251
|
He P, Fu J, Wang DA. Murine pluripotent stem cells derived scaffold-free cartilage grafts from a micro-cavitary hydrogel platform. Acta Biomater 2016; 35:87-97. [PMID: 26911880 DOI: 10.1016/j.actbio.2016.02.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/18/2016] [Accepted: 02/18/2016] [Indexed: 12/31/2022]
Abstract
By means of appropriate cell type and scaffold, tissue-engineering approaches aim to construct grafts for cartilage repair. Pluripotent stem cells especially induced pluripotent stem cells (iPSCs) are of promising cell candidates due to the pluripotent plasticity and abundant cell source. We explored three dimensional (3D) culture and chondrogenesis of murine iPSCs (miPSCs) on an alginate-based micro-cavity hydrogel (MCG) platform in pursuit of fabricating synthetic-scaffold-free cartilage grafts. Murine embryonic stem cells (mESCs) were employed in parallel as the control. Chondrogenesis was fulfilled using a consecutive protocol via mesoderm differentiation followed by chondrogenic differentiation; subsequently, miPSC and mESC-seeded constructs were further respectively cultured in chondrocyte culture (CC) medium. Alginate phase in the constructs was then removed to generate a graft only comprised of induced chondrocytic cells and cartilaginous extracellular matrix (ECMs). We found that from the mESC-seeded constructs, formation of intact grafts could be achieved in greater sizes with relatively fewer chondrocytic cells and abundant ECMs; from miPSC-seeded constructs, relatively smaller sized cartilaginous grafts could be formed by cells with chondrocytic phenotype wrapped by abundant and better assembled collagen type II. This study demonstrated successful creation of pluripotent stem cells-derived cartilage/chondroid graft from a 3D MCG interim platform. By the support of materials and methodologies established from this study, particularly given the autologous availability of iPSCs, engineered autologous cartilage engraftment may be potentially fulfilled without relying on the limited and invasive autologous chondrocytes acquisition. STATEMENT OF SIGNIFICANCE In this study, we explored chondrogenic differentiation of pluripotent stem cells on a 3D micro-cavitary hydrogel interim platform and creation of pluripotent stem cells-derived cartilage/chondroid graft via a consecutive procedure. Our results demonstrated chondrogenic differentiation could be realized on the platform via mesoderm differentiation. The mESCs/miPSCs derived chondrocytic cells were further cultured to finally generate a pluripotent stem cells-derived scaffold-free construct based on the micro-cavitary hydrogel platform, in which alginate hydrogel could be removed finally. Our results showed that miPSC-derived graft could be formed by cells with chondrocytic phenotype wrapped by abundant and assembled collagen type II. To our knowledge, this study is the first study that initials from pluripotent stem cell seeding on 3D scaffold environment and ends with a scaffold-free chondrogenic micro-tissue. By the support of materials and methodologies established from this study, engineered autologous iPSC-derived cartilage engraftment may be potentially developed instead of autologous chondrocytes grafts that have limited source.
Collapse
|
252
|
Characterisation of synovial fluid and infrapatellar fat pad derived mesenchymal stromal cells: The influence of tissue source and inflammatory stimulus. Sci Rep 2016; 6:24295. [PMID: 27073003 PMCID: PMC4829842 DOI: 10.1038/srep24295] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/16/2016] [Indexed: 12/11/2022] Open
Abstract
The infrapatellar fat pad (FP) and synovial fluid (SF) in the knee serve as reservoirs of mesenchymal stromal cells (MSCs) with potential therapeutic benefit. We determined the influence of the donor on the phenotype of donor matched FP and SF derived MSCs and examined their immunogenic and immunomodulatory properties before and after stimulation with the pro-inflammatory cytokine interferon-gamma (IFN-γ). Both cell populations were positive for MSC markers CD73, CD90 and CD105, and displayed multipotency. FP-MSCs had a significantly faster proliferation rate than SF-MSCs. CD14 positivity was seen in both FP-MSCs and SF-MSCs, and was positively correlated to donor age but only for SF-MSCs. Neither cell population was positive for the co-stimulatory markers CD40, CD80 and CD86, but both demonstrated increased levels of human leukocyte antigen-DR (HLA-DR) following IFN-γ stimulation. HLA-DR production was positively correlated with donor age for FP-MSCs but not SF-MSCs. The immunomodulatory molecule, HLA-G, was constitutively produced by both cell populations, unlike indoleamine 2, 3-dioxygenase which was only produced following IFN-γ stimulation. FP and SF are accessible cell sources which could be utilised in the treatment of cartilage injuries, either by transplantation following ex-vivo expansion or endogenous targeting and mobilisation of cells close to the site of injury.
Collapse
|
253
|
Maturavongsadit P, Luckanagul JA, Metavarayuth K, Zhao X, Chen L, Lin Y, Wang Q. Promotion of In Vitro Chondrogenesis of Mesenchymal Stem Cells Using In Situ Hyaluronic Hydrogel Functionalized with Rod-Like Viral Nanoparticles. Biomacromolecules 2016; 17:1930-8. [DOI: 10.1021/acs.biomac.5b01577] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Panita Maturavongsadit
- Department
of Chemistry and Biochemistry, University of South Carolina, 631
Sumter Street, Columbia, South Carolina 29208, United States
| | - Jittima Amie Luckanagul
- Department
of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, 254 Phayathai Road, Wangmai, Pathumwan, Bangkok, 10330, Thailand
| | - Kamolrat Metavarayuth
- Department
of Chemistry and Biochemistry, University of South Carolina, 631
Sumter Street, Columbia, South Carolina 29208, United States
| | - Xia Zhao
- State
Key Laboratory of Polymer Physics and Chemistry, Changchun Institute
of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People’s Republic of China
| | - Limin Chen
- State
Key Laboratory of Polymer Physics and Chemistry, Changchun Institute
of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People’s Republic of China
| | - Yuan Lin
- State
Key Laboratory of Polymer Physics and Chemistry, Changchun Institute
of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People’s Republic of China
| | - Qian Wang
- Department
of Chemistry and Biochemistry, University of South Carolina, 631
Sumter Street, Columbia, South Carolina 29208, United States
| |
Collapse
|
254
|
Roach BL, Kelmendi-Doko A, Balutis EC, Marra KG, Ateshian GA, Hung CT. Dexamethasone Release from Within Engineered Cartilage as a Chondroprotective Strategy Against Interleukin-1α. Tissue Eng Part A 2016; 22:621-32. [PMID: 26956216 DOI: 10.1089/ten.tea.2016.0018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
While significant progress has been made toward engineering functional cartilage constructs with mechanical properties suitable for in vivo loading, the impact on these grafts of inflammatory cytokines, chemical factors that are elevated with trauma or osteoarthritis, is poorly understood. Previous work has shown dexamethasone to be a critical compound for cultivating cartilage with functional properties, while also providing chondroprotection from proinflammatory cytokines. This study tested the hypothesis that the incorporation of poly(lactic-co-glycolic acid) (PLGA) (75:25) microspheres that release dexamethasone from within chondrocyte-seeded agarose hydrogel constructs would promote development of constructs with functional properties and protect constructs from the deleterious effects of interleukin-1α (IL-1α). After 28 days of growth culture, experimental groups were treated with IL-1α (10 ng/mL) for 7 days. Reaching native equilibrium moduli and proteoglycan levels, dexamethasone-loaded microsphere constructs exhibited tissue properties similar to microsphere-free control constructs cultured in dexamethasone-supplemented culture media and were insensitive to IL-1α exposure. These findings are in stark contrast to constructs containing dexamethasone-free microspheres or no microspheres, cultured without dexamethasone, where IL-1α exposure led to significant tissue degradation. These results support the use of dexamethasone delivery from within engineered cartilage, through biodegradable microspheres, as a strategy to produce mechanically functional tissues that can also combat the deleterious effects of local proinflammatory cytokine exposure.
Collapse
Affiliation(s)
- Brendan L Roach
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Arta Kelmendi-Doko
- 2 Department of Bioengineering, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Elaine C Balutis
- 3 Department of Orthopedics and Sports Medicine, Mount Sinai Health System , New York, New York
| | - Kacey G Marra
- 2 Department of Bioengineering, University of Pittsburgh , Pittsburgh, Pennsylvania.,4 McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania.,5 Department of Plastic Surgery, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Gerard A Ateshian
- 1 Department of Biomedical Engineering, Columbia University , New York, New York.,6 Department of Mechanical Engineering, Columbia University , New York, New York
| | - Clark T Hung
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| |
Collapse
|
255
|
Clevenger TN, Hinman CR, Ashley Rubin RK, Smither K, Burke DJ, Hawker CJ, Messina D, Van Epps D, Clegg DO. Vitronectin-Based, Biomimetic Encapsulating Hydrogel Scaffolds Support Adipogenesis of Adipose Stem Cells. Tissue Eng Part A 2016; 22:597-609. [PMID: 26956095 DOI: 10.1089/ten.tea.2015.0550] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Soft tissue defects are relatively common, yet currently used reconstructive treatments have varying success rates, and serious potential complications such as unpredictable volume loss and reabsorption. Human adipose-derived stem cells (ASCs), isolated from liposuction aspirate have great potential for use in soft tissue regeneration, especially when combined with a supportive scaffold. To design scaffolds that promote differentiation of these cells down an adipogenic lineage, we characterized changes in the surrounding extracellular environment during adipogenic differentiation. We found expression changes in both extracellular matrix proteins, including increases in expression of collagen-IV and vitronectin, as well as changes in the integrin expression profile, with an increase in expression of integrins such as αVβ5 and α1β1. These integrins are known to specifically interact with vitronectin and collagen-IV, respectively, through binding to an Arg-Gly-Asp (RGD) sequence. When three different short RGD-containing peptides were incorporated into three-dimensional (3D) hydrogel cultures, it was found that an RGD-containing peptide derived from vitronectin provided strong initial attachment, maintained the desired morphology, and created optimal conditions for in vitro 3D adipogenic differentiation of ASCs. These results describe a simple, nontoxic encapsulating scaffold, capable of supporting the survival and desired differentiation of ASCs for the treatment of soft tissue defects.
Collapse
Affiliation(s)
- Tracy N Clevenger
- 1 Center for Stem Cell Biology and Engineering, University of California , Santa Barbara, Santa Barbara, California.,2 Department of Molecular, Cellular and Developmental Biology, University of California , Santa Barbara, Santa Barbara, California
| | - Cassidy R Hinman
- 1 Center for Stem Cell Biology and Engineering, University of California , Santa Barbara, Santa Barbara, California
| | - Rebekah K Ashley Rubin
- 1 Center for Stem Cell Biology and Engineering, University of California , Santa Barbara, Santa Barbara, California.,2 Department of Molecular, Cellular and Developmental Biology, University of California , Santa Barbara, Santa Barbara, California
| | | | - Daniel J Burke
- 4 Materials Research Laboratory, University of California , Santa Barbara
| | - Craig J Hawker
- 4 Materials Research Laboratory, University of California , Santa Barbara
| | | | | | - Dennis O Clegg
- 1 Center for Stem Cell Biology and Engineering, University of California , Santa Barbara, Santa Barbara, California.,2 Department of Molecular, Cellular and Developmental Biology, University of California , Santa Barbara, Santa Barbara, California
| |
Collapse
|
256
|
Lolli A, Narcisi R, Lambertini E, Penolazzi L, Angelozzi M, Kops N, Gasparini S, van Osch GJ, Piva R. Silencing of Antichondrogenic MicroRNA-221 in Human Mesenchymal Stem Cells Promotes Cartilage Repair In Vivo. Stem Cells 2016; 34:1801-11. [DOI: 10.1002/stem.2350] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 01/21/2016] [Accepted: 02/01/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Andrea Lolli
- Department of Biomedical and Specialty Surgical Sciences; University of Ferrara; Ferrara Italy
| | - Roberto Narcisi
- Department of Orthopaedics; Erasmus MC, University Medical Center; CN Rotterdam The Netherlands
| | - Elisabetta Lambertini
- Department of Biomedical and Specialty Surgical Sciences; University of Ferrara; Ferrara Italy
| | - Letizia Penolazzi
- Department of Biomedical and Specialty Surgical Sciences; University of Ferrara; Ferrara Italy
| | - Marco Angelozzi
- Department of Biomedical and Specialty Surgical Sciences; University of Ferrara; Ferrara Italy
| | - Nicole Kops
- Department of Orthopaedics; Erasmus MC, University Medical Center; CN Rotterdam The Netherlands
| | - Simona Gasparini
- Department of Orthopaedics; Erasmus MC, University Medical Center; CN Rotterdam The Netherlands
| | - Gerjo J.V.M. van Osch
- Department of Orthopaedics; Erasmus MC, University Medical Center; CN Rotterdam The Netherlands
- Department of Otorhinolaryngology; Erasmus MC, University Medical Center; CN Rotterdam The Netherlands
| | - Roberta Piva
- Department of Biomedical and Specialty Surgical Sciences; University of Ferrara; Ferrara Italy
| |
Collapse
|
257
|
Im GI. Regeneration of articular cartilage using adipose stem cells. J Biomed Mater Res A 2016; 104:1830-44. [PMID: 26990234 DOI: 10.1002/jbm.a.35705] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 03/02/2016] [Indexed: 12/16/2022]
Abstract
Articular cartilage (AC) has limited potential for self-regeneration and damage to AC eventually leads to the development and progression of osteoarthritis (OA). Cell implantation strategies have emerged as a new treatment modality to regenerate AC. Adipose stem cells/adipose-derived stromal cells (ASCs) have gained attention due to their abundance, excellent proliferative potential, and minimal morbidity during harvest. These advantages lower the cost of cell therapy by circumventing time-consuming procedure of culture expansion. ASCs have drawn attention as a potential source for cartilage regeneration since the feasibility of chondrogenesis from ASCs was first reported. After several groups reported inferior chondrogenesis from ASCs, numerous methods were devised to overcome the intrinsic properties. Most in vivo animal studies have reported good results using predifferentiated or undifferentiated, autologous or allogeneic ASCs to regenerate cartilage in osteochondral defects or surgically-induced OA. In this review, we summarize literature on the isolation and in vitro differentiation processes of ASCs, in vivo studies to regenerate AC in osteochondral defects and OA using ASCs, and clinical applications of ASCs. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1830-1844, 2016.
Collapse
Affiliation(s)
- Gun-Il Im
- Department of Orthopedics, Dongguk University Ilsan Hospital, Goyang, Korea
| |
Collapse
|
258
|
Finlay S, Seedhom BB, Carey DO, Bulpitt AJ, Treanor DE, Kirkham J. In Vitro Engineering of High Modulus Cartilage-Like Constructs. Tissue Eng Part C Methods 2016; 22:382-97. [PMID: 26850081 PMCID: PMC4827287 DOI: 10.1089/ten.tec.2015.0351] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
To date, the outcomes of cartilage repair have been inconsistent and have frequently yielded mechanically inferior fibrocartilage, thereby increasing the chances of damage recurrence. Implantation of constructs with biochemical composition and mechanical properties comparable to natural cartilage could be advantageous for long-term repair. This study attempted to create such constructs, in vitro, using tissue engineering principles. Bovine synoviocytes were seeded on nonwoven polyethylene terephthalate fiber scaffolds and cultured in chondrogenic medium for 4 weeks, after which uniaxial compressive loading was applied using an in-house bioreactor for 1 h per day, at a frequency of 1 Hz, for a further 84 days. The initial loading conditions, determined from the mechanical properties of the immature constructs after 4 weeks in chondrogenic culture, were strains ranging between 13% and 23%. After 56 days (sustained at 84 days) of loading, the constructs were stained homogenously with Alcian blue and for type-II collagen. Dynamic compressive moduli were comparable to the high end values for native cartilage and proportional to Alcian blue staining intensity. We suggest that these high moduli values were attributable to the bioreactor setup, which caused the loading regime to change as the constructs developed, that is, the applied stress and strain increased with construct thickness and stiffness, providing continued sufficient cell stimulation as further matrix was deposited. Constructs containing cartilage-like matrix with response to load similar to that of native cartilage could produce long-term effective cartilage repair when implanted.
Collapse
Affiliation(s)
- Scott Finlay
- 1 Division of Oral Biology, School of Dentistry, University of Leeds , Leeds, United Kingdom
| | - Bahaa B Seedhom
- 1 Division of Oral Biology, School of Dentistry, University of Leeds , Leeds, United Kingdom
| | - Duane O Carey
- 2 School of Computing, University of Leeds , Leeds, United Kingdom
| | - Andy J Bulpitt
- 2 School of Computing, University of Leeds , Leeds, United Kingdom
| | - Darren E Treanor
- 3 Department of Pathology, Leeds Institute of Cancer and Pathology, University of Leeds , Leeds, United Kingdom .,4 Leeds Teaching Hospitals NHS Trust , Leeds, United Kingdom
| | - Jennifer Kirkham
- 1 Division of Oral Biology, School of Dentistry, University of Leeds , Leeds, United Kingdom
| |
Collapse
|
259
|
McCorry MC, Puetzer JL, Bonassar LJ. Characterization of mesenchymal stem cells and fibrochondrocytes in three-dimensional co-culture: analysis of cell shape, matrix production, and mechanical performance. Stem Cell Res Ther 2016; 7:39. [PMID: 26971202 PMCID: PMC4789279 DOI: 10.1186/s13287-016-0301-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/09/2016] [Accepted: 02/26/2016] [Indexed: 02/06/2023] Open
Abstract
Background Bone marrow mesenchymal stem cells (MSCs) have shown positive therapeutic effects for meniscus regeneration and repair. Preliminary in vitro work has indicated positive results for MSC applications for meniscus tissue engineering; however, more information is needed on how to direct MSC behavior. The objective of this study was to examine the effect of MSC co-culture with primary meniscal fibrochondrocytes (FCCs) in a three-dimensional collagen scaffold in fibrochondrogenic media. Co-culture of MSCs and FCCs was hypothesized to facilitate the transition of MSCs to a FCC cell phenotype as measured by matrix secretion and morphology. Methods MSCs and FCCs were isolated from bovine bone marrow and meniscus, respectively. Cells were seeded in a 20 mg/mL high-density type I collagen gel at MSC:FCC ratios of 0:100, 25:75, 50:50, 75:25, and 100:0. Constructs were cultured for up to 2 weeks and then analyzed for cell morphology, glycosaminoglycan content, collagen content, and production of collagen type I, II, and X. Results Cells were homogeneously mixed throughout the scaffold and cells had limited direct cell–cell contact. After 2 weeks in culture, MSCs transitioned from a spindle-like morphology toward a rounded phenotype, while FCCs remained rounded throughout culture. Although MSC shape changed with culture, the overall size was significantly larger than FCCs throughout culture. While 75:25 and 100:0 (MSC mono-culture) culture groups produced significantly more glycosaminoglycan (GAG)/DNA than FCCs in mono-culture, GAG retention was highest in 50:50 co-cultures. Similarly, the aggregate modulus was highest in 100:0 and 50:50 co-cultures. All samples contained both collagen types I and II after 2 weeks, and collagen type X expression was evident only in MSC mono-culture gels. Conclusions MSCs shift to a FCC morphology in both mono- and co-culture. Co-culture reduced hypertrophy by MSCs, indicated by collagen type X. This study shows that MSC phenotype can be influenced by indirect homogeneous cell culture in a three-dimensional gel, demonstrating the applicability of MSCs in meniscus tissue engineering applications. Electronic supplementary material The online version of this article (doi:10.1186/s13287-016-0301-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mary Clare McCorry
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Jennifer L Puetzer
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Lawrence J Bonassar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA. .,Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
260
|
Arslan E, Guler MO, Tekinay AB. Glycosaminoglycan-Mimetic Signals Direct the Osteo/Chondrogenic Differentiation of Mesenchymal Stem Cells in a Three-Dimensional Peptide Nanofiber Extracellular Matrix Mimetic Environment. Biomacromolecules 2016; 17:1280-91. [PMID: 26840042 DOI: 10.1021/acs.biomac.5b01637] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recent efforts in bioactive scaffold development focus strongly on the elucidation of complex cellular responses through the use of synthetic systems. Designing synthetic extracellular matrix (ECM) materials must be based on understanding of cellular behaviors upon interaction with natural and artificial scaffolds. Hence, due to their ability to mimic both the biochemical and mechanical properties of the native tissue environment, supramolecular assemblies of bioactive peptide nanostructures are especially promising for development of bioactive ECM-mimetic scaffolds. In this study, we used glycosaminoglycan (GAG) mimetic peptide nanofiber gel as a three-dimensional (3D) platform to investigate how cell lineage commitment is altered by external factors. We observed that amount of fetal bovine serum (FBS) presented in the cell media had synergistic effects on the ability of GAG-mimetic nanofiber gel to mediate the differentiation of mesenchymal stem cells into osteogenic and chondrogenic lineages. In particular, lower FBS concentration in the culture medium was observed to enhance osteogenic differentiation while higher amount FBS promotes chondrogenic differentiation in tandem with the effects of the GAG-mimetic 3D peptide nanofiber network, even in the absence of externally administered growth factors. We therefore demonstrate that mesenchymal stem cell differentiation can be specifically controlled by the combined influence of growth medium components and a 3D peptide nanofiber environment.
Collapse
Affiliation(s)
- Elif Arslan
- Institute of Materials Science and Nanotechnology, Nanotechnology Research Center (UNAM), Bilkent University , Ankara 06800, Turkey
| | - Mustafa O Guler
- Institute of Materials Science and Nanotechnology, Nanotechnology Research Center (UNAM), Bilkent University , Ankara 06800, Turkey
| | - Ayse B Tekinay
- Institute of Materials Science and Nanotechnology, Nanotechnology Research Center (UNAM), Bilkent University , Ankara 06800, Turkey
| |
Collapse
|
261
|
Single-cell differences in matrix gene expression do not predict matrix deposition. Nat Commun 2016; 7:10865. [PMID: 26936319 PMCID: PMC4782061 DOI: 10.1038/ncomms10865] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 01/28/2016] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stem cells (MSCs) display substantial cell-to-cell heterogeneity, complicating their use in regenerative medicine. However, conventional bulk assays mask this variability. Here we show that both chondrocytes and chondrogenically induced MSCs exhibit substantial mRNA expression heterogeneity. Single-molecule RNA FISH to measure mRNA expression of differentiation markers in single cells reveals that sister cell pairs have high levels of mRNA variability, suggesting that marker expression is not heritable. Surprisingly, this variability does not correlate with cell-to-cell differences in cartilage-like matrix production. Transcriptome-wide analysis suggests that no combination of markers can predict functional potential. De-differentiating chondrocytes also show a disconnect between mRNA expression of the cartilage marker aggrecan and cartilage-like matrix accumulation. Altogether, these quantitative analyses suggest that sorting subpopulations based on these markers would only marginally enrich the progenitor population for ‘superior' MSCs. Our results suggest that instantaneous mRNA abundance of canonical markers is tenuously linked to the chondrogenic phenotype at the single-cell level. Regenerative tissue engineering with mesenchymal stem cells is hampered by bulk methods of assessing differentiation status and a general assumption that expression of individual markers of stem cell differentiation correlate with functional capacity. Here the authors debunk this assumption by applying single-cell techniques to disassociate aggrecan mRNA abundance and matrix deposition.
Collapse
|
262
|
Li X, Ding J, Zhang Z, Yang M, Yu J, Wang J, Chang F, Chen X. Kartogenin-Incorporated Thermogel Supports Stem Cells for Significant Cartilage Regeneration. ACS APPLIED MATERIALS & INTERFACES 2016; 8:5148-5159. [PMID: 26844837 DOI: 10.1021/acsami.5b12212] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Recently, cartilage tissue engineering (CTE) attracts increasing attention in cartilage defect repair. In this work, kartogenin (KGN), an emerging chondroinductive nonprotein small molecule, was incorporated into a thermogel of poly(L-lactide-co-glycolide)-poly(ethylene glycol)-poly(L-lactide-co-glycolide) (PLGA-PEG-PLGA) to fabricate an appropriate microenvironment of bone marrow mesenchymal stem cells (BMSCs) for effective cartilage regeneration. More integrative and smoother repaired articular surface, more abundant characteristic glycosaminoglycans (GAGs) and collagen II (COL II), and less degeneration of normal cartilage were obtained in the KGN and BMSCs coloaded thermogel group in vivo. In conclusion, the KGN-loaded PLGA-PEG-PLGA thermogel can be utilized as an alternative support for BMSCs to regenerate damaged cartilage in vivo.
Collapse
Affiliation(s)
- Xuezhou Li
- Department of Orthopaedics, The Second Hospital of Jilin University , Changchun 130041, People's Republic of China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, People's Republic of China
| | - Zhengzheng Zhang
- Institute of Sports Medicine, Peking University Third Hospital , Beijing 100191, People's Republic of China
| | - Modi Yang
- Department of Orthopaedics, The Second Hospital of Jilin University , Changchun 130041, People's Republic of China
| | - Jiakuo Yu
- Institute of Sports Medicine, Peking University Third Hospital , Beijing 100191, People's Republic of China
| | - Jincheng Wang
- Department of Orthopaedics, The Second Hospital of Jilin University , Changchun 130041, People's Republic of China
| | - Fei Chang
- Department of Orthopaedics, The Second Hospital of Jilin University , Changchun 130041, People's Republic of China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, People's Republic of China
| |
Collapse
|
263
|
Zhang W, Ouyang H, Dass CR, Xu J. Current research on pharmacologic and regenerative therapies for osteoarthritis. Bone Res 2016; 4:15040. [PMID: 26962464 PMCID: PMC4772471 DOI: 10.1038/boneres.2015.40] [Citation(s) in RCA: 330] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 12/05/2015] [Accepted: 12/06/2015] [Indexed: 01/05/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disorder commonly encountered in clinical practice, and is the leading cause of disability in elderly people. Due to the poor self-healing capacity of articular cartilage and lack of specific diagnostic biomarkers, OA is a challenging disease with limited treatment options. Traditional pharmacologic therapies such as acetaminophen, non-steroidal anti-inflammatory drugs, and opioids are effective in relieving pain but are incapable of reversing cartilage damage and are frequently associated with adverse events. Current research focuses on the development of new OA drugs (such as sprifermin/recombinant human fibroblast growth factor-18, tanezumab/monoclonal antibody against β-nerve growth factor), which aims for more effectiveness and less incidence of adverse effects than the traditional ones. Furthermore, regenerative therapies (such as autologous chondrocyte implantation (ACI), new generation of matrix-induced ACI, cell-free scaffolds, induced pluripotent stem cells (iPS cells or iPSCs), and endogenous cell homing) are also emerging as promising alternatives as they have potential to enhance cartilage repair, and ultimately restore healthy tissue. However, despite currently available therapies and research advances, there remain unmet medical needs in the treatment of OA. This review highlights current research progress on pharmacologic and regenerative therapies for OA including key advances and potential limitations.
Collapse
Affiliation(s)
- Wei Zhang
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou 310058, China
- School of Pathology and Laboratory Medicine, The University of Western Australia, Perth WA 6009, Australia
| | - Hongwei Ouyang
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Crispin R Dass
- School of Pharmacy, Building 306, Curtin University, Bentley, Perth WA 6102, Australia
| | - Jiake Xu
- School of Pathology and Laboratory Medicine, The University of Western Australia, Perth WA 6009, Australia
| |
Collapse
|
264
|
Mansour JM, Lee Z, Welter JF. Nondestructive Techniques to Evaluate the Characteristics and Development of Engineered Cartilage. Ann Biomed Eng 2016; 44:733-49. [PMID: 26817458 PMCID: PMC4792725 DOI: 10.1007/s10439-015-1535-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 12/12/2015] [Indexed: 12/16/2022]
Abstract
In this review, methods for evaluating the properties of tissue engineered (TE) cartilage are described. Many of these have been developed for evaluating properties of native and osteoarthritic articular cartilage. However, with the increasing interest in engineering cartilage, specialized methods are needed for nondestructive evaluation of tissue while it is developing and after it is implanted. Such methods are needed, in part, due to the large inter- and intra-donor variability in the performance of the cellular component of the tissue, which remains a barrier to delivering reliable TE cartilage for implantation. Using conventional destructive tests, such variability makes it near-impossible to predict the timing and outcome of the tissue engineering process at the level of a specific piece of engineered tissue and also makes it difficult to assess the impact of changing tissue engineering regimens. While it is clear that the true test of engineered cartilage is its performance after it is implanted, correlation of pre and post implantation properties determined non-destructively in vitro and/or in vivo with performance should lead to predictive methods to improve quality-control and to minimize the chances of implanting inferior tissue.
Collapse
Affiliation(s)
- Joseph M Mansour
- Departments of Mechanical and Aerospace Engineering, Case Western Reserve University, 2123 Martin Luther King Jr. Drive, Glennan Building Room 616A, Cleveland, OH, 44106, USA.
| | - Zhenghong Lee
- Radiology and Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Jean F Welter
- Biology (Skeletal Research Center), Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
265
|
Blázquez R, Sánchez-Margallo FM, Crisóstomo V, Báez C, Maestre J, Álvarez V, Casado JG. Intrapericardial Delivery of Cardiosphere-Derived Cells: An Immunological Study in a Clinically Relevant Large Animal Model. PLoS One 2016; 11:e0149001. [PMID: 26866919 PMCID: PMC4750976 DOI: 10.1371/journal.pone.0149001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/25/2016] [Indexed: 01/22/2023] Open
Abstract
Introduction The intrapericardial delivery has been defined as an efficient method for pharmacological agent delivery. Here we hypothesize that intrapericardial administration of cardiosphere-derived cells (CDCs) may have an immunomodulatory effect providing an optimal microenvironment for promoting cardiac repair. To our knowledge, this is the first report studying the effects of CDCs for myocardial repair using the intrapericardial delivery route. Material and Methods CDCs lines were isolated, expanded and characterized by flow cytometry and PCR. Their differentiation ability was determined using specific culture media and differential staining. 300,000 CDCs/kg were injected into the pericardial space of a swine myocardial infarcted model. Magnetic resonance imaging, biochemical analysis of pericardial fluid and plasma, cytokine measurements and flow cytometry analysis were performed. Results Our results showed that, phenotype and differentiation behavior of porcine CDCs were equivalent to previously described CDCs. Moreover, the intrapericardial administration of CDCs fulfilled the safety aspects as non-adverse effects were reported. Finally, the phenotypes of resident lymphocytes and TH1 cytokines in the pericardial fluid were significantly altered after CDCs administration. Conclusions The pericardial fluid could be considered as a safe and optimal vehicle for CDCs administration. The observed changes in the studied immunological parameters could exert a modulation in the inflammatory environment of infarcted hearts, indirectly benefiting the endogenous cardiac repair.
Collapse
Affiliation(s)
- Rebeca Blázquez
- Stem Cell Therapy Unit, 'Jesús Usón' Minimally Invasive Surgery Centre, Cáceres, Spain
| | | | - Verónica Crisóstomo
- Endoluminal Therapy and Diagnosis, 'Jesús Usón' Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Claudia Báez
- Endoluminal Therapy and Diagnosis, 'Jesús Usón' Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Juan Maestre
- Endoluminal Therapy and Diagnosis, 'Jesús Usón' Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Verónica Álvarez
- Stem Cell Therapy Unit, 'Jesús Usón' Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Javier G Casado
- Stem Cell Therapy Unit, 'Jesús Usón' Minimally Invasive Surgery Centre, Cáceres, Spain
| |
Collapse
|
266
|
Maerz JK, Roncoroni LP, Goldeck D, Abruzzese T, Kalbacher H, Rolauffs B, DeZwart P, Nieselt K, Hart ML, Klein G, Aicher WK. Bone marrow-derived mesenchymal stromal cells differ in their attachment to fibronectin-derived peptides from term placenta-derived mesenchymal stromal cells. Stem Cell Res Ther 2016; 7:29. [PMID: 26869043 PMCID: PMC4751672 DOI: 10.1186/s13287-015-0243-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/03/2015] [Accepted: 11/18/2015] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Human mesenchymal stromal cells (MSCs) can be isolated from different sources including bone marrow and term placenta. These two populations display distinct patterns of proliferation and differentiation in vitro. Since proliferation and differentiation of cells are modulated by cell-matrix interactions, we investigated the attachment of MSCs to a set of peptide-coated surfaces and explored their interactions with peptides in suspension. METHODS Human MSCs were isolated from bone marrow and term placenta and expanded. Binding of MSCs to peptides was investigated by a cell-attachment spot assay, by blocking experiments and flow cytometry. The integrin expression pattern was explored by a transcript array and corroborated by quantitative reverse transcription polymerase chain reaction and flow cytometry. RESULTS Expanded placenta-derived MSCs (pMSCs) attached well to surfaces coated with fibronectin-derived peptides P7, P15, and P17, whereas bone marrow-derived MSCs (bmMSCs) attached to P7, but barely to P15 and P17. The binding of bmMSCs and pMSCs to the peptides was mediated by β1 integrins. In suspension, expanded bmMSCs barely bind to P7, P13, P15, and less to P14 and P17. Ex vivo, bmMSCs failed to bind P7, but displayed a weak interaction with P13, P14, and P15. In suspension, expanded pMSCs displayed binding to many peptides, including P4, P7, P13, P14, P15, and P17. The differences observed in binding of bmMSCs and pMSCs to the peptides were associated with significant differences in expression of integrin α2-, α4-, and α6-chains. CONCLUSIONS Human bmMSCs and pMSCs show distinct patterns of attachment to defined peptides and maintain differences in expression of integrins in vitro. Interactions of ex vivo bmMSCs with a given peptide yield different staining patterns compared to expanded bmMSCs in suspension. Attachment of expanded MSCs to peptides on surfaces is different from interactions of expanded MSCs with peptides in suspension. Studies designed to investigate the interactions of human MSCs with peptide-augmented scaffolds or peptides in suspension must therefore regard these differences in cell-peptide interactions.
Collapse
Affiliation(s)
- Jan K Maerz
- KFO273, Department of Urology, University of Tübingen Hospital, Paul Ehrlich Str. 15, 72076, Tübingen, Germany.
| | - Lorenzo P Roncoroni
- KFO273, Department of Urology, University of Tübingen Hospital, Paul Ehrlich Str. 15, 72076, Tübingen, Germany.
| | - David Goldeck
- Center for Medical Research, Department of Medicine II, University of Tübingen, Tübingen, Germany.
| | - Tanja Abruzzese
- KFO273, Department of Urology, University of Tübingen Hospital, Paul Ehrlich Str. 15, 72076, Tübingen, Germany.
| | - Hubert Kalbacher
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany.
| | - Bernd Rolauffs
- BG Trauma Center Tübingen, University of Tübingen, Tübingen, Germany.
| | - Peter DeZwart
- BG Trauma Center Tübingen, University of Tübingen, Tübingen, Germany.
| | - Kay Nieselt
- Integrative Transcriptomics, Center for Bioinformatics, University of Tübingen, Tübingen, Germany.
| | - Melanie L Hart
- KFO273, Department of Urology, University of Tübingen Hospital, Paul Ehrlich Str. 15, 72076, Tübingen, Germany.
| | - Gerd Klein
- Center for Medical Research, Department of Medicine II, University of Tübingen, Tübingen, Germany.
| | - Wilhelm K Aicher
- KFO273, Department of Urology, University of Tübingen Hospital, Paul Ehrlich Str. 15, 72076, Tübingen, Germany.
| |
Collapse
|
267
|
Christo SN, Diener KR, Manavis J, Grimbaldeston MA, Bachhuka A, Vasilev K, Hayball JD. Inflammasome components ASC and AIM2 modulate the acute phase of biomaterial implant-induced foreign body responses. Sci Rep 2016; 6:20635. [PMID: 26860464 PMCID: PMC4748295 DOI: 10.1038/srep20635] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 01/06/2016] [Indexed: 01/03/2023] Open
Abstract
Detailing the inflammatory mechanisms of biomaterial-implant induced foreign body responses (FBR) has implications for revealing targetable pathways that may reduce leukocyte activation and fibrotic encapsulation of the implant. We have adapted a model of poly(methylmethacrylate) (PMMA) bead injection to perform an assessment of the mechanistic role of the ASC-dependent inflammasome in this process. We first demonstrate that ASC−/− mice subjected to PMMA bead injections had reduced cell infiltration and altered collagen deposition, suggesting a role for the inflammasome in the FBR. We next investigated the NLRP3 and AIM2 sensors because of their known contributions in recognising damaged and apoptotic cells. We found that NLRP3 was dispensable for the fibrotic encapsulation; however AIM2 expression influenced leukocyte infiltration and controlled collagen deposition, suggesting a previously unexplored link between AIM2 and biomaterial-induced FBR.
Collapse
Affiliation(s)
- Susan N Christo
- Experimental Therapeutics Laboratory, Sansom Institute and Hanson Institute, School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA, 5000, Australia
| | - Kerrilyn R Diener
- Experimental Therapeutics Laboratory, Sansom Institute and Hanson Institute, School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA, 5000, Australia.,Robinson Research Institute, School of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Jim Manavis
- Centre for Neurological Diseases, SA Pathology, Adelaide, SA 5000, Australia
| | - Michele A Grimbaldeston
- Centre for Cancer Biology, University of South Australia and SA Pathology, SA 5000, Australia
| | - Akash Bachhuka
- Mawson Institute, University of South Australia, Adelaide, SA 5095, Australia
| | - Krasimir Vasilev
- Mawson Institute, University of South Australia, Adelaide, SA 5095, Australia
| | - John D Hayball
- Experimental Therapeutics Laboratory, Sansom Institute and Hanson Institute, School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA, 5000, Australia.,Robinson Research Institute, School of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, SA, 5005, Australia.,School of Medicine, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
268
|
Hydrodynamic loading in concomitance with exogenous cytokine stimulation modulates differentiation of bovine mesenchymal stem cells towards osteochondral lineages. BMC Biotechnol 2016; 16:10. [PMID: 26830345 PMCID: PMC4736240 DOI: 10.1186/s12896-016-0240-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 01/18/2016] [Indexed: 02/07/2023] Open
Abstract
Background Mesenchymal stem cells (MSCs) are viewed as a having significant potential for tissue engineering and regenerative medicine therapies. Clinical implementation of MSCs, however, demands that their preparation be stable and reproducible. Given that environmental and bioprocessing parameters such as substrate stiffness, seeding densities, culture medium composition, and mechanical loading can result in undirected differentiation of the MSC population, the objective of this study was to systematically investigate how hydrodynamic loading influences the differentiation of bone marrow-derived mesenchymal stem cells (MSCs) towards the osteochondral lineages both in the presence and absence of exogenous, inductive factors. Methods Expanded bovine MSCs were suspended in 2.5 % agarose, cast in a custom mold, and placed into either static or one of two dynamic culture environments consisting of “high” and “low” magnitude shear conditions. Constructs were supplemented with varying concentrations (0, 1, 10, 100 ng/mL) of either TGF-β3 or BMP-2 throughout cultivation with tissue samples being collected following each week of culture. Results In the absence of exogenous supplementation, hydrodynamic loading had little effect on cell phenotype at either magnitude of stimulation. When cultures were supplemented with BMP-2 and TGF-β3, MSCs gene expression progressed towards the osteogenic and chondrogenic pathways, respectively. This progression was enhanced by the presence of hydrodynamic loading, particularly under high shear conditions, but may point the chondrogenic cultures down a hypertrophic path toward osteogenesis reminiscent of endochondral ossification if TGF-β3 supplementation is insufficient. Conclusions Moving forward, these results suggest bioprocessing conditions which minimize exposure of chondrogenic cultures to fluid shear stress to avoid undesirable differentiation of the MSC population. Electronic supplementary material The online version of this article (doi:10.1186/s12896-016-0240-6) contains supplementary material, which is available to authorized users.
Collapse
|
269
|
Tao K, Frisch J, Rey-Rico A, Venkatesan JK, Schmitt G, Madry H, Lin J, Cucchiarini M. Co-overexpression of TGF-β and SOX9 via rAAV gene transfer modulates the metabolic and chondrogenic activities of human bone marrow-derived mesenchymal stem cells. Stem Cell Res Ther 2016; 7:20. [PMID: 26830674 PMCID: PMC4736112 DOI: 10.1186/s13287-016-0280-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 12/16/2015] [Accepted: 01/13/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Articular cartilage has a limited potential for self-healing. Transplantation of genetically modified progenitor cells like bone marrow-derived mesenchymal stem cells (MSCs) is an attractive strategy to improve the intrinsic repair capacities of damaged articular cartilage. METHODS In this study, we examined the potential benefits of co-overexpressing the pleiotropic transformation growth factor beta (TGF-β) with the cartilage-specific transcription factor SOX9 via gene transfer with recombinant adeno-associated virus (rAAV) vectors upon the biological activities of human MSCs (hMSCs). Freshly isolated hMSCs were transduced over time with separate rAAV vectors carrying either TGF-β or sox9 in chondrogenically-induced aggregate cultures to evaluate the efficacy and duration of transgene expression and to monitor the effects of rAAV-mediated genetic modification upon the cellular activities (proliferation, matrix synthesis) and chondrogenic differentiation potency compared with control conditions (lacZ treatment, sequential transductions). RESULTS Significant, prolonged TGF-β/sox9 co-overexpression was achieved in chondrogenically-induced hMSCs upon co-transduction via rAAV for up to 21 days, leading to enhanced proliferative, biosynthetic, and chondrogenic activities relative to control treatments, especially when co-applying the candidate vectors at the highest vector doses tested. Optimal co-administration of TGF-β with sox9 also advantageously reduced hypertrophic differentiation of the cells in the conditions applied here. CONCLUSION The present findings demonstrate the possibility of modifying MSCs by combined therapeutic gene transfer as potent future strategies for implantation in clinically relevant animal models of cartilage defects in vivo.
Collapse
Affiliation(s)
- Ke Tao
- Institute of Arthritis, Peking University People's Hospital, Beijing, 100044, P.R. China. .,Peking University Health Science Center, Beijing, 100191, P.R. China. .,Center of Experimental Orthopedics, Saarland University Medical Center, Kirrbergerstraße Bldg 37, Homburg/Saar, D-66421, Germany.
| | - Janina Frisch
- Center of Experimental Orthopedics, Saarland University Medical Center, Kirrbergerstraße Bldg 37, Homburg/Saar, D-66421, Germany.
| | - Ana Rey-Rico
- Center of Experimental Orthopedics, Saarland University Medical Center, Kirrbergerstraße Bldg 37, Homburg/Saar, D-66421, Germany.
| | - Jagadeesh K Venkatesan
- Center of Experimental Orthopedics, Saarland University Medical Center, Kirrbergerstraße Bldg 37, Homburg/Saar, D-66421, Germany.
| | - Gertrud Schmitt
- Center of Experimental Orthopedics, Saarland University Medical Center, Kirrbergerstraße Bldg 37, Homburg/Saar, D-66421, Germany.
| | - Henning Madry
- Center of Experimental Orthopedics, Saarland University Medical Center, Kirrbergerstraße Bldg 37, Homburg/Saar, D-66421, Germany. .,Department of Orthopaedic Surgery, Saarland University Medical Center, Kirrbergerstr. Bldg 37, Homburg/Saar, D-66421, Germany.
| | - Jianhao Lin
- Institute of Arthritis, Peking University People's Hospital, Beijing, 100044, P.R. China. .,Peking University Health Science Center, Beijing, 100191, P.R. China.
| | - Magali Cucchiarini
- Center of Experimental Orthopedics, Saarland University Medical Center, Kirrbergerstraße Bldg 37, Homburg/Saar, D-66421, Germany.
| |
Collapse
|
270
|
Frisch J, Rey-Rico A, Venkatesan JK, Schmitt G, Madry H, Cucchiarini M. TGF-β gene transfer and overexpression via rAAV vectors stimulates chondrogenic events in human bone marrow aspirates. J Cell Mol Med 2016; 20:430-40. [PMID: 26808466 PMCID: PMC4759465 DOI: 10.1111/jcmm.12774] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 12/01/2015] [Indexed: 12/12/2022] Open
Abstract
Genetic modification of marrow concentrates may provide convenient approaches to enhance the chondrogenic differentiation processes and improve the repair capacities in sites of cartilage defects following administration in the lesions. Here, we provided clinically adapted recombinant adeno‐associated virus (rAAV) vectors to human bone marrow aspirates to promote the expression of the potent transforming growth factor beta (TGF‐β) as a means to regulate the biological and chondrogenic activities in the samples in vitro. Successful TGF‐β gene transfer and expression viarAAV was reached relative to control (lacZ) treatment (from 511.1 to 16.1 pg rhTGF‐β/mg total proteins after 21 days), allowing to durably enhance the levels of cell proliferation, matrix synthesis, and chondrogenic differentiation. Strikingly, in the conditions applied here, application of the candidate TGF‐β vector was also capable of reducing the hypertrophic and osteogenic differentiation processes in the aspirates, showing the potential benefits of using this particular vector to directly modify marrow concentrates to generate single‐step, effective approaches that aim at improving articular cartilage repair in vivo.
Collapse
Affiliation(s)
- Janina Frisch
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Ana Rey-Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | | | - Gertrud Schmitt
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany.,Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| |
Collapse
|
271
|
Bornes TD, Jomha NM, Mulet-Sierra A, Adesida AB. Optimal Seeding Densities for In Vitro Chondrogenesis of Two- and Three-Dimensional-Isolated and -Expanded Bone Marrow-Derived Mesenchymal Stromal Stem Cells Within a Porous Collagen Scaffold. Tissue Eng Part C Methods 2016; 22:208-20. [PMID: 26651081 DOI: 10.1089/ten.tec.2015.0365] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Bone marrow-derived mesenchymal stromal stem cells (BMSCs) are a promising cell source for treating articular cartilage defects. The objective of this study was to assess the impact of cell seeding density within a collagen I scaffold on in vitro BMSC chondrogenesis following isolation and expansion in two-dimensional (2D) and three-dimensional (3D) environments. It was hypothesized that both expansion protocols would produce BMSCs capable of hyaline-like chondrogenesis with an optimal seeding density of 10 × 10(6) cells/cm(3). Ovine BMSCs were isolated in a 2D environment by plastic adherence, expanded to passage two in flasks containing an expansion medium, and seeded within collagen I scaffolds at densities of 50, 10, 5, 1, and 0.5 × 10(6) BMSCs/cm(3). For 3D isolation and expansion, aspirates containing known quantities of mononucleated cells (bone marrow-derived mononucleated cells [BMNCs]) were seeded on scaffolds at 50, 10, 5, 1, and 0.5 × 10(6) BMNCs/cm(3) and cultured in the expansion medium for an equivalent duration to 2D expansion. Constructs were differentiated in vitro in the chondrogenic medium for 21 days and assessed with reverse-transcription quantitative polymerase chain reaction, safranin O staining, histological scoring using the Bern Score, collagen immunofluorescence, and glycosaminoglycan (GAG) quantification. Two-dimensional-expanded BMSCs seeded at all densities were capable of proteoglycan production and displayed increased expressions of aggrecan and collagen II messenger RNA (mRNA) relative to predifferentiation controls. Collagen II deposition was apparent in scaffolds seeded at 0.5-10 × 10(6) BMSCs/cm(3). Chondrogenesis of 2D-expanded BMSCs was most pronounced in scaffolds seeded at 5-10 × 10(6) BMSCs/cm(3) based on aggrecan and collagen II mRNA, safranin O staining, Bern Score, total GAG, and GAG/deoxyribonucleic acid (DNA). For 3D-expanded BMSC-seeded scaffolds, increased aggrecan and collagen II mRNA expressions relative to controls were noted with all densities. Proteoglycan deposition was present in scaffolds seeded at 0.5-50 × 10(6) BMNCs/cm(3), while collagen II deposition occurred in scaffolds seeded at 10-50 × 10(6) BMNCs/cm(3). The highest levels of aggrecan and collagen II mRNA, Bern Score, total GAG, and GAG/DNA occurred with seeding at 50 × 10(6) BMNCs/cm(3). Within a collagen I scaffold, 2D- and 3D-expanded BMSCs are capable of hyaline-like chondrogenesis with optimal cell seeding densities of 5-10 × 10(6) BMSCs/cm(3) and 50 × 10(6) BMNCs/cm(3), respectively.
Collapse
Affiliation(s)
- Troy D Bornes
- Divisions of Orthopaedic Surgery and Surgical Research, Department of Surgery, University of Alberta , Edmonton, Canada
| | - Nadr M Jomha
- Divisions of Orthopaedic Surgery and Surgical Research, Department of Surgery, University of Alberta , Edmonton, Canada
| | - Aillette Mulet-Sierra
- Divisions of Orthopaedic Surgery and Surgical Research, Department of Surgery, University of Alberta , Edmonton, Canada
| | - Adetola B Adesida
- Divisions of Orthopaedic Surgery and Surgical Research, Department of Surgery, University of Alberta , Edmonton, Canada
| |
Collapse
|
272
|
Abstract
Mesenchymal stem cells (MSCs) were initially characterized as connective tissue progenitors resident in bone marrow, but have now been isolated from a variety of tissues and organs and shown to also exhibit potent tissue regenerative properties mediated largely via paracrine actions. These findings have spurred the development of MSC-based therapies for treating a diverse array of nonskeletal diseases. Although genetic and experimental rodent models of disease represent important tools for developing efficacious MSC-based therapies, development of reliable methods to isolate MSCs from mouse bone marrow has been hampered by the unique biological properties of these cells. Indeed, few isolation schemes afford high yields and purity while maintaining the genomic integrity of cells. We recently demonstrated that mouse MSCs are highly sensitive to oxidative stress, and long-term expansion of these cells in atmospheric oxygen selects for immortalized clones that lack a functional p53 protein. Herein, we describe a protocol for the isolation of primary MSCs from mouse bone marrow that couples immunodepletion with culture in a low-oxygen environment and affords high purity and yield while preserving p53 function.
Collapse
Affiliation(s)
- Siddaraju V Boregowda
- Department of Molecular Therapeutics, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL, 33458, USA
| | - Veena Krishnappa
- Department of Molecular Therapeutics, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL, 33458, USA
| | - Donald G Phinney
- Department of Molecular Therapeutics, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL, 33458, USA.
| |
Collapse
|
273
|
A micro-architecturally biomimetic collagen template for mesenchymal condensation based cartilage regeneration. Acta Biomater 2016; 30:212-221. [PMID: 26602826 DOI: 10.1016/j.actbio.2015.11.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 11/12/2015] [Accepted: 11/16/2015] [Indexed: 11/23/2022]
Abstract
The unique arcade-like orientation of collagen fibers enables cartilage to bear mechanical loads. In this study continuous-length aligned collagen threads were woven to emulate the interdigitated arcade structure of the cartilage. The weaving pattern provided a macropore network within which micromass cell pellets were seeded to take advantage of mesenchymal condensation driven chondrogenesis. Compression tests showed that the baseline scaffold had a modulus of 0.83±0.39MPa at a porosity of 80%. The modulus of pellet seeded scaffolds increased by 60% to 1.33±0.37MPa after 28days of culture, converging to the modulus of the native cartilage. The scaffolds displayed duress under displacement controlled low-cycle fatigue at 15% strain amplitude such that load reduction stabilized at 8% after 4500 cycles of loading. The woven structure demonstrated a substantial elastic recoil where 40% mechanical strain was close to completely recovered following unloading. A robust chondrogenesis was observed as evidenced by positive staining for GAGs and type II collagen and aggrecan. Dimethyl methylene blue and sircol assays showed GAGs and collagen productions to increase from 3.36±1.24 and 31.46±3.22 at day 3 to 56.61±12.12 and 136.70±12.29μg/μg of DNA at day 28 of culture. This woven collagen scaffold holds a significant potential for cartilage regeneration with shorter in vitro culture periods due to functionally sufficient mechanical robustness at the baseline. In conclusion, the mimicry of cartilage's arcade architecture resulted in substantial improvement of mechanical function while enabling one of the first pellet delivery platforms enabled by a macroporous network. STATEMENT OF SIGNIFICANCE Mesenchymal condensation is critical for driving chondrogenesis, making high density cell seeding a standard in cartilage tissue engineering. Efforts to date have utilized scaffold free delivery of MSCs in pellet form. This study developed a macroporous scaffold that is fabricated by weaving highly aligned collagen threads. The scaffold can deliver high density cell condensates while providing mechanical stiffness comparable to that of cartilage. The scaffold also mimicked the arcade-like orientation of collagen fibers in cartilage. A highly robust chondrogenesis was observed in this mesenchymal cell pellet delivery system. Baseline mechanical robustness of this scaffold system will enable delivery of cell pellets as early as three days.
Collapse
|
274
|
Vu MQ, Der Sarkissian S, Borie M, Bessette PO, Noiseux N. Optimization of Mesenchymal Stem Cells to Increase Their Therapeutic Potential. Methods Mol Biol 2016; 1416:275-88. [PMID: 27236678 DOI: 10.1007/978-1-4939-3584-0_16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The heart which has limited renewal and regenerative capacity is a prime target for cellular therapy. Stem cell transplantation has emerged as a promising therapeutic strategy to improve healing of the ischemic heart, repopulate the injured myocardium, and restore cardiac function. However, clinical usefulness is impacted by the quality and quantity of delivered cells, the suboptimal manipulations prior to transplantation, and the general poor viability of the cells transferred particularly to an ischemic microenvironment. Focus is now on developing new ways to enhance stem cell renewal and survival capacity before transplant. This can be done by physical, chemical, pharmacological, or genetic manipulation of cells followed by accurate evaluation of conditioning methods by validated tests.This chapter covers the proper handling of mesenchymal stem cells (human and rat lines) and methodologies to evaluate efficacy and the translational potential of conditioning methods. Specifically, we will cover stem cell culture methods, preconditioning protocols, viability assessment in hypoxic and oxidative challenges as encountered in an ischemic microenvironment, and the proliferative capacity of cells.
Collapse
Affiliation(s)
- Minh Quan Vu
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
| | - Shant Der Sarkissian
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
| | - Melanie Borie
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | | | - Nicolas Noiseux
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada. .,Faculté de Médecine, Université de Montréal, Montreal, QC, Canada. .,Division of Cardiac Surgery, Centre Hospitalier de l'Université de Montréal (CHUM), 3840 Saint-Urbain Street, Montreal, QC, Canada, H2W1T8.
| |
Collapse
|
275
|
Activin Receptor-Like Kinase Receptors ALK5 and ALK1 Are Both Required for TGFβ-Induced Chondrogenic Differentiation of Human Bone Marrow-Derived Mesenchymal Stem Cells. PLoS One 2015; 10:e0146124. [PMID: 26720610 PMCID: PMC4697836 DOI: 10.1371/journal.pone.0146124] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 12/14/2015] [Indexed: 01/01/2023] Open
Abstract
Introduction Bone marrow-derived mesenchymal stem cells (BMSCs) are promising for cartilage regeneration because BMSCs can differentiate into cartilage tissue-producing chondrocytes. Transforming Growth Factor β (TGFβ) is crucial for inducing chondrogenic differentiation of BMSCs and is known to signal via Activin receptor-Like Kinase (ALK) receptors ALK5 and ALK1. Since the specific role of these two TGFβ receptors in chondrogenesis is unknown, we investigated whether ALK5 and ALK1 are expressed in BMSCs and whether both receptors are required for chondrogenic differentiation of BMSCs. Materials & Methods ALK5 and ALK1 gene expression in human BMSCs was determined with RT-qPCR. To induce chondrogenesis, human BMSCs were pellet-cultured in serum-free chondrogenic medium containing TGFβ1. Chondrogenesis was evaluated by aggrecan and collagen type IIα1 RT-qPCR analysis, and histological stainings of proteoglycans and collagen type II. To overexpress constitutively active (ca) receptors, BMSCs were transduced either with caALK5 or caALK1. Expression of ALK5 and ALK1 was downregulated by transducing BMSCs with shRNA against ALK5 or ALK1. Results ALK5 and ALK1 were expressed in in vitro-expanded as well as in pellet-cultured BMSCs from five donors, but mRNA levels of both TGFβ receptors did not clearly associate with chondrogenic induction. TGFβ increased ALK5 and decreased ALK1 gene expression in chondrogenically differentiating BMSC pellets. Neither caALK5 nor caALK1 overexpression induced cartilage matrix formation as efficient as that induced by TGFβ. Moreover, short hairpin-mediated downregulation of either ALK5 or ALK1 resulted in a strong inhibition of TGFβ-induced chondrogenesis. Conclusion ALK5 as well as ALK1 are required for TGFβ-induced chondrogenic differentiation of BMSCs, and TGFβ not only directly induces chondrogenesis, but also modulates ALK5 and ALK1 receptor signaling in BMSCs. These results imply that optimizing cartilage formation by mesenchymal stem cells will depend on activation of both receptors.
Collapse
|
276
|
Effect of Fibroblast Growth Factor 2 on Equine Synovial Fluid Chondroprogenitor Expansion and Chondrogenesis. Stem Cells Int 2015; 2016:9364974. [PMID: 26839571 PMCID: PMC4709790 DOI: 10.1155/2016/9364974] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 09/29/2015] [Accepted: 09/30/2015] [Indexed: 01/22/2023] Open
Abstract
Mesenchymal stem cells have been identified in the synovial fluid of several species. This study was conducted to characterize chondroprogenitor (CP) cells in equine synovial fluid (SF) and to determine the effect of fibroblast growth factor 2 (FGF-2) on SF-CP monolayer proliferation and subsequent chondrogenesis. We hypothesized that FGF-2 would stimulate SF-CP proliferation and postexpansion chondrogenesis. SF aspirates were collected from adult equine joints. Colony-forming unit (CFU) assays were performed during primary cultures. At first passage, SF-cells were seeded at low density, with or without FGF-2. Following monolayer expansion and serial immunophenotyping, cells were transferred to chondrogenic pellet cultures. Pellets were analyzed for chondrogenic mRNA expression and cartilage matrix secretion. There was a mean of 59.2 CFU/mL of SF. FGF-2 increased the number of population doublings during two monolayer passages and halved the population doubling times. FGF-2 did not alter the immunophenotype of SF-CPs during monolayer expansion, nor did FGF-2 compromise chondrogenesis. Hypertrophic phenotypic markers were not expressed in control or FGF-2 groups. FGF-2 did prevent the development of a “fibroblastic” cell layer around pellet periphery. FGF-2 significantly accelerates in vitro SF-CP expansion, the major hurdle to clinical application of this cell population, without detrimentally affecting subsequent chondrogenic capacity.
Collapse
|
277
|
Yang Z, Liu Q, Mannix RJ, Xu X, Li H, Ma Z, Ingber DE, Allen PD, Wang Y. Mononuclear cells from dedifferentiation of mouse myotubes display remarkable regenerative capability. Stem Cells 2015; 32:2492-501. [PMID: 24916688 DOI: 10.1002/stem.1742] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 03/26/2014] [Accepted: 04/04/2014] [Indexed: 02/06/2023]
Abstract
Certain lower organisms achieve organ regeneration by reverting differentiated cells into tissue-specific progenitors that re-enter embryonic programs. During muscle regeneration in the urodele amphibian, postmitotic multinucleated skeletal myofibers transform into mononucleated proliferating cells upon injury, and a transcription factor-msx1 plays a role in their reprograming. Whether this powerful regeneration strategy can be leveraged in mammals remains unknown, as it has not been demonstrated that the dedifferentiated progenitor cells arising from muscle cells overexpressing Msx1 are lineage-specific and possess the same potent regenerative capability as their amphibian counterparts. Here, we show that ectopic expression of Msx1 reprograms postmitotic, multinucleated, primary mouse myotubes to become proliferating mononuclear cells. These dedifferentiated cells reactivate genes expressed by embryonic muscle progenitor cells and generate only muscle tissue in vivo both in an ectopic location and inside existing muscle. More importantly, distinct from adult muscle satellite cells, these cells appear both to fuse with existing fibers and to regenerate myofibers in a robust and time-dependent manner. Upon transplantation into a degenerating muscle, these dedifferentiated cells generated a large number of myofibers that increased over time and replenished almost half of the cross-sectional area of the muscle in only 12 weeks. Our study demonstrates that mammals can harness a muscle regeneration strategy used by lower organisms when the same molecular pathway is activated.
Collapse
Affiliation(s)
- Zhong Yang
- College of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China; Department of Anesthesia Perioperative and Pain Medicine, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
278
|
Green JD, Tollemar V, Dougherty M, Yan Z, Yin L, Ye J, Collier Z, Mohammed MK, Haydon RC, Luu HH, Kang R, Lee MJ, Ho SH, He TC, Shi LL, Athiviraham A. Multifaceted signaling regulators of chondrogenesis: Implications in cartilage regeneration and tissue engineering. Genes Dis 2015; 2:307-327. [PMID: 26835506 PMCID: PMC4730920 DOI: 10.1016/j.gendis.2015.09.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/16/2015] [Indexed: 01/08/2023] Open
Abstract
Defects of articular cartilage present a unique clinical challenge due to its poor self-healing capacity and avascular nature. Current surgical treatment options do not ensure consistent regeneration of hyaline cartilage in favor of fibrous tissue. Here, we review the current understanding of the most important biological regulators of chondrogenesis and their interactions, to provide insight into potential applications for cartilage tissue engineering. These include various signaling pathways, including: fibroblast growth factors (FGFs), transforming growth factor β (TGF-β)/bone morphogenic proteins (BMPs), Wnt/β-catenin, Hedgehog, Notch, hypoxia, and angiogenic signaling pathways. Transcriptional and epigenetic regulation of chondrogenesis will also be discussed. Advances in our understanding of these signaling pathways have led to promising advances in cartilage regeneration and tissue engineering.
Collapse
Affiliation(s)
- Jordan D. Green
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Viktor Tollemar
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Mark Dougherty
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Zhengjian Yan
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Liangjun Yin
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jixing Ye
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- School of Bioengineering, Chongqing University, Chongqing, China
| | - Zachary Collier
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Maryam K. Mohammed
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Rex C. Haydon
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue H. Luu
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Richard Kang
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J. Lee
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Sherwin H. Ho
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Lewis L. Shi
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Aravind Athiviraham
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|
279
|
Gabler J, Ruetze M, Kynast KL, Grossner T, Diederichs S, Richter W. Stage-Specific miRs in Chondrocyte Maturation: Differentiation-Dependent and Hypertrophy-Related miR Clusters and the miR-181 Family. Tissue Eng Part A 2015; 21:2840-51. [DOI: 10.1089/ten.tea.2015.0352] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Jessica Gabler
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Martin Ruetze
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Katharina L. Kynast
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Tobias Grossner
- Department of Orthopaedic and Trauma Surgery, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Solvig Diederichs
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Wiltrud Richter
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
280
|
rAAV-mediated overexpression of sox9, TGF-β and IGF-I in minipig bone marrow aspirates to enhance the chondrogenic processes for cartilage repair. Gene Ther 2015; 23:247-55. [PMID: 26583804 DOI: 10.1038/gt.2015.106] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/13/2015] [Accepted: 10/19/2015] [Indexed: 12/24/2022]
Abstract
Administration of therapeutic gene sequences coding for chondrogenic and chondroreparative factors in bone marrow aspirates using the clinically adapted recombinant adeno-associated virus (rAAV) vector may provide convenient, single-step approaches to improve cartilage repair. Here, we tested the ability of distinct rAAV constructs coding for the potent SOX9, transforming growth factor beta (TGF-β) and insulin-like growth factor I (IGF-I) candidate factors to modify marrow aspirates from minipigs to offer a preclinical large animal model system adapted for a translational evaluation of cartilage repair upon transplantation in sites of injury. Our results demonstrate that high, prolonged rAAV gene transfer efficiencies were achieved in the aspirates (up to 100% for at least 21 days) allowing to produce elevated amounts of the transcription factor SOX9 that led to increased levels of matrix synthesis and chondrogenic differentiation and of the growth factors TGF-β and IGF-I that both increased cell proliferation, matrix synthesis and chondrogenic differentiation (although to a lower level than SOX9) compared with control (lacZ) condition. Remarkably, application of the candidate SOX9 vector also led to reduced levels of hypertrophic differentiation in the aspirates, possibly by modulating the β-catenin, Indian hedgehog and PTHrP pathways. The present findings show the benefits of modifying minipig marrow concentrates via rAAV gene transfer as a future means to develop practical strategies to promote cartilage repair in a large animal model.
Collapse
|
281
|
Abstract
Compared with traditional 2D adherent cell culture, 3D spheroidal cell aggregates, or spheroids, are regarded as more physiological, and this technique has been exploited in the field of oncology, stem cell biology, and tissue engineering. Mesenchymal stem cells (MSCs) cultured in spheroids have enhanced anti-inflammatory, angiogenic, and tissue reparative/regenerative effects with improved cell survival after transplantation. Cytoskeletal reorganization and drastic changes in cell morphology in MSC spheroids indicate a major difference in mechanophysical properties compared with 2D culture. Enhanced multidifferentiation potential, upregulated expression of pluripotency marker genes, and delayed replicative senescence indicate enhanced stemness in MSC spheroids. Furthermore, spheroid formation causes drastic changes in the gene expression profile of MSC in microarray analyses. In spite of these significant changes, underlying molecular mechanisms and signaling pathways triggering and sustaining these changes are largely unknown.
Collapse
|
282
|
Sato Y, Mera H, Takahashi D, Majima T, Iwasaki N, Wakitani S, Takagi M. Synergistic effect of ascorbic acid and collagen addition on the increase in type 2 collagen accumulation in cartilage-like MSC sheet. Cytotechnology 2015; 69:405-416. [PMID: 26572654 DOI: 10.1007/s10616-015-9924-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 10/13/2015] [Indexed: 11/29/2022] Open
Abstract
Aiming to increase the content of type 2 collagen in scaffold-free cartilage-like cell sheets prepared using human bone marrow mesenchymal stem cells, the effect of several kinds of additives in a chondrogenic medium was investigated. Addition of ascorbic acid 2 phosphate (VCP) at a high concentration (250 µg/ml) and type 1 atelocollagen (5 µg/ml) increased the accumulation of type 2 collagen by fourfold and twofold, respectively. On the other hand, an antioxidant, glutathione showed no such effect. The synergistic effect of VCP and type 1 atelocollagen resulted in an eightfold increase in the accumulation level of type 2 collagen. Furthermore, the gene expression level of type 2 collagen increased and that of matrix metalloproteinase-13 (MMP-13) decreased to approximately one-third of the control. The increase in type 2 collagen accumulation in the scaffold-free cartilage-like cell sheet might be due to not only the enhancement of the synthesis but also the suppression of the degradation of type 2 collagen by MMP-13.
Collapse
Affiliation(s)
- Yasushi Sato
- Division of Biotechnology and Macromolecular Chemistry, Graduate School of Engineering, Hokkaido University, Kita-ku, N13W8, Sapporo, 060-8628, Japan
| | - Hisashi Mera
- School of Health and Sports Sciences, Mukogawa Women's University, 6-46 Ikebiraki, Nishinomiya, Hyogo, 663-8558, Japan.,Foundation for Biomedical Research and Innovation, International Medical Device Alliance, 1-6-5, Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Daisuke Takahashi
- Department of Orthopaedic Surgery, Graduate School of Medicine, Hokkaido University, Kita-ku, N15W7, Sapporo, 060-8638, Japan
| | - Tokifumi Majima
- Department of Joint Replacement and Tissue Engineering, Graduate School of Medicine, Hokkaido University, Kita-ku, N15W7, Sapporo, 060-8638, Japan
| | - Norimasa Iwasaki
- Department of Orthopaedic Surgery, Graduate School of Medicine, Hokkaido University, Kita-ku, N15W7, Sapporo, 060-8638, Japan
| | - Shigeyuki Wakitani
- Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 739-8553, Japan
| | - Mutsumi Takagi
- Division of Biotechnology and Macromolecular Chemistry, Graduate School of Engineering, Hokkaido University, Kita-ku, N13W8, Sapporo, 060-8628, Japan.
| |
Collapse
|
283
|
Abstract
Among the surgical options for large full-thickness chondral injuries, cell-based therapy has been practiced and its satisfactory outcomes have been reported. One area that appears promising is cell-based therapies utilizing stem cells. Various tissues within the human body contain mesenchymal stem cells (MSCs) from where these can be harvested. These include bone marrow, adipose, synovium, peripheral blood, and umbilical cord. In this article, both preclinical animal studies and clinical studies dealing with the use of MSCs for cartilage repair of the knee are reviewed. Majority of the clinical papers have shown promising results; however, there are a limited number of studies of high evidence level. Clinical significance of the stem cell therapy as compared to other surgical options as well as optimization of the procedure in terms of cell type and delivery method is still to be determined.
Collapse
Affiliation(s)
- Shinichi Yoshiya
- Department of Orthopaedic Surgery, Hyogo College of Medicine, 1-1, Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan.
| | - Aman Dhawan
- Sports Medicine, Penn State Hershey Bone and Joint Institute, Hershey, PA, 17033-0850, USA.
| |
Collapse
|
284
|
Shao J, Zhang W, Yang T. Using mesenchymal stem cells as a therapy for bone regeneration and repairing. Biol Res 2015; 48:62. [PMID: 26530042 PMCID: PMC4630918 DOI: 10.1186/s40659-015-0053-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 10/22/2015] [Indexed: 02/07/2023] Open
Abstract
Bone is a unique tissue which could regenerate completely after injury rather than heal itself with a scar. Compared with other tissues the difference is that, during bone repairing and regeneration, after the inflammatory phase the mesenchymal stem cells (MSCs) are recruited to the injury site and differentiate into either chondroblasts or osteoblasts precursors, leading to bone repairing and regeneration. Besides these two precursors, the MSCs can also differentiate into adipocyte precursors, skeletal muscle precursors and some other mesodermal cells. With this multilineage potentiality, the MSCs are probably used to cure bone injury and other woundings in the near future. Here we will introduce the recent developments in understanding the mechanism of MSCs action in bone regeneration and repairing.
Collapse
Affiliation(s)
- Jin Shao
- Department of Orthopaedics, Shanghai Pudong New Area Gongli Hospital, Second Military Medical University, Shanghai, 200135, China.
| | - Weiwei Zhang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Tieyi Yang
- Department of Orthopaedics, Shanghai Pudong New Area Gongli Hospital, Second Military Medical University, Shanghai, 200135, China.
| |
Collapse
|
285
|
Leferink AM, Chng YC, van Blitterswijk CA, Moroni L. Distribution and Viability of Fetal and Adult Human Bone Marrow Stromal Cells in a Biaxial Rotating Vessel Bioreactor after Seeding on Polymeric 3D Additive Manufactured Scaffolds. Front Bioeng Biotechnol 2015; 3:169. [PMID: 26557644 PMCID: PMC4617101 DOI: 10.3389/fbioe.2015.00169] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/08/2015] [Indexed: 12/28/2022] Open
Abstract
One of the conventional approaches in tissue engineering is the use of scaffolds in combination with cells to obtain mechanically stable tissue constructs in vitro prior to implantation. Additive manufacturing by fused deposition modeling is a widely used technique to produce porous scaffolds with defined pore network, geometry, and therewith defined mechanical properties. Bone marrow-derived mesenchymal stromal cells (MSCs) are promising candidates for tissue engineering-based cell therapies due to their multipotent character. One of the hurdles to overcome when combining additive manufactured scaffolds with MSCs is the resulting heterogeneous cell distribution and limited cell proliferation capacity. In this study, we show that the use of a biaxial rotating bioreactor, after static culture of human fetal MSCs (hfMSCs) seeded on synthetic polymeric scaffolds, improved the homogeneity of cell and extracellular matrix distribution and increased the total cell number. Furthermore, we show that the relative mRNA expression levels of indicators for stemness and differentiation are not significantly changed upon this bioreactor culture, whereas static culture shows variations of several indicators for stemness and differentiation. The biaxial rotating bioreactor presented here offers a homogeneous distribution of hfMSCs, enabling studies on MSCs fate in additive manufactured scaffolds without inducing undesired differentiation.
Collapse
Affiliation(s)
- Anne M Leferink
- Department of Tissue Regeneration, MIRA Institute, University of Twente , Enschede , Netherlands ; Department of Complex Tissue Regeneration, Faculty of Health, Medicine and Life Sciences, Maastricht University , Maastricht , Netherlands
| | | | - Clemens A van Blitterswijk
- Department of Tissue Regeneration, MIRA Institute, University of Twente , Enschede , Netherlands ; Department of Complex Tissue Regeneration, Faculty of Health, Medicine and Life Sciences, Maastricht University , Maastricht , Netherlands
| | - Lorenzo Moroni
- Department of Tissue Regeneration, MIRA Institute, University of Twente , Enschede , Netherlands ; Department of Complex Tissue Regeneration, Faculty of Health, Medicine and Life Sciences, Maastricht University , Maastricht , Netherlands
| |
Collapse
|
286
|
Lee P, Tran K, Zhou G, Bedi A, Shelke NB, Yu X, Kumbar SG. Guided differentiation of bone marrow stromal cells on co-cultured cartilage and bone scaffolds. SOFT MATTER 2015; 11:7648-7655. [PMID: 26292727 DOI: 10.1039/c5sm01909e] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Focal chondral defects that result from traumatic injuries to the knee remain one of the most common causes of disability in patients. Current solutions for healing focal cartilage defects are mainly limited by the production of inferior cartilage-like tissue and subsequent delamination due to incomplete healing of the subchondral bone. In this experiment a polymeric osteochondral implant for guiding autologous bone marrow stem cells (BMSCs) to populate the scaffold to create distinctive bone and cartilage tissue is used. The cartilage component presents bioactive aligned nanofibers containing chondroitin sulfate and hyaluronic acid while the bone component includes hydroxyapatite to promote chondrogenic and osteogenic differentiation of the rat BMSCs in vitro. The different cartilage and bone components resulted in the elevated expression of osteogenic markers such as bone sialoprotein, runt related transcription factor 2, and bone morphogenetic protein 2 in the deeper bone layer and chondrogenic markers such as collagen type II and aggrecan in the cartilage layer. Through immunofluorescence imaging, the alignment of the secreted collagen type II fibrils and aggrecan was visualized and quantified on the cartilage component of the scaffold. These current studies show that the biodegradable biphasic osteochondral implant may be effective in promoting more hyaline-like tissue to fill in chondral defects of the knee.
Collapse
Affiliation(s)
- Paul Lee
- Department of Chemistry, Chemical Biology and Biomedical Engineering, Stevens Institute of Technology, Hoboken, USA.
| | | | | | | | | | | | | |
Collapse
|
287
|
Lee P, Tran K, Chang W, Fang YL, Zhou G, Junka R, Shelke NB, Yu X, Kumbar SG. Bioactive polymeric scaffolds for osteochondral tissue engineering: in vitro
evaluation of the effect of culture media on bone marrow stromal cells. POLYM ADVAN TECHNOL 2015. [DOI: 10.1002/pat.3680] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Paul Lee
- Department of Chemistry, Chemical Biology and Biomedical Engineering; Stevens Institute of Technology, 1 Castle Point on Hudson; Hoboken NJ 07030 USA
| | - Katelyn Tran
- Department of Chemistry, Chemical Biology and Biomedical Engineering; Stevens Institute of Technology, 1 Castle Point on Hudson; Hoboken NJ 07030 USA
| | - Wei Chang
- Department of Chemistry, Chemical Biology and Biomedical Engineering; Stevens Institute of Technology, 1 Castle Point on Hudson; Hoboken NJ 07030 USA
| | - Ya-Lin Fang
- Department of Chemistry, Chemical Biology and Biomedical Engineering; Stevens Institute of Technology, 1 Castle Point on Hudson; Hoboken NJ 07030 USA
| | - Gan Zhou
- Department of Chemistry, Chemical Biology and Biomedical Engineering; Stevens Institute of Technology, 1 Castle Point on Hudson; Hoboken NJ 07030 USA
| | - Radoslaw Junka
- Department of Chemistry, Chemical Biology and Biomedical Engineering; Stevens Institute of Technology, 1 Castle Point on Hudson; Hoboken NJ 07030 USA
| | - Namdev B. Shelke
- Department of Orthopaedic Surgery; UConn Health; Farmington CT 06030 USA
- Institute for Regenerative Engineering; UConn Health; Farmington CT 06030 USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences; UConn Health; Farmington CT 06030 USA
| | - Xiaojun Yu
- Department of Chemistry, Chemical Biology and Biomedical Engineering; Stevens Institute of Technology, 1 Castle Point on Hudson; Hoboken NJ 07030 USA
| | - Sangamesh G. Kumbar
- Department of Orthopaedic Surgery; UConn Health; Farmington CT 06030 USA
- Institute for Regenerative Engineering; UConn Health; Farmington CT 06030 USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences; UConn Health; Farmington CT 06030 USA
- Department of Biomedical Engineering; University of Connecticut; Storrs CT 06269 USA
| |
Collapse
|
288
|
LeBlon CE, Casey ME, Fodor CR, Zhang T, Zhang X, Jedlicka SS. Correlation between in vitro expansion-related cell stiffening and differentiation potential of human mesenchymal stem cells. Differentiation 2015; 90:1-15. [PMID: 26381795 DOI: 10.1016/j.diff.2015.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 08/10/2015] [Accepted: 08/20/2015] [Indexed: 12/28/2022]
Abstract
Human mesenchymal stem cells (hMSCs) are an attractive cell source for tissue regeneration, given their self-renewal and multilineage potential. However, they are present in only small percentages in human bone marrow, and are generally propagated in vitro prior to downstream use. Previous work has shown that hMSC propagation can lead to alterations in cell behavior and differentiation potency, yet optimization of differentiation based on starting cell elastic modulus is an area still under investigation. To further advance the knowledge in this field, hMSCs were cultured and routinely passaged on tissue-culture polystyrene to investigate the correlation between cell stiffening and differentiation potency during in vitro aging. Local cell elastic modulus was measured at every passage using atomic force microscopy indentation. At each passage, cells were induced to differentiate down myogenic and osteogenic paths. Cells induced to differentiate, as well as undifferentiated cells were assessed for gene and protein expression using quantitative polymerase chain reaction and immunofluorescent staining, respectively, for osteogenic and myogenic markers. Myogenic and osteogenic cell potential are highly reliant on the elastic modulus of the starting cell population (of undifferentiated cells), and this potential appears to peak when the innate cell elastic modulus is close to that of differentiated tissue. However, the latent expression of the same markers in undifferentiated cells also appears to undergo a correlative relationship with cell elastic modulus, indicating some endogenous effects of cell elastic modulus and gene/protein expression. Overall, this study correlates age-related changes with regards to innate cell stiffening and gene/protein expression in commercial hMSCs, providing some guidance as to maintenance and future use of hMSCs in future tissue engineering applications.
Collapse
Affiliation(s)
- Courtney E LeBlon
- Mechanical Engineering & Mechanics, Packard Laboratory, Lehigh University, 19 Memorial Drive, Bethlehem, PA 18015, United States
| | - Meghan E Casey
- Bioengineering Program, Lehigh University, 111 Research Drive, Iacocca Hall, Bethlehem, PA 18015, United States
| | - Caitlin R Fodor
- Bioengineering Program, Lehigh University, 111 Research Drive, Iacocca Hall, Bethlehem, PA 18015, United States
| | - Tony Zhang
- Bioengineering Program, Lehigh University, 111 Research Drive, Iacocca Hall, Bethlehem, PA 18015, United States
| | - Xiaohui Zhang
- Mechanical Engineering & Mechanics, Packard Laboratory, Lehigh University, 19 Memorial Drive, Bethlehem, PA 18015, United States; Bioengineering Program, Lehigh University, 111 Research Drive, Iacocca Hall, Bethlehem, PA 18015, United States
| | - Sabrina S Jedlicka
- Bioengineering Program, Lehigh University, 111 Research Drive, Iacocca Hall, Bethlehem, PA 18015, United States; Materials Science and Engineering, Whitaker Laboratory, Lehigh University, 5 East Packer Ave., Bethlehem, PA 18015, United States; Center for Advanced Materials & Nanotechnology, Whitaker Laboratory, Lehigh University, 5 East Packer Ave., Bethlehem, PA 18015, United States.
| |
Collapse
|
289
|
Beckmann R, Lippross S, Hartz C, Tohidnezhad M, Ferreira MSV, Neuss-Stein S, Seekamp A, Nebelung S, Kweider N, Rath B, Jahr H, Pufe T, Varoga DJ. Abrasion arthroplasty increases mesenchymal stem cell content of postoperative joint effusions. BMC Musculoskelet Disord 2015; 16:250. [PMID: 26364138 PMCID: PMC4567829 DOI: 10.1186/s12891-015-0705-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 09/02/2015] [Indexed: 01/15/2023] Open
Abstract
Background Abrasion arthroplasty (AAP) is a procedure by which intrinsic cartilage healing is believed to be stimulated. Although clinically accepted for degenerative and traumatic cartilage lesions scientific evidence at a molecular level that proves the effect of AAP is scarce. Method Mononuclear cells were extracted from postoperative joint effusions 21.5 h post AAP and simple debridement of cartilage lesions. Luminex, ELISA and FACS experiments were performed. Immunohistochemical stainings of cell cultures for cartilage markers were used to confirm the findings. Results Postoperative joint effusions after AAP showed increased contents of Mononuclear cells compared to Arthroscopic Chondroplasty (ACP). BMP-4 and IGF were increased in AAP as complared to ACP. Mononuclear cells isolated after AAP express the MSC markers CD 73, CD 105, CD 90, CD 44 and are CD34 negative. Chondrogenic differentiation was demonstrated by positive staining for Sox9, collagen II, proteoglycan, chondroitin-4-sulfate. Conclusion Our results support the clinical application of AAP as a procedure that enhances cartilage repair as an alternative to far more complex procedures that have gained popularity. Furthermore the data presented supports clinical investigations that recommend not to use suction drainage as by this procedure a considerable amount of the regeneratory potential of postoperative joint effusions might be extracted.
Collapse
Affiliation(s)
- Rainer Beckmann
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| | | | - Claudia Hartz
- Department of Trauma and Orthopaedics, University Hospital Heidelberg, Bergheim, Germany.
| | - Mersedeh Tohidnezhad
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| | - Mónica S Ventura Ferreira
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen, Aachen, Germany.
| | - Sabine Neuss-Stein
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen, Aachen, Germany.
| | - Andreas Seekamp
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| | - Sven Nebelung
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany. .,Department of Orthopaedic Surgery, RWTH Aachen University, Aachen, Germany.
| | - Nisreen Kweider
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| | - Björn Rath
- Department of Orthopaedic Surgery, RWTH Aachen University, Aachen, Germany.
| | - Holger Jahr
- Department of Orthopaedic Surgery, RWTH Aachen University, Aachen, Germany.
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| | - Deike J Varoga
- Department of Trauma, University Hospital of Kiel, Kiel, Germany.
| |
Collapse
|
290
|
Growth factor directed chondrogenic differentiation of porcine bone marrow-derived progenitor cells. J Craniofac Surg 2015; 24:1026-30. [PMID: 23714939 DOI: 10.1097/scs.0b013e31827ff323] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Despite advances in surgical technique, reconstruction of a mandibular condyle still causes significant donor-site morbidity. The purpose of this study was to compare the effect of 3 different growth factors and define optimal cell culture conditions for bone marrow-derived progenitor cells to differentiate into chondrocytes for mandibular condyle reconstruction. METHODS Porcine bone marrow-derived progenitor cells (pBMPCs) were cultured as a pellet for 2, 3, and 4 weeks under the following conditions: group 1, TGF-β3 + standard medium; group 2, TGF-β3 + BMP-2 + standard medium; group 3, TGF-β3 + IGF-1 + standard medium; and group 4, TGF-β3 + BMP-2 + IGF-1 + standard medium. Chondrogenic differentiation was evaluated using 3 lineage differentiation markers. RESULTS The mean type II collagen positive area increased over weeks 2, 3, and 4 in group 4 compared to all the other groups (ANOVA; P = 0.005). At week 4, there was significantly greater type II collagen production in group 4 compared to all the other groups (ANOVA; P = 0.003). The medium in group 4 produces the greatest amount of cartilage when compared to groups 1, 2, and 3, and that 4 weeks produces the greatest amount of type II collagen. CONCLUSIONS The results of this study indicate that the most efficacious medium for chondrogenic differentiation of pBMPCs was group 4 medium and the most type II collagen was produced at 4 weeks.
Collapse
|
291
|
Mechano-growth factor enhances differentiation of bone marrow-derived mesenchymal stem cells. Biotechnol Lett 2015; 37:2341-8. [DOI: 10.1007/s10529-015-1915-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 07/13/2015] [Indexed: 11/26/2022]
|
292
|
Lin HA, Gupta MS, Varma DM, Gilchrist ML, Nicoll SB. Lower crosslinking density enhances functional nucleus pulposus-like matrix elaboration by human mesenchymal stem cells in carboxymethylcellulose hydrogels. J Biomed Mater Res A 2015; 104:165-77. [DOI: 10.1002/jbm.a.35552] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 07/20/2015] [Accepted: 08/07/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Huizi A. Lin
- Department of Biomedical Engineering; The City College of New York; New York New York
| | - Michelle S. Gupta
- Department of Biomedical Engineering; The City College of New York; New York New York
| | - Devika M. Varma
- Department of Biomedical Engineering; The City College of New York; New York New York
| | - M. Lane Gilchrist
- Department of Chemical Engineering; The City College of New York; New York City New York
| | - Steven B. Nicoll
- Department of Biomedical Engineering; The City College of New York; New York New York
| |
Collapse
|
293
|
da Silva Meirelles L, Malta TM, de Deus Wagatsuma VM, Palma PVB, Araújo AG, Ribeiro Malmegrim KC, Morato de Oliveira F, Panepucci RA, Silva WA, Kashima Haddad S, Covas DT. Cultured Human Adipose Tissue Pericytes and Mesenchymal Stromal Cells Display a Very Similar Gene Expression Profile. Stem Cells Dev 2015; 24:2822-40. [PMID: 26192741 DOI: 10.1089/scd.2015.0153] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are cultured cells that can give rise to mature mesenchymal cells under appropriate conditions and secrete a number of biologically relevant molecules that may play an important role in regenerative medicine. Evidence indicates that pericytes (PCs) correspond to mesenchymal stem cells in vivo and can give rise to MSCs when cultured, but a comparison between the gene expression profiles of cultured PCs (cPCs) and MSCs is lacking. We have devised a novel methodology to isolate PCs from human adipose tissue and compared cPCs to MSCs obtained through traditional methods. Freshly isolated PCs expressed CD34, CD140b, and CD271 on their surface, but not CD146. Both MSCs and cPCs were able to differentiate along mesenchymal pathways in vitro, displayed an essentially identical surface immunophenotype, and exhibited the ability to suppress CD3(+) lymphocyte proliferation in vitro. Microarray expression data of cPCs and MSCs formed a single cluster among other cell types. Further analyses showed that the gene expression profiles of cPCs and MSCs are extremely similar, although MSCs differentially expressed endothelial cell (EC)-specific transcripts. These results confirm, using the power of transcriptomic analysis, that PCs give rise to MSCs and suggest that low levels of ECs may persist in MSC cultures established using traditional protocols.
Collapse
Affiliation(s)
- Lindolfo da Silva Meirelles
- 1 Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil .,2 Laboratory for Stem Cells and Tissue Engineering, PPGBioSaúde, Lutheran University of Brazil , Canoas, Brazil
| | - Tathiane Maistro Malta
- 1 Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil
| | - Virgínia Mara de Deus Wagatsuma
- 1 Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil
| | - Patrícia Viana Bonini Palma
- 1 Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil
| | - Amélia Goes Araújo
- 3 Laboratory of Large-Scale Functional Biology (LLSFBio), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil
| | | | - Fábio Morato de Oliveira
- 1 Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil
| | - Rodrigo Alexandre Panepucci
- 3 Laboratory of Large-Scale Functional Biology (LLSFBio), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil
| | - Wilson Araújo Silva
- 1 Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil .,5 Department of Genetics, School of Medicine of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil
| | - Simone Kashima Haddad
- 1 Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil
| | - Dimas Tadeu Covas
- 1 Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil .,6 Department of Clinical Medicine, School of Medicine of Ribeirão Preto, University of São Paulo , Ribeirão Preto, Brazil
| |
Collapse
|
294
|
Laks M, Freitas-Filho LG, Sayeg K, Leite MTC, Teixeira LC. Penile reconstruction using mesenchymal stem cells. Acta Cir Bras 2015; 30:529-36. [DOI: 10.1590/s0102-865020150080000003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
295
|
Lu Y, Liu J, Liu Y, Qin Y, Luo Q, Wang Q, Duan H. TLR4 plays a crucial role in MSC-induced inhibition of NK cell function. Biochem Biophys Res Commun 2015; 464:541-7. [DOI: 10.1016/j.bbrc.2015.07.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 07/01/2015] [Indexed: 12/30/2022]
|
296
|
Daley EL, Coleman RM, Stegemann JP. Biomimetic microbeads containing a chondroitin sulfate/chitosan polyelectrolyte complex for cell-based cartilage therapy. J Mater Chem B 2015; 3:7920-7929. [PMID: 26693016 DOI: 10.1039/c5tb00934k] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Articular cartilage has a limited healing capacity that complicates the treatment of joint injuries and osteoarthritis. Newer repair strategies have focused on the use of cells and biomaterials to promote cartilage regeneration. In the present study, we developed and characterized bioinspired materials designed to mimic the composition of the cartilage extracellular matrix. Chondroitin sulfate (CS) and chitosan (CH) were used to form physically cross-linked macromolecular polyelectrolyte complexes (PEC) without the use of additional crosslinkers. A single-step water-in-oil emulsification process was used to either directly embed mesenchymal stem cells (MSC) in PEC particles created with a various concentrations of CS and CH, or to co-embed MSC with PEC in agarose-based microbeads. Direct embedding of MSC in PEC resulted in high cell viability but irregular and large particles. Co-embedding of PEC particles with MSC in agarose (Ag) resulted in uniform microbeads 80-90 μm in diameter that maintained high cell viability over three weeks in culture. Increased serum content resulted in more uniform PEC distribution within the microbead matrix, and both high and low CS:CH ratios resulted in more homogeneous microbeads than 1:1 formulations. Under chondrogenic conditions, expression of sulfated GAG and collagen type II was increased in 10:1 CS:CH PEC-Ag microbeads compared to pure Ag beads, indicating a chondrogenic influence of the PEC component. Such PEC-Ag microbeads may have utility in the directed differentiation and delivery of progenitor cell populations for cartilage repair.
Collapse
Affiliation(s)
- Ethan Lh Daley
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Rhima M Coleman
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Jan P Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
297
|
Pustlauk W, Paul B, Brueggemeier S, Gelinsky M, Bernhardt A. Modulation of chondrogenic differentiation of human mesenchymal stem cells in jellyfish collagen scaffolds by cell density and culture medium. J Tissue Eng Regen Med 2015; 11:1710-1722. [PMID: 26178016 DOI: 10.1002/term.2065] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 05/05/2015] [Accepted: 06/04/2015] [Indexed: 12/17/2022]
Abstract
Studies on tissue-engineering approaches for the regeneration of traumatized cartilage focus increasingly on multipotent human mesenchymal stem cells (hMSCs) as an alternative to autologous chondrocytes. The present study applied porous scaffolds made of collagen from the jellyfish Rhopilema esculentum for the in vitro chondrogenic differentiation of hMSCs. Culture conditions in those scaffolds differ from conditions in high-density pellet cultures, making a re-examination of these data necessary. We systematically investigated the influence of seeding density, basic culture media [Dulbecco's modified Eagle's medium (DMEM), α-minimum essential medium (α-MEM)] with varying glucose content and supplementation with fetal calf serum (FCS) or bovine serum albumin (BSA) on the chondrogenic differentiation of hMSCs. Gene expression analyses of selected markers for chondrogenic differentiation and hypertrophic development were conducted. Furthermore, the production of cartilage extracellular matrix (ECM) was analysed by quantification of sulphated glycosaminoglycan and collagen type II contents. The strongest upregulation of chondrogenic markers, along with the highest ECM deposition was observed in scaffolds seeded with 2.4 × 106 cells/cm3 after cultivation in high-glucose DMEM and 0.125% BSA. Lower seeding densities compared to high-density pellet cultures were sufficient to induce in vitro chondrogenic differentiation of hMSCs in collagen scaffolds, which reduces the amount of cells required for the seeding of scaffolds and thus the monolayer expansion period. Furthermore, examination of the impact of FCS and α-MEM on chondrogenic MSC differentiation is an important prerequisite for the development of an osteochondral medium for simultaneous osteogenic and chondrogenic differentiation in biphasic scaffolds for osteochondral tissue regeneration. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- W Pustlauk
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital, and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| | - B Paul
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital, and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| | - S Brueggemeier
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital, and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| | - M Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital, and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| | - A Bernhardt
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital, and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| |
Collapse
|
298
|
Fensky F, Reichert JC, Traube A, Rackwitz L, Siebenlist S, Nöth U. Chondrogenic predifferentiation of human mesenchymal stem cells in collagen type I hydrogels. ACTA ACUST UNITED AC 2015; 59:375-83. [PMID: 24803605 DOI: 10.1515/bmt-2013-0076] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 04/02/2014] [Indexed: 11/15/2022]
Abstract
Hyaline cartilage displays a limited regenerative potential. Consequently, therapeutic approaches have been developed to treat focal cartilage lesions. Larger-sized lesions are commonly treated by osteochondral grafting/mosaicplasty, autologous chondrocyte implantation (ACI) or matrix-induced chondrocyte implantation (MACI). As an alternative cell source to chondrocytes, multipotent mesenchymal stem cells (MSCs) are regarded a promising option. We therefore investigated the feasibility of pre-differentiating human MSCs incorporated in hydrogels clinically applied for MACI (CaReS®). MSC-laden hydrogels were cast and cultured over 10 days in a defined chondrogenic differentiation medium supplemented with TGF-β1. This was followed by an 11-day culture in TGF-β1 free media. After 21 days, considerable contraction of the hydrogels was observed. Histochemistry showed cells of a chondrocyte-like morphology embedded in a proteoglycan-rich extracellular matrix. Real-time polymerase chain reaction (RT-PCR) analysis showed the expression of chondrogenic marker genes, such as collagen type II and aggrecan. In summary, we demonstrate that chondrogenic differentiation of human mesenchymal stem cells embedded in collagen type I hydrogels can be induced under the influence of TGF-β1 over a period of 10 days.
Collapse
|
299
|
Repair of ear cartilage defects with allogenic bone marrow mesenchymal stem cells in rabbits. Cell Biochem Biophys 2015; 70:1137-43. [PMID: 24879616 DOI: 10.1007/s12013-014-0033-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The study aims to investigate the feasibility of repairing cartilaginous defects with chondrocytes induced from allogenic bone marrow mesenchymal stem cells (BMMSC) in rabbits' ear. BMMSCs were isolated and purified from New Zealand rabbits, in vitro amplified, and cultured in chondrocyte induction medium in order to acquire chondrocytes. After 3 weeks of induction, their phenotypes were confirmed as chondrocytes, then they were implanted onto novel polymeric scaffolds made from Poly (dl-lactide-co-glycolide) (PLGA) embedded with chitosan nonwoven cloth. The experimental group was transplanted with tissue engineering cartilaginous grafts composed of chondrogenetic BMMSC/scaffolds; the scaffold group was treated with scaffolds without cells, while in the control group, nothing was implanted. Specimens were taken at 6, 12, and 18 weeks after implantation, and the healing condition was observed by hematoxylin-eosin staining and toluidine blue staining. The right and left ears with cartilage defects of eighteen rabbits were randomly divided into three groups. In the experimental group, after 18 weeks of transplantation, the gross observation indicated that the cartilaginous defects were completely repaired by chondrocytes with smooth surface and similar color with the surrounding tissue. Hematoxylin-eosin staining and toluidine blue staining suggested that the defective area was filled with mature cartilage cells with obvious lacunae but without obvious boundaries with the normal cartilage tissue, and that the new cartilage cells were evenly distributed with homogeneously dyed cytoplasm and smaller in size. The chondrocyte induced from allogenic BMMSC can be used to repair cartilage defects in rabbit's ear.
Collapse
|
300
|
Millan C, Cavalli E, Groth T, Maniura-Weber K, Zenobi-Wong M. Engineered Microtissues Formed by Schiff Base Crosslinking Restore the Chondrogenic Potential of Aged Mesenchymal Stem Cells. Adv Healthc Mater 2015; 4:1348-58. [PMID: 25866187 DOI: 10.1002/adhm.201500102] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 03/12/2015] [Indexed: 01/09/2023]
Abstract
A universal method for reproducibly directing stem cell differentiation remains a major challenge for clinical applications involving cell-based therapies. The standard approach for chondrogenic induction by micromass pellet culture is highly susceptible to interdonor variability. A novel method for the fabrication of condensation-like engineered microtissues (EMTs) that utilizes hydrophilic polysaccharides to induce cell aggregation is reported here. Chondrogenesis of mesenchymal stem cells (MSCs) in EMTs is significantly enhanced compared to micromass pellets made by centrifugation measured by type II collagen gene expression, dimethylmethylene blue assay, and histology. MSCs from aged donors that fail to differentiate in pellet culture are successfully induced to synthesize cartilage-specific matrix in EMTs under identical media conditions. Furthermore, the EMT polysaccharides support the loading and release of the chondroinduction factor transforming growth factor β3 (TGF-β3). TGF-β-loaded EMTs (EMT(+TGF) ) facilitate cartilaginous tissue formation during culture in media not supplemented with the growth factor. The clinical potential of this approach is demonstrated in an explant defect model where EMT(+TGF) from aged MSCs synthesize de novo tissue containing sulfated glycosaminoglycans and type II collagen in situ.
Collapse
Affiliation(s)
- Christopher Millan
- Cartilage Engineering + Regeneration Laboratory; ETH Zürich; Otto-Stern-Weg 7 8093 Zürich Switzerland
| | - Emma Cavalli
- Cartilage Engineering + Regeneration Laboratory; ETH Zürich; Otto-Stern-Weg 7 8093 Zürich Switzerland
| | - Thomas Groth
- Biomedical Materials Group; Martin Luther University Halle-Wittenberg; Heinrich-Damerow-Strasse 4 06120 Halle (Saale) Germany
| | - Katharina Maniura-Weber
- Laboratory for Biointerfaces, Empa; Swiss Federal Laboratories for Materials Science and Technology; Lerchenfeldstrasse 5 9014 St. Gallen Switzerland
| | - Marcy Zenobi-Wong
- Cartilage Engineering + Regeneration Laboratory; ETH Zürich; Otto-Stern-Weg 7 8093 Zürich Switzerland
| |
Collapse
|