and without (n = 5) postoperative joint infection in healthy patients (ASA 1-2) were used as model validation. Inflammation in vitro resulted in an enormous increase in IL-1 and a successive reduction in SFB numbers. CHDR however, maintained metabolic activity and proteoglycan synthesis. While concentrations of bFGF in vivo and in vitro rose consistently, the mRNA increase was only moderate. Concurring with our in vivo data, cartilage-specific IGF-1 steadily increased, while IGF-1 mRNA in the CHDR and SFB did not correlate with protein levels. Similarly, aggrecan (ACAN) protein concentrations increased in vivo and failed to correlate in vitro with gene expression in either the CHDR or the SFB, indicating extracellular matrix breakdown. Anabolic cartilage-specific BMP-7 with highly significant intra-articular levels was significantly elevated in vitro on day 10 following maximum inflammation. Our in vitro model enables us to validate early inflammation of in vivo cell- and cytokine-specific regulatory patterns. This trial is registered with MISSinG, DRKS 00003536.
Collapse
|
45
|
Abstract
Context: Articular cartilage has a unique functional architecture capable of providing a lifetime of pain-free joint motion. This tissue, however, undergoes substantial age-related physiologic, mechanical, biochemical, and functional changes that reduce its ability to overcome the effects of mechanical stress and injury. Many factors affect joint function in the maturing athlete—from chondrocyte survival and metabolism to structural composition and genetic/epigenetic factors governing cartilage and synovium. An evaluation of age-related changes for joint homeostasis and risk for osteoarthritis is important to the development of new strategies to rejuvenate aging joints. Objective: This review summarizes the current literature on the biochemical, cellular, and physiologic changes occurring in aging articular cartilage. Data Sources: PubMed (1969-2013) and published books in sports health, cartilage biology, and aging. Study Selection: Keywords included aging, athlete, articular cartilage, epigenetics, and functional performance with age. Study Design: Systematic review. Level of Evidence: Level 3. Data Extraction: To be included, research questions addressed the effect of age-related changes on performance, articular cartilage biology, molecular mechanism, and morphology. Results: The mature athlete faces challenges in maintaining cartilage health and joint function due to age-related changes to articular cartilage biology, morphology, and physiology. These changes include chondrocyte loss and a decline in metabolic response, alterations to matrix and synovial tissue composition, and dysregulation of reparative responses. Conclusion: Although physical decline has been regarded as a normal part of aging, many individuals maintain overall fitness and enjoy targeted improvement to their athletic capacity throughout life. Healthy articular cartilage and joints are needed to maintain athletic performance and general activities. Genetic and potentially reversible epigenetic factors influence cartilage physiology and its response to mechanical and injurious stimuli. Improved understandings of the physical and molecular changes to articular cartilage with aging are important to develop successful strategies for joint rejuvenation.
Collapse
Affiliation(s)
- Ayala Luria
- Department of Orthopaedic Surgery, Stanford School of Medicine, Stanford, California
| | - Constance R Chu
- Department of Orthopaedic Surgery, Stanford School of Medicine, Stanford, California
| |
Collapse
|
46
|
Milner PI, Smith HC, Robinson R, Wilkins RJ, Gibson JS. Growth factor regulation of intracellular pH homeostasis under hypoxic conditions in isolated equine articular chondrocytes. J Orthop Res 2013; 31:197-203. [PMID: 22987771 DOI: 10.1002/jor.22221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 08/07/2012] [Indexed: 02/04/2023]
Abstract
Hypoxia and acidosis are recognized features of inflammatory arthroses. This study describes the effects of IGF-1 and TGF-β(1) on pH regulatory mechanisms in articular cartilage under hypoxic conditions. Acid efflux, reactive oxygen species (ROS), and mitochondrial membrane potential were measured in equine articular chondrocytes isolated in the presence of serum (10% fetal calf serum), IGF-1 (1, 10, 50, 100 ng/ml) or TGF-β(1) (0.1, 1, 10 ng/ml) and then exposed to a short-term (3 h) hypoxic insult (1% O(2)). Serum and 100 ng/ml IGF-1 but not TGF-β(1) attenuated hypoxic regulation of pH homeostasis. IGF-1 appeared to act through mitochondrial membrane potential stabilization and maintenance of intracellular ROS levels in very low levels of oxygen. Using protein phosphorylation inhibitors PD98059 (25 µM) and wortmannin (200 nM) and Western blotting, ERK1/2 and PI-3 kinase pathways are important for the effect of IGF-1 downstream to ROS generation in normoxia but only PI-3 kinase is implicated in hypoxia. These results show that oxygen and growth factors interact to regulate pH recovery in articular chondrocytes by modulating intracellular oxygen metabolites.
Collapse
Affiliation(s)
- Peter I Milner
- Faculty of Health and Life Sciences, Department of Musculoskeletal Biology, University of Liverpool, Leahurst Campus, Neston CH64 7TE, United Kingdom.
| | | | | | | | | |
Collapse
|
47
|
Insulin is essential for in vitro chondrogenesis of mesenchymal progenitor cells and influences chondrogenesis in a dose-dependent manner. INTERNATIONAL ORTHOPAEDICS 2012; 37:153-8. [PMID: 23229799 DOI: 10.1007/s00264-012-1726-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 11/17/2012] [Indexed: 10/27/2022]
Abstract
PURPOSE Insulin is a commonly used additive in chondrogenic media for differentiating mesenchymal stem cells (MSCs). The indispensability of other bioactive factors like TGF-β or dexamethasone in these medium formulations has been shown, but the role of insulin is unclear. The purpose of this study was to investigate whether insulin is essential for MSC chondrogenesis and if there is a dose-dependent effect of insulin on MSC chondrogenesis. METHODS We cultivated human MSCs in pellet culture in serum-free chondrogenic medium with insulin concentrations between 0 and 50 μg/ml and assessed the grade of chondrogenic differentiation by histological evaluation and determination of glycosaminoglycan (GAG), total collagen and DNA content. We further tested whether insulin can be delivered in an amount sufficient for MSC chondrogenesis via a drug delivery system in insulin-free medium. RESULTS Chondrogenesis was not induced by standard chondrogenic medium without insulin and the expression of cartilage differentiation markers was dose-dependent at insulin concentrations between 0 and 10 μg/ml. An insulin concentration of 50 μg/ml had no additional effect compared with 10 μg/ml. Insulin was delivered by a release system into the cell culture under insulin-free conditions in an amount sufficient to induce chondrogenesis. CONCLUSIONS Insulin is essential for MSC chondrogenesis in this system and chondrogenic differentiation is influenced by insulin in a dose-dependent manner. Insulin can be provided in a sufficient amount by a drug delivery system. Therefore, insulin is a suitable and inexpensive indicator substance for testing drug release systems in vitro.
Collapse
|
48
|
Kisiday JD, McIlwraith CW, Rodkey WG, Frisbie DD, Steadman J. Effects of Platelet-Rich Plasma Composition on Anabolic and Catabolic Activities in Equine Cartilage and Meniscal Explants. Cartilage 2012; 3:245-54. [PMID: 26069637 PMCID: PMC4297115 DOI: 10.1177/1947603511433181] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE To evaluate the effects of single- and double-spin preparations of platelet-rich plasma (PRP) on anabolic and catabolic activities of cartilage and meniscal explants in vitro. METHODS Single- and double-spin PRP was prepared using laboratory processing or commercial kits. The cellular contents were quantified, and each PRP was mixed in equal quantities with cell culture medium and added to cartilage or meniscus explant cultures, with or without interleukin 1 β (IL-1β). Extracellular matrix synthesis was quantified over 24 hours via (35)S-sulfate and (3)H-proline incorporation, while gene expression of catabolic enzymes was evaluated using real-time PCR. RESULTS The platelet concentration in single-spin laboratory PRP was 59% higher than blood. Platelet and white blood cell concentrations in single-spin laboratory and kit PRP were not significantly different, while the double-spin kit resulted in approximately 2.5-fold higher platelet and approximately 400-fold higher white blood cell concentrations. In cartilage cultures without IL-1β, radiolabel incorporation in single-spin PRP cultures was significantly higher than in double-spin cultures. Similar results were obtained for (35)S-sulfate incorporation in meniscus cultures without IL-1β. In IL-1β, radiolabel incorporation was largely similar among all PRPs. After 24 hours of culture, ADAMTS-4 gene expression in cartilage was lowest for single-spin PRP, while expression in the double-spin kit was not significantly different from double-spin laboratory PRP in which platelets were concentrated 6-fold. CONCLUSIONS This study suggests that single-spin PRP preparations may be the most advantageous for intra-articular applications and that double-spin systems should be considered with caution.
Collapse
Affiliation(s)
- John D. Kisiday
- Department of Clinical Science, Orthopaedic Research Center, Colorado State University, Fort Collins, CO, USA
| | - C. Wayne McIlwraith
- Department of Clinical Science, Orthopaedic Research Center, Colorado State University, Fort Collins, CO, USA
| | | | - David D. Frisbie
- Department of Clinical Science, Orthopaedic Research Center, Colorado State University, Fort Collins, CO, USA
| | | |
Collapse
|
49
|
JONITZ ANIKA, LOCHNER KATRIN, TISCHER THOMAS, HANSMANN DORIS, BADER RAINER. TGF-β1 and IGF-1 influence the re-differentiation capacity of human chondrocytes in 3D pellet cultures in relation to different oxygen concentrations. Int J Mol Med 2012; 30:666-72. [DOI: 10.3892/ijmm.2012.1042] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 06/05/2012] [Indexed: 11/06/2022] Open
|
50
|
|