1
|
Théron A, Maumus M, Biron-Andreani C, Sirvent N, Jorgensen C, Noël D. What is the rationale for mesenchymal stromal cells based therapies in the management of hemophilic arthropathies? Osteoarthritis Cartilage 2024; 32:634-642. [PMID: 38160743 DOI: 10.1016/j.joca.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/21/2023] [Accepted: 12/07/2023] [Indexed: 01/03/2024]
Abstract
Hemophilia A and B are rare X-linked genetic bleeding disorders due to a complete or partial deficiency in the coagulation factors VIII or IX, respectively. The main treatment for hemophilia is prophylactic and based on coagulation factor replacement therapies. These treatments have significantly reduced bleeding and improved the patients' quality of life. Nevertheless, repeated joint bleedings (hemarthroses), even subclinical hemarthroses, can lead to hemophilic arthropathy (HA). This disabling condition is characterized by chronic pain due to synovial inflammation, cartilage and bone destruction requiring ultimately joint replacement. HA resembles to rheumatoid arthritis because of synovitis but HA is considered as having similarities with osteoarthritis as illustrated by the migration of immune cells, production of inflammatory cytokines, synovial hypertrophy and cartilage damage. Various drugs have been evaluated for the management of HA with limited success. The objective of the review is to discuss new therapeutic approaches with a special focus on the studies that have investigated the potential of using mesenchymal stromal cells (MSCs) in the management of HA. A systematic review of the literature has been made. Most of the studies have focused on the interest of MSCs for the delivery of missing factors VIII or IX but in some studies, more insight on the effect of MSC injection on synovial inflammation or cartilage structure were provided and put in perspective for possible clinical applications.
Collapse
Affiliation(s)
- Alexandre Théron
- IRMB, University of Montpellier, INSERM, Montpellier, France; Resources and Competence Center for Hereditary Hemorrhagic Diseases, CHU Montpellier, Montpellier, France; Department of Pediatric Oncology and Hematology, CHU Montpellier, Montpellier, France
| | - Marie Maumus
- IRMB, University of Montpellier, INSERM, Montpellier, France
| | - Christine Biron-Andreani
- Resources and Competence Center for Hereditary Hemorrhagic Diseases, CHU Montpellier, Montpellier, France
| | - Nicolas Sirvent
- Department of Pediatric Oncology and Hematology, CHU Montpellier, Montpellier, France
| | - Christian Jorgensen
- IRMB, University of Montpellier, INSERM, Montpellier, France; Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, Montpellier, France
| | - Danièle Noël
- IRMB, University of Montpellier, INSERM, Montpellier, France; Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, Montpellier, France.
| |
Collapse
|
2
|
Stone RN, Reeck JC, Oxford JT. Advances in Cartilage Tissue Engineering Using Bioinks with Decellularized Cartilage and Three-Dimensional Printing. Int J Mol Sci 2023; 24:ijms24065526. [PMID: 36982597 PMCID: PMC10051657 DOI: 10.3390/ijms24065526] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/08/2023] [Accepted: 03/12/2023] [Indexed: 03/16/2023] Open
Abstract
Osteoarthritis, a chronic, debilitating, and painful disease, is one of the leading causes of disability and socioeconomic burden, with an estimated 250 million people affected worldwide. Currently, there is no cure for osteoarthritis and treatments for joint disease require improvements. To address the challenge of improving cartilage repair and regeneration, three-dimensional (3D) printing for tissue engineering purposes has been developed. In this review, emerging technologies are presented with an overview of bioprinting, cartilage structure, current treatment options, decellularization, bioinks, and recent progress in the field of decellularized extracellular matrix (dECM)–bioink composites is discussed. The optimization of tissue engineering approaches using 3D-bioprinted biological scaffolds with dECM incorporated to create novel bioinks is an innovative strategy to promote cartilage repair and regeneration. Challenges and future directions that may lead to innovative improvements to currently available treatments for cartilage regeneration are presented.
Collapse
Affiliation(s)
- Roxanne N. Stone
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Drive, Boise, ID 83725, USA
| | - Jonathon C. Reeck
- Center of Excellence in Biomedical Research, Boise State University, 1910 University Drive, Boise, ID 83725, USA
| | - Julia Thom Oxford
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Drive, Boise, ID 83725, USA
- Center of Excellence in Biomedical Research, Boise State University, 1910 University Drive, Boise, ID 83725, USA
- Department of Biological Sciences, Boise State University, 1910 University Drive, Boise, ID 83725, USA
- Correspondence: ; Tel.: +1-(208)-426-2238
| |
Collapse
|
3
|
Transcriptomic response of bioengineered human cartilage to parabolic flight microgravity is sex-dependent. NPJ Microgravity 2023; 9:5. [PMID: 36658138 PMCID: PMC9852254 DOI: 10.1038/s41526-023-00255-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 01/10/2023] [Indexed: 01/20/2023] Open
Abstract
Spaceflight and simulated spaceflight microgravity induced osteoarthritic-like alterations at the transcriptomic and proteomic levels in the articular and meniscal cartilages of rodents. But little is known about the effect of spaceflight or simulated spaceflight microgravity on the transcriptome of tissue-engineered cartilage developed from human cells. In this study, we investigate the effect of simulated spaceflight microgravity facilitated by parabolic flights on tissue-engineered cartilage developed from in vitro chondrogenesis of human bone marrow mesenchymal stem cells obtained from age-matched female and male donors. The successful induction of cartilage-like tissue was confirmed by the expression of well-demonstrated chondrogenic markers. Our bulk transcriptome data via RNA sequencing demonstrated that parabolic flight altered mostly fundamental biological processes, and the modulation of the transcriptome profile showed sex-dependent differences. The secretome profile analysis revealed that two genes (WNT7B and WNT9A) from the Wnt-signaling pathway, which is implicated in osteoarthritis development, were only up-regulated for female donors. The results of this study showed that the engineered cartilage tissues responded to microgravity in a sex-dependent manner, and the reported data offers a strong foundation to further explore the underlying mechanisms.
Collapse
|
4
|
Xiao K, Yang L, Xie W, Gao X, Huang R, Xie M. Bcl-xL mutant promotes cartilage differentiation of BMSCs by upregulating TGF-β/BMP expression levels. Exp Ther Med 2021; 22:736. [PMID: 34055053 PMCID: PMC8138271 DOI: 10.3892/etm.2021.10168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 03/19/2021] [Indexed: 12/22/2022] Open
Abstract
Bcl-xL is a transmembrane molecule in the mitochondria, with apoptosis-related and pro-metabolic functions, that also plays a role in chondrogenesis and differentiation. A Bcl-xL mutant, in which the GRI sequence is replaced by ELN, has no anti-apoptotic effect, while other biological functions of this mutant remain unchanged. The present study investigated the impact of this Bcl-xL mutant on cartilage differentiation and the expression levels of TGF-β and bone morphogenetic protein (BMP). Human bone marrow mesenchymal stem cells (BMSCs) were transfected with Bcl-xL and Bcl-xL mutant (∆Bcl-xL) overexpression vectors. The cells were divided into four groups: Control (not subjected to any transfection), EV (empty pcDNA3.1-Bcl-xL vector), OV (Bcl-xL overexpression) and ∆OV (∆Bcl-xL overexpression). Saffron and toluidine blue staining was performed to observe cartilage tissue formation. Flow cytometry was conducted to measure BMSC apoptosis. The expression levels of TGF-β and BMP were evaluated using reverse transcription-quantitative PCR (RT-qPCR) and western blotting. Compared with that in the control group, the expression levels of Bcl-xL in the OV group increased significantly (P<0.05). Western blotting and RT-qPCR results revealed that OV and ∆OV treatment increased the expression levels of TGF-β and BMP in transfected cells, compared to their expression in the control and EV groups (P<0.05). Saffron and toluidine blue staining results showed that cartilage formation was increased in the ∆OV and ∆OV + Bax-/Bak-groups to similar degrees. Cell apoptosis in the ∆OV group did not change compared with that in the control group. The Bcl-xL mutant promoted cartilage differentiation of BMSCs and upregulated TGF-β/BMP expression. This enhancement of chondrogenic differentiation was not related to the expression of Bax and Bak. Taken together, these findings provided for improved application of bone tissue engineering technology in the treatment of articular cartilage defects.
Collapse
Affiliation(s)
- Kai Xiao
- Foot and Ankle Surgery, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, P.R. China
| | - Lin Yang
- Department of Allergy, Tongji Hospital of Tongji Medical College of HUST, Wuhan, Hubei 430033, P.R. China
| | - Wei Xie
- Foot and Ankle Surgery, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, P.R. China
| | - Xinfeng Gao
- Foot and Ankle Surgery, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, P.R. China
| | - Ruokun Huang
- Foot and Ankle Surgery, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, P.R. China
| | - Ming Xie
- Foot and Ankle Surgery, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, P.R. China
| |
Collapse
|
5
|
Stone RN, Frahs SM, Hardy MJ, Fujimoto A, Pu X, Keller-Peck C, Oxford JT. Decellularized Porcine Cartilage Scaffold; Validation of Decellularization and Evaluation of Biomarkers of Chondrogenesis. Int J Mol Sci 2021; 22:6241. [PMID: 34207917 PMCID: PMC8230108 DOI: 10.3390/ijms22126241] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/01/2021] [Accepted: 06/04/2021] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis is a major concern in the United States and worldwide. Current non-surgical and surgical approaches alleviate pain but show little evidence of cartilage restoration. Cell-based treatments may hold promise for the regeneration of hyaline cartilage-like tissue at the site of injury or wear. Cell-cell and cell-matrix interactions have been shown to drive cell differentiation pathways. Biomaterials for clinically relevant applications can be generated from decellularized porcine auricular cartilage. This material may represent a suitable scaffold on which to seed and grow chondrocytes to create new cartilage. In this study, we used decellularization techniques to create an extracellular matrix scaffold that supports chondrocyte cell attachment and growth in tissue culture conditions. Results presented here evaluate the decellularization process histologically and molecularly. We identified new and novel biomarker profiles that may aid future cartilage decellularization efforts. Additionally, the resulting scaffold was characterized using scanning electron microscopy, fluorescence microscopy, and proteomics. Cellular response to the decellularized scaffold was evaluated by quantitative real-time PCR for gene expression analysis.
Collapse
Affiliation(s)
- Roxanne N. Stone
- Interdisciplinary Studies Program, Boise State University, Boise, ID 83725, USA;
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
| | - Stephanie M. Frahs
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
- Center of Biomedical Research Excellence in Matrix Biology, Boise State University, Boise, ID 83725, USA
- Biomolecular Sciences Graduate Programs, Boise State University, Boise, ID 83725, USA
| | - Makenna J. Hardy
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
- Center of Biomedical Research Excellence in Matrix Biology, Boise State University, Boise, ID 83725, USA
- Biomolecular Sciences Graduate Programs, Boise State University, Boise, ID 83725, USA
| | - Akina Fujimoto
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
- Center of Biomedical Research Excellence in Matrix Biology, Boise State University, Boise, ID 83725, USA
| | - Xinzhu Pu
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
- Center of Biomedical Research Excellence in Matrix Biology, Boise State University, Boise, ID 83725, USA
- Biomolecular Sciences Graduate Programs, Boise State University, Boise, ID 83725, USA
| | - Cynthia Keller-Peck
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
- Center of Biomedical Research Excellence in Matrix Biology, Boise State University, Boise, ID 83725, USA
| | - Julia Thom Oxford
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
- Center of Biomedical Research Excellence in Matrix Biology, Boise State University, Boise, ID 83725, USA
- Biomolecular Sciences Graduate Programs, Boise State University, Boise, ID 83725, USA
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| |
Collapse
|
6
|
Mesgarzadeh AH, Nasiri I, Jarolmasjed S, Naghibi M, Shafaei H. Evaluation of bone regeneration in mandible large defect using undifferentiated adipose stem cells loaded on gelatin carrier: An animal model case study. J Dent Res Dent Clin Dent Prospects 2021; 15:22-29. [PMID: 33927837 PMCID: PMC8058160 DOI: 10.34172/joddd.2021.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 08/22/2020] [Indexed: 11/09/2022] Open
Abstract
Background. Large mandibular defects are considered difficult reconstructive challenges for oral and maxillofacial surgeons. Cell therapy, as an alternative technique, might increase the speed of bone regeneration. This study aimed to investigate bone regeneration in large defects of dog mandibles using allogenic adipose-derived stem cells on gelatin foam as a cell carrier. Methods. The tissue engineering phase consisted of the sampling of adult dogs' adipose tissue that can easily be isolated from adipose stem cells (ASCs) of the dogs, ASCs were cultured in Dulbecco's Modified Eagle's Medium (DMEM, Gibco, USA) with low glucose, containing 10% fetal bovine serum (FBS) (Sigma, USA) and 1% penicillin-streptomycin (Gibco, USA), with the characterization of dog ASCs and gelatin-transplanted ASCs. Six dogs were included in this experimental study in the next step and randomly assigned to the treatment and control groups. The samples in both groups underwent surgery under general anesthesia to create uniform 3-cm bony defects. The samples in both groups were reconstructed with titanium reconstruction plates and screws. A large bone gap filled with ASCs (5×106 ) was seeded on gelatin (ASCs) in the treatment group. In the control group, bony defects were filled with a cell delivery carrier without ASCs. Six months after transplantation, the animals' mandibles were evaluated by CT scan imaging, and the results were quantified through the Hounsfield unit (HU). The data were analyzed with t-test. Results. Before transplantation, the nature of the stem cells was confirmed by the expression of CD44 and CD105 cell markers at 71.9% and 89.3%, respectively, and a lack of the CD45 cell marker expression at 2.2%. Evaluation of CT scan images showed significantly higher bone repair in the ASCs group (920.25±572.92 HU) than in the control group (-94.746± 08.42). Conclusion. The bone regeneration of the ASCs group was significantly higher than that in the control group.
Collapse
Affiliation(s)
- Ali Hossein Mesgarzadeh
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Islam Nasiri
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyedhosein Jarolmasjed
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehran Naghibi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajar Shafaei
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
7
|
Isolation and Characterization of Multipotent Turkey Tendon-Derived Stem Cells. Stem Cells Int 2018; 2018:3697971. [PMID: 29977306 PMCID: PMC6011053 DOI: 10.1155/2018/3697971] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 04/24/2018] [Indexed: 02/06/2023] Open
Abstract
Tendon injuries are among the most common and severe hand injuries with a high demand for functional recovery. Stem cells have been identified and isolated from different species and a variety of tissues for the sake of regenerative medicine. Recently, turkey has been suggested as a potential new large animal model for flexor tendon-related research. However, turkey tissue-specific stem cells have not been investigated. Here, we presented the isolation and verification of tendon-derived stem cells (TDSCs) from 6- to 8-month-old heritage-breed turkey. TDSCs were isolated from turkey flexor tendon by plating nucleated cells at the determined optimal density. Approximately 4% of the nucleated cells demonstrated clonogenicity, high proliferation rate, and trilineage differentiation potential after induction culturing. These cells expressed surface antigens CD90, CD105, and CD44, but did not express CD45. There was a high level of gene expression of tenogenic markers in TDSCs, including mohawk, collagen type I, tenascin C, and elastin. Turkey TDSCs also expressed transcription factors PouV, Nanog, and Sox2, which are critically involved in the regulation of stemness. The successful isolation of tendon-derived stem cells from turkey was beneficial for future studies in tendon tissue engineering and would help in the development of new treatment for tendon diseases using this novel animal model.
Collapse
|
8
|
IFN α-Expressing Amniotic Fluid-Derived Mesenchymal Stem Cells Migrate to and Suppress HeLa Cell-Derived Tumors in a Mouse Model. Stem Cells Int 2018; 2018:1241323. [PMID: 29760719 PMCID: PMC5901954 DOI: 10.1155/2018/1241323] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/23/2018] [Indexed: 12/26/2022] Open
Abstract
Background Immunotherapy for cervical cancer with type I interferon (IFN) is limited because of the cytotoxicity that accompanies the high doses that are administered. In this study, we investigated the utilization of amniotic fluid-derived mesenchymal stem cells (AF-MSCs) as a means for delivering IFNα to local tumor sites for the suppression of cervical cancer in a mouse model using HeLa cell xenografts. Methods The tumor tropism ability of AF-MSCs and AF-MSCs genetically modified to overexpress IFNα (IFNα-AF-MSCs) was examined through Transwell in vitro and through fluorescent images and immunohistochemistry in a mouse model. Tumor size and tumor apoptosis were observed to evaluate the efficacy of the targeting therapy. Mechanistically, tumor cell apoptosis was detected by cytometry and TUNEL, and oncogenic proteins c-Myc, p53, and Bcl-2 as well as microvessel density were detected by immunohistochemistry. Results In this model, intravenously injected AF-MSCs selectively migrated to the tumor sites, participated in tumor construction, and promoted tumor growth. After being genetically modified to overexpress IFNα, the IFNα-AF-MSCs maintained their tumor tropism but could significantly suppress tumor growth. The restrictive efficacy of IFNα-AF-MSCs was associated with the suppression of angiogenesis, inhibition of tumor cell proliferation, and induction of apoptosis in tumor cells. Neither AF-MSCs nor IFNα-AF-MSCs trigger tumor formation. Conclusions IFNα-AF-MSC-based therapy is feasible and shows potential for treating cervical cancer, suggesting that AF-MSCs may be promising vehicles for delivering targeted anticancer therapy.
Collapse
|
9
|
Effects of Bone Marrow Stromal Cell Transplantation on Repair of Bone Defect in Rats. Trauma Mon 2018. [DOI: 10.5812/traumamon.13701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
10
|
Gupta P, Geris L, Luyten FP, Papantoniou I. An Integrated Bioprocess for the Expansion and Chondrogenic Priming of Human Periosteum-Derived Progenitor Cells in Suspension Bioreactors. Biotechnol J 2017; 13. [PMID: 28987025 DOI: 10.1002/biot.201700087] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/01/2017] [Indexed: 12/12/2022]
Abstract
The increasing use of microcarrier-based suspension bioreactors for scalable expansion of adult progenitor cells in recent years reveals the necessity of such approaches to address bio manufacturing challenges of advanced therapeutic medicinal products. However, the differentiation of progenitor cells within suspension bioreactors for the production of tissue modules is of equal importance but not well investigated. This study reports on the development of a bioreactor-based integrated process for expansion and chondrogenic priming of human periosteum-derived stem cells (hPDCs) using Cultispher S microcarriers. Spinner flask-based expansion and priming of hPDCs were carried out over 12 days for expansion and 14 days for priming. Characterization of the cells were carried out every 3rd day. Our study showed that hPDCs were able to expand till confluency with fold increase of 3.2±0.64 and to be subsequently primed toward a chondrogenic state within spinner flasks. During expansion, the cells maintained their phenotypic markers, trilineage differentiation capabilities and viability. Upon switching to TGF-β containing media the cells were able to differentiate toward chondrogenic lineage by clustering into mm-sized macrotissues containing hundreds of microcarriers. Chondrogenic priming was further evidenced by the expression of relevant markers at the mRNA level while maintaining their viability. Ectopic implantation of macrotissues highlighted that they were able to sustain their chondrogenic properties for 8 weeks in vivo. The method indicated here, suggests that expansion and relevant priming of progenitor cells can be carried out in an integrated bioprocess using spinner flasks and as such could be potentially extrapolated to other stem and progenitor cell populations.
Collapse
Affiliation(s)
- Priyanka Gupta
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1 Herestraat 49, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, O&N1 Herestraat 49, Leuven, Belgium
| | - Liesbet Geris
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1 Herestraat 49, Leuven, Belgium.,Biomechanics Research Unit GIGA-R In Silico Medicine, Université de Liege, Quartier Polytechnique 1, Allée de la découverte 13A, Liège, Belgium.,Biomechanics Section, KU Leuven, Celestijnenlaan 300C (2419), Leuven, Belgium
| | - Frank P Luyten
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1 Herestraat 49, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, O&N1 Herestraat 49, Leuven, Belgium
| | - Ioannis Papantoniou
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1 Herestraat 49, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, O&N1 Herestraat 49, Leuven, Belgium
| |
Collapse
|
11
|
Chou CL, Rivera AL, Williams V, Welter JF, Mansour JM, Drazba JA, Sakai T, Baskaran H. Micrometer scale guidance of mesenchymal stem cells to form structurally oriented large-scale tissue engineered cartilage. Acta Biomater 2017; 60:210-219. [PMID: 28709984 PMCID: PMC5581212 DOI: 10.1016/j.actbio.2017.07.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 07/07/2017] [Accepted: 07/10/2017] [Indexed: 11/17/2022]
Abstract
Current clinical methods to treat articular cartilage lesions provide temporary relief of the symptoms but fail to permanently restore the damaged tissue. Tissue engineering, using mesenchymal stem cells (MSCs) combined with scaffolds and bioactive factors, is viewed as a promising method for repairing cartilage injuries. However, current tissue engineered constructs display inferior mechanical properties compared to native articular cartilage, which could be attributed to the lack of structural organization of the extracellular matrix (ECM) of these engineered constructs in comparison to the highly oriented structure of articular cartilage ECM. We previously showed that we can guide MSCs undergoing chondrogenesis to align using microscale guidance channels on the surface of a two-dimensional (2-D) collagen scaffold, which resulted in the deposition of aligned ECM within the channels and enhanced mechanical properties of the constructs. In this study, we developed a technique to roll 2-D collagen scaffolds containing MSCs within guidance channels in order to produce a large-scale, three-dimensional (3-D) tissue engineered cartilage constructs with enhanced mechanical properties compared to current constructs. After rolling the MSC-scaffold constructs into a 3-D cylindrical structure, the constructs were cultured for 21days under chondrogenic culture conditions. The microstructure architecture and mechanical properties of the constructs were evaluated using imaging and compressive testing. Histology and immunohistochemistry of the constructs showed extensive glycosaminoglycan (GAG) and collagen type II deposition. Second harmonic generation imaging and Picrosirius red staining indicated alignment of neo-collagen fibers within the guidance channels of the constructs. Mechanical testing indicated that constructs containing the guidance channels displayed enhanced compressive properties compared to control constructs without these channels. In conclusion, using a novel roll-up method, we have developed large scale MSC based tissue-engineered cartilage that shows microscale structural organization and enhanced compressive properties compared to current tissue engineered constructs. STATEMENT OF SIGNIFICANCE Tissue engineered cartilage constructs made with human mesenchymal stem cells (hMSCs), scaffolds and bioactive factors are a promising solution to treat cartilage defects. A major disadvantage of these constructs is their inferior mechanical properties compared to the native tissue, which is likely due to the lack of structural organization of the extracellular matrix of the engineered constructs. In this study, we developed three-dimensional (3-D) cartilage constructs from rectangular scaffold sheets containing hMSCs in micro-guidance channels and characterized their mechanical properties and metabolic requirements. The work led to a novel roll-up method to embed 2-D microscale structures in 3-D constructs. Further, micro-guidance channels incorporated within the 3-D cartilage constructs led to the production of aligned cell-produced matrix and enhanced mechanical function.
Collapse
Affiliation(s)
- Chih-Ling Chou
- Department of Chemical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Alexander L Rivera
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Valencia Williams
- Department of Chemical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Jean F Welter
- Skeletal Research Center, Department of Biology, Case Western Reserve University, Cleveland, OH, United States; Center for Multimodal Evaluation of Engineered Cartilage, Case Western Reserve University, Cleveland, OH, United States
| | - Joseph M Mansour
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH, United States; Center for Multimodal Evaluation of Engineered Cartilage, Case Western Reserve University, Cleveland, OH, United States
| | - Judith A Drazba
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Takao Sakai
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Harihara Baskaran
- Department of Chemical Engineering, Case Western Reserve University, Cleveland, OH, United States; Center for Multimodal Evaluation of Engineered Cartilage, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
12
|
Luz-Crawford P, Jorgensen C, Djouad F. Mesenchymal Stem Cells Direct the Immunological Fate of Macrophages. Results Probl Cell Differ 2017; 62:61-72. [PMID: 28455706 DOI: 10.1007/978-3-319-54090-0_4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSC) are multipotent stem cells with a broad well-described immunosuppressive potential. They are able to modulate both the innate and the adaptive immune response. Particularly, MSC are able to regulate the phenotype and function of macrophages that are critical for different biological processes including wound healing, inflammation, pathogenesis of several autoimmune diseases, and tumor growth. These multifunctional roles of macrophages are due to their high plasticity, which enable them to adopt different phenotypes such as a pro-inflammatory M1 and anti-inflammatory M2 phenotype. MSC promote macrophage differentiation toward an M2-like phenotype with a high tissue remodeling potential and anti-inflammatory activity but also a pro-tumorigenic function. MSC regulatory effect on macrophages is mediated through the secretion of different immunomodulatory molecules such as PGE2, IL1RA, and IL-6. Moreover, the presence of macrophages in damaged tissue and inflammation is essential for MSC to exert their therapeutic function. In this chapter, we discuss how the interplay between macrophages and MSC mutually modulates their phenotypes and functions, orchestrates tissue repair, and controls inflammation during autoimmunity and tumor growth.
Collapse
Affiliation(s)
- Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.
| | - Christian Jorgensen
- Inserm U1183, IRMB, Hôpital Saint-Eloi, 80 Avenue Augustin Fliche, 34295, Montpellier Cedex 5, France.,Université de Montpellier, Montpellier, 34000, France.,Service d'Immuno-Rhumatologie, Hôpital Lapeyronie, Montpellier, 34295, France
| | - Farida Djouad
- Inserm U1183, IRMB, Hôpital Saint-Eloi, 80 Avenue Augustin Fliche, 34295, Montpellier Cedex 5, France. .,Université de Montpellier, Montpellier, 34000, France.
| |
Collapse
|
13
|
Hsu YH, Yang YY, Huwang MH, Weng YH, Jou IM, Wu PT, Lin TY, Wu LW, Chang MS. Anti-IL-20 monoclonal antibody inhibited inflammation and protected against cartilage destruction in murine models of osteoarthritis. PLoS One 2017; 12:e0175802. [PMID: 28426699 PMCID: PMC5398531 DOI: 10.1371/journal.pone.0175802] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 03/06/2017] [Indexed: 12/17/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by progressive destruction of articular cartilage. Interleukin (IL)-20 is a proinflammatory cytokine involved in the pathogenesis of rheumatoid arthritis. We investigated the role of IL-20 in OA and evaluated whether anti-IL-20 antibody (7E) treatment attenuates disease severity in murine models of surgery-induced OA. Immunohistochemical staining was used to detect IL-20 and its receptors expression in synovial tissue and cartilage from OA patients, and in OA synovial fibroblasts (OASFs) and chondrocytes (OACCs) from rodents with surgery-induced OA. RTQ-PCR and western blotting were used to determine IL-20-regulated OA-associated gene expression in OASFs and OACCs. OA rats and OA mice were treated with 7E. Arthritis severity was determined based on the degree of cartilage damage and the arthritis severity score. We found that IL-20 and its receptors were expressed in OASFs and OACCs. IL-20 induced TNF-α, IL-1β, MMP-1, and MMP-13 expression by activating ERK-1/2 and JNK signals in OASFs. IL-20 not only upregulated MCP-1, IL-6, MMP-1, and MMP-13 expression, but also downregulated aggrecan, type 2 collagen, TGF-β, and BMP-2 expression in OACCs. Arthritis severity was significantly lower in 7E-treated OA rats, and 7E- or MSC-treated OA mice. Therefore, we concluded that IL-20 was involved in the progression and development of OA through inducing proinflammatory cytokines and OA-associated gene expression in OASFs and OACCs. 7E reduced the severity of arthritis in murine models of surgery-induced OA. Our findings provide evidence that IL-20 is a novel target and that 7E is a potential therapeutic agent for OA.
Collapse
Affiliation(s)
- Yu-Hsiang Hsu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan.,Research Center of New Antibody Drug, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Yu Yang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Man-Hsiang Huwang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yun-Han Weng
- Research Center of New Antibody Drug, National Cheng Kung University, Tainan, Taiwan.,Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - I-Ming Jou
- Department of Orthopedics, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Po-Tin Wu
- Department of Orthopedics, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Tain-Yu Lin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Li-Wha Wu
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ming-Shi Chang
- Research Center of New Antibody Drug, National Cheng Kung University, Tainan, Taiwan.,Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
14
|
Djouad F, Ipseiz N, Luz-Crawford P, Scholtysek C, Krönke G, Jorgensen C. PPARβ/δ: A master regulator of mesenchymal stem cell functions. Biochimie 2016; 136:55-58. [PMID: 27914902 DOI: 10.1016/j.biochi.2016.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/16/2016] [Accepted: 11/22/2016] [Indexed: 02/06/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) have emerged as key regulators of physiological and immunological processes. Recently, one of their members PPARβ/δ has been identified as major player in the maintenance of bone homeostasis, by promoting Wnt signalling activity in osteoblast and mesenchymal stem cells (MSC). PPARβ/δ not only controls the fate of MSC but also regulates their immunosuppressive properties by directly modulating their NF-κB activity. In this review, we discuss how the regulation of PPARβ/δ provides an innovative strategy for an optimisation of MSC-based therapy.
Collapse
Affiliation(s)
- Farida Djouad
- Inserm U1183, Montpellier, F-34295, France; University of Montpellier, Montpellier, F-34000, France.
| | - Natacha Ipseiz
- Cardiff University, Division of Infection and Immunity, Cardiff, United Kingdom
| | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Carina Scholtysek
- Department of Internal Medicine 3 and Institute for Clinical Immunology, University Hospital Erlangen, Erlangen, Germany; Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3 and Institute for Clinical Immunology, University Hospital Erlangen, Erlangen, Germany; Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Jorgensen
- Inserm U1183, Montpellier, F-34295, France; University of Montpellier, Montpellier, F-34000, France; Service d'Immuno-Rhumatologie, Hôpital Lapeyronie, Montpellier, F-34295, France
| |
Collapse
|
15
|
Gobbi A, Whyte GP. One-Stage Cartilage Repair Using a Hyaluronic Acid-Based Scaffold With Activated Bone Marrow-Derived Mesenchymal Stem Cells Compared With Microfracture: Five-Year Follow-up. Am J Sports Med 2016; 44:2846-2854. [PMID: 27474386 DOI: 10.1177/0363546516656179] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [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 injury is frequently encountered, yet treatment options capable of providing durable cartilage repair are limited. PURPOSE To investigate the medium-term clinical outcomes of cartilage repair using a 1-stage technique of a hyaluronic acid-based scaffold with activated bone marrow aspirate concentrate (HA-BMAC) and compare results with those of microfracture. A secondary aim of this study was to identify specific patient demographic factors and cartilage lesion characteristics that are associated with superior outcomes. STUDY DESIGN Cohort study; Level of evidence, 2. METHODS Fifty physically active patients (mean age, 45 years) with grade IV cartilage injury of the knee (lesion size, 1.5-24 cm2) were treated with HA-BMAC or microfracture and were observed prospectively for 5 years. Patients were placed into the HA-BMAC group if the health insurance policy of the treating institution supported this option; otherwise, they were placed into the microfracture group. Objective and subjective clinical assessment tools were used preoperatively and at 2 and 5 years postoperatively to compare treatment outcomes. RESULTS Significant improvements in outcome scores were achieved in both treatment groups at 2 years (P < .001). In the microfracture group, 64% were classified as normal or nearly normal according to the International Knee Documentation Committee (IKDC) objective score at 2 years, compared with 100% of those treated with HA-BMAC (P < .001). Normal or nearly normal objective assessments in the microfracture group declined significantly after 5 years to 28% of patients (P = .004). All patients treated with HA-BMAC maintained improvement at 5 years according to Lysholm, Tegner, IKDC objective, and IKDC subjective scores. Tegner, IKDC objective, and Knee injury and Osteoarthritis Outcome Score (KOOS) assessments demonstrated higher scores in the HA-BMAC treatment group compared with microfracture at 5 years. Lysholm and IKDC subjective scores were similar between treatment groups at 5 years. Poorer outcomes in the microfracture group were demonstrated in cases of lesions larger than 4 cm2 and nonsolitary lesions. Age greater than 45 years, large size of lesion, and treatment of multiple lesions were not associated with poorer outcome in patients treated with HA-BMAC. CONCLUSION Repair of chondral injury using a hyaluronic acid-based scaffold with activated bone marrow aspirate concentrate provides better clinical outcomes and more durable cartilage repair at medium-term follow-up compared with microfracture. Positive short-term clinical outcomes can be achieved with either microfracture or HA-BMAC. Cartilage repair using HA-BMAC leads to successful medium-term outcomes independent of age or lesion size.
Collapse
Affiliation(s)
- Alberto Gobbi
- Orthopaedic Arthroscopic Surgery International (OASI) Bioresearch Foundation Gobbi NPO, Milan, Italy
| | - Graeme P Whyte
- Orthopaedic Arthroscopic Surgery International (OASI) Bioresearch Foundation Gobbi NPO, Milan, Italy
| |
Collapse
|
16
|
Heo JS. Chondrogenic Differentiation of Human Mesenchymal Stem Cells on a Patterned Polymer Surface. KOREAN JOURNAL OF CLINICAL LABORATORY SCIENCE 2015. [DOI: 10.15324/kjcls.2015.47.3.117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- June Seok Heo
- Severance Hospital, Cell Therapy Center, Seoul 03722, Korea
| |
Collapse
|
17
|
Chandra P, Lee SJ. Synthetic Extracellular Microenvironment for Modulating Stem Cell Behaviors. Biomark Insights 2015; 10:105-16. [PMID: 26106260 PMCID: PMC4472032 DOI: 10.4137/bmi.s20057] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 04/12/2015] [Accepted: 04/13/2015] [Indexed: 11/30/2022] Open
Abstract
The innate ability of stem cells to self-renew and differentiate into multiple cell types makes them a promising source for tissue engineering and regenerative medicine applications. Their capacity for self-renewal and differentiation is largely influenced by the combination of physical, chemical, and biological signals found in the stem cell niche, both temporally and spatially. Embryonic and adult stem cells are potentially useful for cell-based approaches; however, regulating stem cell behavior remains a major challenge in their clinical use. Most of the current approaches for controlling stem cell fate do not fully address all of the complex signaling pathways that drive stem cell behaviors in their natural microenvironments. To overcome this limitation, a new generation of biomaterials is being developed for use as three-dimensional synthetic microenvironments that can mimic the regulatory characteristics of natural extracellular matrix (ECM) proteins and ECM-bound growth factors. These synthetic microenvironments are currently being investigated as a substrate with surface immobilization and controlled release of bioactive molecules to direct the stem cell fate in vitro, as a tissue template to guide and improve the neo-tissue formation both in vitro and in vivo, and as a delivery vehicle for cell therapy in vivo. The continued advancement of such an intelligent biomaterial system as the synthetic extracellular microenvironment holds the promise of improved therapies for numerous debilitating medical conditions for which no satisfactory cure exists today.
Collapse
Affiliation(s)
- Prafulla Chandra
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
18
|
Chonanant C, Bambery KR, Jearanaikoon N, Chio-Srichan S, Limpaiboon T, Tobin MJ, Heraud P, Jearanaikoon P. Discrimination of micromass-induced chondrocytes from human mesenchymal stem cells by focal plane array-Fourier transform infrared microspectroscopy. Talanta 2014; 130:39-48. [DOI: 10.1016/j.talanta.2014.05.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/19/2014] [Accepted: 05/20/2014] [Indexed: 12/20/2022]
|
19
|
Bhardwaj N, Devi D, Mandal BB. Tissue-engineered cartilage: the crossroads of biomaterials, cells and stimulating factors. Macromol Biosci 2014; 15:153-82. [PMID: 25283763 DOI: 10.1002/mabi.201400335] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 08/25/2014] [Indexed: 02/06/2023]
Abstract
Damage to cartilage represents one of the most challenging tasks of musculoskeletal therapeutics due to its limited propensity for healing and regenerative capabilities. Lack of current treatments to restore cartilage tissue function has prompted research in this rapidly emerging field of tissue regeneration of functional cartilage tissue substitutes. The development of cartilaginous tissue largely depends on the combination of appropriate biomaterials, cell source, and stimulating factors. Over the years, various biomaterials have been utilized for cartilage repair, but outcomes are far from achieving native cartilage architecture and function. This highlights the need for exploration of suitable biomaterials and stimulating factors for cartilage regeneration. With these perspectives, we aim to present an overview of cartilage tissue engineering with recent progress, development, and major steps taken toward the generation of functional cartilage tissue. In this review, we have discussed the advances and problems in tissue engineering of cartilage with strong emphasis on the utilization of natural polymeric biomaterials, various cell sources, and stimulating factors such as biophysical stimuli, mechanical stimuli, dynamic culture, and growth factors used so far in cartilage regeneration. Finally, we have focused on clinical trials, recent innovations, and future prospects related to cartilage engineering.
Collapse
Affiliation(s)
- Nandana Bhardwaj
- Seri-Biotechnology Unit, Life Science Division, Institute of Advanced Study in Science and Technology, Guwahati, 781035, India
| | | | | |
Collapse
|
20
|
Li C, Wang Q, Wang JF. Transforming growth factor-β (TGF-β) induces the expression of chondrogenesis-related genes through TGF-β receptor II (TGFRII)-AKT-mTOR signaling in primary cultured mouse precartilaginous stem cells. Biochem Biophys Res Commun 2014; 450:646-51. [PMID: 24946212 DOI: 10.1016/j.bbrc.2014.06.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 06/06/2014] [Indexed: 01/08/2023]
Abstract
Precartilaginous stem cells (PSCs) are adult stem cells which could initiate chondrocytes and bone growth. In the current study, we purified PSCs from the neonate mice' perichondrial mesenchyme through immunomagnetic beads with the fibroblast growth factor receptor-3 (FGFR-3) antibody. Mouse PSCs were seeded and cultured, and their phenotype was confirmed by FGFR-3 over-expression. Transforming growth factor-β (TGF-β) was added to induce PSCs differentiation. TGF-β increased mRNA expression of chondrogenesis-related genes (collagen type II, Sox 9, and aggrecan) in the cultured PSCs, which was abolished by TGF-β receptor II (TGFRII) lentiviral shRNA depletion. TGF-β induced AKT activation in mouse PSCs, while the PI3K/AKT inhibitor (LY294002) and the AKT specific inhibitors (perifosine and MK-2206) largely suppressed TGF-β-induced collagen II, Sox 9, and aggrecan mRNA expression. Meanwhile, the mTOR complex 1 (mTORC1) blocker RAD001 or the mTORC1/2 dual inhibitor AZD-2014 also alleviated TGF-β-induced chondrogenesis-associated genes expression. Further, lentiviral shRNA depletion of SIN1 (a mTORC2 component) or mTOR inhibited TGF-β's effect in the mouse PSCs. In conclusion, our evidence suggests that TGF-β induces the expression of chondrogenesis-related genes through TGFRII-AKT-mTOR signaling in cultured mouse PSCs.
Collapse
Affiliation(s)
- Cheng Li
- Department of Orthopedics, Wuxi First People's Hospital Affiliated to Nanjing Medical University, Wuxi City, Jiangsu 214023, China
| | - Qiong Wang
- Department of Orthopedics, Wuxi First People's Hospital Affiliated to Nanjing Medical University, Wuxi City, Jiangsu 214023, China.
| | - Jun-Fang Wang
- Department of Orthopedics, Wuxi First People's Hospital Affiliated to Nanjing Medical University, Wuxi City, Jiangsu 214023, China.
| |
Collapse
|
21
|
Zhou J, Wang D, Liang T, Guo Q, Zhang G. Amniotic fluid-derived mesenchymal stem cells: characteristics and therapeutic applications. Arch Gynecol Obstet 2014; 290:223-31. [DOI: 10.1007/s00404-014-3231-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 03/24/2014] [Indexed: 12/26/2022]
|
22
|
Djouad F, Tejedor G, Toupet K, Maumus M, Bony C, Blangy A, Chuchana P, Jorgensen C, Noël D. Promyelocytic leukemia zinc-finger induction signs mesenchymal stem cell commitment: identification of a key marker for stemness maintenance? Stem Cell Res Ther 2014; 5:27. [PMID: 24564963 PMCID: PMC4055047 DOI: 10.1186/scrt416] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 02/17/2014] [Indexed: 12/30/2022] Open
Abstract
Introduction Mesenchymal stem cells (MSCs) are an attractive cell source for cartilage and bone tissue engineering given their ability to differentiate into chondrocytes and osteoblasts. However, the common origin of these two specialized cell types raised the question about the identification of regulatory pathways determining the differentiation fate of MSCs into chondrocyte or osteoblast. Methods Chondrogenesis, osteoblastogenesis, and adipogenesis of human and mouse MSC were induced by using specific inductive culture conditions. Expression of promyelocytic leukemia zinc-finger (PLZF) or differentiation markers in MSCs was determined by RT-qPCR. PLZF-expressing MSC were implanted in a mouse osteochondral defect model and the neotissue was analyzed by routine histology and microcomputed tomography. Results We found out that PLZF is not expressed in MSCs and its expression at early stages of MSC differentiation is the mark of their commitment toward the three main lineages. PLZF acts as an upstream regulator of both Sox9 and Runx2, and its overexpression in MSC enhances chondrogenesis and osteogenesis while it inhibits adipogenesis. In vivo, implantation of PLZF-expressing MSC in mice with full-thickness osteochondral defects resulted in the formation of a reparative tissue resembling cartilage and bone. Conclusions Our findings demonstrate that absence of PLZF is required for stemness maintenance and its expression is an early event at the onset of MSC commitment during the differentiation processes of the three main lineages.
Collapse
|
23
|
Kondo M, Yamaoka K, Sonomoto K, Fukuyo S, Oshita K, Okada Y, Tanaka Y. IL-17 inhibits chondrogenic differentiation of human mesenchymal stem cells. PLoS One 2013; 8:e79463. [PMID: 24260226 PMCID: PMC3829852 DOI: 10.1371/journal.pone.0079463] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 09/23/2013] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE Mesenchymal stem cells (MSCs) can differentiate into cells of mesenchymal lineages, such as osteoblasts and chondrocytes. Here we investigated the effects of IL-17, a key cytokine in chronic inflammation, on chondrogenic differentiation of human MSCs. METHODS Human bone marrow MSCs were pellet cultured in chondrogenic induction medium containing TGF-β3. Chondrogenic differentiation was detected by cartilage matrix accumulation and chondrogenic marker gene expression. RESULTS Over-expression of cartilage matrix and chondrogenic marker genes was noted in chondrogenic cultures, but was inhibited by IL-17 in a dose-dependent manner. Expression and phosphorylation of SOX9, the master transcription factor for chondrogenesis, were induced within 2 days and phosphorylated SOX9 was stably maintained until day 21. IL-17 did not alter total SOX9 expression, but significantly suppressed SOX9 phosphorylation in a dose-dependent manner. At day 7, IL-17 also suppressed the activity of cAMP-dependent protein kinase A (PKA), which is known to phosphorylate SOX9. H89, a selective PKA inhibitor, also suppressed SOX9 phosphorylation, expression of chondrogenic markers and cartilage matrix, and also decreased chondrogenesis. CONCLUSIONS IL-17 inhibited chondrogenesis of human MSCs through the suppression of PKA activity and SOX9 phosphorylation. These results suggest that chondrogenic differentiation of MSCs can be inhibited by a mechanism triggered by IL-17 under chronic inflammation.
Collapse
Affiliation(s)
- Masahiro Kondo
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Yahatanishi-ku, Kitakyushu, Fukuoka, Japan
- Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma Corporation, Aoba-ku, Yokohama, Kanagawa, Japan
| | - Kunihiro Yamaoka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Yahatanishi-ku, Kitakyushu, Fukuoka, Japan
| | - Koshiro Sonomoto
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Yahatanishi-ku, Kitakyushu, Fukuoka, Japan
| | - Shunsuke Fukuyo
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Yahatanishi-ku, Kitakyushu, Fukuoka, Japan
| | - Koichi Oshita
- Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma Corporation, Aoba-ku, Yokohama, Kanagawa, Japan
| | - Yosuke Okada
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Yahatanishi-ku, Kitakyushu, Fukuoka, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Yahatanishi-ku, Kitakyushu, Fukuoka, Japan
- * E-mail:
| |
Collapse
|
24
|
Betsch M, Schneppendahl J, Thuns S, Herten M, Sager M, Jungbluth P, Hakimi M, Wild M. Bone marrow aspiration concentrate and platelet rich plasma for osteochondral repair in a porcine osteochondral defect model. PLoS One 2013; 8:e71602. [PMID: 23951201 PMCID: PMC3741121 DOI: 10.1371/journal.pone.0071602] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Accepted: 07/08/2013] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Bone marrow aspiration concentrate (BMAC) may possess a high potency for cartilage and osseous defect healing because it contains stem cells and multiple growth factors. Alternatively, platelet rich plasma (PRP), which contains a cocktail of multiple growth factors released from enriched activated thrombocytes may potentially stimulate the mesenchymal stem cells (MSCs) in bone marrow to proliferate and differentiate. METHODS A critical size osteochondral defect (10×6 mm) in both medial femoral condyles was created in 14 Goettinger mini-pigs. All animals were randomized into the following four groups: biphasic scaffold alone (TRUFIT BGS, Smith & Nephew, USA), scaffold with PRP, scaffold with BMAC and scaffold in combination with BMAC and PRP. After 26 weeks all animals were euthanized and histological slides were cut, stained and evaluated using a histological score and immunohistochemistry. RESULTS The thrombocyte number was significantly increased (p = 0.049) in PRP compared to whole blood. In addition the concentration of the measured growth factors in PRP such as BMP-2, BMP-7, VEGF, TGF-β1 and PDGF were significantly increased when compared to whole blood (p<0.05). In the defects of the therapy groups areas of chondrogenic tissue were present, which stained blue with toluidine blue and positively for collagen type II. Adding BMAC or PRP in a biphasic scaffold led to a significant improvement of the histological score compared to the control group, but the combination of BMAC and PRP did not further enhance the histological score. CONCLUSIONS The clinical application of BMAC or PRP in osteochondral defect healing is attractive because of their autologous origin and cost-effectiveness. Adding either PRP or BMAC to a biphasic scaffold led to a significantly better healing of osteochondral defects compared with the control group. However, the combination of both therapies did not further enhance healing.
Collapse
Affiliation(s)
- Marcel Betsch
- Department of Trauma and Hand Surgery, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Johannes Schneppendahl
- Department of Trauma and Hand Surgery, University Hospital Duesseldorf, Duesseldorf, Germany
- * E-mail:
| | - Simon Thuns
- Department of Trauma and Hand Surgery, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Monika Herten
- Clinic for Vascular and Endovascular Surgery, University Hospital Muenster, Muenster, Germany
| | - Martin Sager
- Central Animal Research Facility, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Pascal Jungbluth
- Department of Trauma and Hand Surgery, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Mohssen Hakimi
- Department of Trauma and Hand Surgery, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Michael Wild
- Department of Trauma and Orthopaedic Surgery, Klinikum Darmstadt, Darmstadt, Germany
| |
Collapse
|
25
|
Ma A, Jiang L, Song L, Hu Y, Dun H, Daloze P, Yu Y, Jiang J, Zafarullah M, Chen H. Reconstruction of cartilage with clonal mesenchymal stem cell-acellular dermal matrix in cartilage defect model in nonhuman primates. Int Immunopharmacol 2013; 16:399-408. [PMID: 23499511 DOI: 10.1016/j.intimp.2013.02.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 02/01/2013] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Articular cartilage defects are commonly associated with trauma, inflammation and osteoarthritis. Mesenchymal stem cell (MSC)-based therapy is a promising novel approach for repairing articular cartilage. Direct intra-articular injection of uncommitted MSCs does not regenerate high-quality cartilage. This study explored utilization of a new three-dimensional, selected chondrogenic clonal MSC-loaded monkey acellular dermal matrix (MSC-ADM) scaffold to repair damaged cartilage in an experimental model of knee joint cartilage defect in Cynomolgus monkeys. METHODS MSCs were characterized for cell size, cell yield, phenotypes, proliferation and chondrogenic differentiation capacity. Chondrogenic differentiation assays were performed at different MSC passages by sulfated glycosaminoglycans (sGAG), collagen, and fluorescence activated cell sorter (FACS) analysis. Selected chondrogenic clonal MSCs were seeded onto ADM scaffold with the sandwich model and MSC-loaded ADM grafts were analyzed by confocal microscopy and scanning electron microscopy. Cartilage defects were treated with normal saline, clonal MSCs and clonal MSC-ADM grafts, respectively. The clinical parameters, and histological and immunohistochemical examinations were evaluated at weeks 8, 16, 24 post-treatment, respectively. RESULTS Polyclonal and clonal MSCs could differentiate into the chondrogenic lineage after stimulation with suitable chondrogenic factors. They expressed mesenchymal markers and were negative for hematopoietic markers. Articular cartilage defects were considerably improved and repaired by selected chondrogenic clonal MSC-based treatment, particularly, in MSC-ADM-treated group. The histological scores in MSC-ADM-treated group were consistently higher than those of other groups. CONCLUSION Our results suggest that selected chondrogenic clonal MSC-loaded ADM grafts could improve the cartilage lesions in Cynomolgus monkey model, which may be applicable for repairing similar human cartilage defects.
Collapse
Affiliation(s)
- Anlun Ma
- Department of Surgery, Research Center, CHUM (CRCHUM), Notre-Dame Hospital, University of Montreal, Montreal, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Wei H, Shen G, Deng X, Lou D, Sun B, Wu H, Long L, Ding T, Zhao J. The role of IL-6 in bone marrow (BM)-derived mesenchymal stem cells (MSCs) proliferation and chondrogenesis. Cell Tissue Bank 2013; 14:699-706. [PMID: 23322270 DOI: 10.1007/s10561-012-9354-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2012] [Accepted: 12/11/2012] [Indexed: 01/02/2023]
Abstract
Rheumatoid arthritis (RA) is the most common degenerative arthritic cartilage and represents a disease where the prospect of stem cell therapy offers considerable hope. Currently, bone marrow (BM) represents the major source of mesenchymal stem cells (MSCs) for cell therapy. In the pathology of RA, the pro-inflammatory cytokines, such as interleukin 6 (IL-6) play a pivotal role. To investigate the direct role of IL-6 in the chondrogenic differentiation of murine MSCs (mMSCs), we isolate MSCs from the murine bone marrow, and induce MSCs chondrogenesis with different concentrations of IL-6 in vitro. Through detecting the histological and histochemical qualities of the aggregates, we demonstrate that IL-6 inhibited the differentiation of MSCs into chondrocytes in the dose-dependence manner. These findings suggest that possible strategies for improving the clinical outcome of cartilage repair procedures.
Collapse
Affiliation(s)
- Haixiang Wei
- Orthopedic Department, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Chou CL, Rivera AL, Sakai T, Caplan AI, Goldberg VM, Welter JF, Baskaran H. Micrometer scale guidance of mesenchymal stem cells to form structurally oriented cartilage extracellular matrix. Tissue Eng Part A 2012; 19:1081-90. [PMID: 23157410 DOI: 10.1089/ten.tea.2012.0177] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Tissue engineering is a possible method for long-term repair of cartilage lesions, but current tissue-engineered cartilage constructs have inferior mechanical properties compared to native cartilage. This problem may be due to the lack of an oriented structure in the constructs at the microscale that is present in the native tissue. In this study, we utilize contact guidance to develop constructs with microscale architecture for improved chondrogenesis and function. Stable channels of varying microscale dimensions were formed in collagen-based and polydimethylsiloxane membranes via a combination of microfabrication and soft-lithography. Human mesenchymal stem cells (MSCs) were selectively seeded in these channels. The chondrogenic potential of MSCs seeded in these channels was investigated by culturing them for 3 weeks under differentiating conditions, and then evaluating the subsequent synthesized tissue for mechanical function and by type II collagen immunohistochemistry. We demonstrate selective seeding of viable MSCs within the channels. MSC aligned and produced mature collagen fibrils along the length of the channel in smaller linear channels of widths 25-100 μm compared to larger linear channels of widths 500-1000 μm. Further, substrates with microchannels that led to cell alignment also led to superior mechanical properties compared to constructs with randomly seeded cells or selectively seeded cells in larger channels. The ultimate stress and modulus of elasticity of constructs with cells seeded in smaller channels increased by as much as fourfolds. We conclude that microscale guidance is useful to produce oriented cartilage structures with improved mechanical properties. These findings can be used to fabricate large clinically useful MSC-cartilage constructs with superior mechanical properties.
Collapse
Affiliation(s)
- Chih-Ling Chou
- Department of Chemical Engineering, Case Western Reserve University, Cleveland, Ohio 44106-7217, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Klopp AH, Zhang Y, Solley T, Amaya-Manzanares F, Marini F, Andreeff M, Debeb B, Woodward W, Schmandt R, Broaddus R, Lu K, Kolonin MG. Omental adipose tissue-derived stromal cells promote vascularization and growth of endometrial tumors. Clin Cancer Res 2011; 18:771-82. [PMID: 22167410 DOI: 10.1158/1078-0432.ccr-11-1916] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE Adipose tissue contains a population of tumor-tropic mesenchymal progenitors, termed adipose stromal cells (ASC), which engraft in neighboring tumors to form supportive tumor stroma. We hypothesized that intra-abdominal visceral adipose tissue may contain a uniquely tumor-promoting population of ASC to account for the relationship between excess visceral adipose tissue and mortality of intra-abdominal cancers. EXPERIMENTAL DESIGN To investigate this, we isolated and characterized ASC from intra-abdominal omental adipose tissue (O-ASC) and characterized their effects on endometrial cancer progression as compared with subcutaneous adipose-derived mesenchymal stromal cells (SC-ASC), bone marrow-derived mesenchymal stromal cells (BM-MSC), and lung fibroblasts. To model chronic recruitment of ASC by tumors, cells were injected metronomically into mice bearing Hec1a xenografts. RESULTS O-ASC expressed cell surface markers characteristic of BM-MSC and differentiated into mesenchymal lineages. Coculture with O-ASC increased endometrial cancer cell proliferation in vitro. Tumor tropism of O-ASC and SC-ASC for human Hec1a endometrial tumor xenografts was comparable, but O-ASC more potently promoted tumor growth. Compared with tumors in SC-ASC-injected mice, tumors in O-ASC-injected mice contained higher numbers of large tortuous desmin-positive blood vessels, which correlated with decreased central tumor necrosis and increased tumor cell proliferation. O-ASC exhibited enhanced motility as compared with SC-ASC in response to Hec1a-secreted factors. CONCLUSIONS Visceral adipose tissue contains a population of multipotent MSCs that promote endometrial tumor growth more potently than MSCs from subcutaneous adipose tissue. We propose that O-ASCs recruited to tumors express specific factors that enhance tumor vascularization, promoting survival and proliferation of tumor cells.
Collapse
Affiliation(s)
- Ann H Klopp
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Fu RH, Wang YC, Liu SP, Huang CM, Kang YH, Tsai CH, Shyu WC, Lin SZ. Differentiation of stem cells: strategies for modifying surface biomaterials. Cell Transplant 2010; 20:37-47. [PMID: 21054953 DOI: 10.3727/096368910x532756] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Stem cells are a natural choice for cellular therapy because of their potential to differentiate into a variety of lineages, their capacity for self-renewal in the repair of damaged organs and tissues in vivo, and their ability to generate tissue constructs in vitro. Determining how to efficiently drive stem cell differentiation to a lineage of choice is critical for the success of cellular therapeutics. Many factors are involved in this process, the extracellular microenvironment playing a significant role in controlling cellular behavior. In recent years, researchers have focused on identifying a variety of biomaterials to provide a microenvironment that is conducive to stem cell growth and differentiation and that ultimately mimics the in vivo situation. Appropriate biomaterials support the cellular attachment, proliferation, and lineage-specific differentiation of stem cells. Tissue engineering approaches have been used to incorporate growth factors and morphogenetic factors-factors known to induce lineage commitment of stem cells-into cultures with scaffolding materials, including synthetic and naturally derived biomaterials. This review focuses on various strategies that have been used in stem cell expansion and examines modifications of natural and synthetic materials, as well as various culture conditions, for the maintenance and lineage-specific differentiation of embryonic and adult stem cells.
Collapse
Affiliation(s)
- Ru-Huei Fu
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Jorgensen C. Mesenchymal stem cells in arthritis: role of bone marrow microenvironment. Arthritis Res Ther 2010; 12:135. [PMID: 20804569 PMCID: PMC2945053 DOI: 10.1186/ar3105] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Based on their capacity to suppress immune responses, multipotent mesenchymal stromal cells (MSCs) are intensively studied for regenerative medicine. Moreover, MSCs are potent immunomodulatory cells that occur through the secretion of soluble mediators including nitric oxide, transforming growth factor beta, and HLAG5. The MSCs, however, are also able to express inflammatory mediators such as prostaglandin E2 or IL-6. MSCs in the bone marrow are in close contact with T cells and B cells, and they regulate immunological memory by organizing defined numbers of dedicated survival niches for plasma cells and memory T cells in the bone marrow. The role of MSCs in arthritis remains controversial - in some studies, murine allogeneic MSCs are able to decrease arthritis; in other studies, MSCs worsen the local inflammation. A recent paper in Arthritis Research and Therapy shows that bone marrow MSCs have decreased osteoblastic potential in rheumatoid arthritis, which may be related to chronic inflammation or to loss of expression of IL-1 receptor agonist. That article raises the importance of the bone marrow microenvironment for MSC biology.
Collapse
|
31
|
Gabay O, Sanchez C, Taboas JM. [Not Available]. REVUE DU RHUMATISME (ED. FRANCAISE : 1993) 2010; 77:319-322. [PMID: 21057647 PMCID: PMC2967788 DOI: 10.1016/j.rhum.2010.03.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Affiliation(s)
- Odile Gabay
- Service d’Orthopédie et de Biologie du Cartilage, Groupe de Génétique Moléculaire du Cartilage, NIAMS, Institut National de la Santé, Bethesda, MD
| | - Christelle Sanchez
- Unité de Recherche sur l’Os et le Cartilage, Université de Liège, Liège, Belgique
| | - Juan M Taboas
- Centre d’Ingénierie Cellulaire et Moléculaire, Université de Pittsburgh, Faculté de Médecine, Service de Chirurgie Orthopédique, Pittsburgh, PA
| |
Collapse
|
32
|
Gabay O, Sanchez C, Taboas JM. Update in cartilage bio-engineering. Joint Bone Spine 2010; 77:283-6. [DOI: 10.1016/j.jbspin.2010.02.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2009] [Accepted: 02/10/2010] [Indexed: 10/19/2022]
|
33
|
Milano G, Sanna Passino E, Deriu L, Careddu G, Manunta L, Manunta A, Saccomanno MF, Fabbriciani C. The effect of platelet rich plasma combined with microfractures on the treatment of chondral defects: an experimental study in a sheep model. Osteoarthritis Cartilage 2010; 18:971-80. [PMID: 20433936 DOI: 10.1016/j.joca.2010.03.013] [Citation(s) in RCA: 144] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Revised: 02/22/2010] [Accepted: 03/31/2010] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To evaluate the effect of autologous platelet rich plasma (PRP) combined with microfractures on the treatment of chondral defects. The hypothesis of the study was that PRP can enhance cartilage repair after microfractures. METHODS A chronic full-thickness chondral lesion of the medial femoral condyle was performed in 15 sheep. Animals were divided into three groups, according to treatment: group 1: microfractures; group 2: microfractures+PRP and fibrin glue gel; group 3: microfractures+liquid-PRP injection. Animals were sacrificed at 6 months after treatment. Macroscopic appearance was evaluated according to International Cartilage Repair Society (ICRS) score; cartilage stiffness was analyzed with an electromechanical indenter (Artscan 200); histological appearance was scored according to a modified O'Driscoll score. Comparison between groups for each outcome was performed with Kruskal-Wallis test, and Tukey's test for pairwise comparisons. RESULTS Macroscopic ICRS score of group 2 was significantly better than those of the other groups, and score of group 1 was significantly lower than those of the other groups. Scores of group 1 and 3 were significantly lower than that of normal cartilage. Mean cartilage stiffness of groups 1 and 3 was significantly lower than that of normal cartilage. Histological total scores of group 2 and 3 were significantly better than that of group 1. CONCLUSIONS PRP showed a positive effect on cartilage repair and restoration after microfractures. The procedure was more effective when PRP was used as a gel in comparison with liquid intra-articular injection. Histological analysis revealed that none of experimental treatments produced hyaline cartilage.
Collapse
Affiliation(s)
- G Milano
- Department of Orthopaedics, Catholic University, Rome, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Chondrogenic differentiation of human mesenchymal stem cells: a comparison between micromass and pellet culture systems. Biotechnol Lett 2010; 32:1339-46. [PMID: 20464452 DOI: 10.1007/s10529-010-0293-x] [Citation(s) in RCA: 171] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 04/29/2010] [Indexed: 10/19/2022]
Abstract
High-density cell culture is pivotal for the chondrogenic differentiation of human mesenchymal stem cells (hMSCs). Two high-density cell culture systems, micromass and pellet culture, have been used to induce chondrogenic differentiation of hMSCs. In micromass culture, the induced-cartilage tissues were larger, more homogenous and enriched in cartilage-specific collagen II but the fibrocartilage-like feature, collagen I, and hypertrophic chondrocyte feature, collagen X, were markedly decreased compared to those in pellet culture. Furthermore, real time RT-PCR analysis demonstrated that collagen II and aggrecan mRNA were up-regulated while collagen X and collagen I mRNA were down-regulated in micromass culture. Thus, the micromass culture system is a promising tool for in vitro chondrogenic studies.
Collapse
|
35
|
Lindner U, Kramer J, Rohwedel J, Schlenke P. Mesenchymal Stem or Stromal Cells: Toward a Better Understanding of Their Biology? ACTA ACUST UNITED AC 2010; 37:75-83. [PMID: 20737049 DOI: 10.1159/000290897] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Accepted: 02/24/2010] [Indexed: 01/12/2023]
Abstract
The adult bone marrow has been generally considered to be composed of hematopoietic tissue and the associated supporting stroma. Within the latter compartment, a subset of cells with multipotent differentiation capacity exists, usually referred to as mesenchymal stem cells. Mesenchymal stem cells can easily be expanded ex vivo and induced to differentiate into several cell types, including osteoblasts, adipocytes and chondrocytes. Up to now, mesenchymal stem cells have gained wide popularity. Despite the rapid growth in this field, irritations remain with respect to the defining characteristics of these cells, including their differentiation potency, self-renewal and in vivo properties. As a consequence, there is a growing tendency to challenge the term mesenchymal stem cell, especially with respect to the stem cell characteristics. Here, we revisit the experimental origins of mesenchymal stem cells, their classical differentiation capacity into mesodermal lineages and their immunophenotype in order to assess their stemness and function. Based on these essentials, it has to be revisited if the designation as a stem cell remains an appropriate term.
Collapse
Affiliation(s)
- Ulrich Lindner
- Medical Department I, Division of Nephrology and Transplantation Unit, University of Lübeck, Germany
| | | | | | | |
Collapse
|
36
|
de Girolamo L, Lopa S, Arrigoni E, Sartori MF, Baruffaldi Preis FW, Brini AT. Human adipose-derived stem cells isolated from young and elderly women: their differentiation potential and scaffold interaction during in vitro osteoblastic differentiation. Cytotherapy 2010; 11:793-803. [PMID: 19878065 DOI: 10.3109/14653240903079393] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND AIMS Several authors have demonstrated that adipose tissue contains multipotent cells capable of differentiation into several lineages, including bone, cartilage and fat. METHODS This study compared human adipose-derived stem cells (hASC) isolated from 26 female donors, under 35 and over 45 years old, showing differences in their cell numbers and proliferation, and evaluated their in vitro adipocytic and osteoblastic differentiation potential. RESULTS The cellular yield of hASC from older donors was significantly greater than that from younger donors, whereas their clonogenic potential appeared slightly reduced. There were no significant discrepancies between hASC isolated from young and elderly women regarding their in vitro adipocytic differentiation, whereas the osteoblastic potential was significantly reduced by aging. We also assessed the influence of hydroxyapatite (HAP) and silicon carbide (SiC-PECVD) on hASC. Even when cultured on scaffolds, hASC from younger donors had better differentiation into osteoblast-like cells than hASC from older donors; their differentiation ability was up-regulated by the presence of HAP, whereas SiC-PECVD produced no significant effect on hASC osteoblastic differentiation. CONCLUSIONS The large numbers of hASC resident in adipose tissue and their differentiation features suggest that they could be used for a successful bone regeneration process in vivo. We have shown that age does not seem to affect cell viability and in vitro adipocytic differentiation significantly, whereas it does affects osteoblastic differentiation, in the absence and presence of two-dimensional and three-dimensional scaffolds.
Collapse
Affiliation(s)
- Laura de Girolamo
- Department of Medical Pharmacology, Faculty of Medicine, Università degli Studi di Milano, Milan, Italy
| | | | | | | | | | | |
Collapse
|
37
|
Djouad F, Bouffi C, Ghannam S, Noël D, Jorgensen C. Mesenchymal stem cells: innovative therapeutic tools for rheumatic diseases. Nat Rev Rheumatol 2009; 5:392-9. [PMID: 19568253 DOI: 10.1038/nrrheum.2009.104] [Citation(s) in RCA: 221] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs), or multipotent mesenchymal stromal cells as they are also known, have been identified in bone marrow as well as in other tissues of the joint, including adipose, synovium, periosteum, perichondrium, and cartilage. These cells are characterized by their phenotype and their ability to differentiate into three lineages: chondrocytes, osteoblasts and adipocytes. Importantly, MSCs also potently modulate immune responses, exhibit healing capacities, improve angiogenesis and prevent fibrosis. These properties might be explained at least in part by the trophic effects of MSCs through the secretion of a number of cytokines and growth factors. However, the mechanisms involved in the differentiation potential of MSCs, and their immunomodulatory and paracrine properties, are currently being extensively studied. These unique properties of MSCs confer on them the potential to be used for therapeutic applications in rheumatic diseases, including rheumatoid arthritis, osteoarthritis, genetic bone and cartilage disorders as well as bone metastasis.
Collapse
|
38
|
Vinatier C, Mrugala D, Jorgensen C, Guicheux J, Noël D. Cartilage engineering: a crucial combination of cells, biomaterials and biofactors. Trends Biotechnol 2009; 27:307-14. [DOI: 10.1016/j.tibtech.2009.02.005] [Citation(s) in RCA: 353] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Revised: 02/09/2009] [Accepted: 02/12/2009] [Indexed: 12/13/2022]
|
39
|
Wehling N, Palmer GD, Pilapil C, Liu F, Wells JW, Müller PE, Evans CH, Porter RM. Interleukin-1beta and tumor necrosis factor alpha inhibit chondrogenesis by human mesenchymal stem cells through NF-kappaB-dependent pathways. ACTA ACUST UNITED AC 2009; 60:801-12. [PMID: 19248089 DOI: 10.1002/art.24352] [Citation(s) in RCA: 190] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The differentiation of mesenchymal stem cells (MSCs) into chondrocytes provides an attractive basis for the repair and regeneration of articular cartilage. Under clinical conditions, chondrogenesis will often need to occur in the presence of mediators of inflammation produced in response to injury or disease. The purpose of this study was to examine the effects of 2 important inflammatory cytokines, interleukin-1beta (IL-1beta) and tumor necrosis factor alpha (TNFalpha), on the chondrogenic behavior of human MSCs. METHODS Aggregate cultures of MSCs recovered from the femoral intermedullary canal were used. Chondrogenesis was assessed by the expression of relevant transcripts by quantitative reverse transcription-polymerase chain reaction analysis and examination of aggregates by histologic and immunohistochemical analyses. The possible involvement of NF-kappaB in mediating the effects of IL-1beta was examined by delivering a luciferase reporter construct and a dominant-negative inhibitor of NF-kappaB (suppressor-repressor form of IkappaB [srIkappaB]) with adenovirus vectors. RESULTS Both IL-1beta and TNFalpha inhibited chondrogenesis in a dose-dependent manner. This was associated with a marked activation of NF-kappaB. Delivery of srIkappaB abrogated the activation of NF-kappaB and rescued the chondrogenic response. Although expression of type X collagen followed this pattern, other markers of hypertrophic differentiation responded differently. Matrix metalloproteinase 13 was induced by IL-1beta in a NF-kappaB-dependent manner. Alkaline phosphatase activity, in contrast, was inhibited by IL-1beta regardless of srIkappaB delivery. CONCLUSION Cell-based repair of lesions in articular cartilage will be compromised in inflamed joints. Strategies for enabling repair under these conditions include the use of specific antagonists of individual pyrogens, such as IL-1beta and TNFalpha, or the targeting of important intracellular mediators, such as NF-kappaB.
Collapse
Affiliation(s)
- N Wehling
- Campus Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Camafeita E, Lamas JR, Calvo E, López JA, Fernández-Gutiérrez B. Proteomics: New insights into rheumatic diseases. Proteomics Clin Appl 2009; 3:226-241. [DOI: 10.1002/prca.200800146] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
41
|
Stocum DL, Zupanc GK. Stretching the limits: Stem cells in regeneration science. Dev Dyn 2008; 237:3648-71. [DOI: 10.1002/dvdy.21774] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
42
|
De Girolamo L, Sartori MF, Arrigoni E, Rimondini L, Albisetti W, Weinstein RL, Brini AT. Human adipose-derived stem cells as future tools in tissue regeneration: osteogenic differentiation and cell-scaffold interaction. Int J Artif Organs 2008; 31:467-79. [PMID: 18609499 DOI: 10.1177/039139880803100602] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Tissue engineering is now contributing to new developments in several clinical fields, and mesenchymal stem cells derived from adipose tissue (hASCs) may provide a novel opportunity to replace, repair and promote the regeneration of diseased or damaged musculoskeletal tissue. Our interest was to characterize and differentiate hASCs isolated from twenty-three donors. Proliferation, CFU-F, cytofluorimetric and histochemistry analyses were performed. HASCs differentiate into osteogenic, chondrogenic, and adipogenic lineages, as assessed by tissue-specific markers such as alkaline phosphatase, osteopontin expression and deposition of calcium matrix, lipid-vacuoles formation and Glycosaminoglycans production. We also compared osteo-differentiated hASCs cultured on monolayer and loaded on biomaterials routinely used in the clinic, such as hydroxyapatite, cancellous human bone fragments, deproteinized bovine bone granules, and titanium. Scaffolds loaded with pre-differentiated hASCs do not affect cell proliferation and no cellular toxicity was observed. HASCs tightly adhere to scaffolds and differentiated-hASCs on human bone fragments and bovine bone granules produced, respectively, 3.4- and 2.1-fold more calcified matrix than osteo-differentiated hASCs on monolayer. Moreover, both human and deproteinized bovine bone is able to induce osteogenic differentiation of CTRL-hASCs. Although our in vitro results need to be confirmed in in vivo bone regeneration models, our data suggest that hASCs may be considered suitable biological tools for the screening of innovative scaffolds that would be useful in tissue engineering.
Collapse
Affiliation(s)
- L De Girolamo
- Department of Medical Pharmacology, Faculty of Medicine, University of Milan, Milan, Milan - Italy
| | | | | | | | | | | | | |
Collapse
|
43
|
MacIntosh AC, Kearns VR, Crawford A, Hatton PV. Skeletal tissue engineering using silk biomaterials. J Tissue Eng Regen Med 2008; 2:71-80. [PMID: 18383453 DOI: 10.1002/term.68] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Silks have been proposed as potential scaffold materials for tissue engineering, mainly because of their physical properties. They are stable at physiological temperatures, flexible and resist tensile and compressive forces. Bombyx mori (silkworm) cocoon silk has been used as a suture material for over a century, and has proved to be biocompatible once the immunogenic sericin coating is removed. Spider silks have a similar structure to silkworm silk but do not have a sericin coating. This paper provides a general overview on the use of silk protein in biomaterials, with a focus on skeletal tissue engineering.
Collapse
Affiliation(s)
- Ana C MacIntosh
- Centre for Biomaterials and Tissue Engineering, School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | | | | | | |
Collapse
|
44
|
Schallmoser K, Bartmann C, Rohde E, Reinisch A, Kashofer K, Stadelmeyer E, Drexler C, Lanzer G, Linkesch W, Strunk D. Human platelet lysate can replace fetal bovine serum for clinical-scale expansion of functional mesenchymal stromal cells. Transfusion 2007; 47:1436-46. [PMID: 17655588 DOI: 10.1111/j.1537-2995.2007.01220.x] [Citation(s) in RCA: 376] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Human multipotent mesenchymal stromal cells (MSCs) are promising candidates for a growing spectrum of regenerative and immunomodulatory cellular therapies. Translation of auspicious experimental results into clinical applications has been limited by the dependence of MSC propagation from fetal bovine serum (FBS). STUDY DESIGN AND METHODS The capacity of human platelet lysate (HPL) to replace FBS for clinical-scale MSC propagation was analyzed. RESULTS HPL could be efficiently produced from buffy coats. Multiplex analyses allowed a distinct HPL growth factor profile to be delineated. With a previously established two-step clinical-scale procedure, HPL was reproducibly more efficient than FBS in supporting MSC outgrowth. With only 3 x 10(5) primary culture-derived MSCs, a mean of 4.36 x 10(8) HPL-MSCs (range, 3.01 x 10(8)-5.40 x 10(8)) was obtained within a single secondary 11- to 13-day culture step. Although morphologically distinct, HPL-MSCs and FBS-MSCs did not differ significantly in terms of immunophenotype, differentiation potential in vitro, and lack of tumorigenicity in nude mice in vivo. CONCLUSIONS Replacing FBS with HPL prevents bovine prion, viral, and zoonose contamination of the stem cell product. This new efficient FBS-free two-step procedure for clinical-scale MSC propagation may represent a major step toward challenging new stem cell therapies.
Collapse
Affiliation(s)
- Katharina Schallmoser
- Department of Blood Group Serology and Transfusion Medicine, Division of Hematology and Stem Cell Transplantation, Medical University, Auenbrugger Platz 38, A-8036 Graz, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|