201
|
Abstract
The regenerative potential of injured adult tissue suggests the physiological existence of cells capable of participating in the reparative process. Recent studies indicate that stem-like cells residing in tissues contribute to tissue repair and are replenished by precursor bone marrow-derived cells. Mesenchymal stromal cells (MSC) are among the candidates for reparative cells. These cells can potentially be mobilized into the circulation in response to injury signals and exert their reparative effects at the site of injury. Current therapies for musculoskeletal injuries pose unavoidable risks which can impede full recovery. Trafficking of MSC to the injury site and their subsequent participation in the regenerative process is thought to be a natural healing response that can be imitated or augmented by enhancing the endogenous MSC pool with exogenously administered MSC. Therefore, a promising alternative to the existing strategies employed in the treatment of musculoskeletal injuries is to reinforce the inherent reparative capacity of the body by delivering MSC harvested from the patient's own tissues to the site of injury. The aim of this review is to inform the reader of studies that have evaluated the intrinsic homing and regenerative abilities of MSC, with particular emphasis on the repair of musculoskeletal injuries. Research that supports the direct use of MSC (without in vitro differentiation into tissue-specific cells) will also be reported. Based on accruing evidence that the natural healing mechanism involves the recruitment of MSC and their subsequent reparative actions at the site of injury, as well as documented therapeutic response after the exogenous administration of MSC, the feasibility of the emerging strategy of instant stem-cell therapy will be proposed.
Collapse
|
202
|
Matsumoto T, Okabe T, Ikawa T, Iida T, Yasuda H, Nakamura H, Wakitani S. Articular cartilage repair with autologous bone marrow mesenchymal cells. J Cell Physiol 2010; 225:291-5. [PMID: 20458744 DOI: 10.1002/jcp.22223] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Articular cartilage defects that do not repair spontaneously induce osteoarthritic changes in joints over a long period of observation. In this study, we examined the usefulness of transplanting culture-expanded bone marrow mesenchymal cells into osteochondral defects of joints with cartilage defects. First, we performed experiments on rabbits and up on obtaining good results proceeded to perform the experiments on humans. Macroscopic and histological repair with this method was good, and good clinical results were obtained although there was no significant difference with the control group. Recent reports have indicated that this procedure is comparable to autologous chondrocyte implantation, and concluded that it was a good procedure because it required one step less than that required by surgery, reduced costs for patients, and minimized donor site morbidity. Although some reports have previously shown that progenitor cells formed a tumor when implanted into immune-deficient mice after long term in vitro culture, the safety of the cell transplantation was confirmed by our clinical experience. Thus, this procedure is useful, effective, and safe, but the repaired tissues were not always hyaline cartilage. To obtain better repair with this procedure, treatment approaches using some growth factors during in vitro culture or gene transfection are being explored.
Collapse
Affiliation(s)
- Tomiya Matsumoto
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka, Japan
| | | | | | | | | | | | | |
Collapse
|
203
|
Haleem AM, Singergy AAE, Sabry D, Atta HM, Rashed LA, Chu CR, Shewy MTE, Azzam A, Aziz MTA. The Clinical Use of Human Culture-Expanded Autologous Bone Marrow Mesenchymal Stem Cells Transplanted on Platelet-Rich Fibrin Glue in the Treatment of Articular Cartilage Defects: A Pilot Study and Preliminary Results. Cartilage 2010; 1:253-261. [PMID: 21170288 PMCID: PMC3002255 DOI: 10.1177/1947603510366027] [Citation(s) in RCA: 209] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE: To test the hypothesis that platelet-rich fibrin glue (PR-FG) can be used clinically as a scaffold to deliver autologous culture-expanded bone marrow mesenchymal stem cells (BM-MSCs) for cartilage repair and to report clinical results 1 y after implantation of MSCs PR-FG. PATIENTS AND METHODS: Autologous BM-MSCs were culture expanded, placed on PR-FG intraoperatively, and then transplanted into 5 full-thickness cartilage defects of femoral condyles of 5 patients and covered with an autologous periosteal flap. Patients were evaluated clinically at 6 and 12 mo by the Lysholm and Revised Hospital for Special Surgery Knee (RHSSK) scores and radiographically by x-rays and magnetic resonance imaging (MRI) at the same time points. Repair tissue in 2 patients was rated arthroscopically after 12 mo using the International Cartilage Repair Society (ICRS) Arthroscopic Score. STUDY DESIGN: Case series; level of evidence 4. RESULTS: All patients' symptoms improved over the follow-up period of 12 mo. Average Lysholm and RHSSK scores for all patients showed statistically significant improvement at 6 and 12 mo postoperatively (P < 0.05). There was no statistically significant difference between the 6 and 12 mo postoperative clinical scores (P = 0.18). ICRS arthroscopic scores were 8/12 and 11/12 (nearly normal) for the 2 patients who consented to arthroscopy. MRI of 3 patients at 12 mo postoperatively revealed complete defect fill and complete surface congruity with native cartilage, whereas that of 2 patients showed incomplete congruity. CONCLUSION: Autologous BM-MSC transplantation on PR-FG as a cell scaffold may be an effective approach to promote the repair of articular cartilage defects of the knee in human patients.
Collapse
Affiliation(s)
- Amgad M. Haleem
- Department of Orthopedic Surgery, Cairo University School of Medicine, Cairo, Egypt,Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Amgad M. Haleem, MD, Cairo University Hospital, Saray El Manial Street, El Manial, Cairo 12411, Egypt
| | | | - Dina Sabry
- Department of Medical Biochemistry, Molecular Biology and Tissue Engineering Unit, Cairo University School of Medicine, Cairo, Egypt
| | - Hazem M. Atta
- Department of Medical Biochemistry, Molecular Biology and Tissue Engineering Unit, Cairo University School of Medicine, Cairo, Egypt
| | - Laila A. Rashed
- Department of Medical Biochemistry, Molecular Biology and Tissue Engineering Unit, Cairo University School of Medicine, Cairo, Egypt
| | - Constance R. Chu
- Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mohammed T. El Shewy
- Department of Orthopedic Surgery, Cairo University School of Medicine, Cairo, Egypt
| | - Akram Azzam
- Department of Medical Biochemistry, Molecular Biology and Tissue Engineering Unit, Cairo University School of Medicine, Cairo, Egypt
| | - Mohammed T. Abdel Aziz
- Department of Medical Biochemistry, Molecular Biology and Tissue Engineering Unit, Cairo University School of Medicine, Cairo, Egypt
| |
Collapse
|
204
|
Chondrogenesis in a hyaluronic acid scaffold: comparison between chondrocytes and MSC from bone marrow and adipose tissue. Knee Surg Sports Traumatol Arthrosc 2010; 18:1407-16. [PMID: 20020100 DOI: 10.1007/s00167-009-1017-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Accepted: 12/01/2009] [Indexed: 12/13/2022]
Abstract
Treatment of focal lesions of the articular cartilage of the knee using chondrocytes in a hyaluronic acid (HA) scaffold is already being investigated in clinical trials. An alternative may be to use mesenchymal stem cells (MSC). We have compared articular chondrocytes with MSC from human bone marrow (BM) and adipose tissue (AT), all cultured in HA scaffolds, for their ability to express genes and synthesize proteins associated with chondrogenesis. The cells were expanded in monolayer cultures. After seeding into the scaffold, the chondrocytes were maintained in medium, while the two MSC populations were given a chondrogenic differentiation medium. Chondrogenesis was assessed by real-time RT-PCR for chondrocyte-associated genes, by immunohistochemistry and by ELISA for collagens in the supernatant. Redifferentiation of the dedifferentiated chondrocytes in the HA scaffold was shown by a modest increase in type II collagen mRNA (COL2A1) and reduction in COL1A1. BM-MSC expressed 600-fold higher levels of COL2A1 than chondrocytes after 3 weeks in the scaffold. The levels of aggrecan (AGC1) and COL1A1 were similar for chondrocyte and BM-MSC scaffold cultures, while COL10A1 was higher in the BM-MSC. AT-MSC expressed levels of COL2A1 and COL1A1 similar to chondrocytes, but less AGC1 and COL10A1. Surprisingly, little collagen II protein was observed in the scaffold. Instead, collagen II was found in the culture medium. Chondrogenesis in HA scaffolds was more efficient using BM-MSC than AT-MSC or chondrocytes. Some of the secreted collagen II escaped entrapment in the extracellular space and was detected in the culture medium.
Collapse
|
205
|
Jiang YZ, Zhang SF, Qi YY, Wang LL, Ouyang HW. Cell transplantation for articular cartilage defects: principles of past, present, and future practice. Cell Transplant 2010; 20:593-607. [PMID: 20887665 DOI: 10.3727/096368910x532738] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
As articular cartilage has very limited self-repair capability, the repair and regeneration of damaged cartilage is a major challenge. This review aims to outline the past, present, and future of cell therapies for articular cartilage defect repair. Autologous chondrocyte implantation (ACI) has been used clinically for more than 20 years, and the short, medium, and long-term clinical outcomes of three generation of ACI are extensively overviewed. Also, strategies of clinical outcome evaluation, ACI limitations, and the comparison of ACI clinical outcomes with those of other surgical techniques are discussed. Moreover, mesenchymal stem cells and pluripotent stem cells for cartilage regeneration in vitro, in vivo, and in a few clinical studies are reviewed. This review not only comprehensively analyzes the ACI clinical data but also considers the findings from state-of-the-art stem cell research on cartilage repair from bench and bedside. The conclusion provides clues for the future development of strategies for cartilage regeneration.
Collapse
Affiliation(s)
- Yang Zi Jiang
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, China
| | | | | | | | | |
Collapse
|
206
|
Porada CD, Almeida-Porada G. Mesenchymal stem cells as therapeutics and vehicles for gene and drug delivery. Adv Drug Deliv Rev 2010; 62:1156-66. [PMID: 20828588 DOI: 10.1016/j.addr.2010.08.010] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2010] [Revised: 08/26/2010] [Accepted: 08/27/2010] [Indexed: 12/17/2022]
Abstract
Mesenchymal stem cells (MSCs) possess a set of several fairly unique properties which make them ideally suited both for cellular therapies/regenerative medicine, and as vehicles for gene and drug delivery. These include: 1) relative ease of isolation; 2) the ability to differentiate into a wide variety of seemingly functional cell types of both mesenchymal and non-mesenchymal origin; 3) the ability to be extensively expanded in culture without a loss of differentiative capacity; 4) they are not only hypoimmunogenic, but they produce immunosuppression upon transplantation; 5) their pronounced anti-inflammatory properties; and 6) their ability to home to damaged tissues, tumors, and metastases following in vivo administration. In this review, we summarize the latest research in the use of mesenchymal stem cells in regenerative medicine, as immunomodulatory/anti-inflammatory agents, and as vehicles for transferring both therapeutic genes in genetic disease and genes designed to destroy malignant cells.
Collapse
|
207
|
Sun L, Reagan MR, Kaplan DL. Role of Cartilage Forming Cells in Regenerative Medicine for Cartilage Repair. Orthop Res Rev 2010; 2010:85-94. [PMID: 24049462 PMCID: PMC3773876 DOI: 10.2147/orr.s7194] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Currently, cartilage repair remains a major challenge for researchers and physicians due to its limited healing capacity. Cartilage regeneration requires suitable cells; these must be easily obtained and expanded, able to produce hyaline matrix with proper mechanical properties, and demonstrate sustained integration with native tissue. At present, there is a wide variety of possible cell sources for cartilage regeneration; this review explores the diversity of sources for cartilage forming cells and the distinctive characteristics, advantages, limitations, and potential applications of each cell source. We place emphasis on cell sources used for in vitro and clinical studies.
Collapse
Affiliation(s)
- Lin Sun
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby St, Medford MA., USA
| | - Michaela R. Reagan
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford MA., USA
| | - David L. Kaplan
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby St, Medford MA., USA
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford MA., USA
| |
Collapse
|
208
|
Rastegar F, Shenaq D, Huang J, Zhang W, Zhang BQ, He BC, Chen L, Zuo GW, Luo Q, Shi Q, Wagner ER, Huang E, Gao Y, Gao JL, Kim SH, Zhou JZ, Bi Y, Su Y, Zhu G, Luo J, Luo X, Qin J, Reid RR, Luu HH, Haydon RC, Deng ZL, He TC. Mesenchymal stem cells: Molecular characteristics and clinical applications. World J Stem Cells 2010; 2:67-80. [PMID: 21607123 PMCID: PMC3097925 DOI: 10.4252/wjsc.v2.i4.67] [Citation(s) in RCA: 161] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Revised: 06/26/2010] [Accepted: 07/03/2010] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are non-hematopoietic stem cells with the capacity to differentiate into tissues of both mesenchymal and non-mesenchymal origin. MSCs can differentiate into osteoblastic, chondrogenic, and adipogenic lineages, although recent studies have demonstrated that MSCs are also able to differentiate into other lineages, including neuronal and cardiomyogenic lineages. Since their original isolation from the bone marrow, MSCs have been successfully harvested from many other tissues. Their ease of isolation and ex vivo expansion combined with their immunoprivileged nature has made these cells popular candidates for stem cell therapies. These cells have the potential to alter disease pathophysiology through many modalities including cytokine secretion, capacity to differentiate along various lineages, immune modulation and direct cell-cell interaction with diseased tissue. Here we first review basic features of MSC biology including MSC characteristics in culture, homing mechanisms, differentiation capabilities and immune modulation. We then highlight some in vivo and clinical evidence supporting the therapeutic roles of MSCs and their uses in orthopedic, autoimmune, and ischemic disorders.
Collapse
Affiliation(s)
- Farbod Rastegar
- Farbod Rastegar, Deana Shenaq, Jiayi Huang, Wenli Zhang, Bing-Qiang Zhang, Bai-Cheng He, Liang Chen, Guo-Wei Zuo, Qing Luo, Qiong Shi, Eric R Wagner, Enyi Huang, Yanhong Gao, Jian-Li Gao, Stephanie H Kim, Jian-Zhong Zhou, Yang Bi, Yuxi Su, Gaohui Zhu, Jinyong Luo, Xiaoji Luo, Jiaqiang Qin, Russell R Reid, Hue H Luu, Rex C Haydon, Zhong-Liang Deng, Tong-Chuan He, Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
209
|
Nigro J, White JF, Ramshaw JA, Haylock DN, Nilsson SK, Werkmeister JA. The effect of bovine endosteum-derived particles on the proliferation of human mesenchymal stem cells. Biomaterials 2010; 31:5689-99. [DOI: 10.1016/j.biomaterials.2010.03.054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Accepted: 03/21/2010] [Indexed: 01/22/2023]
|
210
|
Nejadnik H, Hui JH, Feng Choong EP, Tai BC, Lee EH. Autologous bone marrow-derived mesenchymal stem cells versus autologous chondrocyte implantation: an observational cohort study. Am J Sports Med 2010; 38:1110-6. [PMID: 20392971 DOI: 10.1177/0363546509359067] [Citation(s) in RCA: 386] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND First-generation autologous chondrocyte implantation has limitations, and introducing new effective cell sources can improve cartilage repair. PURPOSE This study was conducted to compare the clinical outcomes of patients treated with first-generation autologous chondrocyte implantation to patients treated with autologous bone marrow-derived mesenchymal stem cells (BMSCs). STUDY DESIGN Cohort study; Level of evidence, 3. METHODS Seventy-two matched (lesion site and age) patients underwent cartilage repair using chondrocytes (n = 36) or BMSCs (n = 36). Clinical outcomes were measured before operation and 3, 6, 9, 12, 18, and 24 months after operation using the International Cartilage Repair Society (ICRS) Cartilage Injury Evaluation Package, which included questions from the Short-Form Health Survey, International Knee Documentation Committee (IKDC) subjective knee evaluation form, Lysholm knee scale, and Tegner activity level scale. RESULTS There was significant improvement in the patients' quality of life (physical and mental components of the Short Form-36 questionnaire included in the ICRS package) after cartilage repair in both groups (autologous chondrocyte implantation and BMSCs). However, there was no difference between the BMSC and the autologous chondrocyte implantation group in terms of clinical outcomes except for Physical Role Functioning, with a greater improvement over time in the BMSC group (P = .044 for interaction effect). The IKDC subjective knee evaluation (P = .861), Lysholm (P = .627), and Tegner (P = .200) scores did not show any significant difference between groups over time. However, in general, men showed significantly better improvements than women. Patients younger than 45 years of age scored significantly better than patients older than 45 years in the autologous chondrocyte implantation group, but age did not make a difference in outcomes in the BMSC group. CONCLUSION Using BMSCs in cartilage repair is as effective as chondrocytes for articular cartilage repair. In addition, it required 1 less knee surgery, reduced costs, and minimized donor-site morbidity.
Collapse
Affiliation(s)
- Hossein Nejadnik
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | | | | | | | | |
Collapse
|
211
|
Bae JY, Han DW, Wakitani S, Nawata M, Hyon SH. Biological and Biomechanical Evaluations of Osteochondral Allografts Preserved in Cold Storage Solution Containing Epigallocatechin Gallate. Cell Transplant 2010; 19:681-9. [DOI: 10.3727/096368910x508771] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The beneficial effects of (-)-epigallocatechin-3- O-gallate (EGCG) on the nonfrozen preservation of mammalian cells and tissues are generally not well understood. A storage solution containing EGCG was employed to test the hypothesis that EGCG is capable of extending the storage duration for the cold preservation of articular cartilages. Human articular cartilages were preserved in a storage solution composed of serum-free RPMI-1640 medium with 1% antibiotic-antimycotic solution and 1 mM EGCG at 4°C for 1, 2, and 4 weeks. The chondrocyte viability (CCK-8 assay), biochemical and immunohistochemical composition [glycosaminoglycans (GAG) and (type II) collagen], and biomechanical property (compressive elastic modulus) were assessed. The chondrocyte viability of the cartilages preserved with EGCG was significantly well maintained for at least 2 weeks with high content of GAG and total collagen. These beneficial effects of EGCG were confirmed by the immunohistochemical observations of well-preserved cartilaginous structures and delayed denaturation of the extracellular matrix in preserved cartilages. There was no significant difference in the compressive elastic modulus (MPa) between the cartilages preserved with and without EGCG. These results suggest that EGCG may play an effective role in preserving osteochondral allografts, which can be exploited in devising strategies for the long-term preservation of other tissues under cold storage conditions.
Collapse
Affiliation(s)
- Jung Yoon Bae
- Department of Medical Simulation Engineering, Research Center for Nano Medical Engineering, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Dong-Wook Han
- Department of Nanomedical Engineering, College of Nanoscience & Nanotechnology, Pusan National University, Busan, Korea
| | - Shigeyuki Wakitani
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masashi Nawata
- Department of Orthopaedic Surgery, Marunouchi Hospital, Matsumoto, Japan
| | - Suong Hyu Hyon
- Department of Medical Simulation Engineering, Research Center for Nano Medical Engineering, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
212
|
Haleem AM, Chu CR. Advances in Tissue Engineering Techniques for Articular Cartilage Repair. ACTA ACUST UNITED AC 2010; 20:76-89. [PMID: 29430164 DOI: 10.1053/j.oto.2009.10.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The limited repair potential of human articular cartilage contributes to development of debilitating osteoarthritis and remains a great clinical challenge. This has led to evolution of cartilage treatment strategies from palliative to either reconstructive or reparative methods in an attempt to delay or "bridge the gap" to joint replacement. Further development of tissue engineering-based cartilage repair methods have been pursued to provide a more functional biological tissue. Currently, tissue engineering of articular cartilage has three cornerstones; a cell population capable of proliferation and differentiation into mature chondrocytes, a scaffold that can host these cells, provide a suitable environment for cellular functioning and serve as a sustained-release delivery vehicle of chondrogenic growth factors and thirdly, signaling molecules and growth factors that stimulate the cellular response and the production of a hyaline extracellular matrix (ECM). The aim of this review is to summarize advances in each of these three fields of tissue engineering with specific relevance to surgical techniques and technical notes.
Collapse
Affiliation(s)
- A M Haleem
- Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - C R Chu
- Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
213
|
Wang W, Li B, Li Y, Jiang Y, Ouyang H, Gao C. In vivo restoration of full-thickness cartilage defects by poly(lactide-co-glycolide) sponges filled with fibrin gel, bone marrow mesenchymal stem cells and DNA complexes. Biomaterials 2010; 31:5953-65. [PMID: 20488531 DOI: 10.1016/j.biomaterials.2010.04.029] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 04/14/2010] [Indexed: 10/19/2022]
Abstract
A composite construct comprising of bone marrow mesenchymal stem cells (BMSCs), plasmid DNA encoding transforming growth factor-beta1 (pDNA-TGF-beta1), fibrin gel and poly (lactide-co-glycolide) (PLGA) sponge was designed and employed to repair articular cartilage defects. To improve the gene transfection efficiency, a cationized chitosan derivative N,N,N-trimethyl chitosan chloride (TMC) was employed as the vector. The TMC/DNA complexes had a transfection efficiency of 9% to BMSCs and showed heterogeneous TGF-beta1 expression in a 10-day culture period in vitro. In vivo culture of the composite constructs was performed by implantation into full-thickness cartilage defects of New Zealand white rabbit joints, using the constructs absence of pDNA-TGF-beta1 or BMSCs as controls. Heterogeneous expression of TGF-beta1 in vivo was detected at 4 weeks, but its level was decreased in comparison with that of 2 weeks. After implantation for 12 weeks, the cartilage defects were successfully repaired by the composite constructs of the experimental group, and the neo-cartilage integrated well with its surrounding tissue and subchondral bone. Immunohistochemical and glycosaminoglycans (GAGs) staining confirmed the similar amount and distribution of collagen type II and GAGs in the regenerated cartilage as that of hyaline cartilage. The cartilage special genes expressed in the neo-tissue were closer to those of the normal cartilage. An overall score of 2.83 was obtained according to Wakitani's standard. By contrast, only part of the defects was repaired by the pDNA-TGF-beta1 absence constructs, and no cartilage repair but fibrous tissue was found for the BMSCs absence constructs. Therefore, combination of the PLGA sponge/fibrin gel scaffold with BMSCs and gene therapy is an effective method to restore cartilage defects and may have a great potential for practical applications in the near future.
Collapse
Affiliation(s)
- Wei Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | | | | | | | | | | |
Collapse
|
214
|
Disc regeneration therapy using marrow mesenchymal cell transplantation: a report of two case studies. Spine (Phila Pa 1976) 2010; 35:E475-80. [PMID: 20421856 DOI: 10.1097/brs.0b013e3181cd2cf4] [Citation(s) in RCA: 173] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
STUDY DESIGN Marrow mesenchymal cells (MSCs) contain stem cells and possess the ability to regenerate bone, cartilage, and fibrous tissues. Here, we applied this regenerative ability to intervertebral disc regeneration therapy in an attempt to develop a new spinal surgery technique. OBJECTIVE We analyzed the regenerative restoration ability of autologous MSCs in the markedly degenerated intervertebral discs. SUMMARY OF BACKGROUND DATA Fusion for lumbar intervertebral disc instability improves lumbago. However, fused intervertebral discs lack the natural and physiologic functions of intervertebral discs. If intervertebral discs can be regenerated and repaired, then damage to adjacent intervertebral discs can be avoided. We verified the regenerative ability of MSCs by animal studies, and for the first time, performed therapeutic intervertebral disc regeneration therapy in patients and obtained favorable findings. METHODS Subjects were 2 women aged 70 and 67 years; both patients had lumbago, leg pain, and numbness. Myelography and magnetic resonance imaging showed lumbar spinal canal stenosis, and radiograph confirmed the vacuum phenomenon with instability. From the ilium of each patient, marrow fluid was collected, and MSCs were cultured using the medium containing autogenous serum. In surgery, fenestration was performed on the stenosed spinal canal and then pieces of collagen sponge containing autologous MSCs were grafted percutaneously to degenerated intervertebral discs. RESULTS At 2 years after surgery, radiograph and computed tomography showed improvements in the vacuum phenomenon in both patients. On T2-weighted magnetic resonance imaging, signal intensity of intervertebral discs with cell grafts was high, thus indicating high moisture contents. Roentgenkymography showed that lumbar disc instability improved. Symptom was alleviated in both patients. CONCLUSION The intervertebral disc regeneration therapy using MSC brought about favorable results in these 2 cases. It seems to be a promising minimally invasive treatment.
Collapse
|
215
|
Quintin A, Schizas C, Scaletta C, Jaccoud S, Applegate LA, Pioletti DP. Plasticity of fetal cartilaginous cells. Cell Transplant 2010; 19:1349-57. [PMID: 20447338 DOI: 10.3727/096368910x506854] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Tissue-specific stem cells found in adult tissues can participate in the repair process following injury. However, adult tissues, such as articular cartilage and intervertebral disc, have low regeneration capacity, whereas fetal tissues, such as articular cartilage, show high regeneration ability. The presence of fetal stem cells in fetal cartilaginous tissues and their involvement in the regeneration of fetal cartilage is unknown. The aim of the study was to assess the chondrogenic differentiation and the plasticity of fetal cartilaginous cells. We compared the TGF-β3-induced chondrogenic differentiation of human fetal cells isolated from spine and cartilage tissues to that of human bone marrow stromal cells (BMSC). Stem cell surface markers and adipogenic and osteogenic plasticity of the two fetal cell types were also assessed. TGF-β3 stimulation of fetal cells cultured in high cell density led to the production of aggrecan, type I and II collagens, and variable levels of type X collagen. Although fetal cells showed the same pattern of surface stem cell markers as BMSCs, both type of fetal cells had lower adipogenic and osteogenic differentiation capacity than BMSCs. Fetal cells from femoral head showed higher adipogenic differentiation than fetal cells from spine. These results show that fetal cells are already differentiated cells and may be a good compromise between stem cells and adult tissue cells for a cell-based therapy.
Collapse
Affiliation(s)
- Aurelie Quintin
- Cellular Therapy Unit, Department of Musculoskeletal Medicine, University Hospital Center and University of Lausanne, CHUV-UNIL, Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
216
|
Kalwitz G, Andreas K, Endres M, Neumann K, Notter M, Ringe J, Sittinger M, Kaps C. Chemokine profile of human serum from whole blood: migratory effects of CXCL-10 and CXCL-11 on human mesenchymal stem cells. Connect Tissue Res 2010; 51:113-22. [PMID: 20001843 DOI: 10.3109/03008200903111906] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Autologous human serum is used in cartilage repair and may exert its effect by the recruitment of mesenchymal stem and progenitor cells (MSC). Aim of our study was to analyze the chemokine profile of human serum and to verify chemotactic activity of selected chemokines on MSC. Human MSC were isolated from iliac crest bone marrow aspirates. Chemotactic activity of human serum made from whole blood and pharma grade serum was tested in 96-well chemotaxis assays and chemokine levels were analyzed using human chemokine antibody membrane arrays. The chemotactic potential of selected chemokines on MSC was tested dose dependently using chemotaxis assays. Human serum derived from whole blood significantly attracted human MSC, while pharma grade serum did not recruit MSC. Human chemokine antibody array analysis showed that the level of chemokines CXCL-3, 5, 7-8, 10-12, 16; CCL- 2, 5, 11, 13, 16-20, 24-25, 27; as well as XCL-1 was elevated (fold change >1.5) in serum derived from whole blood compared to nonrecruiting pharma grade serum. Chemotaxis assays showed that the chemokines IP-10/CXCL-10 and I-TAC/CXCL-11 significantly recruit human MSC. PARC/CCL-18, HCC-4/CCL-16, CTACK/CCL-27, and Lymphotactin/XCL-1 showed no chemotactic effect on MSC. Therefore, human serum derived from whole blood contains chemokines that may contribute to serum-mediated recruitment of human mesenchymal progenitors from bone marrow.
Collapse
Affiliation(s)
- Gregor Kalwitz
- TransTissue Technologies GmbH and Tissue Engineering Laboratory, Department of Rheumatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
217
|
Bae JY, Matsumura K, Wakitani S, Kawaguchi A, Tsutsumi S, Hyon SH. Beneficial storage effects of epigallocatechin-3-o-gallate on the articular cartilage of rabbit osteochondral allografts. Cell Transplant 2009; 18:505-12. [PMID: 19775510 DOI: 10.1177/096368970901805-604] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
A fresh osteochondral allograft is one of the most effective treatments for cartilage defects of the knee. Despite the clinical success, fresh osteochondral allografts have great limitations in relation to the short storage time that cartilage tissues can be well-preserved. Fresh osteochondral grafts are generally stored in culture medium at 4 degrees C. While the viability of articular cartilage stored in culture medium is significantly diminished within 1 week, appropriate serology testing to minimize the chances for the disease transmission requires a minimum of 2 weeks. (-)-Epigallocatechin-3-O-gallate (EGCG) has differential effects on the proliferation of cancer and normal cells, thus a cytotoxic effect on various cancer cells, but a cytopreservative effect on normal cells. Therefore, a storage solution containing EGCG might extend the storage duration of articular cartilages. Rabbit osteochondral allografts were performed with osteochondral grafts stored at 4 degrees C in culture medium containing EGCG for 2 weeks and then the clinical effects were examined with macroscopic and histological assessment after 4 weeks. The cartilaginous structure of an osteochondral graft stored with EGCG was well-preserved with high cell viability and glycosaminoglycan (GAG) content of the extracellular matrix (ECM). After an osteochondral allograft, the implanted osteochondral grafts stored with EGCG also provided a significantly better retention of the articular cartilage with viability and metabolic activity. These data suggest that EGCG can be an effective storage agent that allows long-term preservation of articular cartilage under cold storage conditions.
Collapse
Affiliation(s)
- Jung Yoon Bae
- Department of Medical Simulation Engineering, Research Center for Nano Medical Engineering, Institute for Frontier Medical Sciences, Kyoto University, Kyoto 6060-8507, Japan
| | | | | | | | | | | |
Collapse
|
218
|
Mesenchymal stem cell-based therapy for cartilage repair: a review. Knee Surg Sports Traumatol Arthrosc 2009; 17:1289-97. [PMID: 19333576 DOI: 10.1007/s00167-009-0782-4] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Accepted: 03/04/2009] [Indexed: 12/13/2022]
Abstract
Articular cartilage injury remains one of the major concerns in orthopaedic surgery. Mesenchymal stem cell (MSC) transplantation has been introduced to avoid some of the side effects and complications of current techniques. The purpose of this paper is to review the literature on MSC-based cell therapy for articular cartilage repair to determine if it can be an alternative treatment for cartilage injury. MSCs retain both high proliferative potential and multipotentiality, including chondrogenic differentiation potential, and a number of successful results in transplantation of MSCs into cartilage defects have been reported in animal studies. However, the use of MSCs for cartilage repair is still at the stage of preclinical and phase I studies, and no comparative clinical studies have been reported. Therefore, it is difficult to make conclusions in human studies. This requires randomized clinical trials to evaluate the effectiveness of MSC-based cell therapy for cartilage repair.
Collapse
|
219
|
Optimizing the success of cell transplantation therapy for stroke. Neurobiol Dis 2009; 37:275-83. [PMID: 19822211 DOI: 10.1016/j.nbd.2009.10.003] [Citation(s) in RCA: 152] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Revised: 09/30/2009] [Accepted: 10/02/2009] [Indexed: 12/22/2022] Open
Abstract
Stem cell transplantation has evolved as a promising experimental treatment approach for stroke. In this review, we address the major hurdles for successful translation from basic research into clinical applications and discuss possible strategies to overcome these issues. We summarize the results from present pre-clinical and clinical studies and focus on specific areas of current controversy and research: (i) the therapeutic time window for cell transplantation; (ii) the selection of patients likely to benefit from such a therapy; (iii) the optimal route of cell delivery to the ischemic brain; (iv) the most suitable cell types and sources; (v) the potential mechanisms of functional recovery after cell transplantation; and (vi) the development of imaging techniques to monitor cell therapy.
Collapse
|
220
|
Steinert AF, Proffen B, Kunz M, Hendrich C, Ghivizzani SC, Nöth U, Rethwilm A, Eulert J, Evans CH. Hypertrophy is induced during the in vitro chondrogenic differentiation of human mesenchymal stem cells by bone morphogenetic protein-2 and bone morphogenetic protein-4 gene transfer. Arthritis Res Ther 2009; 11:R148. [PMID: 19799789 PMCID: PMC2787261 DOI: 10.1186/ar2822] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Revised: 09/15/2009] [Accepted: 10/02/2009] [Indexed: 01/28/2023] Open
Abstract
INTRODUCTION The present study compares bone morphogenetic protein (BMP)-4 and BMP-2 gene transfer as agents of chondrogenesis and hypertrophy in human primary mesenchymal stem cells (MSCs) maintained as pellet cultures. METHODS Adenoviral vectors carrying cDNA encoding human BMP-4 (Ad.BMP-4) were constructed by cre-lox combination and compared to previously generated adenoviral vectors for BMP-2 (Ad.BMP-2), green fluorescent protein (Ad.GFP), or firefly luciferase (Ad.Luc). Cultures of human bone-marrow derived MSCs were infected with 5 x 10(2) viral particles/cell of Ad.BMP-2, or Ad.BMP-4, seeded into aggregates and cultured for three weeks in a defined, serum-free medium. Untransduced cells or cultures transduced with marker genes served as controls. Expression of BMP-2 and BMP-4 was determined by ELISA, and aggregates were analyzed histologically, immunohistochemically, biochemically and by RT-PCR for chondrogenesis and hypertrophy. RESULTS Levels of BMP-2 and BMP-4 in the media were initially 30 to 60 ng/mL and declined thereafter. BMP-4 and BMP-2 genes were equipotent inducers of chondrogenesis in primary MSCs as judged by lacuna formation, strong staining for proteoglycans and collagen type II, increased levels of GAG synthesis, and expression of mRNAs associated with the chondrocyte phenotype. However, BMP-4 modified aggregates showed a lower tendency to progress towards hypertrophy, as judged by expression of alkaline phosphatase, annexin 5, immunohistochemical staining for type X collagen protein, and lacunar size. CONCLUSIONS BMP-2 and BMP-4 were equally effective in provoking chondrogenesis by primary human MSCs in pellet culture. However, chondrogenesis triggered by BMP-2 and BMP-4 gene transfer showed considerable evidence of hypertrophic differentiation, with, the cells resembling growth plate chondrocytes both morphologically and functionally. This suggests caution when using these candidate genes in cartilage repair.
Collapse
Affiliation(s)
- Andre F Steinert
- Orthopaedic Center for Musculoskeletal Research, Orthopaedic Clinic, König-Ludwig-Haus, Julius-Maximilians-University, Brettreichstrasse 11, 97074 Würzburg, Germany
- Center for Molecular Orthopaedics, Harvard Medical School, 221 Longwood Avenue, BLI 152, Boston, MA 02115, USA
| | - Benedikt Proffen
- Orthopaedic Center for Musculoskeletal Research, Orthopaedic Clinic, König-Ludwig-Haus, Julius-Maximilians-University, Brettreichstrasse 11, 97074 Würzburg, Germany
| | - Manuela Kunz
- Orthopaedic Center for Musculoskeletal Research, Orthopaedic Clinic, König-Ludwig-Haus, Julius-Maximilians-University, Brettreichstrasse 11, 97074 Würzburg, Germany
| | - Christian Hendrich
- Orthopaedic Center for Musculoskeletal Research, Orthopaedic Clinic, König-Ludwig-Haus, Julius-Maximilians-University, Brettreichstrasse 11, 97074 Würzburg, Germany
| | - Steven C Ghivizzani
- Center for Molecular Orthopaedics, Harvard Medical School, 221 Longwood Avenue, BLI 152, Boston, MA 02115, USA
- Department of Orthopaedics and Rehabilitation, University of Florida, 3450 Hull Road, Gainesville, FL 32607, USA
| | - Ulrich Nöth
- Orthopaedic Center for Musculoskeletal Research, Orthopaedic Clinic, König-Ludwig-Haus, Julius-Maximilians-University, Brettreichstrasse 11, 97074 Würzburg, Germany
| | - Axel Rethwilm
- Institut für Virologie und Immunbiologie, Julius-Maximilians-University, Versbacherstrasse 7, 97078 Würzburg, Germany
| | - Jochen Eulert
- Orthopaedic Center for Musculoskeletal Research, Orthopaedic Clinic, König-Ludwig-Haus, Julius-Maximilians-University, Brettreichstrasse 11, 97074 Würzburg, Germany
| | - Christopher H Evans
- Center for Molecular Orthopaedics, Harvard Medical School, 221 Longwood Avenue, BLI 152, Boston, MA 02115, USA
| |
Collapse
|
221
|
Matsumoto T, Cooper GM, Gharaibeh B, Meszaros LB, Li G, Usas A, Fu FH, Huard J. Cartilage repair in a rat model of osteoarthritis through intraarticular transplantation of muscle-derived stem cells expressing bone morphogenetic protein 4 and soluble Flt-1. ACTA ACUST UNITED AC 2009; 60:1390-405. [PMID: 19404941 DOI: 10.1002/art.24443] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVE The control of angiogenesis during chondrogenic differentiation is an important issue affecting the use of stem cells in cartilage repair, especially with regard to the persistence of regenerated cartilage. This study was undertaken to investigate the effect of vascular endothelial growth factor (VEGF) stimulation and the blocking of VEGF with its antagonist, soluble Flt-1 (sFlt-1), on the chondrogenesis of skeletal muscle-derived stem cells (MDSCs) in a rat model of osteoarthritis (OA). METHODS We investigated the effect of VEGF on cartilage repair in an immunodeficiency rat model of OA after intraarticular injection of murine MDSCs expressing bone morphogenetic protein 4 (BMP-4) in combination with MDSCs expressing VEGF or sFlt-1. RESULTS In vivo, a combination of sFlt-1- and BMP-4-transduced MDSCs demonstrated better repair without osteophyte formation macroscopically and histologically following OA induction, when compared with the other groups. Higher differentiation/proliferation and lower levels of chondrocyte apoptosis were also observed in sFlt-1- and BMP-4-transduced MDSCs compared with a combination of VEGF- and BMP-4-transduced MDSCs or with BMP-4-transduced MDSCs alone. In vitro experiments with mixed pellet coculture of MDSCs and OA chondrocytes revealed that BMP-4-transduced MDSCs produced the largest pellets, which had the highest gene expression of not only type II collagen and SOX9 but also type X collagen, suggesting formation of hypertrophic chondrocytes. CONCLUSION Our results demonstrate that MDSC-based therapy involving sFlt-1 and BMP-4 repairs articular cartilage in OA mainly by having a beneficial effect on chondrogenesis by the donor and host cells as well as by preventing angiogenesis, which eventually prevents cartilage resorption, resulting in persistent cartilage regeneration and repair.
Collapse
Affiliation(s)
- Tomoyuki Matsumoto
- Stem Cell Research Center, Children's Hospital of Pittsburgh, and University of Pittsburgh, Pittsburgh, Pennsylvania 15213-2582, USA
| | | | | | | | | | | | | | | |
Collapse
|
222
|
|
223
|
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: 107] [Impact Index Per Article: 6.7] [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
|
224
|
Lee K, Chan CK, Patil N, Goodman SB. Cell therapy for bone regeneration--bench to bedside. J Biomed Mater Res B Appl Biomater 2009; 89:252-63. [PMID: 18777578 DOI: 10.1002/jbm.b.31199] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The concept of bone tissue engineering, which began in the early 1980s, has seen tremendous growth in the numbers of research studies. One of the key areas of research has been in the field of mesenchymal stem cells, where the challenge is to produce the perfect tissue-engineered bone construct. This practical review summarizes basic and applied state-of-the-art research in the area of mesenchymal stem cells, and highlights the important translational research that has already been initiated. The topics that will be covered include the sources of stem cells in use, scaffolds, gene therapy, clinical applications in nonunions, tumors, osteonecrosis, revision arthroplasties, and spine fusion. Although significant challenges remain, there exists an exceptional opportunity to translate basic research in mesenchymal stem cell technologies into viable clinical treatments for bone regeneration.
Collapse
Affiliation(s)
- Kevin Lee
- Department of Orthopaedic Surgery, Stanford University Medical Center, Stanford, California 94305-5326, USA
| | | | | | | |
Collapse
|
225
|
Cell-based therapy in articular cartilage lesions of the knee. Arthroscopy 2009; 25:531-52. [PMID: 19409312 DOI: 10.1016/j.arthro.2009.02.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2009] [Revised: 02/12/2009] [Accepted: 02/12/2009] [Indexed: 02/02/2023]
Abstract
PURPOSE The purpose of this systematic review was to determine the effectiveness of cell-based therapy for articular cartilage defects of the knee. METHODS We performed a literature search in Medline (1994 to 2009) regarding cell-based therapies for chondral lesions. RESULTS We identified 10 Level I or II randomized controlled trials and 3 Level II prospective comparative studies. Although many of these studies had substantial flaws, which could introduce bias, we overall found no difference between the cell-based studies and other interventions. In addition, we identified 26 Level III and IV studies of cell-based therapy. CONCLUSIONS There is insufficient evidence from the studies included in this review to say whether cell-based therapy is superior to other treatment strategies in articular cartilage lesions of the knee.
Collapse
|
226
|
Jung M, Kaszap B, Redöhl A, Steck E, Breusch S, Richter W, Gotterbarm T. Enhanced early tissue regeneration after matrix-assisted autologous mesenchymal stem cell transplantation in full thickness chondral defects in a minipig model. Cell Transplant 2009; 18:923-32. [PMID: 19523325 DOI: 10.3727/096368909x471297] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Adult mesenchymal stem cells (MSCs) are an attractive cell source for new treatment strategies in regenerative medicine. This study investigated the potential effect of matrix assisted MSC transplantation for articular cartilage regeneration in a large-animal model 8 weeks postoperatively. MSCs from bone marrow aspirates of eight Goettingen minipigs were isolated and expanded prior to surgery. Articular cartilage defects of 5.4 mm were created bilaterally in the medial patellar groove without penetrating the subchondral bone plate. Defects were either left empty (n = 4), covered with a collagen type I/III membrane (n = 6) or additionally treated with autologous MSC transplantation (2 x 10(6); n = 6). After 8 weeks animals were euthanized and the defect area was assessed for its gross appearance. Histomorphological analysis of the repair tissue included semiquantitative scoring (O'Driscoll score) and quantitative histomorphometric analysis for its glycosaminoglycan (GAG) and collagen type II content. All membranes were found to cover the defect area 8 weeks postoperatively. Median defect filling was 115.8% (membrane), 117.8% (empty), and 100.4% (MSC), respectively (not significant). Histomorphological scoring revealed significantly higher values in MSC-treated defects (median 16.5) when compared to membrane treatment (median 9.5) or empty defects (median 11.5; p = 0.015 and p = 0.038). Histomorphometric analysis showed larger GAG/collagen type II-positive areas in the MSC-treated group (median 24.6%/29.5% of regeneration tissue) compared to 13.6%/33.1% (empty defects) and 1.7%/6.2% (membrane group; p = 0.066). Cell distribution was more homogeneous in MSC compared to membrane-only group, where cells were found mainly near the subchondral zone. In conclusion, autologous matrix-assisted MSC transplantation significantly increased the histomorphological repair tissue quality during early articular cartilage defect repair and resulted in higher GAG/collagen type II-positive cross-sectional areas of the regenerated tissue.
Collapse
Affiliation(s)
- Martin Jung
- Department of Orthopaedic Surgery, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|
227
|
Kubo S, Cooper GM, Matsumoto T, Phillippi JA, Corsi KA, Usas A, Li G, Fu FH, Huard J. Blocking vascular endothelial growth factor with soluble Flt-1 improves the chondrogenic potential of mouse skeletal muscle-derived stem cells. ACTA ACUST UNITED AC 2009; 60:155-65. [PMID: 19116905 DOI: 10.1002/art.24153] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE To investigate the effect of vascular endothelial growth factor (VEGF) stimulation and the effect of blocking VEGF with its antagonist, soluble Flt-1 (sFlt-1), on chondrogenesis, using muscle-derived stem cells (MDSCs) isolated from mouse skeletal muscle. METHODS The direct effect of VEGF on the in vitro chondrogenic ability of mouse MDSCs was tested using a pellet culture system, followed by real-time quantitative polymerase chain reaction (PCR) and histologic analyses. Next, the effect of VEGF on chondrogenesis within the synovial joint was tested, using genetically engineered MDSCs implanted into rat osteochondral defects. In this model, MDSCs transduced with a retroviral vector to express bone morphogenetic protein 4 (BMP-4) were coimplanted with MDSCs transduced to express either VEGF or sFlt-1 (a VEGF antagonist) to provide a gain- and loss-of-function experimental design. Histologic scoring was used to compare cartilage formation among the treatment groups. RESULTS Hyaline-like cartilage matrix production was observed in both VEGF-treated and VEGF-blocked (sFlt-1-treated) pellet cultures, but quantitative PCR revealed that sFlt-1 treatment improved the expression of chondrogenic genes in MDSCs that were stimulated to undergo chondrogenic differentiation with BMP-4 and transforming growth factor beta3 (TGFbeta3). In vivo testing of articular cartilage repair showed that VEGF-transduced MDSCs caused an arthritic change in the knee joint, and sFlt-1 improved the MDSC-mediated repair of articular cartilage, compared with BMP-4 alone. CONCLUSION Soluble Flt-1 gene therapy improved the BMP-4- and TGFbeta3-induced chondrogenic gene expression of MDSCs in vitro and improved the persistence of articular cartilage repair by preventing vascularization and bone invasion into the repaired articular cartilage.
Collapse
Affiliation(s)
- Seiji Kubo
- Children's Hospital of Pittsburgh, and University of Pittsburgh, Pittsburgh, Pennsylvania 15213-2582, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
228
|
Stoddart MJ, Grad S, Eglin D, Alini M. Cells and biomaterials in cartilage tissue engineering. Regen Med 2009; 4:81-98. [PMID: 19105618 DOI: 10.2217/17460751.4.1.81] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cartilage defects are notoriously difficult to repair and owing to the long-term prognosis of osteoarthritis, and a rapidly aging population, a need for new therapies is pressing. Cell-based therapies for cartilage regeneration were introduced into patients in the early 1990s. Since that time the technology has developed from a simple cell suspension to more complex 3D structures. Cells, both chondrocytes and stem cells, have been incorporated into scaffold material with the aim to better recreate the natural environment of the cell, while providing more structural support to withstand the large forces applied on the de novo tissue. This review aims to provide an overview of potential cell sources and different scaffold materials, which are in development for cartilage tissue engineering.
Collapse
Affiliation(s)
- Martin J Stoddart
- Biomaterials & Tissue Engineering, AO Research Institute, Davos Platz, Switzerland.
| | | | | | | |
Collapse
|
229
|
Tateishi K, Ando W, Higuchi C, Hart DA, Hashimoto J, Nakata K, Yoshikawa H, Nakamura N. Comparison of human serum with fetal bovine serum for expansion and differentiation of human synovial MSC: potential feasibility for clinical applications. Cell Transplant 2009; 17:549-57. [PMID: 18714674 DOI: 10.3727/096368908785096024] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The aim of this study was to evaluate the effect of human serum (HS) on growth and differentiation capacity of human synovium-derived mesenchymal stem cells (MSC) in comparison to cells grown in fetal bovine serum (FBS). Human MSCs were isolated from the synovium of knee joints of three donors and the cells were cultured individually in varying concentrations of allogenic HS or FBS. Bovine MSCs were isolated from synovium and cultured in the same manner. Cell proliferation was assessed by the tetrazolium assay after passage 3. The capacity for chondrogenic and osteogenic differentiation was investigated in specific media followed by 1,9-dimethylmethylene blue assay and alcian blue staining, or by alizarin red staining, respectively. Human MSCs proliferated significantly more rapidly in the presence of HS than with equivalent levels of FBS. Chondrogenic or osteogenic differentiation occurred to nearly identical levels in HS or FBS. The results of this study indicate that HS is superior for the culture of human MSCs compared with FBS in terms of cellular expandability, without losing chondrogenic or osteogenic differentiation capacity. Coupled with the advantage in eliminating the potential risk accompanied with the use of xeno-derived materials, pooled, well-characterized HS could be a useful reagent to promote cellular expansion for clinical synovial stem cell-based therapy.
Collapse
Affiliation(s)
- K Tateishi
- Department of Orthopedics, National Hospital Organization Kure Medical Center, Hiroshima, Japan
| | | | | | | | | | | | | | | |
Collapse
|
230
|
Li Y, Chopp M. Marrow stromal cell transplantation in stroke and traumatic brain injury. Neurosci Lett 2009; 456:120-3. [PMID: 19429146 DOI: 10.1016/j.neulet.2008.03.096] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Revised: 02/24/2008] [Accepted: 03/01/2008] [Indexed: 12/19/2022]
Abstract
There is a paucity of therapies for most central nervous system (CNS) disorders. Bone marrow stromal cells (MSCs) are a mixed cell population, including stem and progenitor cells, and are currently a strong candidate for cell-based therapy in "brain attack", including stroke, and traumatic brain injury (TBI), since they are easily isolated and can be expanded in culture from patients without ethical and technical problems. Although it has been suggested that trans-differentiation of MSCs into cells of neural lineage may occur in vitro, no one has yet observed that MSCs give rise to fully differentiated and functional neurons in vivo. The overwhelming body of data indicate that bioactive factors secreted by MSCs in response to the local environment underlie the tissue restorative effects of MSCs. The MSCs that are employed in this therapy are not necessarily stem cells, but progenitor and differentiated cells that escape immune system surveillance and survive in the CNS even for transplantation of allogeneic or xenogeneic MSCs. The injured CNS is stimulated by the MSCs to amplify its intrinsic restorative processes. Treatment of damaged brain with MSCs promotes functional recovery, and facilitates CNS endogenous plasticity and remodeling. The current mini-review is mainly based on our data and focuses on possible cellular and molecular mechanisms of interaction of MSCs with glia, neurons and vessels after brain attack. The transplantation of MSCs opens up new avenues of cell therapy and may provide an effective treatment for various CNS diseases.
Collapse
Affiliation(s)
- Yi Li
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, United States
| | | |
Collapse
|
231
|
Min BH, Lee HJ, Kim YJ. Cartilage Repair Using Mesenchymal Stem Cells. JOURNAL OF THE KOREAN MEDICAL ASSOCIATION 2009. [DOI: 10.5124/jkma.2009.52.11.1077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Byoung-Hyun Min
- Department of Orthopedic Surgery, Ajou University College of Medicine, Korea.
| | - Hyun Jung Lee
- Cell Therapy Center, Ajou University Medical Center, Korea
| | - Young Jick Kim
- Cell Therapy Center, Ajou University Medical Center, Korea
| |
Collapse
|
232
|
Abstract
Articular cartilage repair and regeneration continue to be largely intractable because of the poor regenerative properties of this tissue. The field of articular cartilage tissue engineering, which aims to repair, regenerate, and/or improve injured or diseased articular cartilage functionality, has evoked intense interest and holds great potential for improving articular cartilage therapy. This review provides an overall description of the current state of and progress in articular cartilage repair and regeneration. Traditional therapies and related problems are introduced. More importantly, a variety of promising cell sources, biocompatible tissue engineered scaffolds, scaffoldless techniques, growth factors, and mechanical stimuli used in current articular cartilage tissue engineering are reviewed. Finally, the technical and regulatory challenges of articular cartilage tissue engineering and possible future directions are also discussed.
Collapse
Affiliation(s)
- Lijie Zhang
- Department of Biomedical Engineering, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Jerry Hu
- Department of Biomedical Engineering, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Kyriacos A. Athanasiou
- Department of Biomedical Engineering, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| |
Collapse
|
233
|
Karow M, Popp T, Egea V, Ries C, Jochum M, Neth P. Wnt signalling in mouse mesenchymal stem cells: impact on proliferation, invasion and MMP expression. J Cell Mol Med 2008; 13:2506-2520. [PMID: 19413884 DOI: 10.1111/j.1582-4934.2008.00619.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Based on the capacity of mesenchymal stem cells (MSC) to differentiate into multiple cell types in vitro and in vivo, MCS may be a suitable source for cell therapy and regeneration strategies. A prerequisite for effective clinical applications of human MSC (hMSC) is a profound knowledge of signal transduction cascades that mediate processes like proliferation, targeted migration and differentiation. Recently, we identified the canonical Wnt signal transduction pathway as a key player in hMSC proliferation and invasion. To evaluate whether those findings are transferable to the equivalent counterparts in mice, we studied important steps in the wingless/int-1 (Wnt) signal transduction pathway in mouse MSC (mMSC) and mMSC carrying a T cell specific transcription factor (TCF)/lymphoid enhancer binding factor (LEF)-reporter transgene. We found that the induction of the canonical Wnt pathway resulted in the up-regulation of the known Wnt target gene cyclin D1, closely associated with an enhanced proliferation capacity of mMSC. Interestingly, the expression of the Wnt target gene membrane type 1-matrix metalloproteinase (MT1-MMP) was diminished in mMSC upon Wnt3a stimulation, which came along with an impaired invasion. In line with these findings, MMP-2 and MMP-9 expression levels in mMSC were also decreased after Wnt3a treatment. In contrast, inhibition of Wnt signalling by the knockdown of the transcriptional activator beta-catenin resulted in an up-regulation of MT1-MMP and mMSC invasion. By comparing these findings with the settings in hMSC, major differences in Wnt-regulated MMP expression were observed in mMSC. Thus, our data advice caution when mouse model systems represent the pre-clinical validation of MSC-mediated therapeutical approaches.
Collapse
Affiliation(s)
- Marisa Karow
- Division of Clinical Chemistry and Clinical Biochemistry, Department of Surgery, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Tanja Popp
- Division of Clinical Chemistry and Clinical Biochemistry, Department of Surgery, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Virginia Egea
- Division of Clinical Chemistry and Clinical Biochemistry, Department of Surgery, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Christian Ries
- Division of Clinical Chemistry and Clinical Biochemistry, Department of Surgery, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Marianne Jochum
- Division of Clinical Chemistry and Clinical Biochemistry, Department of Surgery, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Peter Neth
- Division of Clinical Chemistry and Clinical Biochemistry, Department of Surgery, Ludwig-Maximilians-University of Munich, Munich, Germany
| |
Collapse
|
234
|
Arthur A, Zannettino A, Gronthos S. The therapeutic applications of multipotential mesenchymal/stromal stem cells in skeletal tissue repair. J Cell Physiol 2008; 218:237-45. [PMID: 18792913 DOI: 10.1002/jcp.21592] [Citation(s) in RCA: 243] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Four decades after the first isolation and characterization of clonogenic bone marrow stromal cells or mesenchymal stem cells (MSC) in the laboratory of Dr. Alexander Friedenstien, the therapeutic application of their progeny following ex vivo expansion are only now starting to be realized in the clinic. The multipotency, paracrine effects, and immune-modulatory properties of MSC present them as an ideal stem cell candidate for tissue engineering and regenerative medicine. In recent years it has come to light that MSC encompass plasticity that extends beyond the conventional bone, adipose, cartilage, and other skeletal structures, and has expanded to the differentiation of liver, kidney, muscle, skin, neural, and cardiac cell lineages. This review will specifically focus on the skeletal regenerative capacity of bone marrow derived MSC alone or in combination with growth factors, biocompatible scaffolds, and following genetic modification.
Collapse
Affiliation(s)
- Agnieszka Arthur
- Mesenchymal Stem Cell Group, Division of Haematology, Institute of Medical and Veterinary Science, Hanson Institute/University of Adelaide, Adelaide, South Australia, Australia
| | | | | |
Collapse
|
235
|
Abdallah BM, Kassem M. The use of mesenchymal (skeletal) stem cells for treatment of degenerative diseases: current status and future perspectives. J Cell Physiol 2008; 218:9-12. [PMID: 18726996 DOI: 10.1002/jcp.21572] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Human bone marrow derived-mesenchymal (skeletal) stem (MSC) cells are a group of non-hematopoietic stem cells residing in the perivascular niches in bone marrow. These cells have the capacity to differentiate mainly into mesoderm-type cells such as osteoblasts, chondrocytes and adipocytes and possibly but not proven to non-mesodermal cell types. Recently, there has been an increased interest in understanding the biology of MSC due to their potential use in cell-based therapy for multiple degenerative diseases. Here, we will provide an update on the current status of these novel therapeutic opportunities. J. Cell. Physiol. 218: 9-12, 2009. (c) 2008 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Basem M Abdallah
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital & Medical Biotechnology Centre, University of Southern Denmark, Odense, Denmark.
| | | |
Collapse
|
236
|
|
237
|
Crop M, Baan C, Weimar W, Hoogduijn M. Potential of mesenchymal stem cells as immune therapy in solid-organ transplantation. Transpl Int 2008; 22:365-76. [PMID: 19000235 DOI: 10.1111/j.1432-2277.2008.00786.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over the last decade, there has been a rising interest in the use of mesenchymal stem cells (MSCs) for clinical applications. This interest stems from the beneficial properties of MSCs, which include multi-lineage differentiation and immunosuppressive ability, suggesting there is a role for MSC therapy for tissue regeneration and in immunologic disease. Despite recent clinical trials investigating the use of MSCs in treating immune-mediated disease, their applicability in solid-organ transplantation is still unknown. In this review, we identified topics that are important when considering MSC therapy in clinical organ transplantation. Whereas, from other clinical studies, it would appear that administration of MSCs is safe, issues like dosing, timing, route of administration, and in particular the use of autologous or donor-derived MSCs may be of crucial importance for the functional outcome of MSCs treatment in organ transplantation. We discuss these topics and assess the feasibility of MSCs therapy in organ transplantation.
Collapse
Affiliation(s)
- Meindert Crop
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | | | | | | |
Collapse
|
238
|
Neumann K, Dehne T, Endres M, Erggelet C, Kaps C, Ringe J, Sittinger M. Chondrogenic differentiation capacity of human mesenchymal progenitor cells derived from subchondral cortico-spongious bone. J Orthop Res 2008; 26:1449-56. [PMID: 18464264 DOI: 10.1002/jor.20635] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Microfracture is frequently used to repair articular cartilage defects and allows mesenchymal progenitors to migrate from subchondral bone into the defect and form cartilaginous repair tissue. The aim of our study was to analyze the cell surface antigen pattern and the differentiation capacity of cells derived from human subchondral bone. Human progenitor cells were derived from subchondral cortico-spongious bone and grown in the presence of human serum. Stem cell-related cell surface antigens were analyzed by flowcytometry. Cortico-spongious progenitor (CSP) cells showed presence of CD73, CD90, CD105, and STRO-1. Multilineage differentiation potential of CSP cells was documented by histological staining and by gene expression analysis of osteogenic, adipogenic, and chondrogenic marker genes. CSP cells formed a mineralized matrix as demonstrated by von Kossa staining and showed induction of osteocalcin, independent of osteogenic stimulation. During adipogenic differentiation, the adipogenic marker genes fatty acid binding protein 4 and peroxisome proliferative activated receptor gamma were induced. Immunohistochemical staining of cartilage-specific type II collagen and induction of the chondrocytic marker genes cartilage oligomeric matrix protein, aggrecan, and types II and IX collagen confirmed TGF beta 3-mediated chondrogenic lineage development. CSP cells from subchondral bone, as known from microfracture, are multipotent stem cell-like mesenchymal progenitors with a high chondrogenic differentiation potential.
Collapse
Affiliation(s)
- Katja Neumann
- TransTissue Technologies GmbH, Tucholskystrasse 2, 10117 Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
239
|
Bajada S, Mazakova I, Richardson JB, Ashammakhi N. Updates on stem cells and their applications in regenerative medicine. J Tissue Eng Regen Med 2008; 2:169-83. [PMID: 18493906 DOI: 10.1002/term.83] [Citation(s) in RCA: 234] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Stem cells have the capacity for self-renewal and capability of differentiation to various cell lineages. Thus, they represent an important building block for regenerative medicine and tissue engineering. These cells can be broadly classified into embryonic stem cells (ESCs) and non-embryonic or adult stem cells. ESCs have great potential but their use is still limited by several ethical and scientific considerations. The use of bone marrow-, umbilical cord-, adipose tissue-, skin- and amniotic fluid-derived mesenchymal stem cells might be an adequate alternative for translational practice. In particular, bone marrow-derived stem cells have been used successfully in the clinic for bone, cartilage, spinal cord, cardiac and bladder regeneration. Several preclinical experimental studies are under way for the application of stem cells in other conditions where current treatment options are inadequate. Stem cells can be used to improve healthcare by either augmenting the body's own regenerative potential or developing new therapies. This review is not meant to be exhaustive but gives a brief outlook on the past, present and the future of stem cell-based therapies in clinical practice.
Collapse
Affiliation(s)
- Stefan Bajada
- Institute for Science and Technology in Medicine, Keele University, Staffordshire, UK.
| | | | | | | |
Collapse
|
240
|
Mimura T, Imai S, Kubo M, Isoya E, Ando K, Okumura N, Matsusue Y. A novel exogenous concentration-gradient collagen scaffold augments full-thickness articular cartilage repair. Osteoarthritis Cartilage 2008; 16:1083-91. [PMID: 18378169 DOI: 10.1016/j.joca.2008.02.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Accepted: 02/01/2008] [Indexed: 02/02/2023]
Abstract
OBJECTIVES A collagen scaffold has been long used in order to enhance the regeneration of articular cartilage. In the present study, we investigate the effectiveness of a concentration-gradient (CG) collagen that is designed to recruit efficiently the mesenchymal stem cells (MSCs) to the central region of the full-thickness cartilage defects via haptotaxis. METHODS The present study used Cellmatrix (0.3% type I collagen; Nitta gelatin, Osaka, Japan) as the collagen material. We prepared 33%CG collagen gel and 50%CG collagen gel. No gradient collagen gel served as negative control. Full-thickness cartilage defects were created at the patella groove of the rabbit knee, to which the three different collagen gels were transplanted. Bromodeoxyuridine (BrdU) positive, proliferating cells were enumerated and localized, whereas the histological grading score for cartilage regeneration was counted. The expression of type I and type II collagens was evaluated by immunohistochemistry. We also confirmed that the MSCs migrate toward the collagen substrate of higher concentration in a stringently in vitro haptotactic manner. RESULTS Enumeration of the BrdU-positive cells demonstrated that 33%CG collagen gel recruited a significantly larger number of proliferating cells to the central region of the cartilage defect. The histological grading score for the regenerated cartilage treated with 33%CG collagen gel was superior to the other groups. CONCLUSIONS CG collagen scaffold recruits effectively the MSCs to the center of full-thickness cartilage defect and enhances regeneration of the full-thickness cartilage defect.
Collapse
Affiliation(s)
- T Mimura
- Department of Orthopedic Surgery, Shiga University of Medical Science, Tsukinowa-cho, Seta, Otsu, Shiga 520-2192, Japan.
| | | | | | | | | | | | | |
Collapse
|
241
|
Kalwitz G, Endres M, Neumann K, Skriner K, Ringe J, Sezer O, Sittinger M, Häupl T, Kaps C. Gene expression profile of adult human bone marrow-derived mesenchymal stem cells stimulated by the chemokine CXCL7. Int J Biochem Cell Biol 2008; 41:649-58. [PMID: 18707017 DOI: 10.1016/j.biocel.2008.07.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Revised: 07/21/2008] [Accepted: 07/21/2008] [Indexed: 01/16/2023]
Abstract
A variety of chemokines has been shown to recruit human bone marrow-derived mesenchymal stem cells (MSC) and may be potential candidates for chemokine-based tissue regeneration approaches. The aim of our study was to determine whether the chemokine CXCL7 stimulates migration of human bone marrow-derived MSC and to analyze the effect of CXCL7 on the recruitment of MSC on the broad molecular level. Chemotaxis assays documented that high doses of CXCL7 significantly recruited MSC. Gene expression profiling using oligonucleotide microarrays showed that MSC treated with CXCL7 differentially expressed genes related to cell migration, cell adhesion and extracellular matrix remodeling. Pathway analysis showed that CXCL7 induced the expression of all chemokines binding the interleukin (IL) receptors A and B, CXCR1 and CXCR2, as well as the IL6 signal transducer (gp130) and its ligands IL6 and leukemia inhibitory factor (LIF). Induction of differentially expressed chemokines CXCL1-3, CXCL5, and CXCL6 as well as LIF and gp130 in MSC by CXCL7 was verified by real-time polymerase chain reaction. Immunoassay of cell culture supernatants confirmed elevated levels of the interleukins 6 and 8 in MSC upon treatment with CXCL7. Chemotaxis assays showed that interleukin 6 did not recruit MSC. In conclusion, CXCL7 significantly stimulates the migration of human MSC in vitro. Pathway analysis suggests that recruitment of human MSC by CXCL7 is supported by the induction of ligands of the interleukin 8 receptors, synergistically activating the respective signaling pathways.
Collapse
Affiliation(s)
- Gregor Kalwitz
- TransTissue Technologies GmbH, Tucholskystrasse 2, 10117 Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
242
|
Sakao K, Takahashi KA, Arai Y, Inoue A, Tonomura H, Saito M, Yamamoto T, Kanamura N, Imanishi J, Mazda O, Kubo T. Induction of chondrogenic phenotype in synovium-derived progenitor cells by intermittent hydrostatic pressure. Osteoarthritis Cartilage 2008; 16:805-14. [PMID: 18571101 DOI: 10.1016/j.joca.2007.10.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2007] [Accepted: 10/29/2007] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The aim of this study was to investigate the effect of intermittent hydrostatic pressure (IHP) on chondrogenic differentiation of synovium-derived progenitor cells (SPCs). METHODS SPCs, bone marrow-derived progenitor cells and skin fibroblasts from rabbits were subjected to IHP ranging from 1.0 to 5.0 MPa. The mRNA expression of proteoglycan core protein (PG), collagen type II and SOX-9 was examined using real-time reverse transcriptase-polymerase chain reaction (RT-PCR). The production of SOX-9 protein and glycosaminoglycan (GAG) by SPCs was analyzed by Western blot and the dimethylmethylene blue assay. In addition, mitogen-activated protein (MAP) kinase inhibitors for c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK), and the p38 pathway were used to identify the signal transduction pathways. RESULTS Real-time RT-PCR showed that mRNA expression of PG, collagen type II and SOX-9 was significantly enhanced only in SPCs receiving 5.0 MPa of IHP. The production of SOX-9 protein and GAG by SPCs was also increased by exposure to 5.0 MPa of IHP. These up-regulated expressions were suppressed by pretreatment with an inhibitor of JNK, but not with inhibitors of ERK or p38. CONCLUSION Our results demonstrated that the exposure of SPCs to 5.0 MPa of IHP could facilitate induction of the chondrogenic phenotype by the MAP kinase/JNK pathway. This finding suggests the potential for IHP utilization in regenerative treatments for cartilage injuries or osteoarthritis.
Collapse
Affiliation(s)
- K Sakao
- Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
243
|
Freyria AM, Courtes S, Mallein-Gerin F. Différenciation des cellules souches mésenchymateuses adultes humaines : effet chondrogénique de la BMP-2. ACTA ACUST UNITED AC 2008; 56:326-33. [DOI: 10.1016/j.patbio.2007.09.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Accepted: 09/19/2007] [Indexed: 01/22/2023]
|
244
|
Nagase T, Muneta T, Ju YJ, Hara K, Morito T, Koga H, Nimura A, Mochizuki T, Sekiya I. Analysis of the chondrogenic potential of human synovial stem cells according to harvest site and culture parameters in knees with medial compartment osteoarthritis. ACTA ACUST UNITED AC 2008; 58:1389-98. [PMID: 18438859 DOI: 10.1002/art.23418] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Synovial mesenchymal stem cells (MSCs) are an attractive cell source for cartilage regeneration because of their high chondrogenic ability. In this study, we examined the synovium of patients with medial compartment knee osteoarthritis (OA) to determine the proportion of MSCs in relation to cellular compartmentalization, and to identify the culture parameters that could affect the chondrogenic potential of synovial MSCs. METHODS Human synovium was collected from 4 different harvest sites in the knees of patients with medial compartment OA. Each synovial tissue sample was divided into 2 parts, one for histologic assessment and the other for analysis of the cell size, surface epitopes, and chondrogenic potential of colony-forming cells in vitro. RESULTS The numbers of alpha-smooth muscle actin-positive vessels and CD31+ endothelial cells were higher in the medial outer region than in the other regions of OA synovial tissue. The numbers of these cells correlated with the number of colony-forming cells. In parallel with increasing duration of the preculture period, the size of the cells increased, while the chondrogenic potential decreased, and this was correlated with expression of CD90. CONCLUSION Medial compartment knee OA demonstrates variability in the distribution of vessels, which results in a varying distribution of MSCs. The preculture period should be utilized to assess both the potential for expansion and the chondrogenic potential of MSCs.
Collapse
|
245
|
Technology insight: adult mesenchymal stem cells for osteoarthritis therapy. ACTA ACUST UNITED AC 2008; 4:371-80. [PMID: 18477997 DOI: 10.1038/ncprheum0816] [Citation(s) in RCA: 192] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Accepted: 03/03/2008] [Indexed: 12/13/2022]
Abstract
Despite the high prevalence and morbidity of osteoarthritis (OA), an effective treatment for this disease is currently lacking. Restoration of the diseased articular cartilage in patients with OA is, therefore, a challenge of considerable appeal to researchers and clinicians. Techniques that cause multipotent adult mesenchymal stem cells (MSCs) to differentiate into cells of the chondrogenic lineage have led to a variety of experimental strategies to investigate whether MSCs instead of chondrocytes can be used for the regeneration and maintenance of articular cartilage. MSC-based strategies should provide practical advantages for the patient with OA. These strategies include use of MSCs as progenitor cells to engineer cartilage implants that can be used to repair chondral and osteochondral lesions, or as trophic producers of bioactive factors to initiate endogenous regenerative activities in the OA joint. Targeted gene therapy might further enhance these activities of MSCs. Delivery of MSCs might be attained by direct intra-articular injection or by graft of engineered constructs derived from cell-seeded scaffolds; this latter approach could provide a three-dimensional construct with mechanical properties that are congruous with the weight-bearing function of the joint. Promising experimental and clinical data are beginning to emerge in support of the use of MSCs for regenerative applications.
Collapse
|
246
|
Wakitani S, Nawata M, Tensho K, Okabe T, Machida H, Ohgushi H. Repair of articular cartilage defects in the patello-femoral joint with autologous bone marrow mesenchymal cell transplantation: three case reports involving nine defects in five knees. J Tissue Eng Regen Med 2008; 1:74-9. [PMID: 18038395 DOI: 10.1002/term.8] [Citation(s) in RCA: 242] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
To investigate the effectiveness of autologous culture-expanded bone marrow mesenchymal cell transplantation for repairing articular cartilage defects, we transplanted autologous culture-expanded bone marrow mesenchymal cells into nine full-thickness articular cartilage defects of the patello-femoral joints (including two kissing lesions) in the knees of three patients, a 31 year-old female, a 44 year-old male and a 45 year-old male. Three weeks before transplantation, bone marrow blood was aspirated from the iliac crest. Adherent cells were cultured with media containing autologous serum. Single-passaged cells were collected, embedded in a collagen solution (5 x 10(6) cells/ml), placed on a collagen sheet, gelated, transplanted into the defect and covered with autologous periosteum or synovium. Six months after transplantation, the patients' clinical symptoms had improved and the improvements have been maintained over the follow-up periods (17-27 months). Histology of the first patient 12 months after the transplantation revealed that the defect had been repaired with the fibrocartilaginous tissue. Magnetic resonance imaging of the second patient 1 year after transplantation revealed complete coverage of the defect, but we were unable to determine whether or not the material that covered the defects was hyaline cartilage. Autologous bone marrow mesenchymal cells transplantation may be an effective approach to promote the repair of articular cartilage defects.
Collapse
Affiliation(s)
- Shigeyuki Wakitani
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan.
| | | | | | | | | | | |
Collapse
|
247
|
Abstract
Once articular cartilage is injured, it has a very limited capacity for self repair. Although current surgical therapeutic procedures for cartilage repair are clinically useful, they cannot restore a normal articular surface. Current research offers a growing number of bioactive reagents, including proteins and nucleic acids, that may be used to augment various aspects of the repair process. As these agents are difficult to administer effectively, gene-transfer approaches are being developed to provide their sustained synthesis at sites of repair. To augment regeneration of articular cartilage, therapeutic genes can be delivered to the synovium or directly to the cartilage lesion. Gene delivery to the cells of the synovial lining is generally considered more suitable for chondroprotective approaches, based on the expression of anti-inflammatory mediators. Gene transfer targeted at cartilage defects can be achieved by either direct vector administration to cells located at or surrounding the defects, or by transplantation of genetically modified chondrogenic cells into the defect. Several studies have shown that exogenous cDNAs encoding growth factors can be delivered locally to sites of cartilage damage, where they are expressed at therapeutically relevant levels. Furthermore, data is beginning to emerge indicating that efficient delivery and expression of these genes is capable of influencing a repair response toward the synthesis of a more hyaline cartilage repair tissue in vivo. This review presents the current status of gene therapy for cartilage healing and highlights some of the remaining challenges.
Collapse
Affiliation(s)
- Andre F. Steinert
- Orthopaedic Center for Musculoskeletal Research König-Ludwig-Haus, Julius-Maximilians-University, Würzburg, Germany
| | - Ulrich Nöth
- Orthopaedic Center for Musculoskeletal Research König-Ludwig-Haus, Julius-Maximilians-University, Würzburg, Germany
| | - Rocky S. Tuan
- Cartilage Biology and Orthopaedics Branch National Institute of Arthritis, and Musculoskeletal and Skin Diseases National Institutes of Health, Department of Health and Human Services Bethesda, MD, U.S.A
| |
Collapse
|
248
|
Abstract
The application of autologous chondrocytes in cartilage repair procedures is associated with several disadvantages, including injury of healthy cartilage in a preceding surgery frequently resulting in formation of inferior fibrocartilage at defect sites. In order to improve the quality of regeneration, adult mesenchymal stem cells (MSC) are regarded as a promising alternative. The great challenge, when considering MSC for articular cartilage repair, is to generate cells with features of stable chondrocytes which are resistant to hypertrophy and terminal differentiation, as found in hyaline articular cartilage. Common in vitro protocols for chondrogenic differentiation of MSC successfully induce expression of multiple cartilage-specific molecules, including collagen type II and aggrecan, and result in a chondrocyte-like phenotype. However, in vitro chondrogenesis of MSC additionally promotes induction of fibrocartilage-like features such as expression of collagen type I, and hypertrophy, as demonstrated by up-regulation of collagen type X, MMP13 and ALP-activity. As a consequence, differentiated MSC pellets undergo mineralisation and vascularisation after ectopic transplantation in a process similar to endochondral ossification. This review discusses the complexity and entailed challenges when considering MSC from various sources for clinical application and the necessity to optimise chondrogenesis by repressing hypertrophy to obtain functional and suitable cells for cartilage repair.
Collapse
Affiliation(s)
- Karoliina Pelttari
- Division of Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Schlierbacher Landstrasse 2002, Heidelberg, Germany
| | | | | |
Collapse
|
249
|
Sheth RN, Manzano G, Li X, Levi AD. Transplantation of human bone marrow-derived stromal cells into the contused spinal cord of nude rats. J Neurosurg Spine 2008; 8:153-62. [PMID: 18248287 DOI: 10.3171/spi/2008/8/2/153] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECT Human bone marrow stromal cells (hMSCs) constitute a potential source of pluripotent stem cells. In the present study, hMSCs were transplanted into an area of spinal cord contusion in nude rats to determine their survival, differentiation, potential for neuroprotection, and influence on axonal growth and functional recovery. METHODS Twenty-nine animals received 6 x 10(5) hMSCs in 6 microl medium 1 week after a contusion, while 14 control animals received an injection of 6 microl medium alone. Basso-Beattie-Bresnahan (BBB) tests were performed weekly. The spinal cords were collected at 6 weeks posttransplantation for histological analysis and assessment of tissue injury. RESULTS Immunostaining with anti-human mitochondria antibody and pretransplantation labeling with green fluorescent protein demonstrated that the grafted hMSCs survived and were capable of achieving a flattened appearance in the grafted area; however, none of the transplanted cells stained positively for human-specific neuronal, anti-neurofilament H or glial fibrillary acidic protein within the sites of engraftment. While neuronal or astrocytic differentiation was not seen, cells lining blood vessels in the vicinity of the transplant stained positively for anti-human endothelium CD105 antibody. Staining for anti-neurofilament H antibody demonstrated abundant axonlike structures around the transplanted area in the hMSC group. Tissue sparing analysis showed that animals with grafted hMSCs had a smaller area of contusion cyst compared with controls, but there was no significant difference between the two groups in BBB scores. CONCLUSIONS The grafted hMSCs survived for > or = 6 weeks posttransplantation, although they did not differentiate into neural or glial cells. Cells with human endothelial characteristics were observed. Spinal cord-injured rats grafted with hMSCs had smaller contusion cavities, which did not have a significant influence on functional recovery.
Collapse
Affiliation(s)
- Rishi N Sheth
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | | | | |
Collapse
|
250
|
De Bari C, Dell'Accio F, Karystinou A, Guillot PV, Fisk NM, Jones EA, McGonagle D, Khan IM, Archer CW, Mitsiadis TA, Donaldson AN, Luyten FP, Pitzalis C. A biomarker-based mathematical model to predict bone-forming potency of human synovial and periosteal mesenchymal stem cells. ACTA ACUST UNITED AC 2008; 58:240-50. [PMID: 18163504 DOI: 10.1002/art.23143] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE To develop a biomarker-based model to predict osteogenic potency of human mesenchymal stem cells (MSCs) from synovial membrane and periosteum. METHODS MSC populations were derived from adult synovium and periosteum. Phenotype analysis was performed by fluorescence-activated cell sorting and real-time reverse transcriptase-polymerase chain reaction (RT-PCR). Telomere lengths were determined by Southern blot analysis. In vitro osteogenesis was assessed quantitatively by measurements of alkaline phosphatase activity and calcium deposits. To investigate bone formation in vivo, MSCs were seeded onto osteoinductive scaffolds and implanted subcutaneously in nude mice. Bone was assessed by histology, and the human origin investigated by in situ hybridization for human Alu genomic repeats. Quantitation was achieved by histomorphometry and real-time RT-PCR for human osteocalcin. Analysis at the single-cell level was performed with clonal populations obtained by limiting dilution. Multiple regressions were used to explore the incremental predictive value of the markers. RESULTS Periosteal MSCs had significantly greater osteogenic potency than did synovial MSCs inherent to the single cell. Bone was largely of human origin in vivo. Within the same tissue type, there was variability between different donors. To identify predictors of osteogenic potency, we measured the expression levels of osteoblast lineage genes in synovial and periosteal clonal MSCs prior to osteogenic treatment. We identified biomarkers that correlated with osteogenic outcome and developed a mathematical model based on type I collagen and osteoprotegerin expression that predicts the bone-forming potency of MSC preparations, independent of donor-related variables and tissue source. CONCLUSION Our findings indicate that our quality-control mathematical model estimates the bone-forming potency of MSC preparations for bone repair.
Collapse
|