1
|
Vinod E, Parameswaran R, Ramasamy B, Kachroo U. Pondering the Potential of Hyaline Cartilage-Derived Chondroprogenitors for Tissue Regeneration: A Systematic Review. Cartilage 2021; 13:34S-52S. [PMID: 32840123 PMCID: PMC8804774 DOI: 10.1177/1947603520951631] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Chondroprogenitors have recently gained prominence due to promising results seen in in vitro and animal studies as a potential contender in cell-based therapy for cartilage repair. Lack of consensus regarding nomenclature, isolation techniques, and expansion protocols create substantial limitations for translational research, especially given the absence of distinct markers of identification. The objective of this systematic review was to identify and collate information pertaining to hyaline cartilage-derived chondroprogenitors, with regard to their isolation, culture, and outcome measures. DESIGN As per Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, a web-based search of Scopus and PubMed databases was performed from January 2000 to May 2020, which yielded 509 studies. A total of 65 studies were identified that met the standardized inclusion criteria which comprised of, but was not limited to, progenitors derived from fibronectin adhesion, migrated subpopulation from explant cultures, and single-cell sorting. RESULT Literature search revealed that progenitors demonstrated inherent chondrogenesis and minimal tendency for hypertrophy. Multiple sources also demonstrated significantly better outcomes that bone marrow-derived mesenchymal stem cells and comparable results to chondrocytes. With regard to progenitor subgroups, collated evidence points to better and consistent outcomes with the use of migratory progenitors when compared to fibronectin adhesion assay-derived progenitors, although a direct comparison between the two cell populations is warranted. CONCLUSION Since chondroprogenitors exhibit favorable properties for cartilage repair, efficient characterization of progenitors is imperative, to complete their phenotypic profile, so as to optimize their use in translational research for neocartilage formation.
Collapse
Affiliation(s)
- Elizabeth Vinod
- Department of Physiology, Christian
Medical College, Vellore, India,Centre for Stem Cell Research (a unit of
InStem, Bengaluru), Christian Medical College, Vellore, India,Elizabeth Vinod, Department of Physiology,
Christian Medical College, Vellore 632002, Tamil Nadu, India.
| | | | - Boopalan Ramasamy
- Department of Orthopaedics, Royal Darwin
Hospital, Tiwi, Northern Territory, Australia
| | - Upasana Kachroo
- Department of Physiology, Christian
Medical College, Vellore, India
| |
Collapse
|
2
|
Vahedi P, Moghaddamshahabi R, Webster TJ, Calikoglu Koyuncu AC, Ahmadian E, Khan WS, Jimale Mohamed A, Eftekhari A. The Use of Infrapatellar Fat Pad-Derived Mesenchymal Stem Cells in Articular Cartilage Regeneration: A Review. Int J Mol Sci 2021; 22:ijms22179215. [PMID: 34502123 PMCID: PMC8431575 DOI: 10.3390/ijms22179215] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 08/18/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
Cartilage is frequently damaged with a limited capacity for repair. Current treatment strategies are insufficient as they form fibrocartilage as opposed to hyaline cartilage, and do not prevent the progression of degenerative changes. There is increasing interest in the use of autologous mesenchymal stem cells (MSC) for tissue regeneration. MSCs that are used to treat articular cartilage defects must not only present a robust cartilaginous production capacity, but they also must not cause morbidity at the harvest site. In addition, they should be easy to isolate from the tissue and expand in culture without terminal differentiation. The source of MSCs is one of the most important factors that may affect treatment. The infrapatellar fat pad (IPFP) acts as an important reservoir for MSC and is located in the anterior compartment of the knee joint in the extra-synovial area. The IPFP is a rich source of MSCs, and in this review, we discuss studies that demonstrate that these cells have shown many advantages over other tissues in terms of ease of isolation, expansion, and chondrogenic differentiation. Future studies in articular cartilage repair strategies and suitable extraction as well as cell culture methods will extend the therapeutical application of IPFP-derived MSCs into additional orthopedic fields, such as osteoarthritis. This review provides the latest research concerning the use of IPFP-derived MSCs in the treatment of articular cartilage damage, providing critical information for the field to grow.
Collapse
Affiliation(s)
- Parviz Vahedi
- Department of Anatomical Sciences, Maragheh University of Medical Sciences, Maragheh 78151-55158, Iran;
| | - Rana Moghaddamshahabi
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta 99628, North Cyprus, Turkey;
| | - Thomas J. Webster
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA;
| | - Ayse Ceren Calikoglu Koyuncu
- Materials and Metallurgical Engineering Department, Faculty of Technology, Marmara University, Istanbul 34722, Turkey;
- Center for Nanotechnology & Biomaterials Application and Research (NBUAM), Marmara University, Istanbul 34722, Turkey
| | - Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz 51666-15731, Iran;
| | - Wasim S. Khan
- Division of Trauma & Orthopaedic Surgery, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, UK
- Correspondence: (W.S.K.); (A.E.)
| | - Ali Jimale Mohamed
- Department of Pharmacology, Faculty of Medicine, Somali National University, Mogadishu 801, Somalia;
| | - Aziz Eftekhari
- Department of Toxicology and Pharmacology, Maragheh University of Medical Sciences, Maragheh 78151-55158, Iran
- Department of Synthesis and Characterization of Polymers, Polymer Institute, Slovak Academy of Sciences (SAS), Dúbravská cesta, 9, 845 41 Bratislava, Slovakia
- Correspondence: (W.S.K.); (A.E.)
| |
Collapse
|
3
|
Arakura M, Lee SY, Fukui T, Oe K, Takahara S, Matsumoto T, Hayashi S, Matsushita T, Kuroda R, Niikura T. Endochondral Bone Tissue Engineering Using Human Induced Pluripotent Stem Cells. Tissue Eng Part A 2021; 28:184-195. [PMID: 34309415 DOI: 10.1089/ten.tea.2021.0009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
There has been great interest in the use of induced pluripotent stem cells (iPSCs) in bone regenerative strategies for bone defects. In the present study, we investigated whether the implantation of chondrogenically differentiated iPSC-derived mesenchymal stem cells (iMSCs) could lead to the successful regeneration of bone defects in nude mice. Two clones of human iPSCs (201B7 and 454E2) were used. After the generation of iMSCs, chondrogenic differentiation was achieved using a three-dimensional pellet culture. Then, a 2-mm defect was created in the radius of nude mice and chondrogenically differentiated iMSC pellets were placed in the defect. Micro-computed tomography (μ-CT) imaging analysis was performed 8 weeks after transplantation to assess bone regeneration. Eleven out of 11 (100%) radii in the 201B7 cell-derived-pellet transplantation group and 7 out of 10 (70%) radii in the 454E2 cell-derived-pellet transplantation group showed bone union. On the other hand, only 2 out of 11 radii (18%) in the control group showed bone union. Therefore, the bone union rates in the experimental groups were significantly higher than that in the control group (p < 0.05). Histological analysis 2 weeks post-implantation in the experimental groups revealed hypertrophic chondrocytes within grafted iMSC pellets, and the formation of woven bone around them; this hypertrophic chondrocyte transitioning to the newly formed bone suggests that the cartilaginous template can trigger the process of endochondral bone ossification (ECO). Four weeks post-implantation, the cartilage template was reduced in size; newly formed woven bone predominated at the defect site. New vessels were surrounded by a matrix of woven bone and the hypertrophic chondrocytes transitioning to the newly formed bone indicated the progression of ECO. Eight weeks post-implantation, the pellets were completely resorbed and replaced by bone; complete bone union was overall observed. Dense mature bone developed with evidence of lamellar-like bone formation. Collectively, our results suggest that iMSC-based cartilage grafts recapitulating the morphogenetic process of ECO in the context of embryonic skeletogenesis are a novel and promising strategy for the repair of large bone defects.
Collapse
Affiliation(s)
- Michio Arakura
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Hyogo, Japan;
| | - Sang Yang Lee
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Hyogo, Japan.,Department of Orthopaedic Surgery, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan;
| | - Tomoaki Fukui
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Hyogo, Japan;
| | - Keisuke Oe
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Hyogo, Japan;
| | - Shunsuke Takahara
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Hyogo, Japan;
| | - Tomoyuki Matsumoto
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Hyogo, Japan;
| | - Shinya Hayashi
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Hyogo, Japan;
| | - Takehiko Matsushita
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Hyogo, Japan;
| | - Ryosuke Kuroda
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Hyogo, Japan;
| | - Takahiro Niikura
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Japan;
| |
Collapse
|
4
|
Kouroupis D, Bowles AC, Best TM, Kaplan LD, Correa D. CD10/Neprilysin Enrichment in Infrapatellar Fat Pad-Derived Mesenchymal Stem Cells Under Regulatory-Compliant Conditions: Implications for Efficient Synovitis and Fat Pad Fibrosis Reversal. Am J Sports Med 2020; 48:2013-2027. [PMID: 32427493 DOI: 10.1177/0363546520917699] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Synovitis and infrapatellar fat pad (IFP) fibrosis participate in various conditions of the knee. Substance P (SP), a neurotransmitter secreted within those structures and historically associated with nociception, also modulates local neurogenic inflammatory and fibrotic responses. Exposure of IFP mesenchymal stem cells (IFP-MSCs) to a proinflammatory/profibrotic environment (ex vivo priming with TNFα, IFNγ, and CTGF) induces their expression of CD10/neprilysin, effectively degrading SP in vitro and in vivo. PURPOSE/HYPOTHESIS The purpose was to test the therapeutic effects of IFP-MSCs processed under regulatory-compliant protocols, comparing them side-by-side with standard fetal bovine serum (FBS)-grown cells. The hypothesis was that when processed under such protocols, IFP-MSCs do not require ex vivo priming to acquire a CD10-rich phenotype efficiently degrading SP and reversing synovitis and IFP fibrosis. STUDY DESIGN Controlled laboratory study. METHODS Human IFP-MSCs were processed in FBS or either of 2 alternative conditions-regulatory-compliant pooled human platelet lysate (hPL) and chemically reinforced medium (Ch-R)-and then subjected to proinflammatory/profibrotic priming with TNFα, IFNγ, and CTGF. Cells were assessed for in vitro proliferation, stemness, immunophenotype, differentiation potential, transcriptional and secretory profiles, and SP degradation. Based on a rat model of acute synovitis and IFP fibrosis, the in vivo efficacy of cells degrading SP plus reversing structural signs of inflammation and fibrosis was assessed. RESULTS When compared with FBS, IFP-MSCs processed with either hPL or Ch-R exhibited a CD10High phenotype and showed enhanced proliferation, differentiation, and immunomodulatory transcriptional and secretory profiles (amplified by priming). Both methods recapitulated and augmented the secretion of growth factors seen with FBS plus priming, with some differences between them. Functionally, in vitro SP degradation was more efficient in hPL and Ch-R, confirmed upon intra-articular injection in vivo where CD10-rich IFP-MSCs also dramatically reversed signs of synovitis and IFP fibrosis even without priming or at significantly lower cell doses. CONCLUSION hPL and Ch-R formulations can effectively replace FBS plus priming to induce specific therapeutic attributes in IFP-MSCs. The resulting fine-tuned, regulatory-compliant, cell-based product has potential future utilization as a novel minimally invasive cell therapy for the treatment of synovitis and IFP fibrosis. CLINICAL RELEVANCE The therapeutic enhancement of IFP-MSCs manufactured under regulatory-compliant conditions suggests that such a strategy could accelerate the time from preclinical to clinical phases. The therapeutic efficacy obtained at lower MSC numbers than currently needed and the avoidance of cell priming for efficient results could have a significant effect on the design of clinical protocols to potentially treat conditions involving synovitis and IFP fibrosis.
Collapse
Affiliation(s)
- Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA.,Diabetes Research Institute and Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Annie C Bowles
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA.,Diabetes Research Institute and Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, Florida, USA.,Department of Biomedical Engineering, College of Engineering, University of Miami, Miami, Florida, USA
| | - Thomas M Best
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Lee D Kaplan
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Diego Correa
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA.,Diabetes Research Institute and Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
5
|
Wu Y, Yang Z, Denslin V, Ren X, Lee CS, Yap FL, Lee EH. Repair of Osteochondral Defects With Predifferentiated Mesenchymal Stem Cells of Distinct Phenotypic Character Derived From a Nanotopographic Platform. Am J Sports Med 2020; 48:1735-1747. [PMID: 32191492 DOI: 10.1177/0363546520907137] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Articular cartilage has a zonal architecture and biphasic mechanical properties. The recapitulation of surface lubrication properties with high compressibility of the deeper layers of articular cartilage during regeneration is essential in achieving long-term cartilage integrity. Current clinical approaches for cartilage repair, especially with the use of mesenchymal stem cells (MSCs), have yet to restore the hierarchically organized architecture of articular cartilage. HYPOTHESIS MSCs predifferentiated on surfaces with specific nanotopographic patterns can provide phenotypically stable and defined chondrogenic cells and, when delivered as a bilayered stratified construct at the cartilage defect site, will facilitate the formation of functionally superior cartilage tissue in vivo. STUDY DESIGN Controlled laboratory study. METHODS MSCs were subjected to chondrogenic differentiation on specific nanopatterned surfaces. The phenotype of the differentiated cells was assessed by the expression of cartilage markers. The ability of the 2-dimensional nanopattern-generated chondrogenic cells to retain their phenotypic characteristics after removal from the patterned surface was tested by subjecting the enzymatically harvested cells to 3-dimensional fibrin hydrogel culture. The in vivo efficacy in cartilage repair was demonstrated in an osteochondral rabbit defect model. Repair by bilayered construct with specific nanopattern predifferentiated cells was compared with implantation with cell-free fibrin hydrogel, undifferentiated MSCs, and mixed-phenotype nanopattern predifferentiated MSCs. Cartilage repair was evaluated at 12 weeks after implantation. RESULTS Three weeks of predifferentiation on 2-dimensional nanotopographic patterns was able to generate phenotypically stable chondrogenic cells. Implantation of nanopatterned differentiated MSCs as stratified bilayered hydrogel constructs improved the repair quality of cartilage defects, as indicated by histological scoring, mechanical properties, and polarized microscopy analysis. CONCLUSION Our results indicate that with an appropriate period of differentiation, 2-dimensional nanotopographic patterns can be employed to generate phenotypically stable chondrogenic cells, which, when implanted as stratified bilayered hydrogel constructs, were able to form functionally superior cartilage tissue. CLINICAL RELEVANCE Our approach provides a relatively straightforward method of obtaining large quantities of zone-specific chondrocytes from MSCs to engineer a stratified cartilage construct that could recapitulate the zonal architecture of hyaline cartilage, and it represents a significant improvement in current MSC-based cartilage regeneration.
Collapse
Affiliation(s)
- Yingnan Wu
- Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Zheng Yang
- Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Vinitha Denslin
- Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore
| | - XiaFei Ren
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chang Sheng Lee
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), Singapore
| | - Fung Ling Yap
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), Singapore
| | - Eng Hin Lee
- Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
6
|
Sun Y, Chen S, Pei M. Comparative advantages of infrapatellar fat pad: an emerging stem cell source for regenerative medicine. Rheumatology (Oxford) 2018; 57:2072-2086. [PMID: 29373763 PMCID: PMC6256334 DOI: 10.1093/rheumatology/kex487] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 11/13/2017] [Indexed: 12/15/2022] Open
Abstract
Growing evidence indicates that infrapatellar fat pad (IPFP)-derived stem cells (IPFSCs) exert robust proliferation capacities and multilineage differentiation potentials. However, few papers summarize the advantages that the IPFP and IPFSCs have in regenerative medicine. In this review we delineate the development and anatomy of the IPFP by comparing it with an adjacent fibrous tissue, synovium, and a more frequently harvested fat depot, subcutaneous adipose tissue. Furthermore, we explore the similarities and differences of stem cells from these three tissues in terms of IPFSCs, synovium-derived stem cells and subcutaneous adipose tissue-derived stem cells in proliferation capacity and tri-lineage differentiation potentials, including chondrogenesis, osteogenesis and adipogenesis. Finally, we highlight the advantages of IPFSCs in regenerative medicine, such as the abundant accessibility and the ability to resist inflammation and senescence, two hurdles for cell-based tissue regeneration. Considering the comparative advantages of IPFSCs, the IPFP can serve as an excellent stem cell source for regenerative medicine, particularly for cartilage regeneration.
Collapse
Affiliation(s)
- Yu Sun
- Department of Orthopaedics, Orthopaedics Institute, Subei People’s Hospital of Jiangsu Province, Yangzhou, Jiangsu, China
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
| | - Song Chen
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Exercise Physiology, West Virginia University, Morgantown, WV, USA
- Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
7
|
Repair of Damaged Articular Cartilage: Current Approaches and Future Directions. Int J Mol Sci 2018; 19:ijms19082366. [PMID: 30103493 PMCID: PMC6122081 DOI: 10.3390/ijms19082366] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/07/2018] [Accepted: 08/07/2018] [Indexed: 12/28/2022] Open
Abstract
Articular hyaline cartilage is extensively hydrated, but it is neither innervated nor vascularized, and its low cell density allows only extremely limited self-renewal. Most clinical and research efforts currently focus on the restoration of cartilage damaged in connection with osteoarthritis or trauma. Here, we discuss current clinical approaches for repairing cartilage, as well as research approaches which are currently developing, and those under translation into clinical practice. We also describe potential future directions in this area, including tissue engineering based on scaffolding and/or stem cells as well as a combination of gene and cell therapy. Particular focus is placed on cell-based approaches and the potential of recently characterized chondro-progenitors; progress with induced pluripotent stem cells is also discussed. In this context, we also consider the ability of different types of stem cell to restore hyaline cartilage and the importance of mimicking the environment in vivo during cell expansion and differentiation into mature chondrocytes.
Collapse
|
8
|
Iwasa K, Reddi AH. Optimization of Methods for Articular Cartilage Surface Tissue Engineering: Cell Density and Transforming Growth Factor Beta Are Critical for Self-Assembly and Lubricin Secretion. Tissue Eng Part C Methods 2018; 23:389-395. [PMID: 28578597 DOI: 10.1089/ten.tec.2017.0121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE Lubricin/superficial zone protein (SZP)/proteoglycan4 (PRG4) plays an important role in boundary lubrication in articular cartilage. Lubricin is secreted by superficial zone chondrocytes and synoviocytes of the synovium. The specific objective of this investigation is to optimize the methods for tissue engineering of articular cartilage surface. The aim of this study is to investigate the effect of cell density on the self-assembly of superficial zone chondrocytes and lubricin secretion as a functional assessment. DESIGN Superficial zone chondrocytes were cultivated as a monolayer at low, medium, and high densities. Chondrocytes at the three different densities were treated with transforming growth factor beta (TGF-β)1 twice a week or daily, and the accumulated lubricin in the culture medium was analyzed by immunoblots and quantitated by enzyme-linked immunosorbent assay (ELISA). RESULTS Cell numbers in low and medium densities were increased by TGF-β1; whereas cell numbers in high-density cell cultures were decreased by twice-a-week treatment of TGF-β1. On the other hand, the cell numbers were maintained by daily TGF-β treatment. Immunoblots and quantitation of lubricin by ELISA analysis indicated that TGF-β1 stimulated lubricin secretion by superficial zone chondrocytes at all densities with twice-a-week TGF-β treatment. It is noteworthy that the daily treatment of TGF-β1 increased lubricin much higher compared with twice-a-week treatment. CONCLUSIONS These data demonstrate that daily treatment is optimal for the TGF-β1 response in a higher density of monolayer cultures. These findings have implications for self-assembly of surface zone chondrocytes of articular cartilage for application in tissue engineering of articular cartilage surface.
Collapse
Affiliation(s)
- Kenjiro Iwasa
- Department of Orthopaedic Surgery, Lawrence Ellison Center for Tissue Regeneration and Repair, School of Medicine, University of California , Davis, Sacramento, California
| | - A Hari Reddi
- Department of Orthopaedic Surgery, Lawrence Ellison Center for Tissue Regeneration and Repair, School of Medicine, University of California , Davis, Sacramento, California
| |
Collapse
|
9
|
Vinod E, Boopalan PRJVC, Sathishkumar S. Reserve or Resident Progenitors in Cartilage? Comparative Analysis of Chondrocytes versus Chondroprogenitors and Their Role in Cartilage Repair. Cartilage 2018; 9:171-182. [PMID: 29047310 PMCID: PMC5871122 DOI: 10.1177/1947603517736108] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Introduction Articular cartilage is made up of hyaline tissue embodying chondrocytes, which arise from mesenchymal stromal cells (MSCs) and specialized extracellular matrix. Despite possessing resident progenitors in and around the joint primed for chondrogenesis, cartilage has limited intrinsic capacity of repair and cell turnover. Advances in isolation, culture, and characterization of these progenitors have raised the possibility for their use in cell-based cartilage repair. Chondroprogenitors (CPCs) have been classified as MSCs and have been postulated to play a vital role in injury response and are identified by their colony forming ability, proliferative potential, telomere dynamics, multipotency, and expression of stem cell markers. The combined presence of CPCs and chondrocytes within the same tissue compartments and the ability of chondrocytes to dedifferentiate and acquire stemness during culture expansion has obscured our ability to define and provide clear-cut differences between these 2 cell populations. Objective This review aims to evaluate and summarize the available literature on CPCs in terms of their origin, growth kinetics, molecular characteristics, and differential and therapeutic potential with emphasis on their difference from daughter chondrocytes. Design For this systematic review, a comprehensive electronic search was performed on PubMed and Google Scholar using relevant terms such as chondrocytes, chondroprogenitors, and surface marker expression. Results and Conclusion Our comparative analysis shows that there is an ill-defined distinction between CPCs and chondrocytes with respect to their cell surface expression (MSC markers and CPC-specific markers) and differentiation potential. Accumulating evidence indicates that the 2 subpopulations may be distinguished based on their growth kinetics and chondrogenic marker.
Collapse
Affiliation(s)
- Elizabeth Vinod
- Department of Physiology, Christian Medical College, Vellore, India
| | - P. R. J. V. C. Boopalan
- Department of Orthopaedics, Christian Medical College/Center for Stem Cell Research, Vellore, India,P. R. J. V. C. Boopalan, Department of Orthopaedics, Centre for Stem Cell Research, Christian Medical College & Hospital, Vellore 632002, India.
| | | |
Collapse
|
10
|
Zhao Z, Zhou X, Guan J, Wu M, Zhou J. Co-implantation of bone marrow mesenchymal stem cells and chondrocytes increase the viability of chondrocytes in rat osteo-chondral defects. Oncol Lett 2018; 15:7021-7027. [PMID: 29731871 PMCID: PMC5921083 DOI: 10.3892/ol.2018.8195] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 12/22/2017] [Indexed: 12/11/2022] Open
Abstract
Replacement of chondrocytes by adult stem cells was believed to improve the performance of autologous chondrocytes transplantation, since less chondrocytes were needed. Previous studies have demonstrated that the increased cartilage production in pellet co-cultures of chondrocytes and bone marrow mesenchymal stem cells (BMSCs) is due to the trophic effects of the MSC by stimulating chondrocyte proliferation and matrix production. However, the destination of MSCs or chondrocytes after implanted in osteo-chondral defects is not clear. The aim of the present study is to investigate the viability of MSCs and chondrocytes after co-implantation into a rat osteo-chondral defect model. MSCs were isolated from bone marrow and chondrocytes were extracted from knee joints of neonatal rats. Results of sulfated glycosaminoglycans (GAG) and collagen quantification demonstrated that co-culture pellets of BMSCs and chondrocytes have more GAG deposition than that of BMSCs or chondrocytes alone. Tracking cells with fluorescence protein demonstrated that MSCs disappeared following co-culture. In a rat knee injury model, co-implantation of BMSCs and chondrocytes contained more viable chondrocytes than chondrocytes implanted alone. To conclude, BMSCs were replaced by chondrocytes in pellet co-culture and BMSCs increased the viability of chondrocytes following co-implantation in a osteo-chondral defects model. Co-implantation of BMSCs and chondrocytes may be a promising approach to repairing osteo-chondral defects in the clinical setting.
Collapse
Affiliation(s)
- Zhi Zhao
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical College, Anhui Key Laboratory of Tissue Transplantation, Bengbu, Anhui 233004, P.R. China
| | - Xinshe Zhou
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical College, Anhui Key Laboratory of Tissue Transplantation, Bengbu, Anhui 233004, P.R. China
| | - Jianzhong Guan
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical College, Anhui Key Laboratory of Tissue Transplantation, Bengbu, Anhui 233004, P.R. China
| | - Min Wu
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical College, Anhui Key Laboratory of Tissue Transplantation, Bengbu, Anhui 233004, P.R. China
| | - Jiansheng Zhou
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical College, Anhui Key Laboratory of Tissue Transplantation, Bengbu, Anhui 233004, P.R. China
| |
Collapse
|
11
|
Characterisation and immunosuppressive activity of human cartilage-derived mesenchymal stem cells. Cytotechnology 2018; 70:1037-1050. [PMID: 29497876 DOI: 10.1007/s10616-017-0182-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/07/2017] [Indexed: 01/04/2023] Open
Abstract
Mesenchymal stem cells (MSCs) exert potent immuno-regulatory activities on various immune cells and also differentiate into various mesodermal lineages besides retaining a distinct self-renewal ability. Such exclusive characteristics had enabled MSCs to be recognised as an ideal source for cell-based treatment in regenerative medicine and immunotherapy. Thus, considering MSCs for treating degenerative disease of organs with limited regenerative potential such as cartilage would serve as an ideal therapy. This study explored the feasibility of generating human cartilage-derived MSCs (hC-MSCs) from sports injured patients and characterised based on multipotent differentiation and immunosuppressive activities. Cartilage tissues harvested from a non-weight bearing region during an arthroscopy procedure were used to generate MSCs. Despite the classic morphology of fibroblast-like cells and a defined immunophenotyping, MSCs expressed early embryonic transcriptional markers (SOX2, REX1, OCT4 and NANOG) and differentiated into chondrocytes, adipocytes and osteocytes when induced accordingly. Upon co-culture with PHA-L activated T-cells, hC-MSCs suppressed the proliferation of the T-cells in a dose-dependent manner. Although, hC-MSCs did not alter the activation profile of T cells significantly, yet prevented the entering of activated T cells into S phase of the cell cycle by cell cycle arrest. The present study has strengthened the evidence of tissue-resident mesenchymal stem cells in human cartilage tissue. The endogenous MSCs could be an excellent tool in treating dysregulated immune response that associated with cartilage since hC-MSCs exerted both immunosuppressive and regenerative capabilities.
Collapse
|
12
|
Tribe HC, McEwan J, Taylor H, Oreffo ROC, Tare RS. Mesenchymal Stem Cells: Potential Role in the Treatment of Osteochondral Lesions of the Ankle. Biotechnol J 2017; 12:1700070. [PMID: 29068173 PMCID: PMC5765412 DOI: 10.1002/biot.201700070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 10/13/2017] [Indexed: 12/11/2022]
Abstract
Given articular cartilage has a limited repair potential, untreated osteochondral lesions of the ankle can lead to debilitating symptoms and joint deterioration necessitating joint replacement. While a wide range of reparative and restorative surgical techniques have been developed to treat osteochondral lesions of the ankle, there is no consensus in the literature regarding which is the ideal treatment. Tissue engineering strategies, encompassing stem cells, somatic cells, biomaterials, and stimulatory signals (biological and mechanical), have a potentially valuable role in the treatment of osteochondral lesions. Mesenchymal stem cells (MSCs) are an attractive resource for regenerative medicine approaches, given their ability to self-renew and differentiate into multiple stromal cell types, including chondrocytes. Although MSCs have demonstrated significant promise in in vitro and in vivo preclinical studies, their success in treating osteochondral lesions of the ankle is inconsistent, necessitating further clinical trials to validate their application. This review highlights the role of MSCs in cartilage regeneration and how the application of biomaterials and stimulatory signals can enhance chondrogenesis. The current treatments for osteochondral lesions of the ankle using regenerative medicine strategies are reviewed to provide a clinical context. The challenges for cartilage regeneration, along with potential solutions and safety concerns are also discussed.
Collapse
Affiliation(s)
- Howard C. Tribe
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and RegenerationFaculty of MedicineUniversity of SouthamptonSouthamptonSO16 6YDUK
- Foot and Ankle Orthopaedic DepartmentRoyal Bournemouth HospitalBournemouthBH7 7DWUK
| | - Josephine McEwan
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and RegenerationFaculty of MedicineUniversity of SouthamptonSouthamptonSO16 6YDUK
| | - Heath Taylor
- Foot and Ankle Orthopaedic DepartmentRoyal Bournemouth HospitalBournemouthBH7 7DWUK
| | - Richard O. C. Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and RegenerationFaculty of MedicineUniversity of SouthamptonSouthamptonSO16 6YDUK
| | - Rahul S. Tare
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and RegenerationFaculty of MedicineUniversity of SouthamptonSouthamptonSO16 6YDUK
- Bioengineering Science, Mechanical Engineering DepartmentFaculty of Engineering and the EnvironmentUniversity of SouthamptonSouthamptonSO17 1BJUK
| |
Collapse
|
13
|
Sakata R, Reddi AH. Platelet-Rich Plasma Modulates Actions on Articular Cartilage Lubrication and Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2016; 22:408-419. [DOI: 10.1089/ten.teb.2015.0534] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Ryosuke Sakata
- Department of Orthopedic Surgery, Center for Tissue Regeneration and Repair, University of California, Davis, Sacramento, California
| | - A. Hari Reddi
- Department of Orthopedic Surgery, Center for Tissue Regeneration and Repair, University of California, Davis, Sacramento, California
| |
Collapse
|
14
|
Schagemann JC, Rudert N, Taylor ME, Sim S, Quenneville E, Garon M, Klinger M, Buschmann MD, Mittelstaedt H. Bilayer Implants: Electromechanical Assessment of Regenerated Articular Cartilage in a Sheep Model. Cartilage 2016; 7:346-60. [PMID: 27688843 PMCID: PMC5029563 DOI: 10.1177/1947603515623992] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To compare the regenerative capacity of 2 distinct bilayer implants for the restoration of osteochondral defects in a preliminary sheep model. METHODS Critical sized osteochondral defects were treated with a novel biomimetic poly-ε-caprolactone (PCL) implant (Treatment No. 2; n = 6) or a combination of Chondro-Gide and Orthoss (Treatment No. 1; n = 6). At 19 months postoperation, repair tissue (n = 5 each) was analyzed for histology and biochemistry. Electromechanical mappings (Arthro-BST) were performed ex vivo. RESULTS Histological scores, electromechanical quantitative parameter values, dsDNA and sGAG contents measured at the repair sites were statistically lower than those obtained from the contralateral surfaces. Electromechanical mappings and higher dsDNA and sGAG/weight levels indicated better regeneration for Treatment No. 1. However, these differences were not significant. For both treatments, Arthro-BST revealed early signs of degeneration of the cartilage surrounding the repair site. The International Cartilage Repair Society II histological scores of the repair tissue were significantly higher for Treatment No. 1 (10.3 ± 0.38 SE) compared to Treatment No. 2 (8.7 ± 0.45 SE). The parameters cell morphology and vascularization scored highest whereas tidemark formation scored the lowest. CONCLUSION There was cell infiltration and regeneration of bone and cartilage. However, repair was incomplete and fibrocartilaginous. There were no significant differences in the quality of regeneration between the treatments except in some histological scoring categories. The results from Arthro-BST measurements were comparable to traditional invasive/destructive methods of measuring quality of cartilage repair.
Collapse
Affiliation(s)
- Jan C. Schagemann
- University Medical Center Schleswig-Holstein Campus Lübeck, Clinic for Orthopedics and Trauma Surgery, Lübeck, Germany,Mayo Clinic, Orthopedic Surgery, Rochester, MN, USA,Jan C. Schagemann, University Medical Center Schleswig Holstein Campus Lübeck, Clinic for Orthopedics and Trauma Surgery, Ratzeburger Allee 160, 23538 Lübeck, Germany. Email
| | - Nicola Rudert
- University Medical Center Schleswig-Holstein Campus Lübeck, Clinic for Orthopedics and Trauma Surgery, Lübeck, Germany
| | | | - Sotcheadt Sim
- Biomedical and Chemical Engineering, Polytechnique Montreal, Montreal, Canada,Biomomentum Inc., Laval, Quebec, Canada
| | | | | | | | | | - Hagen Mittelstaedt
- University Medical Center Schleswig-Holstein Campus Lübeck, Clinic for Orthopedics and Trauma Surgery, Lübeck, Germany
| |
Collapse
|
15
|
Miyatake K, Iwasa K, McNary SM, Peng G, Reddi AH. Modulation of Superficial Zone Protein/Lubricin/PRG4 by Kartogenin and Transforming Growth Factor-β1 in Surface Zone Chondrocytes in Bovine Articular Cartilage. Cartilage 2016; 7:388-97. [PMID: 27688846 PMCID: PMC5029568 DOI: 10.1177/1947603516630789] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE Superficial zone protein (SZP)/lubricin/PRG4 functions as a boundary lubricant in articular cartilage to decrease friction and wear. As articular cartilage lubrication is critical for normal joint function, the accumulation of SZP at the surface of cartilage is important for joint homeostasis. Recently, a heterocyclic compound called kartogenin (KGN) was found to induce chondrogenic differentiation and enhance mRNA expression of lubricin. The objective of this study was to determine whether KGN can stimulate synthesis of SZP in superficial zone, articular chondrocytes. DESIGN We investigated the effects of KGN and transforming growth factor-β1 (TGF-β1) on articular cartilage and synovium of the bovine knee joint by evaluating SZP secretion by enzyme-linked immunosorbent assay analysis. Monolayer, micromass, and explant cultures of articular cartilage, and monolayer culture of synoviocytes, were treated with KGN. SZP accumulation in the medium was evaluated and mRNA expression was measured through quantitative polymerase chain reaction. RESULTS TGF-β1 stimulated SZP secretion by superficial zone chondrocytes in monolayer, explant, and micromass cultures as expected. In addition, SZP secretion was inhibited by IL-1β in explant cultures, and enhanced by TGF-β1 in synoviocyte monolayer cultures. Although KGN elicited a 1.2-fold increase in SZP mRNA expression in combination with TGF-β1, KGN neither stimulated any significant increases in SZP synthesis nor prevented catabolic decreases in SZP production from IL-1β. CONCLUSIONS These data suggest that the chondrogenic effects of KGN depend on cellular phenotype and differentiation status, as KGN did not alter SZP synthesis in differentiated, superficial zone articular chondrocytes.
Collapse
Affiliation(s)
- Kazumasa Miyatake
- Department of Orthopaedic Surgery, Lawrence Ellison Center for Tissue Regeneration and Repair, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Kenjiro Iwasa
- Department of Orthopaedic Surgery, Lawrence Ellison Center for Tissue Regeneration and Repair, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Sean M. McNary
- Department of Orthopaedic Surgery, Lawrence Ellison Center for Tissue Regeneration and Repair, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Gordon Peng
- Department of Orthopaedic Surgery, Lawrence Ellison Center for Tissue Regeneration and Repair, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - A. Hari Reddi
- Department of Orthopaedic Surgery, Lawrence Ellison Center for Tissue Regeneration and Repair, School of Medicine, University of California, Davis, Sacramento, CA, USA,A. Hari Reddi, Department of Orthopaedic Surgery, School of Medicine, University of California, Davis, Research Building I, Room 2000, Sacramento, CA 95817, USA.
| |
Collapse
|
16
|
Nakagawa Y, Muneta T, Otabe K, Ozeki N, Mizuno M, Udo M, Saito R, Yanagisawa K, Ichinose S, Koga H, Tsuji K, Sekiya I. Cartilage Derived from Bone Marrow Mesenchymal Stem Cells Expresses Lubricin In Vitro and In Vivo. PLoS One 2016; 11:e0148777. [PMID: 26867127 PMCID: PMC4750963 DOI: 10.1371/journal.pone.0148777] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 01/22/2016] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Lubricin expression in the superficial cartilage will be a crucial factor in the success of cartilage regeneration. Mesenchymal stem cells (MSCs) are an attractive cell source and the use of aggregates of MSCs has some advantages in terms of chondrogenic potential and efficiency of cell adhesion. Lubricin expression in transplanted MSCs has not been fully elucidated so far. Our goals were to determine (1) whether cartilage pellets of human MSCs expressed lubricin in vitro chondrogenesis, (2) whether aggregates of human MSCs promoted lubricin expression, and (3) whether aggregates of MSCs expressed lubricin in the superficial cartilage after transplantation into osteochondral defects in rats. METHODS For in vitro analysis, human bone marrow (BM) MSCs were differentiated into cartilage by pellet culture, and also aggregated using the hanging drop technique. For an animal study, aggregates of BM MSCs derived from GFP transgenic rats were transplanted to the osteochondral defect in the trochlear groove of wild type rat knee joints. Lubricin expression was mainly evaluated in differentiated and regenerated cartilages. RESULTS In in vitro analysis, lubricin was detected in the superficial zone of the pellets and conditioned medium. mRNA expression of Proteoglycan4 (Prg4), which encodes lubricin, in pellets was significantly higher than that of undifferentiated MSCs. Aggregates showed different morphological features between the superficial and deep zone, and the Prg4 mRNA expression increased after aggregate formation. Lubricin was also found in the aggregate. In a rat study, articular cartilage regeneration was significantly better in the MSC group than in the control group as shown by macroscopical and histological analysis. The transmission electron microscope showed that morphology of the superficial cartilage in the MSC group was closer to that of the intact cartilage than in the control group. GFP positive cells remained in the repaired tissue and expressed lubricin in the superficial cartilage. CONCLUSION Cartilage derived from MSCs expressed lubricin protein both in vitro and in vivo. Aggregation promoted lubricin expression of MSCs in vitro and transplantation of aggregates of MSCs regenerated cartilage including the superficial zone in a rat osteochondral defect model. Our results indicate that aggregated MSCs could be clinically relevant for therapeutic approaches to articular cartilage regeneration with an appropriate superficial zone in the future.
Collapse
Affiliation(s)
- Yusuke Nakagawa
- Department of Joint Surgery and Sports Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takeshi Muneta
- Department of Joint Surgery and Sports Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Koji Otabe
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Nobutake Ozeki
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mitsuru Mizuno
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mio Udo
- Department of Joint Surgery and Sports Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ryusuke Saito
- Department of Joint Surgery and Sports Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Katsuaki Yanagisawa
- Department of Joint Surgery and Sports Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shizuko Ichinose
- Research Center for Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hideyuki Koga
- Department of Joint Surgery and Sports Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kunikazu Tsuji
- Department of Cartilage Regeneration, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ichiro Sekiya
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Tokyo, Japan
- * E-mail:
| |
Collapse
|
17
|
Wu L, Prins HJ, Leijten J, Helder MN, Evseenko D, Moroni L, van Blitterswijk CA, Lin Y, Karperien M. Chondrocytes Cocultured with Stromal Vascular Fraction of Adipose Tissue Present More Intense Chondrogenic Characteristics Than with Adipose Stem Cells. Tissue Eng Part A 2016; 22:336-48. [PMID: 26732248 DOI: 10.1089/ten.tea.2015.0269] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Partial replacement of chondrocytes by stem cells has been proposed to improve the performance of autologous chondrocyte implantation (ACI). Our previous studies showed that the increased cartilage production in pellet cocultures of chondrocytes and mesenchymal stem cells (MSCs) is due to a trophic role of the MSCs by stimulating chondrocyte proliferation and matrix production rather than MSCs actively undergoing chondrogenic differentiation. The aim of this study is to compare the trophic effects of stromal vascular fraction cells (SVF) and in vitro expanded adipose stem cells (ASC). SVF and culture-expanded ASC (n = 9) were cocultured with primary human chondrocytes in pellets. By glycosaminoglycan (GAG) and DNA assays, we showed that coculture pellets of SVF and chondrocytes have more GAG deposition than that of ASC and chondrocytes. Results of the short tandem repeats analysis indicated that the increase in the chondrocyte proportion in the coculture pellets is more pronounced in the SVF coculture group than in the ASC coculture group. Using flow cytometry and microarray, we demonstrated that SVF and ASC have different characteristics in cell surface markers and gene expression profiles. SVF is more heterogeneous than ASC, whereas ASC is more enriched in cells from the mesenchymal lineage than SVF. By subcutaneous implantation into nude mice, we showed that constructs of SVF and chondrocytes are better in depositing cartilage matrix than the mixture of ASC and chondrocytes. Taken together, SVF is better than ASC in terms of forming cartilage matrix in pellet coculture and in coimplantation models omitting the need for prior cell expansion. Our study suggests that the SVF in combination with primary human chondrocytes may be a good cell combination for one-stage cartilage repair.
Collapse
Affiliation(s)
- Ling Wu
- 1 Department of Developmental BioEngineering, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands .,2 Department of Orthopedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California , Los Angeles, California
| | - Henk-Jan Prins
- 3 Department of Oral Cell Biology, Academic Center for Dentistry , Amsterdam, The Netherlands .,4 Department of Oral & Maxillofacial Surgery, VU Medical Center , Amsterdam, The Netherlands
| | - Jeroen Leijten
- 1 Department of Developmental BioEngineering, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands
| | - Marco N Helder
- 5 Department of Orthopedics, VU Medical Center , Amsterdam, The Netherlands
| | - Denis Evseenko
- 2 Department of Orthopedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California , Los Angeles, California
| | - Lorenzo Moroni
- 6 Department of Tissue Regeneration, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands
| | - Clemens A van Blitterswijk
- 6 Department of Tissue Regeneration, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands
| | - Yunfeng Lin
- 7 State Key Laboratory for Oral Diseases, West China School of Stomatology, Sichuan University , Chengdu, China
| | - Marcel Karperien
- 1 Department of Developmental BioEngineering, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands
| |
Collapse
|
18
|
Scavenger receptor class A member 5 (SCARA5) and suprabasin (SBSN) are hub genes of coexpression network modules associated with peripheral vein graft patency. J Vasc Surg 2015; 64:202-209.e6. [PMID: 25935274 DOI: 10.1016/j.jvs.2014.12.052] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 12/18/2014] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Approximately 30% of autogenous vein grafts develop luminal narrowing and fail because of intimal hyperplasia or negative remodeling. We previously found that vein graft cells from patients who later develop stenosis proliferate more in vitro in response to growth factors than cells from patients who maintain patent grafts. To discover novel determinants of vein graft outcome, we have analyzed gene expression profiles of these cells using a systems biology approach to cluster the genes into modules by their coexpression patterns and to correlate the results with growth data from our prior study and with new studies of migration and matrix remodeling. METHODS RNA from 4-hour serum- or platelet-derived growth factor (PDGF)-BB-stimulated human saphenous vein cells obtained from the outer vein wall (20 cell lines) was used for microarray analysis of gene expression, followed by weighted gene coexpression network analysis. Cell migration in microchemotaxis chambers in response to PDGF-BB and cell-mediated collagen gel contraction in response to serum were also determined. Gene function was determined using short-interfering RNA to inhibit gene expression before subjecting cells to growth or collagen gel contraction assays. These cells were derived from samples of the vein grafts obtained at surgery, and the long-term fate of these bypass grafts was known. RESULTS Neither migration nor cell-mediated collagen gel contraction showed a correlation with graft outcome. Although 1188 and 1340 genes were differentially expressed in response to treatment with serum and PDGF, respectively, no single gene was differentially expressed in cells isolated from patients whose grafts stenosed compared with those that remained patent. Network analysis revealed four unique groups of genes, which we term modules, associated with PDGF responses, and 20 unique modules associated with serum responses. The "yellow" and "skyblue" modules, from PDGF and serum analyses, respectively, correlated with later graft stenosis (P = .005 and P = .02, respectively). In response to PDGF, yellow was also associated with increased cell growth. For serum, skyblue was also associated with inhibition of collagen gel contraction. The hub genes for yellow and skyblue (ie, the gene most connected to other genes in the module), scavenger receptor class A member 5 (SCARA5) and suprabasin (SBSN), respectively, were tested for effects on proliferation and collagen contraction. Knockdown of SCARA5 increased proliferation by 29.9% ± 7.8% (P < .01), whereas knockdown of SBSN had no effect. Knockdown of SBSN increased collagen gel contraction by 24.2% ± 8.6% (P < .05), whereas knockdown of SCARA5 had no effect. CONCLUSIONS Using weighted gene coexpression network analysis of cultured vein graft cell gene expression, we have discovered two small gene modules, which comprise 42 genes, that are associated with vein graft failure. Further experiments are needed to delineate the venous cells that express these genes in vivo and the roles these genes play in vein graft healing, starting with the module hub genes SCARA5 and SBSN, which have been shown to have modest effects on cell proliferation or collagen gel contraction.
Collapse
|
19
|
Jingsheng S, Yibing W, Jun X, Siqun W, Jianguo W, Feiyan C, Gangyong H, Jie C. MicroRNAs are potential prognostic and therapeutic targets in diabetic osteoarthritis. J Bone Miner Metab 2015; 33:1-8. [PMID: 25245120 DOI: 10.1007/s00774-014-0628-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 08/18/2014] [Indexed: 12/21/2022]
Abstract
Osteoarthritis is an aging-related degenerative disease that severely influences the elders' life quality. However, there have been few clinical approaches available until now. Currently, more knowledge of the pathology of osteoarthritis has been illustrated. Especially, diabetes can be the only predictor of osteoarthritis. Due to its outstanding characteristics, MicroRNA has been considered as an efficient target in treating diseases. In this review, we will discuss a new insight focusing on the roles of microRNA in the progression of osteoarthritis-induced by diabetes, especially type II diabetes mellitus.
Collapse
Affiliation(s)
- Shi Jingsheng
- Department of Orthopedics, Huashan Hospital, Fudan University, 12 Urumqi Road, Shanghai, 200040, China
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Efficient derivation of osteoprogenitor cells from induced pluripotent stem cells for bone regeneration. INTERNATIONAL ORTHOPAEDICS 2014; 38:1779-85. [DOI: 10.1007/s00264-014-2440-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 06/20/2014] [Indexed: 10/25/2022]
|
21
|
Harris Q, Seto J, O'Brien K, Lee PS, Kondo C, Heard BJ, Hart DA, Krawetz RJ. Monocyte chemotactic protein-1 inhibits chondrogenesis of synovial mesenchymal progenitor cells: an in vitro study. Stem Cells 2014; 31:2253-65. [PMID: 23836536 DOI: 10.1002/stem.1477] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 06/14/2013] [Indexed: 12/17/2022]
Abstract
Osteoarthritis (OA) is a multifactorial, often progressive, painful disease. OA often progresses with an apparent irreversible loss of articular cartilage, exposing underlying bone, resulting in pain and loss of mobility. This cartilage loss is thought to be permanent due to ineffective repair and apparent lack of stem/progenitor cells in that tissue. However, the adjacent synovial lining and synovial fluid are abundant with mesenchymal progenitor/stem cells (synovial mesenchymal progenitor cells [sMPCs]) capable of differentiating into cartilage both in vitro and in vivo. Previous studies have demonstrated that MPCs can home to factors such as monocyte chemotactic protein 1 (MCP-1/CCL2) expressed after injury. While MCP-1 (and its corresponding receptors) appears to play a role in recruiting stem cells to the site of injury, in this study, we have demonstrated that MCP-1 is upregulated in OA synovial fluid and that exposure to MCP-1 activates sMPCs, while concurrently inhibiting these cells from undergoing chondrogenesis in vitro. Furthermore, exposure to physiological (OA knee joint synovial fluid) levels of MCP-1 triggers changes in the transcriptome of sMPCs and prolonged exposure to the chemokine induces the expression of MCP-1 in sMPCs, resulting in a positive feedback loop from which sMPCs cannot apparently escape. Therefore, we propose a model where MCP-1 (normally expressed after joint injury) recruits sMPCs to the area of injury, but concurrently triggers changes in sMPC transcriptional regulation, leading to a blockage in the chondrogenic program. These results may open up new avenues of research into the lack of endogenous repair observed after articular cartilage injury and/or arthritis.
Collapse
Affiliation(s)
- Quinn Harris
- Department of Surgery, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Somoza RA, Welter JF, Correa D, Caplan AI. Chondrogenic differentiation of mesenchymal stem cells: challenges and unfulfilled expectations. TISSUE ENGINEERING PART B-REVIEWS 2014; 20:596-608. [PMID: 24749845 DOI: 10.1089/ten.teb.2013.0771] [Citation(s) in RCA: 222] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Articular cartilage repair and regeneration provides a substantial challenge in Regenerative Medicine because of the high degree of morphological and mechanical complexity intrinsic to hyaline cartilage due, in part, to its extracellular matrix. Cartilage remains one of the most difficult tissues to heal; even state-of-the-art regenerative medicine technology cannot yet provide authentic cartilage resurfacing. Mesenchymal stem cells (MSCs) were once believed to be the panacea for cartilage repair and regeneration, but despite years of research, they have not fulfilled these expectations. It has been observed that MSCs have an intrinsic differentiation program reminiscent of endochondral bone formation, which they follow after exposure to specific reagents as a part of current differentiation protocols. Efforts have been made to avoid the resulting hypertrophic fate of MSCs; however, so far, none of these has recreated a fully functional articular hyaline cartilage without chondrocytes exhibiting a hypertrophic phenotype. We reviewed the current literature in an attempt to understand why MSCs have failed to regenerate articular cartilage. The challenges that must be overcome before MSC-based tissue engineering can become a front-line technology for successful articular cartilage regeneration are highlighted.
Collapse
Affiliation(s)
- Rodrigo A Somoza
- Department of Biology, Skeletal Research Center, Case Western Reserve University , Cleveland, Ohio
| | | | | | | |
Collapse
|
23
|
Ballard GA, Warnock JJ, Bobe G, Duesterdieck-Zellmer KF, Baker L, Baltzer WI, Ott J. Comparison of meniscal fibrochondrocyte and synoviocyte bioscaffolds toward meniscal tissue engineering in the dog. Res Vet Sci 2014; 97:400-8. [PMID: 24856453 DOI: 10.1016/j.rvsc.2014.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Revised: 02/03/2014] [Accepted: 05/04/2014] [Indexed: 02/06/2023]
Abstract
Tissue engineering is a promising field of study toward curing the meniscal deficient stifle; however the ideal cell type for this task is not known. We describe here the extraction of synoviocytes and meniscal fibrochondrocytes from arthroscopic debris from six dogs, which were cultured as tensioned bioscaffolds to synthesize meniscal-like fibrocartilage sheets. Despite the diseased status of the original tissues, synoviocytes and meniscal fibrochondrocytes had high viability at the time of removal from the joint. Glycosaminoglycan and collagen content of bioscaffolds did not differ. Meniscal fibrochondrocyte bioscaffolds contained more type II collagen, but collagen deposition was disorganized, with only 30-40% of cells viable. The collagen of synoviocyte bioscaffolds was organized into sheets and bands and 80-90% of cells were viable. Autologous, diseased meniscal fibrochondrocytes and synoviocytes are plausible cell sources for future meniscal tissue engineering research, however cell viability of meniscal fibrochondrocytes in the tensioned bioscaffolds was low.
Collapse
Affiliation(s)
- George A Ballard
- College of Veterinary Medicine, Oregon State University, 105 Magruder Hall, 700 SW 30th St., Corvallis, OR 97331, USA
| | - Jennifer J Warnock
- College of Veterinary Medicine, Oregon State University, 105 Magruder Hall, 700 SW 30th St., Corvallis, OR 97331, USA.
| | - Gerd Bobe
- Linus Pauling Institute, Oregon State University, 307 Linus Pauling Science Center, Corvallis, OR 97331, USA
| | - Katja F Duesterdieck-Zellmer
- College of Veterinary Medicine, Oregon State University, 105 Magruder Hall, 700 SW 30th St., Corvallis, OR 97331, USA
| | - Lindsay Baker
- College of Veterinary Medicine, Oregon State University, 105 Magruder Hall, 700 SW 30th St., Corvallis, OR 97331, USA
| | - Wendy I Baltzer
- College of Veterinary Medicine, Oregon State University, 105 Magruder Hall, 700 SW 30th St., Corvallis, OR 97331, USA
| | - Jesse Ott
- College of Veterinary Medicine, Oregon State University, 105 Magruder Hall, 700 SW 30th St., Corvallis, OR 97331, USA
| |
Collapse
|
24
|
The pilot study of fibrin with temporomandibular joint derived synovial stem cells in repairing TMJ disc perforation. BIOMED RESEARCH INTERNATIONAL 2014; 2014:454021. [PMID: 24822210 PMCID: PMC4009306 DOI: 10.1155/2014/454021] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 03/08/2014] [Accepted: 03/11/2014] [Indexed: 11/23/2022]
Abstract
TMJ disc related diseases are difficult to be cured due to the poor repair ability of the disc. TMJ-SDSCs were ideal cell sources for cartilage tissue engineering which have been widely used in hyaline cartilage regeneration. Fibrin gel has been demonstrated as a potential scaffold for neocartilage formation. The aim of this study was to repair the TMJ disc perforation using fibrin/chitosan hybrid scaffold combined with TMJ-SDSCs. Rat TMJ-SDSCs were cultured on hybrid scaffold or pure chitosan scaffolds. The cell seeding efficiency, distribution, proliferation, and chondrogenic differentiation capacity were investigated. To evaluate the in vivo repair ability of cell/scaffold construct, rat TMJ disc explants were punched with a defect to mimic TMJ disc perforation. Cell seeded scaffolds were inserted into the defect of TMJ disc explants and then were implanted subcutaneously in nude mice for 4 weeks. Results demonstrated that fibrin may improve cell seeding, proliferation, and chondrogenic induction in vitro. The in vivo experiments showed more cartilage ECM deposition in fibrin/chitosan scaffold, which suggested an enhanced reparative ability. This pilot study demonstrated that the regenerative ability of TMJ-SDSCs seeded in fibrin/chitosan scaffold could be applied for repairing TMJ disc perforation.
Collapse
|
25
|
Coates EE, Fisher JP. Engineering superficial zone chondrocytes from mesenchymal stem cells. Tissue Eng Part C Methods 2014; 20:630-40. [PMID: 24279336 DOI: 10.1089/ten.tec.2013.0224] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Recent cartilage engineering efforts have focused on development of zonally organized tissue. However, there remains a need for protocols that differentiate progenitor populations into chondrocytes of zonal phenotype. Here, we evaluate the potential of coculture of bovine mesenchymal stem cells (MSCs) and zonal explants of bovine cartilage tissue to drive MSC differentiation to chondrocytes with the superficial zone phenotype. Two coculture systems were set up: one between alginate encapsulated MSCs and superficial zone cartilage explants, and one between MSCs and middle/deep zone cartilage explants. Chondrogenic and superficial zone markers were monitored over a 21-day differentiation period via gene and protein expression. A control conditioned media study was used to determine the impact of communication via soluble factors between cell populations during differentiation. At day 21, results show superficial zone explant coculture without transforming-growth factor β3 supplementation induces upregulation of chondrogenic gene expression markers SOX9 and type II collagen 3.4-fold and 11.4-fold, respectively, over standard chondrogenic control media. Further, coculture of MSCs and superficial zone explants can be used to upregulate mRNA expression of the superficial zone marker proteoglycan-4 in MSCs (1.75-fold over chondrogenic control at day 21), indicating the superficial zone chondrocyte phenotype. Gene expression data show middle/deep zone explant and MSC coculture did not induce the chondrogenesis observed in superficial zone explant coculture. Likewise, poor chondrogenesis was observed in all conditioned media groups. Results highlight the importance of superficial zone cartilage and cells in guiding stem cell fate and regulating differentiation of MSCs to chondrocytes of the superficial zone type.
Collapse
Affiliation(s)
- Emily E Coates
- Fischell Department of Bioengineering, University of Maryland , College Park, Maryland
| | | |
Collapse
|
26
|
Santhagunam A, Dos Santos F, Madeira C, Salgueiro JB, Cabral JMS. Isolation and ex vivo expansion of synovial mesenchymal stromal cells for cartilage repair. Cytotherapy 2013; 16:440-53. [PMID: 24364906 DOI: 10.1016/j.jcyt.2013.10.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 10/21/2013] [Accepted: 10/22/2013] [Indexed: 01/01/2023]
Abstract
BACKGROUND AIMS Hyaline articular cartilage is a highly specialized tissue that offers a low-friction and wear-resistant interface for weight-bearing surface articulation in diarthrodial joints, but it lacks vascularity. It displays an inherent inability to heal when injured in a skeletally mature individual. Joint-preserving treatment procedures such as mosaicplasty, débridement, perichondrium transplantation and autologous chondrocyte implantation have shown variable results, and the average long-term result is sub-standard. Because of these limitations of the treatment methods and lack of intrinsic repair capacity of mature cartilage tissue, an alternative treatment approach is needed, and synovial mesenchymal stromal cells (SMSCs) represent an attractive therapeutic alternative because of their ex vivo proliferation capacity, multipotency and ability to undergo chondrogenesis. METHODS SMSCs were isolated from tissues obtained by arthroscopy using two types of biopsies. Ex vivo cell expansion was accomplished under static and dynamic culture followed by characterization of cells according to the International Society for Cellular Therapy guidelines. Kinetic growth models and metabolite analysis were used for understanding the growth profile of these cells. RESULTS For the first time, SMSCs were expanded in stirred bioreactors and achieved higher cell density in a shorter period of time compared with static culture or with other mesenchymal stromal cell sources. CONCLUSIONS In this study we were able to achieve (8.8 ± 0.2) × 10(5) cells within <2 weeks in dynamic culture under complete xeno-free conditions. Our results also provided evidence that after dynamic culture these cells had an up-regulation of chondrogenic genes, which can be a potential factor for articular cartilage regeneration in clinical settings.
Collapse
Affiliation(s)
- Aruna Santhagunam
- Department of Bioengineering and Institute for Biotechnology and Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Francisco Dos Santos
- Department of Bioengineering and Institute for Biotechnology and Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Catarina Madeira
- Department of Bioengineering and Institute for Biotechnology and Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - João B Salgueiro
- Centro Hospitalar de Lisboa Ocidental, E.P.E, Hospital São Francisco Xavier, Lisboa, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering and Institute for Biotechnology and Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
27
|
Iwakura T, Sakata R, Reddi AH. Induction of chondrogenesis and expression of superficial zone protein in synovial explants with TGF-β1 and BMP-7. Tissue Eng Part A 2013; 19:2638-44. [PMID: 23848497 DOI: 10.1089/ten.tea.2013.0047] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Superficial zone protein (SZP) functions as a boundary lubricant in articular cartilage and decreases the coefficient of friction. As lubrication of articular cartilage is critical for normal joint function, the ability to secrete SZP at the surface of tissue-engineered cartilage is a prerequisite for optimal lubrication. Synovium-derived mesenchymal stem cells (MSCs) are thought to be an attractive cell source for cartilage regeneration. However, optimization of a three-dimensional environment is necessary for tissue engineering. In this study, we investigated whether synovial explants, which would preserve the physiologic microenvironment for MSCs therein, have the potential of SZP secretion after chondrogenic differentiation by treatment with transforming growth factor-β1 (TGF-β1) and bone morphogenetic protein-7 (BMP-7). Immunostaining and enzyme-linked immunosorbent assay analysis demonstrated that synovial explants can synthesize and secrete SZP following chondrogenic differentiation in response to TGF-β1 and BMP-7. Interestingly, the combined treatment with TGF-β1 and BMP-7 or treatment first with TGF-β1 followed by BMP-7 was more effective than other treatment groups in both chondrogenic differentiation and SZP secretion. In conclusion, synovial explants represent not only a superb source of progenitors/stem cells for the regeneration of the surface zone of articular cartilage, but also a useful model system for the in vitro differentiation into mature articular cartilage phenotypes in response to morphogens for tissue engineering of articular cartilage.
Collapse
Affiliation(s)
- Takashi Iwakura
- Department of Orthopaedic Surgery, Center for Tissue Regeneration and Repair, University of California , Sacramento, California
| | | | | |
Collapse
|
28
|
Coates EE, Riggin CN, Fisher JP. Matrix molecule influence on chondrocyte phenotype and proteoglycan 4 expression by alginate-embedded zonal chondrocytes and mesenchymal stem cells. J Orthop Res 2012; 30:1886-97. [PMID: 22674584 DOI: 10.1002/jor.22166] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 05/17/2012] [Indexed: 02/06/2023]
Abstract
Articular cartilage resists load and provides frictionless movement at joint surfaces. The tissue is organized into the superficial, middle, deep, and calcified zones throughout its depth, each which serve distinct functions. Proteoglycan 4 (PRG4), found in the superficial zone, is a critical component of the joint's lubricating mechanisms. Maintenance of both the chondrocyte and zonal chondrocyte phenotype remain challenges for in vitro culture and tissue engineering. Here we investigate the expression of PRG4 mRNA and protein by primary bovine superficial zone chondrocytes, middle/deep zone chondrocytes, and mesenchymal stem cells encapsulated in alginate hydrogels with hyaluronic acid (HA) and chondroitin sulfate (CS) additives. Chondrogenic phenotype and differentiation markers are evaluated by mRNA expression, histochemical, and immunohistochemical staining. Results show middle/deep cells express no measurable PRG4 mRNA by day 7. In contrast, superficial zone cells express elevated PRG4 mRNA throughout culture time. This expression can be significantly enhanced up to 15-fold by addition of both HA and CS to scaffolds. Conversely, PRG4 mRNA expression is downregulated (up to 5-fold) by CS and HA in differentiating MSCs, possibly due to build up of entrapped protein. HA and CS demonstrate favorable effects on chondrogenesis by upregulating transcription factor Sox9 mRNA (up to 4.6-fold) and downregulating type I collagen mRNA (up to 18-fold). Results highlight the important relationship between matrix components and expression of critical lubricating proteins in an engineered cartilage scaffold.
Collapse
Affiliation(s)
- Emily E Coates
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building, College Park, Maryland 20742, USA
| | | | | |
Collapse
|
29
|
O'hEireamhoin S, Buckley CT, Jones E, McGonagle D, Mulhall KJ, Kelly DJ. Recapitulating aspects of the oxygen and substrate environment of the damaged joint milieu for stem cell-based cartilage tissue engineering. Tissue Eng Part C Methods 2012; 19:117-27. [PMID: 22834895 DOI: 10.1089/ten.tec.2012.0142] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Human infrapatellar fat pad contains a source of mesenchymal stem cells (FPSCs) that potentially offer a novel population for the treatment of damaged or diseased articular cartilage. Existing cartilage repair strategies such as microfracture harness the presence of a low-oxygen microenvironment, fibrin clot formation at sites of microfracture, and elevations in growth factors in the damaged joint milieu. Bearing this in mind, the objective of this study was to determine the chondrogenic potential of diseased human FPSCs in a model system that recapitulates some of these features. In the first phase of the study, the role of transforming growth factor beta-3 (TGF-β3) and fibroblast growth factor-2 (FGF-2), in addition to an altered oxygen-tension environment, on the colony-forming unit-fibroblast (CFU-F) capacity and growth kinetics of human FPSCs during monolayer expansion was evaluated. The subsequent chondrogenic capacity of these cells was quantified in both normoxic (20%) and low- (5%) oxygen conditions. Expansion in FGF-2 was shown to reduce CFU-F numbers, but simultaneously increase both the colony size and the cell yield compared to standard expansion conditions. Supplementation with both FGF-2 and TGF-β3 significantly reduced cell-doubling time. Expansion in FGF-2, followed by differentiation at 5% oxygen tension, was observed to synergistically enhance subsequent sulfated glycosaminoglycan (sGAG) accumulation after chondrogenic induction. FPSCs expanded in FGF-2 were then encapsulated in either agarose or fibrin hydrogels in an attempt to engineer cartilaginous grafts. sGAG synthesis was higher in fibrin constructs, and was further enhanced by differentiation at 5% oxygen tension, accumulating 2.7% (ww) sGAG after 42 days in culture. These results indicate that FPSCs, a readily accessible cell population, form cartilage in an in vitro environment that recapitulates several key biological features of cartilage repair during microfracture and also point toward the potential utility of such cells when combined with fibrin hydrogel scaffolds.
Collapse
Affiliation(s)
- Sven O'hEireamhoin
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | | | | | | |
Collapse
|
30
|
Jones BA, Pei M. Synovium-Derived Stem Cells: A Tissue-Specific Stem Cell for Cartilage Engineering and Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2012; 18:301-11. [PMID: 22429320 DOI: 10.1089/ten.teb.2012.0002] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Brendan A. Jones
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, West Virginia
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, West Virginia
- Division of Exercise Physiology, West Virginia University, Morgantown, West Virginia
- Division of Mechanical and Aerospace Engineering, West Virginia University, Morgantown, West Virginia
| |
Collapse
|
31
|
Wu L, Prins HJ, Helder MN, van Blitterswijk CA, Karperien M. Trophic effects of mesenchymal stem cells in chondrocyte co-cultures are independent of culture conditions and cell sources. Tissue Eng Part A 2012; 18:1542-51. [PMID: 22429306 DOI: 10.1089/ten.tea.2011.0715] [Citation(s) in RCA: 166] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Earlier, we have shown that the increased cartilage production in pellet co-cultures of chondrocytes and bone marrow-derived mesenchymal stem cells (BM-MSCs) is due to a trophic role of the MSC in stimulating chondrocyte proliferation and matrix production rather than MSCs actively undergoing chondrogenic differentiation. These studies were performed in a culture medium that was not compatible with the chondrogenic differentiation of MSCs. In this study, we tested whether the trophic role of the MSCs is dependent on culturing co-culture pellets in a medium that is compatible with the chondrogenic differentiation of MSCs. In addition, we investigated whether the trophic role of the MSCs is dependent on their origins or is a more general characteristic of MSCs. Human BM-MSCs and bovine primary chondrocytes were co-cultured in a medium that was compatible with the chondrogenic differentiation of MSCs. Enhanced matrix production was confirmed by glycosaminoglycans (GAG) quantification. A species-specific quantitative polymerase chain reaction demonstrated that the cartilage matrix was mainly of bovine origin, indicative of a lack of the chondrogenic differentiation of MSCs. In addition, pellet co-cultures were overgrown by bovine cells over time. To test the influence of origin on MSCs' trophic effects, the MSCs isolated from adipose tissue and the synovial membrane were co-cultured with human primary chondrocytes, and their activity was compared with BM-MSCs, which served as control. GAG quantification again confirmed increased cartilage matrix production, irrespective of the source of the MSCs. EdU staining combined with cell tracking revealed an increased proliferation of chondrocytes in each condition. Irrespective of the MSC source, a short tandem repeat analysis of genomic DNA showed a decrease in MSCs in the co-culture over time. Our results clearly demonstrate that in co-culture pellets, the MSCs stimulate cartilage formation due to a trophic effect on the chondrocytes rather than differentiating into chondrocytes, irrespective of culture condition or origin. This implies that the trophic effect of MSCs in co-cultures is a general phenomenon with potential implications for use in cartilage repair strategies.
Collapse
Affiliation(s)
- Ling Wu
- Department of Tissue Regeneration, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | | | | | | | | |
Collapse
|
32
|
Khan WS, Adesida AB, Tew SR, Longo UG, Hardingham TE. Fat pad-derived mesenchymal stem cells as a potential source for cell-based adipose tissue repair strategies. Cell Prolif 2012; 45:111-20. [PMID: 22260253 DOI: 10.1111/j.1365-2184.2011.00804.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells are able to undergo adipogenic differentiation and present a possible alternative cell source for regeneration and replacement of adipose tissue. The human infrapatellar fat pad is a promising source of mesenchymal stem cells with many source advantages over from bone marrow. It is important to determine whether a potential mesenchymal stem-cell exhibits tri-lineage differentiation potential and is able to maintain its proliferation potential and cell-surface characterization on expansion in tissue culture. We have previously shown that mesenchymal stem cells derived from the fat pad can undergo chondrogenic and osteogenic differentiation, and we characterized these cells at early passage. In the study described here, proliferation potential and characterization of fat pad-derived mesenchymal stem cells were assessed at higher passages, and cells were allowed to undergo adipogenic differentiation. MATERIALS AND METHODS Infrapatellar fat pad tissue was obtained from six patients undergoing total knee replacement. Cells isolated were expanded to passage 18 and proliferation rates were measured. Passage 10 and 18 cells were characterized for cell-surface epitopes using a range of markers. Passage 2 cells were allowed to undergo differentiation in adipogenic medium. RESULTS The cells maintained their population doubling rates up to passage 18. Cells at passage 10 and passage 18 had cell-surface epitope expression similar to other mesenchymal stem cells previously described. By staining it was revealed that they highly expressed CD13, CD29, CD44, CD90 and CD105, and did not express CD34 or CD56, they were also negative for LNGFR and STRO1. 3G5 positive cells were noted in cells from both passages. These fat pad-derived cells had adipogenic differentiation when assessed using gene expression for peroxisome proliferator-activated receptor γ2 and lipoprotein lipase, and oil red O staining. DISCUSSION These results indicate that the cells maintained their proliferation rate, and continued expressing mesenchymal stem-cell markers and pericyte marker 3G5 at late passages. These results also show that the cells were capable of adipogenic differentiation and thus could be a promising source for regeneration and replacement of adipose tissue in reconstructive surgery.
Collapse
Affiliation(s)
- W S Khan
- United Kingdom Centre for Tissue Engineering, University of Manchester, Manchester, UK
| | | | | | | | | |
Collapse
|
33
|
McNary SM, Athanasiou KA, Reddi AH. Engineering lubrication in articular cartilage. TISSUE ENGINEERING PART B-REVIEWS 2012; 18:88-100. [PMID: 21955119 DOI: 10.1089/ten.teb.2011.0394] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Despite continuous progress toward tissue engineering of functional articular cartilage, significant challenges still remain. Advances in morphogens, stem cells, and scaffolds have resulted in enhancement of the bulk mechanical properties of engineered constructs, but little attention has been paid to the surface mechanical properties. In the near future, engineered tissues will be able to withstand and support the physiological compressive and tensile forces in weight-bearing synovial joints such as the knee. However, there is an increasing realization that these tissue-engineered cartilage constructs will fail without the optimal frictional and wear properties present in native articular cartilage. These characteristics are critical to smooth, pain-free joint articulation and a long-lasting, durable cartilage surface. To achieve optimal tribological properties, engineered cartilage therapies will need to incorporate approaches and methods for functional lubrication. Steady progress in cartilage lubrication in native tissues has pushed the pendulum and warranted a shift in the articular cartilage tissue-engineering paradigm. Engineered tissues should be designed and developed to possess both tribological and mechanical properties mirroring natural cartilage. In this article, an overview of the biology and engineering of articular cartilage structure and cartilage lubrication will be presented. Salient progress in lubrication treatments such as tribosupplementation, pharmacological, and cell-based therapies will be covered. Finally, frictional assays such as the pin-on-disk tribometer will be addressed. Knowledge related to the elements of cartilage lubrication has progressed and, thus, an opportune moment is provided to leverage these advances at a critical step in the development of mechanically and tribologically robust, biomimetic tissue-engineered cartilage. This article is intended to serve as the first stepping stone toward future studies in functional tissue engineering of articular cartilage that begins to explore and incorporate methods of lubrication.
Collapse
Affiliation(s)
- Sean M McNary
- Department of Orthopaedic Surgery, Lawrence Ellison Center for Tissue Regeneration and Repair, School of Medicine, University of California, Davis, Sacramento, California, USA
| | | | | |
Collapse
|
34
|
He F, Pei M. Extracellular matrix enhances differentiation of adipose stem cells from infrapatellar fat pad toward chondrogenesis. J Tissue Eng Regen Med 2011; 7:73-84. [PMID: 22095700 DOI: 10.1002/term.505] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 03/29/2011] [Accepted: 07/18/2011] [Indexed: 12/13/2022]
Abstract
The objective was to improve proliferation and chondrogenic potential of adipose stem cells (ASCs) by expansion on extracellular matrix (ECM) deposited by either ASCs or synovium-derived stem cells (SDSCs). ASCs isolated from porcine infrapatellar fat pad were separately expanded on conventional plastic flasks, ASC-deposited ECM and SDSC-deposited ECM. ASCs were centrifuged to form pellets and cultured in a serum-free chondrogenic medium with either TGFβ3 or TGFβ3 combined with BMP-6. Cell number yielded on ECM expansion did not show a significant difference in deposition between ASCs and SDSCs but was 6-10 times that grown on non-coated flasks. ECM-expanded ASCs exhibited a lower level of intracellular reactive oxygen species (ROS) compared to those grown on non-coated flasks. Typical chondrogenic markers, including type II collagen and glycosaminoglycans (GAGs), were intensively distributed in the pellets from ECM-expanded ASCs instead of those from flask-grown cells. ASCs expanded on ECM, either from ASCs or SDSCs, exhibited a similar chondrogenic index (GAG:DNA), which was significantly higher than that from ASCs grown on non-coated flasks. The combination of TGFβ3 and BMP-6 increased 36% more in ASC chondrogenic index than the treatment with TGFβ3 alone. Interestingly, ECM pretreatment also decreased expanded ASC hypertrophic marker genes. ECM deposited by either ASCs or SDSCs did not exhibit enhanced adipogenic differentiation of ASCs. Our study indicates that the sequential application of ECM for cell expansion and combined TGFβ3 with BMP-6 for chondrogenic differentiation may be a promising approach for ASC-based cartilage tissue engineering and regeneration.
Collapse
Affiliation(s)
- Fan He
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics and Division of Exercise Physiology, West Virginia University, Morgantown, WV 26506, USA
| | | |
Collapse
|
35
|
Plaas A, Velasco J, Gorski DJ, Li J, Cole A, Christopherson K, Sandy JD. The relationship between fibrogenic TGFβ1 signaling in the joint and cartilage degradation in post-injury osteoarthritis. Osteoarthritis Cartilage 2011; 19:1081-90. [PMID: 21624477 DOI: 10.1016/j.joca.2011.05.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 05/05/2011] [Accepted: 05/07/2011] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To review the literature on modulation of chondrocyte activities in the osteoarthritic joint, and to discuss these changes in relation to established hard and soft tissue repair paradigms, with an emphasis on transforming growth factor beta (TGFβ1)-mediated signaling which can promote either a chondrogenic or fibrogenic phenotype. METHODS Papers addressing the close relationship between repair in general, and the specific post-injury response of joint tissues are summarized. Different interpretations of the role of TGFβ1 in the emergence of an "osteoarthritic" chondrocyte are compared and the phenotypic plasticity of "reparative" progenitor cells is examined. Lastly, emerging data on a central role for A-Disintegrin-And-Metalloproteinase-with-Thrombospondin-like-Sequences-5 (ADAMTS5) activity in modulating TGFβ1 signaling through activin receptor-like kinase 1 (ALK1) and activin receptor-like kinase 5 (ALK5) pathways is discussed. RESULTS The review illustrates how a transition from ALK5-mediated fibrogenic signaling to ALK1-mediated chondrogenic signaling in joint cells represents the critical transition from a non-reparative to a reparative cell phenotype. Data from cell and in vivo studies illustrates the mechanism by which ablation of ADAMTS5 activity allows the transition to reparative chondrogenesis. Multiple large gene expression studies of normal and osteoarthritis (OA) human cartilages (CAs) also support an important role for TGFβ1-mediated pro-fibrogenic activities during disease progression. CONCLUSIONS We conclude that progressive articular CA damage in post-injury OA results primarily from biomechanical, cell biologic and mediator changes that promote a fibroblastic phenotype in joint cells. Since ADAMTS5 and TGFβ1 appear to control this process, agents which interfere with their activities may not only enhance endogenous CA repair in vivo, but also improve the properties of tissue-engineered CA for implantation.
Collapse
Affiliation(s)
- A Plaas
- Department of Internal Medicine (Rheumatology), Rush University Medical Center, Chicago, IL, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Coleman CM, Curtin C, Barry FP, O'Flatharta C, Murphy JM. Mesenchymal stem cells and osteoarthritis: remedy or accomplice? Hum Gene Ther 2011; 21:1239-50. [PMID: 20649459 DOI: 10.1089/hum.2010.138] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Multipotent mesenchymal stromal or stem cells (MSCs) are likely to be agents of connective tissue homeostasis and repair. Because the hallmark of osteoarthritis (OA) is degeneration and failure to repair connective tissues it is compelling to think that these cells have a role to play in OA. Indeed, MSCs have been implicated in the pathogenesis of OA and, in turn, progression of the disease has been shown to be therapeutically modulated by MSCs. This review discusses current knowledge on the potential of both marrow- and local joint-derived MSCs in OA, the mode of action of the cells, and possible effects of the osteoarthritic niche on the function of MSCs. The use of stem cells for repair of isolated cartilage lesions and strategies for modulation of OA using local cell delivery are discussed as well as therapeutic options for the future to recruit and appropriately activate endogenous progenitors and/or locally systemically administered MSCs in the early stages of the disease. The use of gene therapy protocols, particularly as they pertain to modulation of inflammation associated with the osteoarthritic niche, offer an additional option in the treatment of this chronic disease. In summary, elucidation of the etiology of OA and development of technologies to detect early disease, allied to an increased understanding of the role MSCs in aging and OA, should lead to more targeted and efficacious treatments for this debilitating chronic disease in the future.
Collapse
Affiliation(s)
- Cynthia M Coleman
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | | | | | | | | |
Collapse
|
37
|
Hollander AP, Dickinson SC, Kafienah W. Stem cells and cartilage development: complexities of a simple tissue. Stem Cells 2011; 28:1992-6. [PMID: 20882533 PMCID: PMC3003945 DOI: 10.1002/stem.534] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cartilage is considered to be a simple tissue that should be easy to engineer because it is avascular and contains just one cell type, the chondrocyte. Despite this apparent simplicity, regenerating cartilage in a form that can function effectively after implantation in the joint has proven difficult. This may be because we have not fully appreciated the importance of different structural regions of articular cartilage or of understanding the origins of chondrocytes and how this cell population is maintained in the normal tissue. This review considers what is known about different regions of cartilage and the types of stem cells in articulating joints and emphasizes the potential importance of regeneration of the lamina splendens at the joint surface and calcified cartilage at the junction with bone for long-term survival of regenerated tissue in vivo.
Collapse
Affiliation(s)
- Anthony P Hollander
- Department of Cellular & Molecular Medicine, University of Bristol, Bristol, United Kingdom.
| | | | | |
Collapse
|
38
|
Boeuf S, Richter W. Chondrogenesis of mesenchymal stem cells: role of tissue source and inducing factors. Stem Cell Res Ther 2010; 1:31. [PMID: 20959030 PMCID: PMC2983444 DOI: 10.1186/scrt31] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) are an attractive cell source for cell therapy in cartilage. Although their therapeutic potential is clear, the requirements and conditions for effective induction of chondrogenesis in MSCs and for the production of a stable cartilaginous tissue by these cells are far from being understood. Different sources of MSCs have been considered for cartilage tissue engineering, mainly based on criteria of availability, as for adipose tissue, or of proximity to cartilage and the joint environment in vivo, as for bone marrow and synovial tissues. Focussing on human MSCs, this review will provide an overview of studies featuring comparative analysis of the chondrogenic differentiation of MSCs from different sources. In particular, it will examine the influence of the cells' origin on the requirements for the induction of chondrogenesis and on the phenotype achieved by the cells after differentiation.
Collapse
Affiliation(s)
- Stephane Boeuf
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany.
| | | |
Collapse
|