1
|
Kuroiwa T, Tsuboi Y, Michikawa T, Tajima K, Uraya Y, Maeda A, Shizu K, Suzuki K, Suzuki K, Kawano Y, Fujita N. DNA methylation of bone morphogenetic protein 7 in leukocytes as a possible biomarker for hand osteoarthritis: A pilot study. J Orthop Res 2024. [PMID: 39182186 DOI: 10.1002/jor.25963] [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: 04/23/2024] [Revised: 08/01/2024] [Accepted: 08/08/2024] [Indexed: 08/27/2024]
Abstract
Hand osteoarthritis (HOA), characterized by an earlier onset age and reduced susceptibility to mechanical stress compared with knee and hip osteoarthritis, is considered a suitable disease for identifying predictive biomarkers of osteoarthritis. In particular, DNA methylation variants, expected to contribute to HOA susceptibility, hold potential as osteoarthritis biomarkers. In this study, leukocyte DNA methylation patterns were analyzed in blood samples from patients with HOA, aiming to identify disease-specific biomarkers for osteoarthritis. Using DNA methylation microarrays, we analyzed samples from three subjects with HOA and three age- and gender-matched healthy individuals. For validation, pyrosequencing analysis was conducted using samples from 16 to 9 subjects with and without HOA, respectively. From 735,026 probes in the DNA methylation array, the Top 100 CpG sites associated with HOA, based on low adjusted P-values, including those targeting bone morphogenetic protein 7 (BMP7), SBF2-AS1, PLOD2, ICOS, and CSF1R were identified. Validation analysis revealed significantly higher methylation levels in the BMP7-related site in the HOA group compared with the control group, even after adjusting for age, gender, and body mass index (p = 0.037). In contrast, no significant difference was observed in the other selected CpG sites between the HOA and control groups. This study highlights the significantly increased frequency of methylation at the specific BMP7 site in leukocytes of patients with HOA, suggesting its potential as a biomarker for HOA. Measurement of methylation levels at the CpG sites identified in this study offers a potential approach to prevent future osteoarthritis progression, providing valuable insights into disease management.
Collapse
Affiliation(s)
- Takashi Kuroiwa
- Department of Orthopaedic Surgery, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Yoshiki Tsuboi
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
| | - Takehiro Michikawa
- Department of Environmental and Occupational Health, School of Medicine, Toho University, Tokyo, Japan
| | - Kaori Tajima
- Department of Orthopaedic Surgery, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Yuki Uraya
- Department of Orthopaedic Surgery, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Atsushi Maeda
- Department of Orthopaedic Surgery, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Kanae Shizu
- Department of Orthopaedic Surgery, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Katsuji Suzuki
- Department of Orthopaedic Surgery, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Koji Suzuki
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
| | - Yusuke Kawano
- Department of Orthopaedic Surgery, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Nobuyuki Fujita
- Department of Orthopaedic Surgery, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| |
Collapse
|
2
|
Zhang C, Jiang C, Jin J, Lei P, Cai Y, Wang Y. Cartilage fragments combined with BMSCs-Derived exosomes can promote tendon-bone healing after ACL reconstruction. Mater Today Bio 2023; 23:100819. [PMID: 37810754 PMCID: PMC10550801 DOI: 10.1016/j.mtbio.2023.100819] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/11/2023] [Accepted: 09/23/2023] [Indexed: 10/10/2023] Open
Abstract
Anterior cruciate ligament reconstruction (ACLR) often fails due to the inability of tendon-bone integration to regenerate normal tissues and formation of fibrous scar tissues in the tendon-bone interface. Cartilage fragments and exosomes derived from bone mesenchymal stromal cells (BMSCs-Exos) can enhance enthesis healing. Nevertheless, the effects on the tendon-bone healing of ACLR remain unknown. This study found that BMSCs-Exos can promote the proliferation of chondrocytes in cartilage fragments, and activated the expression of chondro-related genes SOX9 and Aggrecan. The optimal effect concentration was 1012 events/uL. Besides, BMSCs-Exos could significantly upregulated the expression of BMP7 and Smad5 in cartilage fragments, and further enhanced the expression of chondrogenic genes. Moreover, this study established a rat model of ACLR and implanted the BMSCs-Exos/cartilage fragment complex into the femoral bone tunnel. Results demonstrated that the mean diameters of the femoral bone tunnels were significantly smaller in the BE-CF group than those in the CF group (p = 0.038) and control group (p = 0.007) at 8 weeks after surgery. Besides, more new bone formation was observed in the femoral tunnels in the BE-CF group, as demonstrated by a larger BV/TV ratio based on the reconstructed CT scans. Histological results also revealed the regeneration of tendon-bone structures, especially fibrocartilage. Thus, these findings provide a promising result that BMSCs-Exos/cartilage fragment complex can prevent the enlargement of bone tunnel and promote tendon-bone healing after ACLR, which may have resulted from the regulation of the BMP7/Smad5 signaling axis.
Collapse
Affiliation(s)
- Chi Zhang
- Center for Sports Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310008, China
- Institute of Sports Medicine of Zhejiang University, 388 Yuhangtang Road, Hangzhou, 310030, China
| | - Chao Jiang
- Spine Lab, Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiale Jin
- Center for Sports Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310008, China
| | - Pengfei Lei
- Center for Sports Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310008, China
| | - Youzhi Cai
- Center for Sports Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310008, China
- Institute of Sports Medicine of Zhejiang University, 388 Yuhangtang Road, Hangzhou, 310030, China
| | - Yue Wang
- Spine Lab, Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
3
|
Smith C, du Toit R, Ollewagen T. Potential of bone morphogenetic protein-7 in treatment of lupus nephritis: addressing the hurdles to implementation. Inflammopharmacology 2023; 31:2161-2172. [PMID: 37626268 PMCID: PMC10518293 DOI: 10.1007/s10787-023-01321-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023]
Abstract
Up to 50% of systemic lupus erythematosus (SLE) patients world-wide develop lupus nephritis (LN). In low to middle income countries and in particular in sub-Saharan Africa, where SLE is prevalent with a more aggressive course, LN and end stage renal disease is a major cause of mortality. While developed countries have the funding to invest in SLE and LN research, patients of African descent are often underrepresented in clinical trials. Thus, the complex influence of ethnicity and genetic background on outcome of LN and SLE as a whole, is not fully understood. Several pathophysiological mechanisms including major role players driving LN have been identified. A large body of literature suggest that prevention of fibrosis-which contributes to chronicity of LN-may significantly improve long-term prognosis. Bone morphogenetic protein-7 (BMP-7) was first identified as a therapeutic option in this context decades ago and evidence of its benefit in various conditions, including LN, is ever-increasing. Despite these facts, BMP-7 is not being implemented as therapy in the context of renal disease. With this review, we briefly summarise current understanding of LN pathology and discuss the evidence in support of therapeutic potential of BMP-7 in this context. Lastly, we address the obstacles that need to be overcome, before BMP-7 may become available as LN treatment.
Collapse
Affiliation(s)
- Carine Smith
- Experimental Medicine Research Group, Department Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Parow, South Africa.
| | - Riette du Toit
- Division Rheumatology, Department Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Parow, South Africa
| | - Tracey Ollewagen
- Experimental Medicine Research Group, Department Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Parow, South Africa
| |
Collapse
|
4
|
Makarczyk MJ, Hines S, Yagi H, Li ZA, Aguglia AM, Zbikowski J, Padget AM, Gao Q, Bunnell BA, Goodman SB, Lin H. Using Microphysiological System for the Development of Treatments for Joint Inflammation and Associated Cartilage Loss-A Pilot Study. Biomolecules 2023; 13:384. [PMID: 36830751 PMCID: PMC9952916 DOI: 10.3390/biom13020384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/19/2023] Open
Abstract
Osteoarthritis (OA) is a painful and disabling joint disease affecting millions worldwide. The lack of clinically relevant models limits our ability to predict therapeutic outcomes prior to clinical trials, where most drugs fail. Therefore, there is a need for a model that accurately recapitulates the whole-joint disease nature of OA in humans. Emerging microphysiological systems provide a new opportunity. We recently established a miniature knee joint system, known as the miniJoint, in which human bone-marrow-derived mesenchymal stem cells (hBMSCs) were used to create an osteochondral complex, synovial-like fibrous tissue, and adipose tissue analogs. In this study, we explored the potential of the miniJoint in developing novel treatments for OA by testing the hypothesis that co-treatment with anti-inflammation and chondroinducing agents can suppress joint inflammation and associated cartilage degradation. Specifically, we created a "synovitis"-relevant OA model in the miniJoint by treating synovial-like tissues with interleukin-1β (IL-1β), and then a combined treatment of oligodeoxynucleotides (ODNs) suppressing the nuclear factor kappa beta (NF-κB) genetic pathway and bone morphogenic protein-7 (BMP-7) was introduced. The combined treatment with BMP-7 and ODNs reduced inflammation in the synovial-like fibrous tissue and showed an increase in glycosaminoglycan formation in the cartilage portion of the osteochondral complex. For the first time, this study demonstrated the potential of the miniJoint in developing disease-modifying OA drugs. The therapeutic efficacy of co-treatment with NF-κB ODNs and BMP-7 can be further validated in future clinical studies.
Collapse
Affiliation(s)
- Meagan J. Makarczyk
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Rm 217, Pittsburgh, PA 15219, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, 450 Technology Drive, Rm 217, Pittsburgh, PA 15219, USA
| | - Sophie Hines
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Rm 217, Pittsburgh, PA 15219, USA
| | - Haruyo Yagi
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Rm 217, Pittsburgh, PA 15219, USA
| | - Zhong Alan Li
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Rm 217, Pittsburgh, PA 15219, USA
| | - Alyssa M. Aguglia
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Rm 217, Pittsburgh, PA 15219, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, 450 Technology Drive, Rm 217, Pittsburgh, PA 15219, USA
| | - Justin Zbikowski
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Rm 217, Pittsburgh, PA 15219, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, 450 Technology Drive, Rm 217, Pittsburgh, PA 15219, USA
| | - Anne-Marie Padget
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Rm 217, Pittsburgh, PA 15219, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94350, USA
| | - Bruce A. Bunnell
- Department of Microbiology, Immunology, and Genetics University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94350, USA
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Rm 217, Pittsburgh, PA 15219, USA
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94350, USA
| |
Collapse
|
5
|
Whitty C, Pernstich C, Marris C, McCaskie A, Jones M, Henson F. Sustained delivery of the bone morphogenetic proteins BMP-2 and BMP-7 for cartilage repair and regeneration in osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2022; 4:100240. [DOI: 10.1016/j.ocarto.2022.100240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/17/2022] [Accepted: 01/26/2022] [Indexed: 10/19/2022] Open
|
6
|
Application of Alginate Hydrogels for Next-Generation Articular Cartilage Regeneration. Int J Mol Sci 2022; 23:ijms23031147. [PMID: 35163071 PMCID: PMC8835677 DOI: 10.3390/ijms23031147] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 12/28/2022] Open
Abstract
The articular cartilage has insufficient intrinsic healing abilities, and articular cartilage injuries often progress to osteoarthritis. Alginate-based scaffolds are attractive biomaterials for cartilage repair and regeneration, allowing for the delivery of cells and therapeutic drugs and gene sequences. In light of the heterogeneity of findings reporting the benefits of using alginate for cartilage regeneration, a better understanding of alginate-based systems is needed in order to improve the approaches aiming to enhance cartilage regeneration with this compound. This review provides an in-depth evaluation of the literature, focusing on the manipulation of alginate as a tool to support the processes involved in cartilage healing in order to demonstrate how such a material, used as a direct compound or combined with cell and gene therapy and with scaffold-guided gene transfer procedures, may assist cartilage regeneration in an optimal manner for future applications in patients.
Collapse
|
7
|
Caron MMJ, Ripmeester EGJ, van den Akker G, Wijnands NKAP, Steijns J, Surtel DAM, Cremers A, Emans PJ, van Rhijn LW, Welting TJM. Discovery of bone morphogenetic protein 7-derived peptide sequences that attenuate the human osteoarthritic chondrocyte phenotype. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:247-261. [PMID: 33850953 PMCID: PMC8022858 DOI: 10.1016/j.omtm.2021.03.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 03/09/2021] [Indexed: 01/18/2023]
Abstract
Treatment of osteoarthritis (OA) is mainly symptomatic by alleviating pain to postpone total joint replacement. Bone morphogenetic protein 7 (BMP7) is a candidate morphogen for experimental OA treatment that favorably alters the chondrocyte and cartilage phenotype. Intra-articular delivery and sustained release of a recombinant growth factor for treating OA are challenging, whereas the use of peptide technology potentially circumvents many of these challenges. In this study, we screened a high-resolution BMP7 peptide library and discovered several overlapping peptide sequences from two regions in BMP7 with nanomolar bioactivity that attenuated the pathological OA chondrocyte phenotype. A single exposure of OA chondrocytes to peptides p[63-82] and p[113-132] ameliorated the OA chondrocyte phenotype for up to 8 days, and peptides were bioactive on chondrocytes in OA synovial fluid. Peptides p[63-82] and p[113-132] required NKX3-2 for their bioactivity on chondrocytes and provoke changes in SMAD signaling activity. The bioactivity of p[63-82] depended on specific evolutionary conserved sequence elements common to BMP family members. Intra-articular injection of a rat medial meniscal tear (MMT) model with peptide p[63-82] attenuated cartilage degeneration. Together, this study identified two regions in BMP7 from which bioactive peptides are able to attenuate the OA chondrocyte phenotype. These BMP7-derived peptides provide potential novel disease-modifying treatment options for OA.
Collapse
Affiliation(s)
- Marjolein M J Caron
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Ellen G J Ripmeester
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Guus van den Akker
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Nina K A P Wijnands
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Jessica Steijns
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Don A M Surtel
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Andy Cremers
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Pieter J Emans
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands.,Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, the Netherlands
| | - Lodewijk W van Rhijn
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands.,Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, the Netherlands
| | - Tim J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands.,Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, the Netherlands
| |
Collapse
|
8
|
Primorac D, Molnar V, Rod E, Jeleč Ž, Čukelj F, Matišić V, Vrdoljak T, Hudetz D, Hajsok H, Borić I. Knee Osteoarthritis: A Review of Pathogenesis and State-Of-The-Art Non-Operative Therapeutic Considerations. Genes (Basel) 2020; 11:E854. [PMID: 32722615 PMCID: PMC7464436 DOI: 10.3390/genes11080854] [Citation(s) in RCA: 172] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/11/2020] [Accepted: 07/23/2020] [Indexed: 02/07/2023] Open
Abstract
Being the most common musculoskeletal progressive condition, osteoarthritis is an interesting target for research. It is estimated that the prevalence of knee osteoarthritis (OA) among adults 60 years of age or older is approximately 10% in men and 13% in women, making knee OA one of the leading causes of disability in elderly population. Today, we know that osteoarthritis is not a disease characterized by loss of cartilage due to mechanical loading only, but a condition that affects all of the tissues in the joint, causing detectable changes in tissue architecture, its metabolism and function. All of these changes are mediated by a complex and not yet fully researched interplay of proinflammatory and anti-inflammatory cytokines, chemokines, growth factors and adipokines, all of which can be measured in the serum, synovium and histological samples, potentially serving as biomarkers of disease stage and progression. Another key aspect of disease progression is the epigenome that regulates all the genetic expression through DNA methylation, histone modifications, and mRNA interference. A lot of work has been put into developing non-surgical treatment options to slow down the natural course of osteoarthritis to postpone, or maybe even replace extensive surgeries such as total knee arthroplasty. At the moment, biological treatments such as platelet-rich plasma, bone marrow mesenchymal stem cells and autologous microfragmented adipose tissue containing stromal vascular fraction are ordinarily used. Furthermore, the latter two mentioned cell-based treatment options seem to be the only methods so far that increase the quality of cartilage in osteoarthritis patients. Yet, in the future, gene therapy could potentially become an option for orthopedic patients. In the following review, we summarized all of the latest and most important research in basic sciences, pathogenesis, and non-operative treatment.
Collapse
Affiliation(s)
- Dragan Primorac
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- Eberly College of Science, The Pennsylvania State University, University Park, State College, PA 16802, USA
- The Henry C. Lee College of Criminal Justice and Forensic Sciences, University of New Haven, West Haven, CT 06516, USA
- Medical School, University of Split, 21000 Split, Croatia
- School of Medicine, Faculty of Dental Medicine and Health, University “Josip Juraj Strossmayer”, 31000 Osijek, Croatia
- School of Medicine, JJ Strossmayer University of Osijek, 31000 Osijek, Croatia
- Medical School, University of Rijeka, 51000 Rijeka, Croatia
- Medical School REGIOMED, 96 450 Coburg, Germany
- Medical School, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
| | - Vilim Molnar
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- School of Medicine, JJ Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Eduard Rod
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- School of Medicine, JJ Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Željko Jeleč
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- School of Medicine, JJ Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Nursing, University North, 48 000 Varaždin, Croatia
| | - Fabijan Čukelj
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- Medical School, University of Split, 21000 Split, Croatia
| | - Vid Matišić
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
| | - Trpimir Vrdoljak
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- Department of Orthopedics, Clinical Hospital “Sveti Duh”, 10000 Zagreb, Croatia
| | - Damir Hudetz
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- School of Medicine, JJ Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Orthopedics, Clinical Hospital “Sveti Duh”, 10000 Zagreb, Croatia
| | - Hana Hajsok
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- Medical School, University of Zagreb, 10000 Zagreb, Croatia
| | - Igor Borić
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- Medical School, University of Split, 21000 Split, Croatia
- Medical School, University of Rijeka, 51000 Rijeka, Croatia
- Medical School, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
| |
Collapse
|
9
|
Abstract
Being the most common musculoskeletal progressive condition, osteoarthritis is an interesting target for research. It is estimated that the prevalence of knee osteoarthritis (OA) among adults 60 years of age or older is approximately 10% in men and 13% in women, making knee OA one of the leading causes of disability in elderly population. Today, we know that osteoarthritis is not a disease characterized by loss of cartilage due to mechanical loading only, but a condition that affects all of the tissues in the joint, causing detectable changes in tissue architecture, its metabolism and function. All of these changes are mediated by a complex and not yet fully researched interplay of proinflammatory and anti-inflammatory cytokines, chemokines, growth factors and adipokines, all of which can be measured in the serum, synovium and histological samples, potentially serving as biomarkers of disease stage and progression. Another key aspect of disease progression is the epigenome that regulates all the genetic expression through DNA methylation, histone modifications, and mRNA interference. A lot of work has been put into developing non-surgical treatment options to slow down the natural course of osteoarthritis to postpone, or maybe even replace extensive surgeries such as total knee arthroplasty. At the moment, biological treatments such as platelet-rich plasma, bone marrow mesenchymal stem cells and autologous microfragmented adipose tissue containing stromal vascular fraction are ordinarily used. Furthermore, the latter two mentioned cell-based treatment options seem to be the only methods so far that increase the quality of cartilage in osteoarthritis patients. Yet, in the future, gene therapy could potentially become an option for orthopedic patients. In the following review, we summarized all of the latest and most important research in basic sciences, pathogenesis, and non-operative treatment.
Collapse
|
10
|
Yan X, Zhou Z, Guo L, Zeng Z, Guo Z, Shao Q, Xu W. BMP7-overexpressing bone marrow-derived mesenchymal stem cells (BMSCs) are more effective than wild-type BMSCs in healing fractures. Exp Ther Med 2018; 16:1381-1388. [PMID: 30112066 DOI: 10.3892/etm.2018.6339] [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: 09/25/2017] [Accepted: 01/09/2018] [Indexed: 12/20/2022] Open
Abstract
Bone fractures are a worldwide public health concern. Previous studies have demonstrated that bone morphogenetic protein-7 (BMP7) gene transfer or mesenchymal stem cells (MSCs) transplantation may be a promising novel therapeutic approach. Therefore, the aim of the present study was to observe the effect of bone BMP7 transfer to MSCs on fracture healing. Bone marrow-derived MSCs (BMSCs) from New Zealand white rabbits were isolated and identified using flow cytometry. A recombinant BMP7 overexpressing adenovirus vector (Adv) was constructed and transfected into BMSCs. The expression of BMP7 was detected by reverse transcription-polymerase chain reaction, immunofluorescence and western blotting. The present study additionally investigated the effect of BMP7 on the differentiation capacity of BMSCs. Finally, tissue-engineered bone was created with support material to verify the effect of BMP7-BMSCs on fracture healing. The results demonstrated that the expression of BMP7 was increased at the mRNA and protein levels in BMSCs following transfection with BMP7 overexpressing Adv. The results additionally demonstrated that the expression of BMP7 enhanced the differentiation capacity of bone marrow mesenchymal stem cells and had a promotional effect on fracture healing. Overall, these data suggest that Adv-BMP7 is useful for introducing foreign genes into BMSCs and will be a powerful gene therapy tool for bone regeneration and other tissue engineering applications in the future.
Collapse
Affiliation(s)
- Xu Yan
- Department of Orthopedics, The 455th Hospital of PLA, Shanghai 200052, P.R. China
| | - Zhenhua Zhou
- Department of Orthopedic Oncology, Changzheng Hospital, The Second Military Medical University, Shanghai 200003, P.R. China.,Department of Orthopedics, The 169th Hospital of PLA, Hengyang, Hunan 421002, P.R. China.,Department of Orthopedics, Xiangnan Hospital, College of Medicine, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Lixin Guo
- Department of Orthopedics, The 169th Hospital of PLA, Hengyang, Hunan 421002, P.R. China.,Department of Orthopedics, Xiangnan Hospital, College of Medicine, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Zhaochi Zeng
- Department of Orthopedics, The 169th Hospital of PLA, Hengyang, Hunan 421002, P.R. China.,Department of Orthopedics, Xiangnan Hospital, College of Medicine, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Zhongkai Guo
- Department of Orthopedics, The 169th Hospital of PLA, Hengyang, Hunan 421002, P.R. China.,Department of Orthopedics, Xiangnan Hospital, College of Medicine, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Qingdong Shao
- Department of Orthopedics, The 455th Hospital of PLA, Shanghai 200052, P.R. China
| | - Weidong Xu
- Department of Orthopedics, Changhai Hospital Affiliated to The Second Military Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
11
|
Gavenis K, Schneider U, Groll J, Schmidt-Rohlfing B. BMP-7-Loaded PGLA Microspheres as a New Delivery System for the Cultivation of Human Chondrocytes in a Collagen Type I Gel: The Common Nude Mouse Model. Int J Artif Organs 2018. [DOI: 10.1177/039139881003300107] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Purpose Bone morphogenic protein 7 (BMP-7) released from polylactide (PLGA) microspheres has proven to be a potent system in cartilage tissue engineering in vitro. However, in vivo data are still lacking. The aim of this study was to investigate this BMP-7 release system utilizing the nude mouse as a small animal model. Methods Human osteoarthritic chondrocytes of 10 patients were enzymatically released and transferred into a collagen type-I gel. A concentration of 2×105 cells/mL was used. BMP-7 encapsulated in PGLA microspheres was added at an initial concentration of 500 ng BMP-7/mL gel. Untreated specimens and specimens with empty microspheres served as control. Samples were cultivated subcutaneously in nude mice for 6 weeks. Results After recovery, chondrocytes of all groups displayed a spheroid morphology without signs of dedifferentiation. The proteoglycan and collagen type II content of the control groups was restricted to the immediate pericellular region, whereas treatment group samples showed enhanced collagen type II production. Collagen type II and aggrecan gene expression was enhanced in treatment group samples with respect to the two control groups (mean ± SD: 0.268 ± 0.450 to 0.152 ± 0.129 and 0.155 ± 0.216 ng/ng β-actin for collagen type II; 0.535 ± 0.731 to 0.367 ± 0.651 and 0.405 ± 0.326 ng/ng β-actin for aggrecan), whereas collagen type I gene expression decreased by a factor of 10. Relative protein quantification of collagen type II, collagen type I and proteoglycan was in accordance. Conclusions Our data suggest that BMP-7 release from PGLA microspheres led to an improved tissue-engineered cartilage analogue in vivo with an increase in hyaline-cartilage-specific components.
Collapse
Affiliation(s)
- Karsten Gavenis
- Department of Orthopedic and Trauma Surgery, Aachen University Hospital, Aachen
| | | | - Jürgen Groll
- Deutsches Wollforschungsinstitut (DWI), Aachen - Germany
| | | |
Collapse
|
12
|
Gigout A, Guehring H, Froemel D, Meurer A, Ladel C, Reker D, Bay-Jensen AC, Karsdal MA, Lindemann S. Sprifermin (rhFGF18) enables proliferation of chondrocytes producing a hyaline cartilage matrix. Osteoarthritis Cartilage 2017; 25:1858-1867. [PMID: 28823647 DOI: 10.1016/j.joca.2017.08.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 07/24/2017] [Accepted: 08/08/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Fibroblast growth factor (FGF) 18 has been shown to increase cartilage volume when injected intra-articularly in animal models of osteoarthritis (OA) and in patients with knee OA (during clinical development of the recombinant human FGF18, sprifermin). However, the exact nature of this effect is still unknown. In this study, we aimed to investigate the effects of sprifermin at the cellular level. DESIGN A combination of different chondrocyte culture systems was used and the effects of sprifermin on proliferation, the phenotype and matrix production were evaluated. The involvement of MAPKs in sprifermin signalling was also studied. RESULTS In monolayer, we observed that sprifermin promoted a round cell morphology and stimulated both cellular proliferation and Sox9 expression while strongly decreasing type I collagen expression. In 3D culture, sprifermin increased the number of matrix-producing chondrocytes, improved the type II:I collagen ratio and enabled human OA chondrocytes to produce a hyaline extracellular matrix (ECM). Furthermore, we found that sprifermin displayed a 'hit and run' mode of action, with intermittent exposure required for the compound to fully exert its anabolic effect. Finally, sprifermin appeared to signal through activation of ERK. CONCLUSIONS Our results indicate that intermittent exposure to sprifermin leads to expansion of hyaline cartilage-producing chondrocytes. These in vitro findings are consistent with the increased cartilage volume observed in the knees of OA patients after intra-articular injection with sprifermin in clinical studies.
Collapse
Affiliation(s)
- A Gigout
- Osteoarthritis Research, Merck KGaA, Darmstadt, Germany.
| | - H Guehring
- Osteoarthritis Research, Merck KGaA, Darmstadt, Germany.
| | - D Froemel
- Orthopaedic University Hospital Friedrichsheim, Frankfurt, Germany.
| | - A Meurer
- Orthopaedic University Hospital Friedrichsheim, Frankfurt, Germany.
| | - C Ladel
- Osteoarthritis Research, Merck KGaA, Darmstadt, Germany.
| | - D Reker
- Rheumatology, Biomarkers and Research, Nordic Bioscience, Herlev, Denmark.
| | - A C Bay-Jensen
- Rheumatology, Biomarkers and Research, Nordic Bioscience, Herlev, Denmark.
| | - M A Karsdal
- Rheumatology, Biomarkers and Research, Nordic Bioscience, Herlev, Denmark.
| | - S Lindemann
- Osteoarthritis Research, Merck KGaA, Darmstadt, Germany.
| |
Collapse
|
13
|
Crecente-Campo J, Borrajo E, Vidal A, Garcia-Fuentes M. New scaffolds encapsulating TGF-β3/BMP-7 combinations driving strong chondrogenic differentiation. Eur J Pharm Biopharm 2017; 114:69-78. [PMID: 28087378 DOI: 10.1016/j.ejpb.2016.12.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/07/2016] [Accepted: 12/09/2016] [Indexed: 11/28/2022]
Abstract
The regeneration of articular cartilage remains an unresolved question despite the current access to a variety of tissue scaffolds activated with growth factors relevant to this application. Further advances might result from combining more than one of these factors; here, we propose a scaffold composition optimized for the dual delivery of BMP-7 and TGF-β3, two proteins with described chondrogenic activity. First, we tested in a mesenchymal stem cell micromass culture with TGF-β3 whether the exposure to microspheres loaded with BMP-7 would improve cartilage formation. Histology and qRT-PCR data confirmed that the sustained release of BMP-7 cooperates with TGF-β3 towards chondrogenic differentiation. Then, we optimized a scaffold prototype for tissue culture and dual encapsulation of BMP-7 and TGF-β3. The scaffolds were prepared from poly(lactic-co-glycolic acid), and BMP-7/TGF-β3 were loaded as nanocomplexes with heparin and Tetronic 1107. The scaffolds showed the sustained release of both proteins over four weeks, with minimal burst effect. We finally cultured human mesenchymal stem cells on these scaffolds, in the absence of exogenous chondrogenic factor supplementation. The cells cultured on the scaffolds loaded with BMP-7 and TGF-β3 showed clear signs of cartilage formation macroscopically and histologically. RT-PCR studies confirmed a clear upregulation of cartilage markers SOX9 and Aggrecan. In summary, scaffolds encapsulating BMP-7 and TGF-β3 can efficiently deliver a cooperative growth factor combination that drives efficient cartilage formation in human mesenchymal stem cell cultures. These results open attractive perspectives towards in vivo translation of this technology in cartilage regeneration experiments.
Collapse
Affiliation(s)
- Jose Crecente-Campo
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Avda. Barcelona s/n, 15782 Santiago de Compostela, Spain
| | - Erea Borrajo
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Avda. Barcelona s/n, 15782 Santiago de Compostela, Spain
| | - Anxo Vidal
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Avda. Barcelona s/n, 15782 Santiago de Compostela, Spain
| | - Marcos Garcia-Fuentes
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Avda. Barcelona s/n, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
14
|
Xia X, Li J, Xia B, Yang H, Zhang D, Zhou B, Zhang J, Zhou M, Liu F. Matrigel scaffold combined with Ad-hBMP7-transfected chondrocytes improves the repair of rabbit cartilage defect. Exp Ther Med 2016; 13:542-550. [PMID: 28352329 PMCID: PMC5348698 DOI: 10.3892/etm.2016.3972] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 04/27/2016] [Indexed: 12/03/2022] Open
Abstract
The aim of this study was to explore an effective method for the repair of cartilage defects using chitosan/glycerophosphate (C/GP) gel- and Matrigel-engineered human bone morphogenetic protein 7 (hBMP7)-expressing chondrocytes. Rabbit chondrocytes were obtained, cultured in vitro and transfected with an adenovirus containing hBMP7 and green fluorescent protein (Ad-hBMP7-GFP). The expression of hBMP7 in the transfected cells was tested by reverse transcription-polymerase chain reaction (RT-PCR) and western blotting. The phenotype of the transfected cells was evaluated by detecting the yields of collagen II and hyaluronic acid using RT-PCR and enzyme-linked immunosorbent assay (ELISA). The growth of chondrocytes in the C/GP gel and Matrigel was accessed by measuring the cell growth rate, hematoxylin and eosin (H&E) staining and observation under a scanning microscope. Twelve adult male New Zealand white rabbits were randomly divided into three groups. Two cartilage defects were created in the rabbits' knees by aseptic surgery. Group A (n=4) did not receive any treatment, group B (n=4) were treated with C/GP gel and Matrigel-engineered Ad-mock-GFP-transfected chondrocytes, and group C (n=4) were treated with C/GP gel and Matrigel-engineered Ad-hBMP7-GFP-transfected chondrocytes. Rabbits were sacrificed at 4 weeks after transplantation, and the repair effect was measured by the Wakitani scoring method. On the basis of the RT-PCR and western blot results, hBMP7 was efficiently overexpressed in the Ad-hBMP7-GFP-transfected chondrocytes. The ELISA results showed that the yields of collagen II and hyaluronic acid in Ad-hBMP7-GFP-transfected chondrocytes were significantly higher than those in Ad-mock-GFP-transfected chondrocytes. Chondrocytes have a better morphology and arrangement in a Matrigel scaffold than in C/GP, as assessed by H&E staining and scanning microscopy. According to the Wakitani score, Matrigel combined with Ad-hBMP7-GFP-transfected chondrocytes successfully promoted the repair of cartilage defects in rabbit knees.
Collapse
Affiliation(s)
- Xiaopeng Xia
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China; Department of Orthopedics, Nantong Hospital of Traditional Chinese Medicine, Nantong, Jiangsu 215000, P.R. China
| | - Jing Li
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA
| | - Bo Xia
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA
| | - Huilin Yang
- Department of Orthopedics, Nantong Hospital of Traditional Chinese Medicine, Nantong, Jiangsu 215000, P.R. China
| | - Dongmei Zhang
- Department of Immunology, Medical College of Nantong University, Nantong, Jiangsu 226019, P.R. China
| | - Bin Zhou
- Department of Orthopedics, Nantong Hospital of Traditional Chinese Medicine, Nantong, Jiangsu 215000, P.R. China
| | - Jie Zhang
- Department of Immunology, Medical College of Nantong University, Nantong, Jiangsu 226019, P.R. China
| | - Man Zhou
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA
| | - Fan Liu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China; Department of Orthopedics, Nantong Hospital of Traditional Chinese Medicine, Nantong, Jiangsu 215000, P.R. China
| |
Collapse
|
15
|
Ellman MB, Kim J, An HS, Chen D, Kc R, Li X, Xiao G, Yan D, Suh J, van Wijnen AJ, Wang JHC, Kim SG, Im HJ. Lactoferricin enhances BMP7-stimulated anabolic pathways in intervertebral disc cells. Gene 2013; 524:282-91. [PMID: 23644135 PMCID: PMC3679319 DOI: 10.1016/j.gene.2013.04.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 04/01/2013] [Accepted: 04/03/2013] [Indexed: 01/07/2023]
Abstract
Bone-morphogenetic protein-7 (BMP7) is a well-known anabolic and anti-catabolic growth factor on intervertebral disc (IVD) matrix and cell homeostasis. Similarly, Lactoferricin B (LfcinB) has recently been shown to have pro-anabolic, anti-catabolic, anti-oxidative and/or anti-inflammatory effects in bovine disc cells in vitro. In this study, we investigated the potential benefits of using combined peptide therapy with LfcinB and BMP7 for intervertebral disc matrix repair and to understand cellular and signaling mechanisms controlled by these factors. We studied the effects of BMP7 and LfcinB as individual treatments and combined therapy on bovine nucleus pulposus (NP) cells by assessing proteoglycan (PG) accumulation and synthesis, and the gene expression of matrix protein aggrecan and transcription factor SOX-9. We also analyzed the role of Noggin, a BMP antagonist, in IVD tissue and examined its effect after stimulation with LfcinB. To understand the molecular mechanisms by which LfcinB synergizes with BMP7, we investigated the ERK-SP1 axis as a downstream intracellular signaling regulator involved in BMP7 and LfcinB-mediated activities. Treatment of bovine NP cells cultured in alginate with LfcinB plus BMP7 synergistically stimulates PG synthesis and accumulation in part by upregulation of aggrecan gene expression. The synergism results from LfcinB-mediated activation of Sp1 and SMAD signaling pathways by (i) phosphorylation of SMAD 1/5/8; (ii) downregulation of SMAD inhibitory factors [i.e., noggin and SMAD6 (inhibitory SMAD)]; and (iii) upregulation of SMAD4 (universal co-SMAD). These data indicate that LfcinB-suppression of Noggin may eliminate the negative feedback of BMP7, thereby maximizing biological activity of BMP7 and ultimately shifting homeostasis to a pro-anabolic state in disc cells. We propose that combination growth factor therapy using BMP7 and LfcinB may be beneficial for treatment of disc degeneration.
Collapse
Affiliation(s)
- Michael B Ellman
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612
| | - Jaesung Kim
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612
| | - Howard S An
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612
| | - Di Chen
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612
| | - Ranjan Kc
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612
| | - Xin Li
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612
| | - Guozhi Xiao
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612
| | - Dongyao Yan
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612
| | - Joon Suh
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612
| | - Andre J. van Wijnen
- Center of Regenerative Medicine and Departments of Orthopedic Surgery & Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN 55905
| | - James H-C Wang
- MechanoBiology Laboratory, Departments of Orthopedic Surgery and Bioengineering, University of Pittsburgh, PA 15213, USA
| | - Su-Gwan Kim
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Chosun University, GwangJu City, Republic of Korea, 501-759
| | - Hee-Jeong Im
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612
- Department of Internal Medicine, Section of Rheumatology, Rush University Medical Center, Chicago, IL 60612
- Department of Bioengineering, University of Illinois at Chicago, IL 60612
| |
Collapse
|
16
|
Gavenis K, Heussen N, Hofman M, Andereya S, Schneider U, Schmidt-Rohlfing B. Cell-free repair of small cartilage defects in the Goettinger minipig: The effects of BMP-7 continuously released by poly(lactic-co-glycolid acid) microspheres. J Biomater Appl 2013; 28:1008-15. [DOI: 10.1177/0885328213491440] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Objective Cartilage repair of full-thickness chondral defects in the knees of Goettinger minipigs was assessed after treatment with cell-free collagen type-I gel with or without additional BMP-7 loaded poly(lactic-co-glycolid acid) microspheres. Methods Two full-thickness chondral defects were created in the trochlear groove of one hind leg knee in six Goettinger minipigs. Six defects were treated with a cell-free collagen type-I gel plug of 10 mm, the corresponding six defects were treated with a cell-free collagen type-I plug with poly(lactic-co-glycolid acid) microspheres loaded with recombinant BMP-7 (100 ng/ml gel). After 1 year, the animals were sacrificed. Immediately after recovery, non-destructive biomechanical testing was performed. The repair tissue quality was evaluated by immunohistochemistry and the O’Driscoll score was calculated. Results After 1 year, a robust cellular migration into the cell-free collagen gel plugs occurred and a hyaline-like repair tissue was found. Collagen type-II production and cellular organisation were higher in the BMP-7 microsphere group. The determination of the E-modulus, creep and relaxation revealed that mechanical properties of the BMP-7 microsphere group in summary were closer to control hyaline cartilage. Conclusions While all specimens revealed a homogeneous cellular distribution, ECM production, cellular organisation and mechanical properties were enhanced by continuous BMP-7 release.
Collapse
Affiliation(s)
- Karsten Gavenis
- Department of Orthopaedic and Trauma Surgery, Aachen University Hospital, Aachen, Germany
| | | | - Martijn Hofman
- Department of Orthopaedic and Trauma Surgery, Aachen University Hospital, Aachen, Germany
| | - Stefan Andereya
- Department of Orthopaedic and Trauma Surgery, Aachen University Hospital, Aachen, Germany
| | - Ulrich Schneider
- Institute of Medical Statistics, Aachen University Hospital, Aachen, Germany
| | | |
Collapse
|
17
|
Castro-Chavez F, Vickers KC, Lee JS, Tung CH, Morrisett JD. Effect of lyso-phosphatidylcholine and Schnurri-3 on osteogenic transdifferentiation of vascular smooth muscle cells to calcifying vascular cells in 3D culture. Biochim Biophys Acta Gen Subj 2013; 1830:3828-34. [PMID: 23500015 DOI: 10.1016/j.bbagen.2013.02.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 02/13/2013] [Accepted: 02/15/2013] [Indexed: 12/22/2022]
Abstract
BACKGROUND In vitro cell culture is a widely used technique for investigating a range of processes such as stem cell behavior, regenerative medicine, tissue engineering, and drug discovery. Conventional cell culture is performed in Petri dishes or flasks where cells typically attach to a flat glass or plastic surface as a cell monolayer. However, 2D cell monolayers do not provide a satisfactory representation of in vivo conditions. A 3D culture could be a much better system for representing the conditions that prevail in vivo. METHODS AND RESULTS To simulate 3D conditions, vascular smooth muscle cells (VSMCs) were loaded with gold-polyvmer-iron oxide hydrogel, enabling levitation of the cells by using spatially varying magnetic fields. These magnetically levitated 3D cultures appeared as freely suspended, clustered cells which proliferated 3-4 times faster than cells in conventional 2D cultures. When the levitated cells were treated with 10nM lysophosphatidylcholine (LPC), for 3days, cell clusters exhibited translucent extensions/rods 60-80μm wide and 200-250μm long. When 0.5μg/μl Schnurri-3 was added to the culture containing LPC, these extensions were smaller or absent. When excited with 590-650nm light, these extensions emitted intrinsic fluorescence at >667nm. When the 3D cultures were treated with a fluorescent probe specific for calcium hydroxyapatite (FITC-HABP-19), the cell extensions/rods emitted intensely at 518nm, the λmax for FITC emission. Pellets of cells treated with LPC were more enriched in calcium, phosphate, and glycosaminoglycans than cells treated with LPC and Schnurri-3. CONCLUSIONS In 3D cultures, VSMCs grow more rapidly and form larger calcification clusters than cells in 2D cultures. Transdifferentiation of VSMC into calcifying vascular cells is enhanced by LPC and attenuated by Schnurri-3. GENERAL SIGNIFICANCE The formation of calcified structures in 3D VSMC cultures suggests that similar structures may be formed in vivo.
Collapse
|
18
|
Gokce A, Yilmaz I, Bircan R, Tonbul M, Gokay NS, Gokce C. Synergistic Effect of TGF-β1 And BMP-7 on Chondrogenesis and Extracellular Matrix Synthesis: An In Vitro Study. Open Orthop J 2012; 6:406-13. [PMID: 23002411 PMCID: PMC3447182 DOI: 10.2174/1874325001206010406] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Revised: 07/13/2012] [Accepted: 07/15/2012] [Indexed: 11/22/2022] Open
Abstract
Introduction: The purpose of the present study seeks to determine the signal timing of BMP–7 and TGF-β1 from a novel chitosan based hydrogel system that may affect chondrocyte proliferation resulting in the presence of a synergism seen conspicuously in consecutive controlled delivery. Methods: Four groups of cultured chondrocytes were seeded on a novel designed chitosan based hydrogel. The hydrogel was left empty (control) in one group and loaded with BMP–7, TGF-β1 and their combination in the other groups, respectively. Hydrogel structure was analyzed with scanning electron microscope. The release kinetics of Growth Factors (GFs) was determined with ELISA. Chondrocyte viability and toxicity after being tested with MTS and collagen type II synthesis, were quantified with western blotting. Canonical regression analysis was used for measuring statistical evaluation. Results: Chitosan based hydrogel allowed controlled release of GFs in different time intervals for BMP–7 and TGF-β1. Double peak concentration gradient was found to be present in the group loaded with both GFs. In this group, substantially higher chondrocyte growth and collagen synthesis were also detected. Conclusions: We concluded that, chitosan based hydrogel systems may be adjusted to release GFs consecutively during biodegradation at the layers of surface, which may increase the cell number and enhance collagen type II synthesis.
Collapse
Affiliation(s)
- Alper Gokce
- Department of Orthopaedics and Traumatology, Namik Kemal University, School of Medicine, Tekirdag, Turkey
| | | | | | | | | | | |
Collapse
|
19
|
Smith RL, Lindsey DP, Dhulipala L, Harris AHS, Goodman SB, Maloney WJ. Effects of intermittent hydrostatic pressure and BMP-2 on osteoarthritic human chondrocyte metabolism in vitro. J Orthop Res 2011; 29:361-8. [PMID: 20882590 DOI: 10.1002/jor.21250] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Accepted: 07/23/2010] [Indexed: 02/04/2023]
Abstract
PURPOSE This study examined effects of intermittent hydrostatic pressure (IHP) and a chondrogenic growth factor, bone morphogenetic protein-2 (BMP-2), on anabolic, catabolic, and other metabolic markers in human osteoarthritic (OA) chondrocytes in vitro. METHODS Articular chondrocytes, isolated from femoral OA cartilage and maintained in high-density monolayer culture, were examined for effects of BMP-2 and IHP on gene expression of matrix-associated proteins (aggrecan, type II collagen, and SOX9) and catabolic matrix metalloproteinases (MMP-2 and MMP-3) and culture medium levels of the metabolic markers MMP-2, nitric oxide (NO), and glycosaminoglycan (GAG). The results were analyzed using a mixed linear regression model to investigate the effects of load and growth factor concentration. RESULTS IHP and BMP-2 modulated OA chondrocyte metabolism in accordance with growth factor concentration independently, without evidence of synergism or antagonism. Each type of stimulus acted independently on anabolic matrix gene expression. Type II collagen and SOX9 gene expression were stimulated by both IHP and BMP-2 whereas aggrecan was increased only by BMP-2. IHP exhibited a trend to decrease MMP-2 gene expression as a catabolic marker whereas BMP-2 did not. NO production was increased by addition of BMP-2 and IHP exhibited a trend for increased levels. GAG production was increased by BMP-2. CONCLUSIONS This study confirmed the hypothesis that human OA chondrocytes respond to a specific type of mechanical load, IHP, through enhanced articular cartilage macromolecule gene expression and that IHP, in combination with a chondrogenic growth factor BMP-2, additively enhanced matrix gene expression without interactive effects.
Collapse
Affiliation(s)
- R L Smith
- Bone and Joint RR&D Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, USA.
| | | | | | | | | | | |
Collapse
|
20
|
Effects of recombinant adeno-associated viral vectors on angiopoiesis and osteogenesis in cultured rabbit bone marrow stem cells via co-expressing hVEGF and hBMP genes: a preliminary study in vitro. Tissue Cell 2011; 42:314-21. [PMID: 20817238 DOI: 10.1016/j.tice.2010.07.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Revised: 06/30/2010] [Accepted: 07/08/2010] [Indexed: 11/24/2022]
Abstract
OBJECTIVE VEGF and BMP play important roles in angiogenesis and osteogenesis. Combining these two factors may be a promising therapeutic strategy for avascular necrosis of the femoral head (ANFH). METHODS Rabbit bone marrow-derived mesenchymal stem cells (BMSCs) were isolated and purified by density gradient centrifugation combined with attachment culture methods. The purity and characteristics of the BMSCs were detected by cell surface antigen identification. The best MOI of BMSCs transfected with rAAV was detected by fluorescent cell counting, and cell viability was determined by MTT assay. Expression of the genes of interest was detected by GFP gene expression, RT-PCR assay, and ELISA assay. The biological activities of VEGF and BMP were detected by angiogenic and osteogenic assays. RESULTS The best MOI of BMSCs transfected with rAAV was 5 x 10(4)v.g./cell. Cell growth curves showed vigorous cell viability. Expressions of the GFP, VEGF165, and BMP(7) genes were detected 1 day post-transfection and peaked 14 days post-transfection. Expression of the genes of interest was sustained over 1 month. VEGF and BMP proteins secreted from BMSCs transfected with rAAV-hVEGF(165)-IRES-hBMP(7) enhanced angiogenesis and osteogenesis in vitro. CONCLUSION Recombinant adeno-associated viral vectors co-expressing the hVEGF(165) and hBMP(7) genes showed efficient gene expression ability. The VEGF(165) and BMP(7) proteins expressed from the vector have efficient biological activity in vitro.
Collapse
|
21
|
Gavenis K, Heussen N, Schmidt-Rohlfing B. Effects of Low Concentration BMP-7 on Human Osteoarthritic Chondrocytes: Comparison of Different Applications. J Biomater Appl 2010; 26:845-59. [DOI: 10.1177/0885328210388439] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
While BMP-7 (OP-1) is one of the most potent growth factors in cartilage tissue engineering, the effects of exogenous low concentration BMP-7 on osteoarthritic chondrocytes are still unknown. Human osteoarthritic chondrocytes obtained from the femoral condyles of 10 patients were grown either in monolayer or in 3D collagen type-I gel culture in vitro. The growth factor was either given as a single dose of 50 ng/mL, a repeated dose, or continuously released from PGLA microspheres. Matrix formation was monitored by immunohistochemical staining and real-time PCR. In contrast to monolayer culture, the differentiated phenotype was prevailed in 3D culture. Collagen type-II protein production in the 3D group with a continuous BMP-7 release was enhanced in comparison to all other groups. Gene expression of collagen type-II and aggrecan was elevated in all treatment groups, with the highest extent in the BMP-7 microsphere group. In summary, treatment of articular chondrocytes with a low dose of BMP-7 leads to an elevated production of extracellular matrix components. This effect is further increased when BMP-7 is given repeatedly or continuously, which proved to be the most effective form of application.
Collapse
Affiliation(s)
- Karsten Gavenis
- Department of Orthopaedic Surgery, Aachen University Hospital, Aachen, Germany
| | - Nicole Heussen
- Institute of Medical Statistics, RWTH Aachen University, Germany
| | | |
Collapse
|
22
|
Hayashi M, Muneta T, Takahashi T, Ju YJ, Tsuji K, Sekiya I. Intra-articular injections of bone morphogenetic protein-7 retard progression of existing cartilage degeneration. J Orthop Res 2010; 28:1502-6. [PMID: 20872588 DOI: 10.1002/jor.21165] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We investigated the effect of weekly intra-articular injections of bone morphogenetic protein-7 (BMP-7) on prevention of progression of existing cartilage degeneration in an osteoarthritis model in rabbits. An anterior cruciate ligament transection (ACLT) model was used to create a progressive osteoarthritis model. BMP-7 was intra-articular injected weekly into the right knee and PBS into the left knee from 4 weeks after ACLT. Both sides of the knees were compared macroscopically, histologically, immunohistochemically, and by micro CT. Macroscopically, fibrillation in the femoral condyle was observed 4 weeks after ACLT. In the control knees, cartilage degeneration further progressed throughout the 12-week period. In the BMP-7 treated knee, osteoarthritis progression was milder than in the control knees. Histologically, safranin-O staining was decreased in the surgical knees at 4 weeks. Obvious erosions in both medial and lateral condyles were revealed in the control knees at 12 weeks, while cartilage matrix was predominantly retained in the BMP-7 treated knees. The macroscopic and microscopic OA score in the BMP-7 treated knee was better than that in the control in each rabbit. Immunohistochemical analysis demonstrated that both type II collagen and BMP-7 were more expressed in cartilage treated with BMP-7. Micro CT analysis showed that osteophytes were smaller in the BMP-7 treated knee compared to that of the control. Weekly intra-articular injections of BMP-7 inhibited progression of existing cartilage degeneration.
Collapse
Affiliation(s)
- Masaya Hayashi
- Section of Orthopaedic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | | | | | | | | | | |
Collapse
|
23
|
Shen B, Wei A, Whittaker S, Williams LA, Tao H, Ma DDF, Diwan AD. The role of BMP-7 in chondrogenic and osteogenic differentiation of human bone marrow multipotent mesenchymal stromal cells in vitro. J Cell Biochem 2010; 109:406-16. [PMID: 19950204 DOI: 10.1002/jcb.22412] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
This study addresses the role of bone morphogenetic protein-7 (BMP-7) in chondrogenic and osteogenic differentiation of human bone marrow multipotent mesenchymal stromal cells (BM MSCs) in vitro. BM MSCs were expanded and differentiated in the presence or absence of BMP-7 in monolayer and three-dimensional cultures. After 3 days of stimulation, BMP-7 significantly inhibited MSC growth in expansion cultures. When supplemented in commonly used induction media for 7-21 days, BMP-7 facilitated both chondrogenic and osteogenic differentiation of MSCs. This was evident by specific gene and protein expression analyses using real-time PCR, Western blot, histological, and immunohistochemical staining. BMP-7 supplementation appeared to enhance upregulation of lineage-specific markers, such as type II and type IX collagens (COL2A1, COL9A1) in chondrogenic and secreted phosphoprotein 1 (SPP1), osteocalcin (BGLAP), and osterix (SP7) in osteogenic differentiation. BMP-7 in the presence of TGF-beta3 induced superior chondrocytic proteoglycan accumulation, type II collagen, and SOX9 protein expression in alginate and pellet cultures compared to either factor alone. BMP-7 increased alkaline phosphatase activity and dose-dependently accelerated calcium mineralization of osteogenic differentiated MSCs. The potential of BMP-7 to promote adipogenesis of MSCs was restricted under osteogenic conditions, despite upregulation of adipocyte gene expression. These data suggest that BMP-7 is not a singular lineage determinant, rather it promotes both chondrogenic and osteogenic differentiation of MSCs by co-ordinating with initial lineage-specific signals to accelerate cell fate determination. BMP-7 may be a useful enhancer of in vitro differentiation of BM MSCs for cell-based tissue repair.
Collapse
Affiliation(s)
- Bojiang Shen
- Orthopaedic Research Institute, Department of Orthopaedic Surgery, St. George Hospital, University of New South Wales, Sydney, Australia
| | | | | | | | | | | | | |
Collapse
|
24
|
Angiopoiesis and bone regeneration via co-expression of the hVEGF and hBMP genes from an adeno-associated viral vector in vitro and in vivo. Acta Pharmacol Sin 2010; 31:821-30. [PMID: 20581855 DOI: 10.1038/aps.2010.67] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
AIM To investigate the therapeutic potential of adeno-associated virus (AAV)-mediated expression of vascular endothelial growth factor (VEGF) and bone morphogenetic protein (BMP). METHODS Four experimental groups were administered the following AAV vector constructs: rAAV-hVEGF(165)-internal ribosome entry site (IRES)-hBMP-7 (AAV-VEGF/BMP), rAAV-hVEGF(165)-GFP (AAV-VEGF), rAAV-hBMP-7-GFP (AAV-BMP), and rAAV-IRES-GFP (AAV-GFP). VEGF(165) and BMP-7 gene expression was detected using RT-PCR. The VEGF(165) and BMP-7 protein expression was determined by Western blotting and ELISA. The rabbit ischemic hind limb model was adopted and rAAV was administered intramuscularly into the ischemic limb. RESULTS Rabbit bone marrow-derived mesenchymal stem cells (BMSCs) were cultured and infected with the four viral vectors. The expression of GFP increased from the 7th day of infection and could be detected on the 28th day post-infection. In the AAV-VEGF/BMP group, the levels of VEGF165 and BMP-7 increased with prolonged infection time. The VEGF(165) and BMP-7 secreted from BMSCs in the AAV-VEGF/BMP group enhanced HUVEC tube formation and resulted in a stronger osteogenic ability, respectively. In rabbit ischemic hind limb model, GFP expression increased from the 4th week and could be detected at 8 weeks post-injection. The rAAV vector had superior gene expressing activity. Eight weeks after gene transfer, the mean blood flow was significantly higher in the AAV-VEGF/BMP group. Orthotopic ossification was radiographically evident, and capillary growth and calcium deposits were obvious in this group. CONCLUSION AAV-mediated VEGF and BMP gene transfer stimulates angiogenesis and bone regeneration and may be a new therapeutic technique for the treatment of avascular necrosis of the femoral head (ANFH).
Collapse
|
25
|
Gavenis K, Schneider U, Wallich R, Mueller-Rath R, Schmidt-Rohlfing B, Andereya S. Effects of Low Concentrated BMP-7 Administered by co-Cultivation or Plasmid Transfection on Human Osteoarthritic Chondrocytes. Int J Artif Organs 2010. [DOI: 10.1177/039139881003300602] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Introduction While BMP-7 has proven to be one of the most potent growth factors in cartilage tissue engineering, protein concentration and route of administration remain a matter of debate. Here we investigated the effects of a low concentration of BMP-7 on human osteoarthritic chondrocytes administered by protein co-cultivation and plasmid transfection. Methods Freshly released (P0) or in vitro propagated chondrocytes (P2) were cultivated in a collagen type-I gel for 3 weeks in vitro or in nude mice. Seeded chondrocytes were treated with 50 ng/mL BMP-7 directly added to the medium or were subject to transient BMP-7 plasmid transfection prior to gel cultivation. Untreated specimens served as a control. After recovery, samples were investigated by histological and immunohistochemical staining and real-time PCR. Results In vitro, collagen type-II protein production was enhanced, and it was stored mainly pericellularly. Collagen type-II and aggrecan gene expression were enhanced in both treatment groups. After nude mouse cultivation, col-II protein production was further enhanced, but specimens of the BMP-7 transfection group revealed a clustering of col-II positive cells. Gene expression was strongly up-regulated, chondrocyte number was increased and the differentiated phenotype prevailed. In general, freshly released chondrocytes (P0) proved to be superior to chondrocytes pre-amplified in vitro (P2). Conclusions Both BMP-7 co-cultivation and plasmid transfection of human osteoarthritic chondrocytes led to improved cartilage repair tissue. Nevertheless, the col-II distribution following BMP-7 co-cultivation was homogeneous, while samples produced by transient transfection revealed a col-II clustering.
Collapse
Affiliation(s)
- Karsten Gavenis
- Department of Orthopedic Surgery, Aachen University Hospital, Aachen - Germany
| | | | - Reinhard Wallich
- Institute of Immunology, University of Heidelberg, Heidelberg - Germany
| | | | | | - Stefan Andereya
- Department of Orthopedic Surgery, Aachen University Hospital, Aachen - Germany
| |
Collapse
|
26
|
Sekiya I, Tang T, Hayashi M, Morito T, Ju YJ, Mochizuki T, Muneta T. Periodic knee injections of BMP-7 delay cartilage degeneration induced by excessive running in rats. J Orthop Res 2009; 27:1088-92. [PMID: 19170094 DOI: 10.1002/jor.20840] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Strenuous running of rats enhances mechanical stress on the knee, thereby inducing degeneration of articular cartilage. Bone morphogenetic protein-7 (BMP-7) has an inhibitory effect on cartilage degeneration, suggesting its usefulness for human osteoarthritis patients. However, its mode of administration should be investigated. We examined whether weekly knee injections of BMP-7 delayed the progression of cartilage degeneration. Wistar rats were forced to run 30 km in 6 weeks on a rodent treadmill, and BMP-7 was injected weekly into the knee. Macroscopically and histologically, this strenuous running regimen induced cartilage degeneration. Weekly injections of 250 ng BMP-7 delayed the progression of cartilage degeneration. Immunohistochemically, in the control knee, type II collagen expression decreased, while BMP-7 expression in chondrocytes slightly increased. Interestingly, weekly injection of BMP-7 increased BMP-7 expression even 9 days after the final injection. Disulfate disaccharide keratan sulfate in serum transiently increased in the control group, while it remained at a low level in the BMP-7 group. Weekly BMP-7 injection increased BMP-7 expression in chondrocytes and its effect seemed to last more than 7 days. The effect of BMP-7 could be monitored by serum keratan sulfate concentration. Periodical injections of BMP-7 delayed progression of cartilage degeneration induced by excessive running in rats.
Collapse
Affiliation(s)
- Ichiro Sekiya
- Section of Cartilage Regeneration, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.
| | | | | | | | | | | | | |
Collapse
|
27
|
van Osch GJVM, Brittberg M, Dennis JE, Bastiaansen-Jenniskens YM, Erben RG, Konttinen YT, Luyten FP. Cartilage repair: past and future--lessons for regenerative medicine. J Cell Mol Med 2009; 13:792-810. [PMID: 19453519 PMCID: PMC3823400 DOI: 10.1111/j.1582-4934.2009.00789.x] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Since the first cell therapeutic study to repair articular cartilage defects in the knee in 1994, several clinical studies have been reported. An overview of the results of clinical studies did not conclusively show improvement over conventional methods, mainly because few studies reach level I of evidence for effects on middle or long term. However, these explorative trials have provided valuable information about study design, mechanisms of repair and clinical outcome and have revealed that much is still unknown and further improvements are required. Furthermore, cellular and molecular studies using new technologies such as cell tracking, gene arrays and proteomics have provided more insight in the cell biology and mechanisms of joint surface regeneration. Besides articular cartilage, cartilage of other anatomical locations as well as progenitor cells are now considered as alternative cell sources. Growth Factor research has revealed some information on optimal conditions to support cartilage repair. Thus, there is hope for improvement. In order to obtain more robust and reproducible results, more detailed information is needed on many aspects including the fate of the cells, choice of cell type and culture parameters. As for the clinical aspects, it becomes clear that careful selection of patient groups is an important input parameter that should be optimized for each application. In addition, the study outcome parameters should be improved. Although reduced pain and improved function are, from the patient's perspective, the most important outcomes, there is a need for more structure/tissue-related outcome measures. Ideally, criteria and/or markers to identify patients at risk and responders to treatment are the ultimate goal for these more sophisticated regenerative approaches in joint surface repair in particular, and regenerative medicine in general.
Collapse
Affiliation(s)
- Gerjo J V M van Osch
- Department of Orthopaedics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands.
| | | | | | | | | | | | | |
Collapse
|
28
|
Hayashi M, Muneta T, Ju YJ, Mochizuki T, Sekiya I. Weekly intra-articular injections of bone morphogenetic protein-7 inhibits osteoarthritis progression. Arthritis Res Ther 2008; 10:R118. [PMID: 18826579 PMCID: PMC2592805 DOI: 10.1186/ar2521] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Revised: 09/04/2008] [Accepted: 09/30/2008] [Indexed: 11/11/2022] Open
Abstract
Introduction We investigated the ability of a weekly intra-articular injection of bone morphogenetic protein (BMP)-7 to prevent osteoarthritis in rabbits with anterior cruciate ligament transections. Methods First, 36 knee joints were randomly divided into four groups: 50, 500, 5,000 ng BMP-7, and control. Knee cartilage was evaluated at 4, 8, and 12 weeks. Then, in order to control for individual differences, 500 ng BMP-7 was injected into one knee and phosphate-buffered saline (PBS) into the other, and the two knees were compared at 4, 8, and 12 weeks (n = 5). For pharmacokinetic analysis, cartilage was harvested at 1 hour and 1, 2, 4, and 7 days after knee injection of 500 ng BMP-7 or PBS (n = 3). Results Histological scores in the 500 and 5,000 ng BMP-7 groups were significantly better than those in the other groups at 12 weeks. Matched pair analysis demonstrated that both macroscopic and histological scores in the 500 ng BMP-7 group were better than those in the control group. Immunohistochemical analysis revealed higher BMP-7 expression by chondrocytes in the BMP-7 injected knees. Histology of whole knee and quantitative micro computed tomography analysis showed that weekly injections of 500 ng BMP-7 did not induce synovial fibrosis, ectopic bone, or osteophyte formation. As detected by enzyme-linked immunosorbent assay, BMP-7 concentration in the cartilage tissue was still higher in the BMP-7 treated group 7 days after the injection. Conclusions Weekly intra-articular injections of BMP-7 inhibited progression of osteoarthritis. Obvious adverse effects were not observed. BMP-7 concentration and expression in cartilage were still higher 7 days after injection.
Collapse
Affiliation(s)
- Masaya Hayashi
- Section of Orthopaedic Surgery, Graduate School, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo, 113-8519 Japan.
| | | | | | | | | |
Collapse
|