301
|
Abstract
Despite substantial clinical advances over the past 65 years, cardiovascular disease remains the leading cause of death in America. The past 15 years has witnessed major basic and translational interest in the use of stem and precursor cells as a therapeutic agent for chronically injured organs. Among the cell types under investigation, adult mesenchymal stem cells are widely studied, and in early stage, clinical studies show promise for repair and regeneration of cardiac tissues. The ability of mesenchymal stem cells to differentiate into mesoderm- and nonmesoderm-derived tissues, their immunomodulatory effects, their availability, and their key role in maintaining and replenishing endogenous stem cell niches have rendered them one of the most heavily investigated and clinically tested type of stem cell. Accumulating data from preclinical and early phase clinical trials document their safety when delivered as either autologous or allogeneic forms in a range of cardiovascular diseases, but also importantly define parameters of clinical efficacy that justify further investigation in larger clinical trials. Here, we review the biology of mesenchymal stem cells, their interaction with endogenous molecular and cellular pathways, and their modulation of immune responses. Additionally, we discuss factors that enhance their proliferative and regenerative ability and factors that may hinder their effectiveness in the clinical setting.
Collapse
Affiliation(s)
- Vasileios Karantalis
- From the University of Miami Miller School of Medicine, Interdisciplinary Stem Cell Institute, FL
| | - Joshua M Hare
- From the University of Miami Miller School of Medicine, Interdisciplinary Stem Cell Institute, FL.
| |
Collapse
|
302
|
Hu P, Pu Y, Li X, Zhu Z, Zhao Y, Guan W, Ma Y. Isolation, in vitro culture and identification of a new type of mesenchymal stem cell derived from fetal bovine lung tissues. Mol Med Rep 2015; 12:3331-3338. [PMID: 26016556 PMCID: PMC4526039 DOI: 10.3892/mmr.2015.3854] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 04/30/2015] [Indexed: 01/08/2023] Open
Abstract
Lung‑derived mesenchymal stem cells (LMSCs) are considered to be important in lung tissue repair and regenerative processes. However, the biological characteristics and differentiation potential of LMSCs remain to be elucidated. In the present study, fetal lung‑derived mesenchymal stem cells (FLMSCs) were isolated from fetal bovine lung tissues by collagenase digestion. The in vitro culture conditions were optimized and stabilized and the self‑renewal ability and differentiation potential were evaluated. The results demonstrated that the FLMSCs were morphologically consistent with fibroblasts, were able to be cultured and passaged for at least 33 passages and the cell morphology and proliferative ability were stable during the first 10 passages. In addition, FLMSCs were found to express CD29, CD44, CD73 and CD166, however, they did not express hematopoietic cell specific markers, including CD34, CD45 and BOLA‑DRα. The growth kinetics of FLMSCs consisted of a lag phase, a logarithmic phase and a plateau phase, and as the passages increased, the proliferative ability of cells gradually decreased. The majority of FLMSCs were in G0/G1 phase. Following osteogenic induction, FLMSCs were positive for the expression of osteopontin and collagen type I α2. Following neurogenic differentiation, the cells were morphologically consistent with neuronal cells and positive for microtubule‑associated protein 2 and nestin expression. It was concluded that the isolated FLMSCs exhibited typical characteristics of mesenchymal stem cells and that the culture conditions were suitable for their proliferation and the maintenance of stemness. The present study illustrated the potential application of lung tissue as an adult stem cell source for regenerative therapies.
Collapse
Affiliation(s)
- Pengfei Hu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Yabin Pu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Xiayun Li
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Zhiqiang Zhu
- Harbin Institute of Physical Education, Harbin, Heilongjiang 150008, P.R. China
| | - Yuhua Zhao
- Harbin Institute of Physical Education, Harbin, Heilongjiang 150008, P.R. China
| | - Weijun Guan
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Yuehui Ma
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| |
Collapse
|
303
|
Antoniou J, Wang HT, Hadjab I, Aldebeyan S, Alaqeel MA, Meij BP, Tryfonidou MA, Mwale F. The Effects of Naproxen on Chondrogenesis of Human Mesenchymal Stem Cells. Tissue Eng Part A 2015; 21:2136-46. [PMID: 25873236 DOI: 10.1089/ten.tea.2014.0668] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Currently, there are no established treatments to prevent, stop, or even retard the degeneration of articular cartilage in osteoarthritis (OA). Biological repair of the degenerating articular cartilage would be preferable to surgery. There is no benign site where autologous chondrocytes can be harvested and used as a cell source for cartilage repair, leaving mesenchymal stem cells (MSCs) as an attractive option. However, MSCs from OA patients have been shown to constitutively express collagen type X (COL-X), a marker of late-stage chondrocyte hypertrophy. We recently found that naproxen (Npx), but not other nonsteroidal anti-inflammatory drugs, can induce collagen type X alpha 1 (COL10A1) gene expression in bone marrow-derived MSCs from healthy and OA donors. In this study, we determined the effect of Npx on COL10A1 expression and investigated the intracellular signaling pathways that mediate such effect in normal human MSCs during chondrogenesis. MSCs were cultured in standard chondrogenic differentiation media supplemented with or without Npx. Our results show that Npx can regulate chondrogenic differentiation by affecting the gene expression of both Indian hedgehog and parathyroid hormone/parathyroid hormone-related protein signaling pathways in a time-dependent manner, suggesting a complex interaction of different signaling pathways during the process.
Collapse
Affiliation(s)
- John Antoniou
- 1 Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec, Canada .,2 Division of Orthopedic Surgery, McGill University , Montreal, Quebec, Canada
| | - Hong Tian Wang
- 1 Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec, Canada
| | - Insaf Hadjab
- 1 Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec, Canada .,3 École Polytechnique , Montreal, Quebec, Canada
| | - Sultan Aldebeyan
- 1 Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec, Canada .,4 Department of Orthopaedic Surgery, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Motaz A Alaqeel
- 1 Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec, Canada .,5 Department of Orthopedics, College of Medicine, King Saud University , Riyadh, Saudi Arabia
| | - Björn P Meij
- 6 Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University , Utrecht, The Netherlands
| | - Marianna A Tryfonidou
- 6 Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University , Utrecht, The Netherlands
| | - Fackson Mwale
- 1 Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec, Canada .,2 Division of Orthopedic Surgery, McGill University , Montreal, Quebec, Canada
| |
Collapse
|
304
|
Increased levels of p21((CIP1/WAF1)) correlate with decreased chondrogenic differentiation potential in synovial membrane progenitor cells. Mech Ageing Dev 2015; 149:31-40. [PMID: 25987237 DOI: 10.1016/j.mad.2015.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 04/27/2015] [Accepted: 05/06/2015] [Indexed: 12/20/2022]
Abstract
Cartilage injuries are a major concern in the field of orthopedics. They occur following trauma, as well as from a variety of pathological conditions including Osteoarthritis (OA). Although cartilage does not exhibit robust endogenous repair, it has been demonstrated that modulating the activity of p21 can increase the regenerative abilities of cartilage in vitro and in vivo. Since the synovial membrane is abundant with mesenchymal progenitor cells (MPCs) capable of differentiating into cartilage both in vitro and in vivo, we examined if p21 expression levels varied between MPCs derived from normal vs. OA knee joints. Analysis of p21 at the mRNA and protein levels within normal and OA MPCs demonstrated differential levels of expression between these two groups, with OA MPCs having higher p21 expression levels. The higher levels of p21 in OA MPCs are also correlated with a decreased chondrogenic differentiation capacity and synovial inflammation, however, there was no evidence of senescence in the OA cells. The results of this study suggest that cell cycle regulation in MPCs may be altered in OA and that modulation of this pathway may have therapeutic potential once the mechanism by which this regulates stem/progenitor cells is better understood.
Collapse
|
305
|
Generation of articular chondrocytes from human pluripotent stem cells. Nat Biotechnol 2015; 33:638-45. [PMID: 25961409 DOI: 10.1038/nbt.3210] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Accepted: 03/25/2015] [Indexed: 12/19/2022]
Abstract
The replacement of articular cartilage through transplantation of chondrogenic cells or preformed cartilage tissue represents a potential new avenue for the treatment of degenerative joint diseases. Although many studies have described differentiation of human pluripotent stem cells (hPSCs) to the chondrogenic lineage, the generation of chondrocytes able to produce stable articular cartilage in vivo has not been demonstrated. Here we show that activation of the TGFβ pathway in hPSC-derived chondrogenic progenitors promotes the efficient development of articular chondrocytes that can form stable cartilage tissue in vitro and in vivo. In contrast, chondrocytes specified by BMP4 signaling display characteristics of hypertrophy and give rise to cartilage tissues that initiate the endochondral ossification process in vivo. These findings provide a simple serum-free and efficient approach for the routine generation of hPSC-derived articular chondrocytes for modeling diseases of the joint and developing cell therapy approaches to treat them.
Collapse
|
306
|
Santos JM, Camões SP, Filipe E, Cipriano M, Barcia RN, Filipe M, Teixeira M, Simões S, Gaspar M, Mosqueira D, Nascimento DS, Pinto-do-Ó P, Cruz P, Cruz H, Castro M, Miranda JP. Three-dimensional spheroid cell culture of umbilical cord tissue-derived mesenchymal stromal cells leads to enhanced paracrine induction of wound healing. Stem Cell Res Ther 2015; 6:90. [PMID: 25956381 PMCID: PMC4448539 DOI: 10.1186/s13287-015-0082-5] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 01/19/2015] [Accepted: 04/21/2015] [Indexed: 12/20/2022] Open
Abstract
Introduction The secretion of trophic factors by mesenchymal stromal cells has gained increased interest given the benefits it may bring to the treatment of a variety of traumatic injuries such as skin wounds. Herein, we report on a three-dimensional culture-based method to improve the paracrine activity of a specific population of umbilical cord tissue-derived mesenchymal stromal cells (UCX®) towards the application of conditioned medium for the treatment of cutaneous wounds. Methods A UCX® three-dimensional culture model was developed and characterized with respect to spheroid formation, cell phenotype and cell viability. The secretion by UCX® spheroids of extracellular matrix proteins and trophic factors involved in the wound-healing process was analysed. The skin regenerative potential of UCX® three-dimensional culture-derived conditioned medium (CM3D) was also assessed in vitro and in vivo against UCX® two-dimensional culture-derived conditioned medium (CM2D) using scratch and tubulogenesis assays and a rat wound splinting model, respectively. Results UCX® spheroids kept in our three-dimensional system remained viable and multipotent and secreted considerable amounts of vascular endothelial growth factor A, which was undetected in two-dimensional cultures, and higher amounts of matrix metalloproteinase-2, matrix metalloproteinase-9, hepatocyte growth factor, transforming growth factor β1, granulocyte-colony stimulating factor, fibroblast growth factor 2 and interleukin-6, when compared to CM2D. Furthermore, CM3D significantly enhanced elastin production and migration of keratinocytes and fibroblasts in vitro. In turn, tubulogenesis assays revealed increased capillary maturation in the presence of CM3D, as seen by a significant increase in capillary thickness and length when compared to CM2D, and increased branching points and capillary number when compared to basal medium. Finally, CM3D-treated wounds presented signs of faster and better resolution when compared to untreated and CM2D-treated wounds in vivo. Although CM2D proved to be beneficial, CM3D-treated wounds revealed a completely regenerated tissue by day 14 after excisions, with a more mature vascular system already showing glands and hair follicles. Conclusions This work unravels an important alternative to the use of cells in the final formulation of advanced therapy medicinal products by providing a proof of concept that a reproducible system for the production of UCX®-conditioned medium can be used to prime a secretome for eventual clinical applications. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0082-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jorge M Santos
- ECBio - Investigação e Desenvolvimento em Biotecnologia S.A., Rua Henrique Paiva Couceiro, N° 27, 2700-451, Amadora, Portugal.
| | - Sérgio P Camões
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Elysse Filipe
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Madalena Cipriano
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Rita N Barcia
- ECBio - Investigação e Desenvolvimento em Biotecnologia S.A., Rua Henrique Paiva Couceiro, N° 27, 2700-451, Amadora, Portugal.
| | - Mariana Filipe
- ECBio - Investigação e Desenvolvimento em Biotecnologia S.A., Rua Henrique Paiva Couceiro, N° 27, 2700-451, Amadora, Portugal.
| | - Mariana Teixeira
- ECBio - Investigação e Desenvolvimento em Biotecnologia S.A., Rua Henrique Paiva Couceiro, N° 27, 2700-451, Amadora, Portugal.
| | - Sandra Simões
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Manuela Gaspar
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Diogo Mosqueira
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal. .,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre, N° 823, 4150-180, Porto, Portugal.
| | - Diana S Nascimento
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal. .,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre, N° 823, 4150-180, Porto, Portugal.
| | - Perpétua Pinto-do-Ó
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal. .,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre, N° 823, 4150-180, Porto, Portugal. .,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, N° 228, 4050-313, Porto, Portugal. .,Unit for Lymphopoiesis, Immunology Department, INSERM U668, University Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Institut Pasteur, Paris, 75015, France.
| | - Pedro Cruz
- ECBio - Investigação e Desenvolvimento em Biotecnologia S.A., Rua Henrique Paiva Couceiro, N° 27, 2700-451, Amadora, Portugal.
| | - Helder Cruz
- ECBio - Investigação e Desenvolvimento em Biotecnologia S.A., Rua Henrique Paiva Couceiro, N° 27, 2700-451, Amadora, Portugal.
| | - Matilde Castro
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Joana P Miranda
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| |
Collapse
|
307
|
Abstract
Stem cells are cells specialized cell, capable of renewing themselves through cell division and can differentiate into multi-lineage cells. These cells are categorized as embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs) and adult stem cells. Mesenchymal stem cells (MSCs) are adult stem cells which can be isolated from human and animal sources. Human MSCs (hMSCs) are the non-haematopoietic, multipotent stem cells with the capacity to differentiate into mesodermal lineage such as osteocytes, adipocytes and chondrocytes as well ectodermal (neurocytes) and endodermal lineages (hepatocytes). MSCs express cell surface markers like cluster of differentiation (CD)29, CD44, CD73, CD90, CD105 and lack the expression of CD14, CD34, CD45 and HLA (human leucocyte antigen)-DR. hMSCs for the first time were reported in the bone marrow and till now they have been isolated from various tissues, including adipose tissue, amniotic fluid, endometrium, dental tissues, umbilical cord and Wharton's jelly which harbours potential MSCs. hMSCs have been cultured long-term in specific media without any severe abnormalities. Furthermore, MSCs have immunomodulatory features, secrete cytokines and immune-receptors which regulate the microenvironment in the host tissue. Multilineage potential, immunomodulation and secretion of anti-inflammatory molecules makes MSCs an effective tool in the treatment of chronic diseases. In the present review, we have highlighted recent research findings in the area of hMSCs sources, expression of cell surface markers, long-term in vitro culturing, in vitro differentiation potential, immunomodulatory features, its homing capacity, banking and cryopreservation, its application in the treatment of chronic diseases and its use in clinical trials.
Collapse
|
308
|
Khalilzad-Sharghi V, Han Z, Xu H, Othman SF. MR elastography for evaluating regeneration of tissue-engineered cartilage in an ectopic mouse model. Magn Reson Med 2015; 75:1209-17. [PMID: 25918870 DOI: 10.1002/mrm.25745] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/30/2015] [Accepted: 03/31/2015] [Indexed: 12/20/2022]
Abstract
PURPOSE The purpose of the present study was to apply noninvasive methods for monitoring regeneration and mechanical properties of tissue-engineered cartilage in vivo at different growth stages using MR elastography (MRE). METHODS Three types of scaffolds, including silk, collagen, and gelatin seeded by human mesenchymal stem cells, were implanted subcutaneously in mice and imaged at 9.4T where the shear stiffness and transverse MR relaxation time (T2 ) were measured for the regenerating constructs for 8 wk. An MRE phase contrast spin echo-based sequence was used for collecting MRE images. At the conclusion of the in vivo study, constructs were excised and transcript levels of cartilage-specific genes were quantitated using reverse-transcription polymerase chain reaction. RESULTS Tissue-engineered constructs showed a cartilage-like construct with progressive tissue formation characterized by increase in shear stiffness and decrease in T2 that can be correlated with increased cartilage transcript levels including aggrecan, type II collagen, and cartilage oligomeric matrix protein after 8 wk of in vivo culture. CONCLUSION Altogether, the outcome of this research demonstrates the feasibility of MRE and MRI for noninvasive monitoring of engineered cartilage construct's growth after implantation and provides noninvasive biomarkers for regeneration, which may be translated into treatment of tissue defects.
Collapse
Affiliation(s)
- Vahid Khalilzad-Sharghi
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Zhongji Han
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Huihui Xu
- School of Engineering and Computer Science, University of the Pacific, Stockton, California, USA
| | - Shadi F Othman
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| |
Collapse
|
309
|
Sampson S, Botto-van Bemden A, Aufiero D. Stem Cell Therapies for Treatment of Cartilage and Bone Disorders: Osteoarthritis, Avascular Necrosis, and Non-union Fractures. PM R 2015; 7:S26-S32. [DOI: 10.1016/j.pmrj.2015.01.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 01/21/2015] [Accepted: 01/23/2015] [Indexed: 12/16/2022]
|
310
|
Bhattacharjee M, Coburn J, Centola M, Murab S, Barbero A, Kaplan DL, Martin I, Ghosh S. Tissue engineering strategies to study cartilage development, degeneration and regeneration. Adv Drug Deliv Rev 2015; 84:107-22. [PMID: 25174307 DOI: 10.1016/j.addr.2014.08.010] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 08/01/2014] [Accepted: 08/20/2014] [Indexed: 01/09/2023]
Abstract
Cartilage tissue engineering has primarily focused on the generation of grafts to repair cartilage defects due to traumatic injury and disease. However engineered cartilage tissues have also a strong scientific value as advanced 3D culture models. Here we first describe key aspects of embryonic chondrogenesis and possible cell sources/culture systems for in vitro cartilage generation. We then review how a tissue engineering approach has been and could be further exploited to investigate different aspects of cartilage development and degeneration. The generated knowledge is expected to inform new cartilage regeneration strategies, beyond a classical tissue engineering paradigm.
Collapse
|
311
|
Bhumiratana S, Vunjak-Novakovic G. Engineering physiologically stiff and stratified human cartilage by fusing condensed mesenchymal stem cells. Methods 2015; 84:109-14. [PMID: 25828645 DOI: 10.1016/j.ymeth.2015.03.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 03/21/2015] [Accepted: 03/23/2015] [Indexed: 01/13/2023] Open
Abstract
For a long time, clinically sized and mechanically functional cartilage could be engineered from young animal chondrocytes, but not from adult human mesenchymal stem cells that are of primary clinical interest. The approaches developed for primary chondrocytes were not successful when used with human mesenchymal cells. The method discussed here was designed to employ a mechanism similar to pre-cartilaginous condensation and fusion of mesenchymal stem cells at a precisely defined time. The formation of cartilage was initiated by press-molding the mesenchymal bodies onto the surface of a bone substrate. By image-guided fabrication of the bone substrate and the molds, the osteochondral constructs were engineered in anatomically precise shapes and sizes. After 5 weeks of cultivation, the cartilage layer assumed physiologically stratified histomorphology, and contained lubricin at the surface, proteoglycans and type II collagen in the bulk phase, collagen type X at the interface with the bone substrate, and collagen type I within the bone phase. For the first time, the Young's modulus and the friction coefficient of human cartilage engineered from mesenchymal stem cells reached physiological levels for adult human cartilage. We propose that this method can be effective for generating human osteochondral tissue constructs.
Collapse
Affiliation(s)
- Sarindr Bhumiratana
- Department of Biomedical Engineering, Columbia University, New York, NY, USA(1,)(2)
| | | |
Collapse
|
312
|
Engineered cartilaginous tubes for tracheal tissue replacement via self-assembly and fusion of human mesenchymal stem cell constructs. Biomaterials 2015; 52:452-62. [PMID: 25818451 DOI: 10.1016/j.biomaterials.2015.01.073] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 01/22/2015] [Accepted: 01/25/2015] [Indexed: 01/14/2023]
Abstract
There is a critical need to engineer a neotrachea because currently there are no long-term treatments for tracheal stenoses affecting large portions of the airway. In this work, a modular tracheal tissue replacement strategy was developed. High-cell density, scaffold-free human mesenchymal stem cell-derived cartilaginous rings and tubes were successfully generated through employment of custom designed culture wells and a ring-to-tube assembly system. Furthermore, incorporation of transforming growth factor-β1-delivering gelatin microspheres into the engineered tissues enhanced chondrogenesis with regard to tissue size and matrix production and distribution in the ring- and tube-shaped constructs, as well as luminal rigidity of the tubes. Importantly, all engineered tissues had similar or improved biomechanical properties compared to rat tracheas, which suggests they could be transplanted into a small animal model for airway defects. The modular, bottom up approach used to grow stem cell-based cartilaginous tubes in this report is a promising platform to engineer complex organs (e.g., trachea), with control over tissue size and geometry, and has the potential to be used to generate autologous tissue implants for human clinical applications.
Collapse
|
313
|
Lee WC, Lim CH, Su C, Loh KP, Lim CT. Cell-assembled graphene biocomposite for enhanced chondrogenic differentiation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015; 11:963-9. [PMID: 25320042 DOI: 10.1002/smll.201401635] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 09/02/2014] [Indexed: 05/10/2023]
Abstract
Graphene-based nanomaterials are increasingly being explored for use as biomaterials for drug delivery and tissue engineering applications due to their exceptional physicochemical and mechanical properties. However, the two-dimensional nature of graphene makes it difficult to extend its applications beyond planar tissue culture. Here, graphene-cell biocomposites are used to pre-concentrate growth factors for chondrogenic differentiation. Bone marrow-derived mesenchymal stem cells (MSCs) are assembled with graphene flakes in the solution to form graphene-cell biocomposites. Increasing concentrations of graphene (G) and porous graphene oxide (pGO) are found to correlate positively with the extent of differentiation. However, beyond a certain concentration, especially in the case of graphene oxide, it will lead to decreased chondrogenesis due to increased diffusional barrier and cytotoxic effects. Nevertheless, these findings indicate that both G and pGO could serve as effective pre-concentration platforms for the construction of tissue-engineered cartilage and suspension-based cultures in vitro.
Collapse
Affiliation(s)
- Wong Cheng Lee
- Department of Biomedical Engineering and Department of Mechanical Engineering, National University of Singapore, Singapore, 117575, Singapore; NUS Graduate School of Integrative Sciences and Engineering, National University of Singapore, Singapore, 117456, Singapore
| | | | | | | | | |
Collapse
|
314
|
Gao G, Yonezawa T, Hubbell K, Dai G, Cui X. Inkjet-bioprinted acrylated peptides and PEG hydrogel with human mesenchymal stem cells promote robust bone and cartilage formation with minimal printhead clogging. Biotechnol J 2015; 10:1568-77. [PMID: 25641582 DOI: 10.1002/biot.201400635] [Citation(s) in RCA: 184] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/10/2014] [Accepted: 01/02/2015] [Indexed: 12/14/2022]
Abstract
Inkjet bioprinting is one of the most promising additive manufacturing approaches for tissue fabrication with the advantages of high speed, high resolution, and low cost. The limitation of this technology is the potential damage to the printed cells and frequent clogging of the printhead. Here we developed acrylated peptides and co-printed with acrylated poly(ethylene glycol) (PEG) hydrogel with simultaneous photopolymerization. At the same time, the bone marrow-derived human mesenchymal stem cells (hMSCs) were precisely printed during the scaffold fabrication process so the cells were delivered simultaneously with minimal UV exposure. The multiple steps of scaffold synthesis and cell encapsulation were successfully combined into one single step using bioprinting. The resulted peptide-conjugated PEG scaffold demonstrated excellent biocompatibility, with a cell viability of 87.9 ± 5.3%. Nozzle clogging was minimized due to the low viscosity of the PEG polymer. With osteogenic and chondrogenic differentiation, the bioprinted bone and cartilage tissue demonstrated excellent mineral and cartilage matrix deposition, as well as significantly increased mechanical properties. Strikingly, the bioprinted PEG-peptide scaffold dramatically inhibited hMSC hypertrophy during chondrogenic differentiation. Collectively, bioprinted PEG-peptide scaffold and hMSCs significantly enhanced osteogenic and chondrogenic differentiation for robust bone and cartilage formation with minimal printhead clogging.
Collapse
Affiliation(s)
- Guifang Gao
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China.,Stemorgan Therapeutics, Albany, NY, USA
| | - Tomo Yonezawa
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA.,Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | | | - Guohao Dai
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.
| | - Xiaofeng Cui
- Stemorgan Therapeutics, Albany, NY, USA. .,Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.
| |
Collapse
|
315
|
Hoffmann W, Feliciano S, Martin I, de Wild M, Wendt D. Novel Perfused Compression Bioreactor System as an in vitro Model to Investigate Fracture Healing. Front Bioeng Biotechnol 2015; 3:10. [PMID: 25699254 PMCID: PMC4313709 DOI: 10.3389/fbioe.2015.00010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 01/16/2015] [Indexed: 01/08/2023] Open
Abstract
Secondary bone fracture healing is a physiological process that leads to functional tissue regeneration via endochondral bone formation. In vivo studies have demonstrated that early mobilization and the application of mechanical loads enhances the process of fracture healing. However, the influence of specific mechanical stimuli and particular effects during specific phases of fracture healing remain to be elucidated. In this work, we have developed and provided proof-of-concept of an in vitro human organotypic model of physiological loading of a cartilage callus, based on a novel perfused compression bioreactor (PCB) system. We then used the fracture callus model to investigate the regulatory role of dynamic mechanical loading. Our findings provide a proof-of-principle that dynamic mechanical loading applied by the PCB can enhance the maturation process of mesenchymal stromal cells toward late hypertrophic chondrocytes and the mineralization of the deposited extracellular matrix. The PCB provides a promising tool to study fracture healing and for the in vitro assessment of alternative fracture treatments based on engineered tissue grafts or pharmaceutical compounds, allowing for the reduction of animal experiments.
Collapse
Affiliation(s)
- Waldemar Hoffmann
- Department of Biomedicine, University Hospital Basel , Basel , Switzerland ; Department of Surgery, University Hospital Basel , Basel , Switzerland ; School of Life Sciences, Institute for Medical and Analytical Technologies, University of Applied Sciences Northwestern Switzerland , Muttenz , Switzerland
| | - Sandra Feliciano
- Department of Biomedicine, University Hospital Basel , Basel , Switzerland ; Department of Surgery, University Hospital Basel , Basel , Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel , Basel , Switzerland ; Department of Surgery, University Hospital Basel , Basel , Switzerland
| | - Michael de Wild
- School of Life Sciences, Institute for Medical and Analytical Technologies, University of Applied Sciences Northwestern Switzerland , Muttenz , Switzerland
| | - David Wendt
- Department of Biomedicine, University Hospital Basel , Basel , Switzerland ; Department of Surgery, University Hospital Basel , Basel , Switzerland
| |
Collapse
|
316
|
Muhonen V, Narcisi R, Nystedt J, Korhonen M, van Osch GJVM, Kiviranta I. Recombinant human type II collagen hydrogel provides a xeno-free 3D micro-environment for chondrogenesis of human bone marrow-derived mesenchymal stromal cells. J Tissue Eng Regen Med 2015; 11:843-854. [PMID: 25643647 DOI: 10.1002/term.1983] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 10/02/2014] [Accepted: 12/09/2014] [Indexed: 12/12/2022]
Abstract
Recombinant human type II collagen (rhCII) hydrogel was tested as a xeno-free micro-environment for the chondrogenesis of human bone marrow-derived mesenchymal stromal cells (BM-MSCs). The rhCII hydrogels were seeded with BM-MSCs and cultured in a xeno-free chondro-inductive medium for 14, 28 and 84 days. High-density pellet cultures served as controls. The samples were subjected to biochemical, histological and gene expression analyses. Although the cells deposited glycosaminoglycans into the extracellular space significantly more slowly in the rhCII hydrogels compared to the high-density pellets, a similar potential of matrix deposition was reached by the end of the 84-day culture. At day 28 of culture, the gene expression level for cartilage marker genes (i.e. genes encoding for Sox9 transcription factor, Collagen type II and Aggrecan) were considerably lower in the rhCII hydrogels than in the high-density pellets, but at the end of the 84-day culture period, all the cartilage marker genes analysed were expressed at a similar level. Interestingly, the expression of the matrix metallopeptidases (MMP)-13, MMP-14 and MMP-8, i.e. extracellular collagen network-degrading enzymes, were transiently upregulated in the rhCII hydrogel, indicating active matrix reorganization. This study demonstrated that the rhCII hydrogel functions as a xeno-free platform for BM-MSC chondrogenesis, although the process is delayed. The reversible catabolic reaction evoked by the rhCII hydrogel might be beneficial in graft integration in vivo and pinpoints the need to further explore the use of hydrogels containing recombinant extracellular matrix (ECM) proteins to induce the chondrogenesis of MSCs. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Virpi Muhonen
- Department of Surgery, Institute of Clinical Medicine, University of Helsinki, Finland.,Department of Orthopaedics and Traumatology, Hospital District of Helsinki and Uusimaa, Helsinki, Finland
| | - Roberto Narcisi
- Department of Orthopaedics, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Johanna Nystedt
- Finnish Red Cross Blood Service, Research and Cell Therapy Services, Helsinki, Finland
| | - Matti Korhonen
- Finnish Red Cross Blood Service, Research and Cell Therapy Services, Helsinki, Finland
| | - Gerjo J V M van Osch
- Department of Orthopaedics, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands.,Department of Otorhinolaryngology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Ilkka Kiviranta
- Department of Surgery, Institute of Clinical Medicine, University of Helsinki, Finland.,Department of Orthopaedics and Traumatology, Hospital District of Helsinki and Uusimaa, Helsinki, Finland
| |
Collapse
|
317
|
Lee JH, Shim W, Choolakadavil Khalid N, Kang WS, Lee M, Kim HS, Choi J, Lee G, Kim JH. Random networks of single-walled carbon nanotubes promote mesenchymal stem cell's proliferation and differentiation. ACS APPLIED MATERIALS & INTERFACES 2015; 7:1560-7. [PMID: 25546303 DOI: 10.1021/am506833q] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Studies on the interaction of cells with single-walled carbon nanotubes (SWCNTs) have been receiving increasing attention owing to their potential for various cellular applications. In this report, we investigated the interactions between biological cells and nanostructured SWCNTs films and focused on how morphological structures of SWCNT films affected cellular behavior such as cell proliferation and differentiation. One directionally aligned SWCNT Langmuir-Blodgett (LB) film and random network SWCNT film were fabricated by LB and vacuum filteration methods, respectively. We demonstrate that our SWCNT LB and network film based scaffolds do not show any cytotoxicity, while on the other hand, these scaffolds promote differentiation property of rat mesenchymal stem cells (rMSCs) when compared with that on conventional tissue culture polystyrene substrates. Especially, the SWCNT network film with average thickness and roughness values of 95 ± 5 and 9.81 nm, respectively, demonstrated faster growth rate and higher cell thickness for rMSCs. These results suggest that systematic manipulation of the thickness, roughness, and directional alignment of SWCNT films would provide the convenient strategy for controlling the growth and maintenance of the differentiation property of stem cells. The SWCNT film could be an alternative culture substrate for various stem cells, which often require close control of the growth and differentiation properties.
Collapse
Affiliation(s)
- Jae-Hyeok Lee
- Department of Molecular Science and Technology, Ajou University , Suwon 443-749, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
318
|
Sathi GA, Kenmizaki K, Yamaguchi S, Nagatsuka H, Yoshida Y, Matsugaki A, Ishimoto T, Imazato S, Nakano T, Matsumoto T. Early initiation of endochondral ossification of mouse femur cultured in hydrogel with different mechanical stiffness. Tissue Eng Part C Methods 2015; 21:567-75. [PMID: 25381834 DOI: 10.1089/ten.tec.2014.0475] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mineralization is one of the most important processes in normal bone tissue development and in disease condition. Developing a novel and standardized in vitro model system that can readily monitor both cellular dynamics and mineralization is crucial for better understanding the bone tissue development and growth. Recent studies indicated that the mechanical environment is a critical condition in mineralization. We hypothesized that hydrogel with different mechanical stiffness can provide a biomimetic mechanical environment that can modulate bone tissue growth and mineralization. A femur of mouse embryo (embryonic day 16) was embedded in agarose hydrogel (2-60 kPa) and cultured in an osteogenic medium for a week. Microcomputed tomography (μCT) results revealed enhanced mineralization was detected in the femur head cultured in the gel condition, whereas no mineralization in the femur head cultured in the control (floating culture) condition. The mineralized region was corresponding to the region of secondary ossification center. Both histological and quantitative analyses indicated that the mineralized region of femur head cultured in 10 kPa gel condition was the highest and the mineralized area was significantly larger than that cultured in 2, 40, and 60 kPa gel condition. Immunofluorescence results indicated the enhanced mineralization caused by the higher chondrogenic differentiation at that region. This enhancement mainly relating to the mechanical forces and not to the oxygen tension was also confirmed. Since this system enhances and shortens the mineralization procedure compared with the conventional two-dimensional or three-dimensional cell culture system, this hydrogel system would be one of the unique models for better understanding the mineralized tissue development.
Collapse
Affiliation(s)
- Gulsan Ara Sathi
- 1Department of Biomaterials, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | | | - Satoshi Yamaguchi
- 3Department of Biomaterials, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Hitoshi Nagatsuka
- 4Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yasuhiro Yoshida
- 5Division of Oral Health Science, Department of Biomaterials and Bioengineering, Graduate School of Dental Medicine, Hokkaido University, Hokkaido, Japan
| | | | | | - Satoshi Imazato
- 3Department of Biomaterials, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | | | - Takuya Matsumoto
- 1Department of Biomaterials, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
319
|
Cao FY, Yin WN, Fan JX, Zhuo RX, Zhang XZ. A novel function of BMHP1 and cBMHP1 peptides to induce the osteogenic differentiation of mesenchymal stem cells. Biomater Sci 2015. [DOI: 10.1039/c4bm00300d] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BMHP1 or cBMHP1 peptide is found to induce MSCs towards the osteogenic lineage when tethered to modified quartz substrates.
Collapse
Affiliation(s)
- Feng-Yi Cao
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- PR China
| | - Wei-Na Yin
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- PR China
| | - Jin-Xuan Fan
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- PR China
| | - Ren-Xi Zhuo
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- PR China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- PR China
| |
Collapse
|
320
|
Jayakumar GC, Mehta A, Rao JR, Fathima NN. Ionic liquids: new age materials for eco-friendly leather processing. RSC Adv 2015. [DOI: 10.1039/c5ra02167g] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The manufacture of leather is a challenging and complicated process, which converts natural biomaterial to various high end applications.
Collapse
Affiliation(s)
| | - Ami Mehta
- CSIR-Central Leather Research Institute
- Chennai-600 020
- India
| | | | | |
Collapse
|
321
|
Fitzpatrick LE, McDevitt TC. Cell-derived matrices for tissue engineering and regenerative medicine applications. Biomater Sci 2015; 3:12-24. [PMID: 25530850 PMCID: PMC4270054 DOI: 10.1039/c4bm00246f] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The development and application of decellularized extracellular matrices (ECM) has grown rapidly in the fields of cell biology, tissue engineering and regenerative medicine in recent years. Similar to decellularized tissues and whole organs, cell-derived matrices (CDMs) represent bioactive, biocompatible materials consisting of a complex assembly of fibrillar proteins, matrix macromolecules and associated growth factors that often recapitulate, at least to some extent, the composition and organization of native ECM microenvironments. The unique ability to engineer CDMs de novo based on cell source and culture methods makes them an attractive alternative to conventional allogeneic and xenogeneic tissue-derived matrices that are currently harvested from cadaveric sources, suffer from inherent heterogeneity, and have limited ability for customization. Although CDMs have been investigated for a number of biomedical applications, including adhesive cell culture substrates, synthetic scaffold coatings, and tissue engineered products, such as heart valves and vascular grafts, the state of the field is still at a relatively nascent stage of development. In this review, we provide an overview of the various applications of CDM and discuss successes to date, current limitations and future directions.
Collapse
Affiliation(s)
| | - Todd C. McDevitt
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, Georgia, USA
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
322
|
Leferink AM, Santos D, Karperien M, Truckenmüller RK, van Blitterswijk CA, Moroni L. Differentiation capacity and maintenance of differentiated phenotypes of human mesenchymal stromal cells cultured on two distinct types of 3D polymeric scaffolds. Integr Biol (Camb) 2015; 7:1574-86. [DOI: 10.1039/c5ib00177c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
This study shows that the classical validation of hMSC differentiation potential on 3D scaffolds might not be sufficient to ensure the maintenance of the cells functionality in the absence of differentiation inducing soluble factors.
Collapse
Affiliation(s)
- A. M. Leferink
- Department of Tissue Regeneration and MIRA Institute for Biomedical Technology and Technical Medicine
- University of Twente
- Enschede
- The Netherlands
- Department of Complex Tissue Regeneration
| | - D. Santos
- Department of Tissue Regeneration and MIRA Institute for Biomedical Technology and Technical Medicine
- University of Twente
- Enschede
- The Netherlands
| | - M. Karperien
- Department of Developmental Bioengineering
- MIRA Institute for Biomedical Technology and Technical Medicine
- University of Twente
- Enschede
- The Netherlands
| | - R. K. Truckenmüller
- Department of Tissue Regeneration and MIRA Institute for Biomedical Technology and Technical Medicine
- University of Twente
- Enschede
- The Netherlands
- Department of Complex Tissue Regeneration
| | - C. A. van Blitterswijk
- Department of Tissue Regeneration and MIRA Institute for Biomedical Technology and Technical Medicine
- University of Twente
- Enschede
- The Netherlands
- Department of Complex Tissue Regeneration
| | - L. Moroni
- Department of Tissue Regeneration and MIRA Institute for Biomedical Technology and Technical Medicine
- University of Twente
- Enschede
- The Netherlands
- Department of Complex Tissue Regeneration
| |
Collapse
|
323
|
Chen AX, Hoffman MD, Chen CS, Shubin AD, Reynolds DS, Benoit DSW. Disruption of cell-cell contact-mediated notch signaling via hydrogel encapsulation reduces mesenchymal stem cell chondrogenic potential: winner of the Society for Biomaterials Student Award in the Undergraduate Category, Charlotte, NC, April 15 to 18, 2015. J Biomed Mater Res A 2014; 103:1291-302. [PMID: 25504509 DOI: 10.1002/jbm.a.35383] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 11/17/2014] [Accepted: 12/04/2014] [Indexed: 12/20/2022]
Abstract
Cell-cell contact-mediated Notch signaling is essential for mesenchymal stem cell (MSC) chondrogenesis during development. However, subsequent deactivation of Notch signaling is also required to allow for stem cell chondrogenic progression. Recent literature has shown that Notch signaling can also influence Wnt/β-catenin signaling, critical for MSC differentiation, through perturbations in cell-cell contacts. Traditionally, abundant cell-cell contacts, consistent with development, are emulated in vitro using pellet cultures for chondrogenesis. However, cells are often encapsulated within biomaterials-based scaffolds, such as hydrogels, to improve therapeutic cell localization in vivo. To explore the role of Notch and Wnt/β-catenin signaling in the context of hydrogel-encapsulated MSC chondrogenesis, we compared signaling and differentiation capacity of MSCs in both hydrogels and traditional pellet cultures. We demonstrate that encapsulation within poly(ethylene glycol) hydrogels reduces cell-cell contacts, and both Notch (7.5-fold) and Wnt/β-catenin (84.7-fold) pathway activation. Finally, we demonstrate that following establishment of cell-cell contacts and transient Notch signaling in pellet cultures, followed by Notch signaling deactivation, resulted in a 1.5-fold increase in MSC chondrogenesis. Taken together, these findings support that cellular condensation, and establishment of initial cell-cell contacts is critical for MSC chondrogenesis, and this process is inhibited by hydrogel encapsulation.
Collapse
Affiliation(s)
- Amanda X Chen
- Department of Biomedical Engineering, University of Rochester, 207 Robert B. Goergen Hall, Box 270168, Rochester, New York, 14627-0168
| | | | | | | | | | | |
Collapse
|
324
|
Pan JF, Yuan L, Guo CA, Geng XH, Fei T, Fan WS, Li S, Yuan HF, Yan ZQ, Mo XM. Fabrication of modified dextran-gelatin in situ forming hydrogel and application in cartilage tissue engineering. J Mater Chem B 2014; 2:8346-8360. [PMID: 32262005 DOI: 10.1039/c4tb01221f] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hydrogels play a very important role in cartilage tissue engineering. Here, we oxidized dextran (Odex) and modified gelatin (Mgel) to fabricate a fast forming hydrogel without the addition of a chemical crosslinking agent. The dynamic gelling process was measured through rheological measurements. The microstructure was examined by lyophilizing to get porous scaffolds. Biological assessment was performed through CCK-8 assays by using synovium-derived mesenchymal cells (SMSCs) at 1, 3, 7 and 14 days. In vivo evaluation for application in cartilage tissue engineering was performed 8 weeks after subcutaneous injection of SMSC-loaded Odex/Mgel hydrogels combined with TGF-β3 in the dorsa of nude mice. According to the results, a fast forming hydrogel was obtained by simply modifying dextran and gelatin. Moreover, the Odex/Mgel hydrogel exhibited good biocompatibility in cultures of SMSCs and a homogeneous distribution of live cells was achieved inside the hydrogels. After 8 weeks, newly formed cartilage was achieved in the dorsa of nude mice; no inflammatory reaction was observed and high production of GAGs was shown. The method provides a strategy for the design and fabrication of fast in situ forming hydrogels. The Odex/Mgel hydrogel could be used for the regeneration of cartilage in tissue engineering.
Collapse
Affiliation(s)
- Jian-Feng Pan
- Department of Orthopedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
325
|
Nagahara T, Yoshimatsu S, Shiba H, Kawaguchi H, Takeda K, Iwata T, Mizuno N, Fujita T, Kurihara H. Introduction of a mixture of β-tricalcium phosphate into a complex of bone marrow mesenchymal stem cells and type I collagen can augment the volume of alveolar bone without impairing cementum regeneration. J Periodontol 2014; 86:456-64. [PMID: 25494830 DOI: 10.1902/jop.2014.140384] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND The purpose of this study is to evaluate whether β-tricalcium phosphate (β-TCP) could be a promising modality to help augment alveolar bone in periodontal tissue regeneration by bone marrow mesenchymal stem cells (BMMSCs). METHODS Expanded BMMSCs and atelocollagen (Col) were mixed together (MSC/Col). A combination of β-TCP with MSC/Col was also prepared (MSC/Col/TCP). MSC/Col/TCP or MSC/Col was transplanted into experimental periodontal Class III furcation defects that had been exposed to inflammation in beagle dogs. Periodontal tissue regeneration was evaluated by histologic and morphometric analyses at 4 and 8 weeks after transplantation. RESULTS MSC/Col and MSC/Col/TCP enhanced periodontal tissue regeneration compared to Col and TCP/Col according to hematoxylin and eosin staining. The percentage of new cementum length in the MSC/Col/TCP group was not significantly different from that in the MSC/Col group at 4 and 8 weeks. On the other hand, the percentage of new bone area in the MSC/Col/TCP group was much higher than that in the MSC/TCP group at 4 weeks. However, at 8 weeks, no significant difference in new bone area was found between the two groups. In the MSC/Col/TCP group, β-TCP was surrounded by newly formed bone. Multinucleated cells, which were positive for osteopontin and tartrate-resistant acid phosphatase, were present in the interconnected macropores of β-TCP. CONCLUSION These findings suggest that β-TCP is applicable as a scaffold for BMMSCs transplantation and helps augment alveolar bone without impairing regeneration of cementum.
Collapse
Affiliation(s)
- Takayosi Nagahara
- Department of Periodontal Medicine, Applied Life Science, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
326
|
Frisch J, Venkatesan JK, Rey-Rico A, Schmitt G, Madry H, Cucchiarini M. Determination of the Chondrogenic Differentiation Processes in Human Bone Marrow-Derived Mesenchymal Stem Cells Genetically Modified to Overexpress Transforming Growth Factor-β via Recombinant Adeno-Associated Viral Vectors. Hum Gene Ther 2014; 25:1050-60. [DOI: 10.1089/hum.2014.091] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Janina Frisch
- Center of Experimental Orthopedics, Saarland University Medical Center, D-66421 Homburg, Germany
| | | | - Ana Rey-Rico
- Center of Experimental Orthopedics, Saarland University Medical Center, D-66421 Homburg, Germany
| | - Gertrud Schmitt
- Center of Experimental Orthopedics, Saarland University Medical Center, D-66421 Homburg, Germany
| | - Henning Madry
- Center of Experimental Orthopedics, Saarland University Medical Center, D-66421 Homburg, Germany
- Department of Orthopedic Surgery, Saarland University Medical Center, D-66421 Homburg, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopedics, Saarland University Medical Center, D-66421 Homburg, Germany
| |
Collapse
|
327
|
Goude MC, McDevitt TC, Temenoff JS. Chondroitin sulfate microparticles modulate transforming growth factor-β1-induced chondrogenesis of human mesenchymal stem cell spheroids. Cells Tissues Organs 2014; 199:117-30. [PMID: 25413333 DOI: 10.1159/000365966] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2014] [Indexed: 11/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been previously explored as a part of cell-based therapies for the repair of damaged cartilage. Current MSC chondrogenic differentiation strategies employ large pellets; however, we have developed a technique to form small MSC aggregates (500-1,000 cells) that can reduce transport barriers while maintaining a multicellular structure analogous to cartilaginous condensations. The objective of this study was to examine the effects of incorporating chondroitin sulfate methacrylate (CSMA) microparticles (MPs) within small MSC spheroids cultured in the presence of transforming growth factor (TGF)-β1 on chondrogenesis. Spheroids with MPs induced earlier increases in collagen II and aggrecan gene expression (chondrogenic markers) than spheroids without MPs, although no large differences in immunostaining for these matrix molecules were observed by day 21 between these groups. Collagen I and X were also detected in the extracellular matrix (ECM) of all spheroids by immunostaining. Interestingly, histology revealed that CSMA MPs clustered together near the center of the MSC spheroids and induced circumferential alignment of cells and ECM around the material core. This study demonstrates the use of CSMA materials to further examine the effects of matrix molecules on MSC phenotype as well as potentially direct differentiation in a more spatially controlled manner that better mimics the architecture of specific musculoskeletal tissues.
Collapse
Affiliation(s)
- Melissa C Goude
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Ga., USA
| | | | | |
Collapse
|
328
|
Pretzel D, Linss S, Ahrem H, Endres M, Kaps C, Klemm D, Kinne RW. A novel in vitro bovine cartilage punch model for assessing the regeneration of focal cartilage defects with biocompatible bacterial nanocellulose. Arthritis Res Ther 2014; 15:R59. [PMID: 23673274 PMCID: PMC4060236 DOI: 10.1186/ar4231] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 02/04/2013] [Accepted: 05/14/2013] [Indexed: 12/21/2022] Open
Abstract
Introduction Current therapies for articular cartilage defects fail to achieve qualitatively sufficient tissue regeneration, possibly because of a mismatch between the speed of cartilage rebuilding and the resorption of degradable implant polymers. The present study focused on the self-healing capacity of resident cartilage cells in conjunction with cell-free and biocompatible (but non-resorbable) bacterial nanocellulose (BNC). This was tested in a novel in vitro bovine cartilage punch model. Methods Standardized bovine cartilage discs with a central defect filled with BNC were cultured for up to eight weeks with/without stimulation with transforming growth factor-β1 (TGF-β1. Cartilage formation and integrity were analyzed by histology, immunohistochemistry and electron microscopy. Content, release and neosynthesis of the matrix molecules proteoglycan/aggrecan, collagen II and collagen I were also quantified. Finally, gene expression of these molecules was profiled in resident chondrocytes and chondrocytes migrated onto the cartilage surface or the implant material. Results Non-stimulated and especially TGF-β1-stimulated cartilage discs displayed a preserved structural and functional integrity of the chondrocytes and surrounding matrix, remained vital in long-term culture (eight weeks) without signs of degeneration and showed substantial synthesis of cartilage-specific molecules at the protein and mRNA level. Whereas mobilization of chondrocytes from the matrix onto the surface of cartilage and implant was pivotal for successful seeding of cell-free BNC, chondrocytes did not immigrate into the central BNC area, possibly due to the relatively small diameter of its pores (2 to 5 μm). Chondrocytes on the BNC surface showed signs of successful redifferentiation over time, including increase of aggrecan/collagen type II mRNA, decrease of collagen type I mRNA and initial deposition of proteoglycan and collagen type II in long-term high-density pellet cultures. Although TGF-β1 stimulation showed protective effects on matrix integrity, effects on other parameters were limited. Conclusions The present bovine cartilage punch model represents a robust, reproducible and highly suitable tool for the long-term culture of cartilage, maintaining matrix integrity and homoeostasis. As an alternative to animal studies, this model may closely reflect early stages of cartilage regeneration, allowing the evaluation of promising biomaterials with/without chondrogenic factors.
Collapse
|
329
|
Gupta MS, Nicoll SB. Duration of TGF-β3 Exposure Impacts the Chondrogenic Maturation of Human MSCs in Photocrosslinked Carboxymethylcellulose Hydrogels. Ann Biomed Eng 2014; 43:1145-57. [DOI: 10.1007/s10439-014-1179-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 10/29/2014] [Indexed: 12/11/2022]
|
330
|
Smith E, Kanczler J, Gothard D, Roberts C, Wells J, White L, Qutachi O, Sawkins M, Peto H, Rashidi H, Rojo L, Stevens M, El Haj A, Rose F, Shakesheff K, Oreffo R. Evaluation of skeletal tissue repair, part 1: assessment of novel growth-factor-releasing hydrogels in an ex vivo chick femur defect model. Acta Biomater 2014; 10:4186-96. [PMID: 24937137 DOI: 10.1016/j.actbio.2014.06.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 05/21/2014] [Accepted: 06/09/2014] [Indexed: 01/08/2023]
Abstract
Current clinical treatments for skeletal conditions resulting in large-scale bone loss include autograft or allograft, both of which have limited effectiveness. In seeking to address bone regeneration, several tissue engineering strategies have come to the fore, including the development of growth factor releasing technologies and appropriate animal models to evaluate repair. Ex vivo models represent a promising alternative to simple in vitro systems or complex, ethically challenging in vivo models. We have developed an ex vivo culture system of whole embryonic chick femora, adapted in this study as a critical size defect model to investigate the effects of novel bone extracellular matrix (bECM) hydrogel scaffolds containing spatio-temporal growth factor-releasing microparticles and skeletal stem cells on bone regeneration, to develop a viable alternative treatment for skeletal degeneration. Alginate/bECM hydrogels combined with poly (d,l-lactic-co-glycolic acid) (PDLLGA)/triblock copolymer (10-30% PDLLGA-PEG-PDLLGA) microparticles releasing VEGF, TGF-β3 or BMP-2 were placed, with human adult Stro-1+ bone marrow stromal cells, into 2mm central segmental defects in embryonic chick femurs. Alginate/bECM hydrogels loaded with HSA/VEGF or HSA/TGF-β3 demonstrated a cartilage-like phenotype, with minimal collagen I deposition, comparable to HSA-only control hydrogels. The addition of BMP-2 releasing microparticles resulted in enhanced structured bone matrix formation, evidenced by increased Sirius red-stained matrix and collagen expression within hydrogels. This study demonstrates delivery of bioactive growth factors from a novel alginate/bECM hydrogel to augment skeletal tissue formation and the use of an organotypic chick femur defect culture system as a high-throughput test model for scaffold/cell/growth factor therapies for regenerative medicine.
Collapse
|
331
|
Effect of intra-articular injection of mesenchymal stem cells in cartilage repair in experimental animals. EGYPTIAN RHEUMATOLOGIST 2014. [DOI: 10.1016/j.ejr.2014.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
332
|
Jagielski M, Wolf J, Marzahn U, Völker A, Lemke M, Meier C, Ertel W, Godkin O, Arens S, Schulze-Tanzil G. The influence of IL-10 and TNFα on chondrogenesis of human mesenchymal stromal cells in three-dimensional cultures. Int J Mol Sci 2014; 15:15821-44. [PMID: 25207597 PMCID: PMC4200793 DOI: 10.3390/ijms150915821] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 08/25/2014] [Accepted: 09/02/2014] [Indexed: 12/17/2022] Open
Abstract
Chondrogenic differentiated mesenchymal stromal cells (MSCs) are a promising cell source for articular cartilage repair. This study was undertaken to determine the effectiveness of two three-dimensional (3D) culture systems for chondrogenic MSC differentiation in comparison to primary chondrocytes and to assess the effect of Interleukin (IL)-10 and Tumor Necrosis Factor (TNF)α on chondrogenesis by MSCs in 3D high-density (H-D) culture. MSCs were isolated from femur spongiosa, characterized using a set of typical markers and introduced in scaffold-free H-D cultures or non-woven polyglycolic acid (PGA) scaffolds for chondrogenic differentiation. H-D cultures were stimulated with recombinant IL-10, TNFα, TNFα + IL-10 or remained untreated. Gene and protein expression of type II collagen, aggrecan, sox9 and TNFα were examined. MSCs expressed typical cell surface markers and revealed multipotency. Chondrogenic differentiated cells expressed cartilage-specific markers in both culture systems but to a lower extent when compared with articular chondrocytes. Chondrogenesis was more pronounced in PGA compared with H-D culture. IL-10 and/or TNFα did not impair the chondrogenic differentiation of MSCs. Moreover, in most of the investigated samples, despite not reaching significance level, IL-10 had a stimulatory effect on the type II collagen, aggrecan and TNFα expression when compared with the respective controls.
Collapse
Affiliation(s)
- Michal Jagielski
- Department for Orthopedic, Trauma and Reconstructive Surgery, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 14195 Berlin, Garystrasse 5, Germany.
| | - Johannes Wolf
- Department for Orthopedic, Trauma and Reconstructive Surgery, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 14195 Berlin, Garystrasse 5, Germany.
| | - Ulrike Marzahn
- Department for Orthopedic, Trauma and Reconstructive Surgery, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 14195 Berlin, Garystrasse 5, Germany.
| | - Anna Völker
- Department for Orthopedic, Trauma and Reconstructive Surgery, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 14195 Berlin, Garystrasse 5, Germany.
| | - Marion Lemke
- Department for Orthopedic, Trauma and Reconstructive Surgery, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 14195 Berlin, Garystrasse 5, Germany.
| | - Carola Meier
- Department for Orthopedic, Trauma and Reconstructive Surgery, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 14195 Berlin, Garystrasse 5, Germany.
| | - Wolfgang Ertel
- Department for Orthopedic, Trauma and Reconstructive Surgery, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 14195 Berlin, Garystrasse 5, Germany.
| | - Owen Godkin
- Department for Orthopedic, Trauma and Reconstructive Surgery, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 14195 Berlin, Garystrasse 5, Germany.
| | - Stephan Arens
- Department for Orthopedic, Trauma and Reconstructive Surgery, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 14195 Berlin, Garystrasse 5, Germany.
| | - Gundula Schulze-Tanzil
- Department for Orthopedic, Trauma and Reconstructive Surgery, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 14195 Berlin, Garystrasse 5, Germany.
| |
Collapse
|
333
|
Frisch J, Venkatesan JK, Rey-Rico A, Schmitt G, Madry H, Cucchiarini M. Influence of insulin-like growth factor I overexpression via recombinant adeno-associated vector gene transfer upon the biological activities and differentiation potential of human bone marrow-derived mesenchymal stem cells. Stem Cell Res Ther 2014; 5:103. [PMID: 25163769 PMCID: PMC4164762 DOI: 10.1186/scrt491] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 08/18/2014] [Indexed: 01/25/2023] Open
Abstract
Introduction The transplantation of genetically modified progenitor cells such as bone marrow-derived mesenchymal stem cells (MSCs) is an attractive strategy to improve the natural healing of articular cartilage defects. In the present study, we examined the potential benefits of sustained overexpression of the mitogenic and pro-anabolic insulin-like growth factor I (IGF-I) via gene transfer upon the biological activities of human MSCs (hMSCs). Methods Recombinant adeno-associated vectors (rAAV) were used to deliver a human IGF-I coding sequence in undifferentiated and chondrogenically-induced primary hMSCs in order to determine the efficacy and duration of transgene expression and the subsequent effects of the genetic modification upon the chondrogenic versus osteogenic differentiation profiles of the cells relative to control (lacZ) treatment after 21 days in vitro. Results Significant and prolonged expression of IGF-I was evidenced in undifferentiated and most importantly in chondrogenically-induced hMSCs transduced with the candidate rAAV-hIGF-I vector for up to 21 days, leading to enhanced proliferative, biosynthetic, and chondrogenic activities compared with rAAV-lacZ treatment. Overexpression of IGF-I as achieved in the conditions applied here also increased the expression of hypertrophic and osteogenic markers in the treated cells. Conclusions These results suggest that a tight regulation of rAAV expression may be necessary for further translation of the approach in clinically relevant animal models in vivo. However, the current findings support the concept of using this type of vector as an effective tool to treat articular cartilage defects via gene- and stem cell-based procedures.
Collapse
|
334
|
Li S, Sengers BG, Oreffo ROC, Tare RS. Chondrogenic potential of human articular chondrocytes and skeletal stem cells: a comparative study. J Biomater Appl 2014; 29:824-36. [PMID: 25145989 PMCID: PMC4274334 DOI: 10.1177/0885328214548604] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Regenerative medicine strategies have increasingly focused on skeletal stem cells (SSCs), in response to concerns such as donor site morbidity, dedifferentiation and limited lifespan associated with the use of articular chondrocytes for cartilage repair. The suitability of SSCs for cartilage regeneration, however, remains to be fully determined. This study has examined the chondrogenic potential of human STRO-1-immunoselected SSCs (STRO-1+ SSCs), in comparison to human articular chondrocytes (HACs), by utilising two bioengineering strategies, namely “scaffold-free” three-dimensional (3-D) pellet culture and culture using commercially available, highly porous, 3-D scaffolds with interconnected pore networks. STRO-1+ SSCs were isolated by magnetic-activated cell sorting from bone marrow samples of haematologically normal osteoarthritic individuals following routine hip replacement procedures. Chondrocytes were isolated by sequential enzymatic digestion of deep zone articular cartilage pieces dissected from femoral heads of the same individuals. After expansion in monolayer cultures, the harvested cell populations were centrifuged to form high-density 3-D pellets and also seeded in the 3-D scaffold membranes, followed by culture in serum-free chondrogenic media under static conditions for 21 and 28 days, respectively. Chondrogenic differentiation was determined by gene expression, histological and immunohistochemical analyses. Robust cartilage formation and expression of hyaline cartilage-specific markers were observed in both day-21 pellets and day-28 explants generated using HACs. In comparison, STRO-1+ SSCs demonstrated significantly lower chondrogenic differentiation potential and a tendency for hypertrophic differentiation in day-21 pellets. Culture of STRO-1+ SSCs in the 3-D scaffolds improved the expression of hyaline cartilage-specific markers in day-28 explants, however, was unable to prevent hypertrophic differentiation of the SSC population. The advantages of application of SSCs in tissue engineering are widely recognised; the results of this study, however, highlight the need for further development of cell culture protocols that may otherwise limit the application of this stem cell population in cartilage bioengineering strategies.
Collapse
Affiliation(s)
- Siwei Li
- Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Bram G Sengers
- Faculty of Engineering and the Environment, Bioengineering Science, University of Southampton, Southampton, UK
| | - Richard O C Oreffo
- Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Rahul S Tare
- Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, UK Faculty of Engineering and the Environment, Bioengineering Science, University of Southampton, Southampton, UK
| |
Collapse
|
335
|
Gupta MS, Nicoll SB. Functional nucleus pulposus-like matrix assembly by human mesenchymal stromal cells is directed by macromer concentration in photocrosslinked carboxymethylcellulose hydrogels. Cell Tissue Res 2014; 358:527-39. [PMID: 25092545 DOI: 10.1007/s00441-014-1962-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 07/08/2014] [Indexed: 02/07/2023]
Abstract
Intervertebral disc (IVD) degeneration is associated with several pathophysiologic changes of the IVD, including dehydration of the nucleus pulposus (NP). Tissue engineering strategies may be used to restore both biological and mechanical function of the IVD following removal of NP tissue during surgical intervention. Recently, photocrosslinked carboxymethylcellulose (CMC) hydrogels were shown to support chondrogenic, NP-like extracellular matrix (ECM) elaboration by human mesenchymal stromal cells (hMSCs) when supplemented with TGF-β3; however, mechanical properties of these constructs did not reach native values. Fabrication parameters (i.e., composition, crosslinking density) can influence the bulk mechanical properties of hydrogel scaffolds, as well as cellular behavior and differentiation patterns. The objective of this study was to evaluate the influence of CMC macromer concentration (1.5, 2.5 and 3.5 % weight/volume) on bulk hydrogel properties and NP-like matrix elaboration by hMSCs. The lowest macromer concentration of 1.5 % exhibited the highest gene expression levels of aggrecan and collagen II at day 7, corresponding with the largest accumulation of glycosaminoglycans and collagen II by day 42. The ECM elaboration in the 1.5 % constructs was more homogeneously distributed compared to primarily pericellular localization in 3.5 % gels. The 1.5 % gels also displayed significant improvements in mechanical functionality by day 42 compared to earlier time points, which was not seen in the other groups. The effects of macromer concentration on matrix accumulation and organization are likely attributed to quantifiable differences in polymer crosslinking density and diffusive properties between the various hydrogel formulations. Taken together, these results demonstrate that macromer concentration of CMC hydrogels can direct hMSC matrix elaboration, such that a lower polymer concentration allows for greater NP-like ECM assembly and improvement of mechanical properties over time.
Collapse
Affiliation(s)
- Michelle S Gupta
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | | |
Collapse
|
336
|
Wang T, Lai JH, Han LH, Tong X, Yang F. Chondrogenic Differentiation of Adipose-Derived Stromal Cells in Combinatorial Hydrogels Containing Cartilage Matrix Proteins with Decoupled Mechanical Stiffness. Tissue Eng Part A 2014; 20:2131-9. [DOI: 10.1089/ten.tea.2013.0531] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Tianyi Wang
- Department of Bioengineering, Stanford University, Stanford, California
| | - Janice H. Lai
- Department of Mechanical Engineering, Stanford University, Stanford, California
| | - Li-Hsin Han
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Xinming Tong
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Fan Yang
- Department of Bioengineering, Stanford University, Stanford, California
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| |
Collapse
|
337
|
Caminal M, Fonseca C, Peris D, Moll X, Rabanal RM, Barrachina J, Codina D, García F, Cairó JJ, Gòdia F, Pla A, Vives J. Use of a chronic model of articular cartilage and meniscal injury for the assessment of long-term effects after autologous mesenchymal stromal cell treatment in sheep. N Biotechnol 2014; 31:492-8. [PMID: 25063342 DOI: 10.1016/j.nbt.2014.07.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 06/28/2014] [Accepted: 07/02/2014] [Indexed: 10/25/2022]
Abstract
Regenerative therapies using adult stem cells have attracted great interest in the recent years and offer a promising alternative to current surgical practices. In this report, we evaluated the safety and efficacy of an autologous cell-based treatment of osteoarthritis using mesenchymal stromal cells expanded from bone marrow aspirates that were administered intra-articularly. Ten 2-year old ewes were divided in two groups (for analysis at 6 and 12 months, respectively). Full thickness articular cartilage defects of approximately 60mm(2) were created arthroscopically in the medial femorotibial condyles and a meniscal tear in the anterior horn of the medial meniscus in the 20 hind legs. Intra-articular injection of 4 mL of either treatment (a suspension of cells) or control (same as treatment, without cells) were applied one month after generating a chronic condition similar to human pathology. Animals were monitored radiographically, by MRI and ultrasound scanning; and macroscopic and histological analyses were conducted at 6 and 12 months. Furthermore a full necropsy was performed at 12 months post-treatment. The intra-articular injection of autologous MSC was safe, as judged by the lack of local or systemic adverse effects during the clinical follow-up and by a full necropsy performed at 12 months post-treatment. Evidence of regeneration of articular cartilage and meniscus was case-dependent but statistically significant improvement was found in specific macroscopic and histological parameters. Such parameters included colour, rigidity, cell distribution and hyaline quality of the refill tissue as well as the structure of subchondral bone.
Collapse
Affiliation(s)
- Marta Caminal
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, Barcelona 08005, Spain
| | - Carla Fonseca
- Departament de Medicina i Cirurgia Animals, Àrea de Medicina i Cirurgia Animal, Facultat de Veterinària, Universitat Autònoma de Barcelona, Edifici V, Campus de la UAB, 08193 Bellaterra, Cerdanyola del Vallès, Spain
| | - David Peris
- Grup d'Enginyeria Cel·lular i Tissular, Departament d'Enginyeria Química, Escola d'Enginyers, Universitat Autònoma de Barcelona, Edifici Q, Campus de la UAB, 08193 Bellaterra, Cerdanyola del Vallès, Spain
| | - Xavier Moll
- Departament de Medicina i Cirurgia Animals, Àrea de Medicina i Cirurgia Animal, Facultat de Veterinària, Universitat Autònoma de Barcelona, Edifici V, Campus de la UAB, 08193 Bellaterra, Cerdanyola del Vallès, Spain
| | - Rosa M Rabanal
- Departament de Medicina i Cirurgia Animals, Àrea de Medicina i Cirurgia Animal, Facultat de Veterinària, Universitat Autònoma de Barcelona, Edifici V, Campus de la UAB, 08193 Bellaterra, Cerdanyola del Vallès, Spain
| | - Josep Barrachina
- Hospital ASEPEYO, Avinguda Alcalde Barnils, 54-60, Sant Cugat del Vallès, Barcelona 08174, Spain
| | - David Codina
- Hospital ASEPEYO, Avinguda Alcalde Barnils, 54-60, Sant Cugat del Vallès, Barcelona 08174, Spain
| | - Félix García
- Departament de Medicina i Cirurgia Animals, Àrea de Medicina i Cirurgia Animal, Facultat de Veterinària, Universitat Autònoma de Barcelona, Edifici V, Campus de la UAB, 08193 Bellaterra, Cerdanyola del Vallès, Spain
| | - Jordi J Cairó
- Grup d'Enginyeria Cel·lular i Tissular, Departament d'Enginyeria Química, Escola d'Enginyers, Universitat Autònoma de Barcelona, Edifici Q, Campus de la UAB, 08193 Bellaterra, Cerdanyola del Vallès, Spain
| | - Francesc Gòdia
- Grup d'Enginyeria Cel·lular i Tissular, Departament d'Enginyeria Química, Escola d'Enginyers, Universitat Autònoma de Barcelona, Edifici Q, Campus de la UAB, 08193 Bellaterra, Cerdanyola del Vallès, Spain
| | - Arnau Pla
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, Barcelona 08005, Spain
| | - Joaquim Vives
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, Barcelona 08005, Spain.
| |
Collapse
|
338
|
Alexander PG, Gottardi R, Lin H, Lozito TP, Tuan RS. Three-dimensional osteogenic and chondrogenic systems to model osteochondral physiology and degenerative joint diseases. Exp Biol Med (Maywood) 2014; 239:1080-95. [PMID: 24994814 DOI: 10.1177/1535370214539232] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Tissue engineered constructs have the potential to function as in vitro pre-clinical models of normal tissue function and disease pathogenesis for drug screening and toxicity assessment. Effective high throughput assays demand minimal systems with clearly defined performance parameters. These systems must accurately model the structure and function of the human organs and their physiological response to different stimuli. Musculoskeletal tissues present unique challenges in this respect, as they are load-bearing, matrix-rich tissues whose functionality is intimately connected to the extracellular matrix and its organization. Of particular clinical importance is the osteochondral junction, the target tissue affected in degenerative joint diseases, such as osteoarthritis (OA), which consists of hyaline articular cartilage in close interaction with subchondral bone. In this review, we present an overview of currently available in vitro three-dimensional systems for bone and cartilage tissue engineering that mimic native physiology, and the utility and limitations of these systems. Specifically, we address the need to combine bone, cartilage and other tissues to form an interactive microphysiological system (MPS) to fully capture the biological complexity and mechanical functions of the osteochondral junction of the articular joint. The potential applications of three-dimensional MPSs for musculoskeletal biology and medicine are highlighted.
Collapse
Affiliation(s)
- Peter G Alexander
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, 15219 USA
| | - Riccardo Gottardi
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, 15219 USA Ri.MED Foundation, Palermo, I-90133 Italy
| | - Hang Lin
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, 15219 USA
| | - Thomas P Lozito
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, 15219 USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, 15219 USA Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15261, USA Department of Mechanical Engineering and Materials Science, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15261, USA
| |
Collapse
|
339
|
Fischer J, Aulmann A, Dexheimer V, Grossner T, Richter W. Intermittent PTHrP(1-34) exposure augments chondrogenesis and reduces hypertrophy of mesenchymal stromal cells. Stem Cells Dev 2014; 23:2513-23. [PMID: 24836507 DOI: 10.1089/scd.2014.0101] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Phenotype instability and premature hypertrophy prevent the use of human mesenchymal stromal cells (MSCs) for cartilage regeneration. Aim of this study was to investigate whether intermittent supplementation of parathyroid hormone-related protein (PTHrP), as opposed to constant treatment, can beneficially influence MSC chondrogenesis and to explore molecular mechanisms below catabolic and anabolic responses. Human MSCs subjected to chondrogenic induction in high-density culture received PTHrP(1-34), forskolin, dbcAMP, or PTHrP(7-34) either constantly or via 6-h pulses (three times weekly), before proteoglycan, collagen type II, and X deposition; gene expression; and alkaline phosphatase (ALP) activity were assessed. While constant application of PTHrP(1-34) suppressed chondrogenesis of MSCs, pulsed application significantly increased collagen type 2 (COL2A1) gene expression and the collagen type II, proteoglycan, and DNA content of pellets after 6 weeks. Collagen type 10 (COL10A1) gene expression was little affected but Indian hedgehog (IHH) expression and ALP activity were significantly downregulated by pulsed PTHrP. A faster response to PTHrP exposure was recorded for ALP activity over COL2A1 regulation, suggesting that signal duration is critical for catabolic versus anabolic reactions. Stimulation of cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) signaling by forskolin reproduced major effects of both treatment modes, whereas application of PTHrP(7-34) capable of protein kinase C (PKC) signaling was ineffective. Pulsed PTHrP exposure of MSCs stimulated chondrogenesis and reduced endochondral differentiation apparently uncoupling chondrogenic matrix deposition from hypertrophic marker expression. cAMP/PKA was the major signaling pathway triggering the opposing effects of both treatment modes. Intermittent application of PTHrP represents an important novel means to improve chondrogenesis of MSCs and may be considered as a supporting clinical-treatment mode for MSC-based cartilage defect regeneration.
Collapse
Affiliation(s)
- Jennifer Fischer
- Research Center for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg , Heidelberg, Germany
| | | | | | | | | |
Collapse
|
340
|
Uehara Y, Hirose J, Yamabe S, Okamoto N, Okada T, Oyadomari S, Mizuta H. Endoplasmic reticulum stress-induced apoptosis contributes to articular cartilage degeneration via C/EBP homologous protein. Osteoarthritis Cartilage 2014; 22:1007-17. [PMID: 24795271 DOI: 10.1016/j.joca.2014.04.025] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 04/18/2014] [Accepted: 04/23/2014] [Indexed: 02/02/2023]
Abstract
OBJECTIVE When endoplasmic reticulum (ER) stress, i.e., the excessive accumulation of unfolded proteins in ER, endangers homeostasis, apoptosis is induced by C/EBP homologous protein (Chop). In osteoarthritis (OA) cartilage, Chop expression and apoptosis increase as degeneration progresses. We investigated the role of Chop in murine chondrocyte apoptosis and in the progression of cartilage degeneration. METHOD We induced experimental OA in Chop-knockout (Chop(-/-)) mice by medial collateral ligament transection and meniscectomy and compared cartilage degeneration, apoptosis, and ER stress in Chop(-/-)- and wild-type (Chop(+/+)) mice. In our in vitro experiments we treated murine Chop(-/-) chondrocytes with the ER stress inducer tunicamycin (TM) and evaluated apoptosis, ER stress, and chondrocyte function. RESULTS In vivo, the degree of ER stress was similar in Chop(-/-)- and Chop(+/+) mice. However, in Chop(-/-) mice apoptosis and cartilage degeneration were lower by 26.4% and 42.4% at 4 weeks, by 26.8% and 44.9% at 8 weeks, and by 26.9% and 32.3% at 12 weeks after surgery than Chop(+/+) mice, respectively. In vitro, the degree of ER stress induction by TM was similar in Chop(-/-)- and Chop(+/+) chondrocytes. On the other hand, apoptosis was 55.3% lower and the suppression of collagen type II and aggrecan mRNA was 21.0% and 23.3% less, and the increase of matrix metalloproteinase-13 mRNA was 20.0% less in Chop(-/-)- than Chop(+/+) chondrocytes. CONCLUSION Our results indicate that Chop plays a direct role in chondrocyte apoptosis and that Chop-mediated apoptosis contributes to the progression of cartilage degeneration in mice.
Collapse
Affiliation(s)
- Y Uehara
- Department of Orthopaedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan.
| | - J Hirose
- Department of Orthopaedic Surgery, Kumamoto University Hospital, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan.
| | - S Yamabe
- Department of Orthopaedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan.
| | - N Okamoto
- Department of Orthopaedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan.
| | - T Okada
- Department of Orthopaedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan.
| | - S Oyadomari
- Division of Molecular Biology, Institute for Genome Research, The University of Tokushima, 3-18-15 Kuramoto, Tokushima 770-8503, Japan.
| | - H Mizuta
- Department of Orthopaedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan.
| |
Collapse
|
341
|
Hubka KM, Dahlin RL, Meretoja VV, Kasper FK, Mikos AG. Enhancing chondrogenic phenotype for cartilage tissue engineering: monoculture and coculture of articular chondrocytes and mesenchymal stem cells. TISSUE ENGINEERING PART B-REVIEWS 2014; 20:641-54. [PMID: 24834484 DOI: 10.1089/ten.teb.2014.0034] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Articular cartilage exhibits an inherently low rate of regeneration. Consequently, damage to articular cartilage often requires surgical intervention. However, existing treatments generally result in the formation of fibrocartilage tissue, which is inferior to native articular cartilage. As a result, cartilage engineering strategies seek to repair or replace damaged cartilage with an engineered tissue that restores full functionality to the impaired joint. These strategies often involve the use of chondrocytes, yet in vitro expansion and culture can lead to undesirable changes in chondrocyte phenotype. This review focuses on the use of articular chondrocytes and mesenchymal stem cells (MSCs) in either monoculture or coculture for the enhancement of chondrogenesis. Coculture strategies increasingly outperform their monoculture counterparts with regard to chondrogenesis and present unique opportunities to attain chondrocyte phenotype stability in vitro. Methods to prevent chondrocyte dedifferentiation and promote chondrocyte redifferentiation as well as to promote the chondrogenic differentiation of MSCs while preventing MSC hypertrophy are discussed.
Collapse
Affiliation(s)
- Kelsea M Hubka
- Department of Bioengineering, Rice University , Houston, Texas
| | | | | | | | | |
Collapse
|
342
|
Sobh MA. Adipogenesis of Sprague Dawely rats mesenchymal stem cells: a morphological, immunophenotyping and gene expression follow-up study. Anat Cell Biol 2014; 47:83-90. [PMID: 24987544 PMCID: PMC4076425 DOI: 10.5115/acb.2014.47.2.83] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 05/21/2014] [Accepted: 05/28/2014] [Indexed: 01/14/2023] Open
Abstract
Mesenchymal stem cells (MSCs) offer significant promise as a multipotent source for cell-based therapies and could form the basis for the differentiation and cultivation of tissue grafts to replace damaged tissue. However, no gene expression follow up analysis has been undertaken to characterize the in vitro adipogenic differentiated MSCs. The main goal of this study was to focus on MSCs and to analyze their differentiation capacity. To achieve this aim, bone marrow MSCs from sprague dawely rats were isolated, expanded in monolayer culture and characterized with respect to their cluster of differentiation (CD) and ability for adipogenic differentiation capacity. The expression of CD44, CD45, CD29, CD34, and CD90 on bone marrow derived MSCs was characterized using flow cytometry. Adipogenesis was determined by staining with oil-red O and reverse transcription polymerase chain reaction assessments of lipoprotein lipase, leptin, adiponectin and adipocyte genes at different time intervals, after 4, 7, 14, and 21 days. Our results revealed that the pattern of CD marker expression was highly positive significant with CD29, CD44, and CD90 when compared with CD34 and CD45. MSCs showed proliferative potential and were capable of adipogenic differentiation characterized by reddish brown-droplets following staining with oil-red O and expression of molecular bands of genes. These results demonstrate, at the morphological, immunophenotyping and gene expression levels, the multipotency of MSCs and thus highlight their potential therapeutic value for cell-based tissue engineering.
Collapse
Affiliation(s)
- Mohamed A Sobh
- Department of Zoology, Urology and Nephrology Center, Research Center, Mansoura University, Cairo, Egypt
| |
Collapse
|
343
|
Harris Q, Seto J, O'Brien K, Lee PS, Kondo C, Heard BJ, Hart DA, Krawetz RJ. Monocyte chemotactic protein-1 inhibits chondrogenesis of synovial mesenchymal progenitor cells: an in vitro study. Stem Cells 2014; 31:2253-65. [PMID: 23836536 DOI: 10.1002/stem.1477] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 06/14/2013] [Indexed: 12/17/2022]
Abstract
Osteoarthritis (OA) is a multifactorial, often progressive, painful disease. OA often progresses with an apparent irreversible loss of articular cartilage, exposing underlying bone, resulting in pain and loss of mobility. This cartilage loss is thought to be permanent due to ineffective repair and apparent lack of stem/progenitor cells in that tissue. However, the adjacent synovial lining and synovial fluid are abundant with mesenchymal progenitor/stem cells (synovial mesenchymal progenitor cells [sMPCs]) capable of differentiating into cartilage both in vitro and in vivo. Previous studies have demonstrated that MPCs can home to factors such as monocyte chemotactic protein 1 (MCP-1/CCL2) expressed after injury. While MCP-1 (and its corresponding receptors) appears to play a role in recruiting stem cells to the site of injury, in this study, we have demonstrated that MCP-1 is upregulated in OA synovial fluid and that exposure to MCP-1 activates sMPCs, while concurrently inhibiting these cells from undergoing chondrogenesis in vitro. Furthermore, exposure to physiological (OA knee joint synovial fluid) levels of MCP-1 triggers changes in the transcriptome of sMPCs and prolonged exposure to the chemokine induces the expression of MCP-1 in sMPCs, resulting in a positive feedback loop from which sMPCs cannot apparently escape. Therefore, we propose a model where MCP-1 (normally expressed after joint injury) recruits sMPCs to the area of injury, but concurrently triggers changes in sMPC transcriptional regulation, leading to a blockage in the chondrogenic program. These results may open up new avenues of research into the lack of endogenous repair observed after articular cartilage injury and/or arthritis.
Collapse
Affiliation(s)
- Quinn Harris
- Department of Surgery, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | | | | | |
Collapse
|
344
|
Padmashali RM, Mistriotis P, Liang MS, Andreadis ST. Lentiviral arrays for live-cell dynamic monitoring of gene and pathway activity during stem cell differentiation. Mol Ther 2014; 22:1971-82. [PMID: 24895998 DOI: 10.1038/mt.2014.103] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 05/02/2014] [Indexed: 02/07/2023] Open
Abstract
Uncovering the complexity of mesenchymal stem cell (MSC) differentiation requires novel methods to capture the dynamics of the process in a quantitative and high-throughput manner. To this end, we developed a lentiviral array (LVA) of reporters to capture the dynamics of gene and pathway activity during MSC differentiation into adipogenic, chondrogenic, and osteogenic lineages. Our results identified signature promoters and pathways with unique activation profile for each MSC lineage. In combination with chemical inhibitors, lineage-specific reporters predicted the effects of signaling pathway perturbations on MSC differentiation. Interestingly, some pathways were critical for differentiation into all lineages, while others had differential effects on each lineage. Our study suggests that when combined with large chemical or siRNA libraries, the reporter LVA can be used to uncover novel genes and signaling pathways affecting complex biological processes such as stem cell differentiation or reprogramming.
Collapse
Affiliation(s)
- Roshan M Padmashali
- Department of Chemical and Biological Engineering, Bioengineering Laboratory, University at Buffalo, The State University of New York, Amherst, New York, USA
| | - Panagiotis Mistriotis
- Department of Chemical and Biological Engineering, Bioengineering Laboratory, University at Buffalo, The State University of New York, Amherst, New York, USA
| | - Mao-shih Liang
- Department of Chemical and Biological Engineering, Bioengineering Laboratory, University at Buffalo, The State University of New York, Amherst, New York, USA
| | - Stelios T Andreadis
- 1] Department of Chemical and Biological Engineering, Bioengineering Laboratory, University at Buffalo, The State University of New York, Amherst, New York, USA [2] Department of Biomedical Engineering, University at Buffalo, The State University of New York, New York, Amherst, USA [3] Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York, USA
| |
Collapse
|
345
|
Green DE, Rubin CT. Consequences of irradiation on bone and marrow phenotypes, and its relation to disruption of hematopoietic precursors. Bone 2014; 63:87-94. [PMID: 24607941 PMCID: PMC4005928 DOI: 10.1016/j.bone.2014.02.018] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 01/07/2014] [Accepted: 02/26/2014] [Indexed: 02/06/2023]
Abstract
The rising levels of radiation exposure, specifically for medical treatments and accidental exposures, have added great concern for the long term risks of bone fractures. Both the bone marrow and bone architecture are devastated following radiation exposure. Even sub-lethal doses cause a deficit to the bone marrow microenvironment, including a decline in hematopoietic cells, and this deficit occurs in a dose dependent fashion. Certain cell phenotypes though are more susceptible to radiation damage, with mesenchymal stem cells being more resilient than the hematopoietic stem cells. The decline in total bone marrow hematopoietic cells is accompanied with elevated adipocytes into the marrow cavity, thereby inhibiting hematopoiesis and recovery of the bone marrow microenvironment. Poor bone marrow is also associated with a decline in bone architectural quality. Therefore, the ability to maintain the bone marrow microenvironment would hinder much of the trabecular bone loss caused by radiation exposure, ultimately decreasing some comorbidities in patients exposed to radiation.
Collapse
Affiliation(s)
- Danielle E Green
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794-5281, USA.
| | - Clinton T Rubin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794-5281, USA
| |
Collapse
|
346
|
Ahmed N, Iu J, Brown CE, Taylor DW, Kandel RA. Serum- and growth-factor-free three-dimensional culture system supports cartilage tissue formation by promoting collagen synthesis via Sox9-Col2a1 interaction. Tissue Eng Part A 2014; 20:2224-33. [PMID: 24606204 DOI: 10.1089/ten.tea.2013.0559] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE One of the factors preventing clinical application of regenerative medicine to degenerative cartilage diseases is a suitable source of cells. Chondrocytes, the only cell type of cartilage, grown in vitro under culture conditions to expand cell numbers lose their phenotype along with the ability to generate hyaline cartilaginous tissue. In this study we determine that a serum- and growth-factor-free three-dimensional (3D) culture system restores the ability of the passaged chondrocytes to form cartilage tissue in vitro, a process that involves sox9. METHODS Bovine articular chondrocytes were passaged twice to allow for cell number expansion (P2) and cultured at high density on 3D collagen-type-II-coated membranes in high glucose content media supplemented with insulin and dexamethasone (SF3D). The cells were characterized after monolayer expansion and following 3D culture by flow cytometry, gene expression, and histology. The early changes in signaling transduction pathways during redifferentiation were characterized. RESULTS The P2 cells showed a progenitor-like antigen profile of 99% CD44(+) and 40% CD105(+) and a gene expression profile suggestive of interzone cells. P2 in SF3D expressed chondrogenic genes and accumulated extracellular matrix. Downregulating insulin receptor (IR) with HNMPA-(AM3) or the PI-3/AKT kinase pathway (activated by insulin treatment) with Wortmannin inhibited collagen synthesis. HNMPA-(AM3) reduced expression of Col2, Col11, and IR genes as well as Sox6 and -9. Co-immunoprecipitation and chromatin immunoprecipitation analyses of HNMPA-(AM3)-treated cells showed binding of the coactivators Sox6 and Med12 with Sox9 but reduced Sox9-Col2a1 binding. CONCLUSIONS We describe a novel culture method that allows for increase in the number of chondrocytes and promotes hyaline-like cartilage tissue formation in part by insulin-mediated Sox9-Col2a1 binding. The suitability of the tissue generated via this approach for use in joint repair needs to be examined in vivo.
Collapse
Affiliation(s)
- Nazish Ahmed
- 1 CIHR-BioEngineering of Skeletal Tissues Team, Mount Sinai Hospital, University of Toronto , Toronto, Canada
| | | | | | | | | |
Collapse
|
347
|
Kalpakci KN, Brown WE, Hu JC, Athanasiou KA. Cartilage tissue engineering using dermis isolated adult stem cells: the use of hypoxia during expansion versus chondrogenic differentiation. PLoS One 2014; 9:e98570. [PMID: 24867063 PMCID: PMC4035316 DOI: 10.1371/journal.pone.0098570] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 05/04/2014] [Indexed: 11/25/2022] Open
Abstract
Dermis isolated adult stem (DIAS) cells, a subpopulation of dermis cells capable of chondrogenic differentiation in the presence of cartilage extracellular matrix, are a promising source of autologous cells for tissue engineering. Hypoxia, through known mechanisms, has profound effects on in vitro chondrogenesis of mesenchymal stem cells and could be used to improve the expansion and differentiation processes for DIAS cells. The objective of this study was to build upon the mechanistic knowledge of hypoxia and translate it to tissue engineering applications to enhance chondrogenic differentiation of DIAS cells through exposure to hypoxic conditions (5% O2) during expansion and/or differentiation. DIAS cells were isolated and expanded in hypoxic (5% O2) or normoxic (20% O2) conditions, then differentiated for 2 weeks in micromass culture on chondroitin sulfate-coated surfaces in both environments. Monolayer cells were examined for proliferation rate and colony forming efficiency. Micromasses were assessed for cellular, biochemical, and histological properties. Differentiation in hypoxic conditions following normoxic expansion increased per cell production of collagen type II 2.3 fold and glycosaminoglycans 1.2 fold relative to continuous normoxic culture (p<0.0001). Groups expanded in hypoxia produced 51% more collagen and 23% more GAGs than those expanded in normoxia (p<0.0001). Hypoxia also limited cell proliferation in monolayer and in 3D culture. Collectively, these data show hypoxic differentiation following normoxic expansion significantly enhances chondrogenic differentiation of DIAS cells, improving the potential utility of these cells for cartilage engineering.
Collapse
Affiliation(s)
- Kerem N. Kalpakci
- Medtronic Spine & Biologics, Memphis, Tennessee, United States of America
| | - Wendy E. Brown
- UC Davis, Department of Biomedical Engineering, Davis, California, United States of America
| | - Jerry C. Hu
- UC Davis, Department of Biomedical Engineering, Davis, California, United States of America
| | - Kyriacos A. Athanasiou
- UC Davis, Department of Biomedical Engineering, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
348
|
Somoza RA, Welter JF, Correa D, Caplan AI. Chondrogenic differentiation of mesenchymal stem cells: challenges and unfulfilled expectations. TISSUE ENGINEERING PART B-REVIEWS 2014; 20:596-608. [PMID: 24749845 DOI: 10.1089/ten.teb.2013.0771] [Citation(s) in RCA: 252] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Articular cartilage repair and regeneration provides a substantial challenge in Regenerative Medicine because of the high degree of morphological and mechanical complexity intrinsic to hyaline cartilage due, in part, to its extracellular matrix. Cartilage remains one of the most difficult tissues to heal; even state-of-the-art regenerative medicine technology cannot yet provide authentic cartilage resurfacing. Mesenchymal stem cells (MSCs) were once believed to be the panacea for cartilage repair and regeneration, but despite years of research, they have not fulfilled these expectations. It has been observed that MSCs have an intrinsic differentiation program reminiscent of endochondral bone formation, which they follow after exposure to specific reagents as a part of current differentiation protocols. Efforts have been made to avoid the resulting hypertrophic fate of MSCs; however, so far, none of these has recreated a fully functional articular hyaline cartilage without chondrocytes exhibiting a hypertrophic phenotype. We reviewed the current literature in an attempt to understand why MSCs have failed to regenerate articular cartilage. The challenges that must be overcome before MSC-based tissue engineering can become a front-line technology for successful articular cartilage regeneration are highlighted.
Collapse
Affiliation(s)
- Rodrigo A Somoza
- Department of Biology, Skeletal Research Center, Case Western Reserve University , Cleveland, Ohio
| | | | | | | |
Collapse
|
349
|
New strategies for cartilage regeneration exploiting selected glycosaminoglycans to enhance cell fate determination. Biochem Soc Trans 2014; 42:703-9. [DOI: 10.1042/bst20140031] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Most research strategies for cartilage tissue engineering use extended culture with complex media loaded with costly GFs (growth factors) to drive tissue assembly and yet they result in the production of cartilage with inferior mechanical and structural properties compared with the natural tissue. Recent evidence suggests that GAGs (glycosaminoglycans) incorporated into tissue engineering scaffolds can sequester and/or activate GFs and thereby more effectively mimic the natural ECM (extracellular matrix). Such approaches may have potential for the improvement of cartilage engineering. However, natural GAGs are structurally complex and heterogeneous, making structure–function relationships hard to determine and clinical translation difficult. Importantly, subfractions of GAGs with specific chain lengths and sulfation patterns have been shown to activate key signalling processes during stem cell differentiation. In addition, recently, GAGs have been bound to synthetic biomaterials, such as electrospun scaffolds and hydrogels, in biologically active conformations, and methods to purify and select affinity-matched GAGs for specific GFs have also been developed. The identification and use of specific GAG moieties to promote chondrogenesis is therefore an exciting new avenue of research. Combining these with synthetic biomaterials may allow a more effective mimicry of the natural ECM, reduction in the need for expensive GFs, and perhaps the deposition of an articular cartilage-like matrix in a clinically relevant manner.
Collapse
|
350
|
Qin H, Zhu C, An Z, Jiang Y, Zhao Y, Wang J, Liu X, Hui B, Zhang X, Wang Y. Silver nanoparticles promote osteogenic differentiation of human urine-derived stem cells at noncytotoxic concentrations. Int J Nanomedicine 2014; 9:2469-78. [PMID: 24899804 PMCID: PMC4038528 DOI: 10.2147/ijn.s59753] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
In tissue engineering, urine-derived stem cells are ideal seed cells and silver nanoparticles (AgNPs) are perfect antimicrobial agents. Due to a distinct lack of information on the effects of AgNPs on urine-derived stem cells, a study was conducted to evaluate the effects of silver ions and AgNPs upon the cytotoxicity and osteogenic differentiation of urine-derived stem cells. Initially, AgNPs or AgNO3 were exposed to urine-derived stem cells for 24 hours. Cytotoxicity was measured using the Cell Counting kit-8 (CCK-8) test. The effects of AgNPs or AgNO3 at the maximum safety concentration determined by the CCK-8 test on osteogenic differentiation of urine-derived stem cells were assessed by alkaline phosphatase activity, Alizarin Red S staining, and the quantitative reverse transcription polymerase chain reaction. Lastly, the effects of AgNPs or AgNO3 on “urine-derived stem cell actin cytoskeleton organization” and RhoA activity were assessed by rhodamine-phalloidin staining and Western blotting. Concentration-dependent toxicity was observed starting at an AgNO3 concentration of 2 μg/mL and at an AgNP concentration of 4 μg/mL. At these concentrations, AgNPs were observed to promote osteogenic differentiation of urine-derived stem cells, induce actin polymerization and increase cytoskeletal tension, and activate RhoA; AgNO3 had no such effects. In conclusion, AgNPs can promote osteogenic differentiation of urine-derived stem cells at a suitable concentration, independently of silver ions, and are suitable for incorporation into tissue-engineered scaffolds that utilize urine-derived stem cells as seed cells.
Collapse
Affiliation(s)
- Hui Qin
- Department of Orthopedics, Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Chen Zhu
- Department of Orthopaedic Surgery, Provincial Hospital Affiliated to Anhui Medical University, HeFei, People's Republic of China
| | - Zhiquan An
- Department of Orthopedics, Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yao Jiang
- Department of Orthopedics, Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yaochao Zhao
- Department of Orthopedics, Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jiaxin Wang
- Department of Orthopedics, Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xin Liu
- Department of Orthopedics, Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Bing Hui
- Department of Orthopedics, Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xianlong Zhang
- Department of Orthopedics, Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yang Wang
- Department of Orthopedics, Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|