451
|
Li X, Zhang M, Bai L, Bai W, Xu W, Zhu H. Effects of 50 Hz pulsed electromagnetic fields on the growth and cell cycle arrest of mesenchymal stem cells: an in vitro study. Electromagn Biol Med 2012; 31:356-64. [PMID: 22676915 DOI: 10.3109/15368378.2012.662194] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Xinping Li
- Department of Rehabilitation, Guangdong Geriatric Institute, Guangdong Academy of Medical Sciences and Guangdong General Hospital
Guangzhou, China
| | - Mingsheng Zhang
- Department of Rehabilitation, Guangdong Geriatric Institute, Guangdong Academy of Medical Sciences and Guangdong General Hospital
Guangzhou, China
| | - Liming Bai
- Department of Rehabilitation, Guangdong Geriatric Institute, Guangdong Academy of Medical Sciences and Guangdong General Hospital
Guangzhou, China
| | - Wenfang Bai
- Department of Rehabilitation, Guangdong Geriatric Institute, Guangdong Academy of Medical Sciences and Guangdong General Hospital
Guangzhou, China
| | - Weicheng Xu
- Department of Rehabilitation, Guangdong Geriatric Institute, Guangdong Academy of Medical Sciences and Guangdong General Hospital
Guangzhou, China
| | - Hongxiang Zhu
- Department of Rehabilitation, Guangdong Geriatric Institute, Guangdong Academy of Medical Sciences and Guangdong General Hospital
Guangzhou, China
| |
Collapse
|
452
|
Lubelski D, Abdullah KG, Benzel EC, Mroz TE. The Utility of Allograft Mesenchymal Stem Cells for Spine Fusion: A Literature Review. Global Spine J 2012; 2:109-14. [PMID: 27054055 PMCID: PMC4813091 DOI: 10.1055/s-0032-1307263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
More than 50% of patients complain of postoperative donor site morbidity following iliac crest bone graft harvest, and recent discoveries have identified adverse outcomes following bone morphogenetic protein use in spine fusion. This has led the spine community to turn toward alternative methods to promote fusion following spine surgery. The present article reviews numerous studies that have shown the osteogenic potential of mesenchymal stem cells (MSCs). MSCs have been used with both in vitro and in vivo models and have involved animal studies ranging from rats to macaque monkeys to successfully induce bone regeneration in lesions of the tibia and spine. There is no fear of graft rejection, as there may be with other allograft materials, because neither undifferentiated nor differentiated MSCs elicit lymphocyte response when transplanted; they tend to alter the cytokine profile to an anti-inflammatory state. Early clinical trials are underway with various commercially available MSC formulations. Although there is much enthusiasm, it is integral that the spine surgery community carefully evaluate the use of MSCs in spine fusion through well-designed and executed studies to determine the efficacy and safety profiles in spine surgery patients.
Collapse
Affiliation(s)
- Daniel Lubelski
- Department of Neurological Surgery, Cleveland Clinic Center for Spine Health,
Cleveland, Ohio
| | - Kalil G. Abdullah
- Department of Neurological Surgery, Cleveland Clinic Center for Spine Health,
Cleveland, Ohio
| | - Edward C. Benzel
- Department of Neurological Surgery, Cleveland Clinic Center for Spine Health,
Cleveland, Ohio
| | - Thomas E. Mroz
- Department of Neurological Surgery, Cleveland Clinic Center for Spine Health,
Cleveland, Ohio
| |
Collapse
|
453
|
Fierro FA, Kalomoiris S, Sondergaard CS, Nolta JA. Effects on proliferation and differentiation of multipotent bone marrow stromal cells engineered to express growth factors for combined cell and gene therapy. Stem Cells 2012; 29:1727-37. [PMID: 21898687 DOI: 10.1002/stem.720] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A key mechanism for mesenchymal stem cells/bone marrow stromal cells (MSCs) to promote tissue repair is by secretion of soluble growth factors (GFs). Therefore, clinical application could be optimized by a combination of cell and gene therapies, where MSCs are genetically modified to express higher levels of a specific factor. However, it remains unknown how this overexpression may alter the fate of the MSCs. Here, we show effects of overexpressing the growth factors, such as basic fibroblast growth factor (bFGF), platelet derived growth factor B (PDGF-BB), transforming growth factor β(1) (TGF-β(1) ), and vascular endothelial growth factor (VEGF), in human bone marrow-derived MSCs. Ectopic expression of bFGF or PDGF-B lead to highly proliferating MSCs and lead to a robust increase in osteogenesis. In contrast, adipogenesis was strongly inhibited in MSCs overexpressing PDGF-B and only mildly affected in MSCs overexpressing bFGF. Overexpression of TGF-β(1) blocked both osteogenic and adipogenic differentiation while inducing the formation of stress fibers and increasing the expression of the smooth muscle marker calponin-1 and the chondrogenic marker collagen type II. In contrast, MSCs overexpressing VEGF did not vary from control MSCs in any parameters, likely due to the lack of VEGF receptor expression on MSCs. MSCs engineered to overexpress VEGF strongly induced the migration of endothelial cells and enhanced blood flow restoration in a xenograft model of hind limb ischemia. These data support the rationale for genetically modifying MSCs to enhance their therapeutically relevant trophic signals, when safety and efficacy can be demonstrated, and when it can be shown that there are no unwanted effects on their proliferation and differentiation.
Collapse
Affiliation(s)
- Fernando A Fierro
- Institute for Regenerative Cures, University of California, Davis, California 95817, USA.
| | | | | | | |
Collapse
|
454
|
Xue JX, Gong YY, Zhou GD, Liu W, Cao Y, Zhang WJ. Chondrogenic differentiation of bone marrow-derived mesenchymal stem cells induced by acellular cartilage sheets. Biomaterials 2012; 33:5832-40. [PMID: 22608213 DOI: 10.1016/j.biomaterials.2012.04.054] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 04/23/2012] [Indexed: 10/28/2022]
Abstract
Acellular cartilage sheets (ACSs) have been used as scaffolds for engineering cartilage with mature chondrocytes. In this study we investigated whether ACSs possess a chondrogenic induction activity that may benefit cartilage engineering with multipotent stem cells. Bone marrow-derived mesenchymal stem cells (BMSCs) isolated from newborn pigs were expanded in vitro and seeded on ACSs that were then stacked layer-by-layer to form BMSC-ACS constructs. Cells seeded on polyglycolic acid/polylactic acid (PGA/PLA) scaffolds served as a control. After 4 weeks of culture with or without additional chondrogenic factors, constructs were subcutaneously implanted into nude mice for another 4 weeks. Cartilage-like tissues were formed after 4 weeks of culture. However, formation of cartilage with a typical lacunar structure was only observed in induced groups. RT-PCR showed that aggrecan, COMP, type II collagen and Sox9 were expressed in all groups except the non-induced BMSC-PGA/PLA group. At 4 weeks post-implantation, cartilage formation was achieved in the induced BMSC-ACS group and partial cartilage formation was achieved in the non-induced BMSC-ACS group, confirmed by safranin O staining, toluidine blue staining and type II collagen immunostaining. In addition, enzyme-linked immunosorbent assay demonstrated the presence of transforming growth factor-β1, insulin-like growth factor-1 and bone morphogenic protein-2 in ACSs. These results indicate that ACSs possess a chondrogenic induction activity that promotes BMSC differentiation.
Collapse
Affiliation(s)
- Ji Xin Xue
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, 639 Zhi Zao Ju Road, Shanghai 200011, China
| | | | | | | | | | | |
Collapse
|
455
|
Diederichs S, Baral K, Tanner M, Richter W. Interplay between local versus soluble transforming growth factor-beta and fibrin scaffolds: role of cells and impact on human mesenchymal stem cell chondrogenesis. Tissue Eng Part A 2012; 18:1140-50. [PMID: 22480213 DOI: 10.1089/ten.tea.2011.0426] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Structural extracellular matrix molecules gain increasing attention as scaffolds for cartilage tissue engineering owing to their natural role as a growth factor repository. We recently observed that a collagen-type I/III (Col-I/III) matrix, human recombinant transforming growth factor-beta (TGF-β) protein, and fibrin hydrogel (FG) combined to a biphasic construct provided sufficient long-term TGF-β support to drive in vitro chondrogenesis of human mesenchymal stem cells (hMSC). Here we ask whether FG and Col-I/III can both retain TGF-β, describe the influence of cell seeding on TGF-β release, and compare the molecular path of hMSC chondrogenic differentiation under soluble versus local TGF-β supply. Release of growth factor from scaffolds augmented with increasing amounts of TGF-β was analyzed over 7 days and chondrogenesis was assessed over 42 days. Low TGF-β release rates from Col-I/III as opposed to higher release from FG indicated that both molecules retained TGF-β, with Col-I/III being the superior storage component. Cell seeding enhanced TGF-β retention in FG by about threefold and almost stopped release beyond 24 h. TGF-β remained bioactive and supported MSC chondrogenesis without impairing the amount of proteoglycan and collagen-type II deposition per cell and per construct compared to standard scaffold-free MSC pellets supplied with soluble TGF-β. Local TGF-β, however, mediated lower cell content, less collagen-type X relative to collagen-type II deposition and no matrix metalloproteinase-13 up-regulation. In conclusion, cells quickly halted release of local TGF-β from FG, turning FG and Col-I/III into attractive TGF-β repositories capable to drive full hMSC chondrogenesis, but via a modulated differentiation pathway. Since only part of the changes was reproduced by transient soluble TGF-β supply, release kinetics alone could not explain the molecular differences, suggesting that local TGF-β acts distinct from its soluble counterpart.
Collapse
Affiliation(s)
- Solvig Diederichs
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | | | | | | |
Collapse
|
456
|
Liu YL, Wang YD, Zhuang F, Xian SL, Fang JY, Su W, Zhang W. Immunosuppression effects of bone marrow mesenchymal stem cells on renal interstitial injury in rats with unilateral ureteral obstruction. Cell Immunol 2012; 276:144-52. [PMID: 22632816 DOI: 10.1016/j.cellimm.2012.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 04/20/2012] [Accepted: 05/02/2012] [Indexed: 01/16/2023]
Abstract
We investigated the effects of intravenously administered bone marrow mesenchymal stem cells (BMSCs) on renal interstitial inflammation and fibrosis. In unilateral ureteral obstruction (UUO) rats, the CD4(+)CD25(+) regulatory T-cell (Treg) cell, macrophage population and some inflammation related cytokines were tested. In the BMSCs -treated rats, renal exhibited lower renal Masson scores, decreased macrophage infiltration and interferon gamma (IFNγ) expression, and increased forkhead transcription factor (Foxp3) and interleukin-10 (IL-10) expression. No significant differences in the CD4(+)CD25(+) Treg population and renal transforming growth factor-β1 (TGFβ1) expression were observed between BMSCs-treated group and control group (p>0.05). In conclusion, BMSCs infusion leads to an anti-inflammation response in the early stage of UUO which may related to paracine mechanism.
Collapse
Affiliation(s)
- Ying-Li Liu
- The Department of Nephrology, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, Shanghai 200011, China
| | | | | | | | | | | | | |
Collapse
|
457
|
Takamoto T, Ichinohe N, Tabata Y. Proliferation of rat mesenchymal stem cells in collagen sponges reinforced with poly(ethylene terephthalate) fibers by stirring culture method. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2012; 23:1741-53. [PMID: 21943688 DOI: 10.1163/156856211x598184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The objective of this study is to investigate the effect of medium stirring conditions on the proliferation of rat mesenchymal stem cells (MSC) in collagen sponges reinforced by the incorporation of poly(ethylene terephthalate) (PET) fibers. A collagen solution with PET fibers homogeneously dispersed was freeze-dried, followed by dehydrothermal cross-linking to obtain a collagen sponge incorporating PET fibers. MSC were proliferated in the sponge by a stirring culture method. The PET fibers reinforcement significantly suppressed the sponge deformation in culture. The MSC proliferation was enhanced by the stirring culture to a significantly higher extent than that of a static one. Homogeneous distribution of cells proliferated was observed at the stirring rate of 50 rpm and compared with that at lower and higher rates. Combination of the PET fiber-reinforced sponge with the stirring culture method is a promising way to allow cells to homogeneously proliferate in the sponge.
Collapse
Affiliation(s)
- Tomoaki Takamoto
- a Department of Biomaterials , Field of Tissue Engineering, Institute for Frontier Medical Sciences, Kyoto University , 53 Kawara-cho Shogoin , Sakyo-ku Kyoto , 606-8507 , Japan
| | | | | |
Collapse
|
458
|
Lewis G. Nucleus pulposus replacement and regeneration/repair technologies: present status and future prospects. J Biomed Mater Res B Appl Biomater 2012; 100:1702-20. [PMID: 22566484 DOI: 10.1002/jbm.b.32712] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 02/19/2012] [Accepted: 03/18/2012] [Indexed: 12/28/2022]
Abstract
Degenerative disc disease is implicated in the pathogenesis of many painful conditions of the back, chief among which is low back pain. Acute and/or chronic low back pain (A/CLBP) afflicts a large number of people, thus making it a major healthcare issue with concomitant cost ramifications. When conservative treatments for A/CLBP, such as bed rest, anti-inflammatory medications, and physical therapy, prove to be ineffectual, surgical options are recommended. The most popular of these is discectomy followed by fusion. Although there are many reports of good to excellent outcomes with this method, there are concerns, such as long-term adverse biomechanical consequences to adjacent functional spinal unit(s). A surgical option that has been attracting much attention recently is replacement or regeneration/repair of the nucleus pulposus, an approach that holds the prospect of not compromising either mobility or function and causing no adjacent-level injury. There is a sizeable body of literature highlighting this option, comprising in vitro biomechanical studies, finite element analyses, animal-model studies, and limited clinical evaluations. This work is a review of this body of literature and is organized into four parts, with the focus being on replacement technologies, regeneration/repair technologies, and detailed expositions on 14 areas for future study. This review ends with a summary of the salient points made.
Collapse
Affiliation(s)
- Gladius Lewis
- Department of Mechanical Engineering, The University of Memphis, Memphis, Tennessee 38152, USA.
| |
Collapse
|
459
|
Yun YR, Jang JH, Jeon E, Kang W, Lee S, Won JE, Kim HW, Wall I. Administration of growth factors for bone regeneration. Regen Med 2012; 7:369-85. [DOI: 10.2217/rme.12.1] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Growth factors (GFs) such as BMPs, FGFs, VEGFs and IGFs have significant impacts on osteoblast behavior, and thus have been widely utilized for bone tissue regeneration. Recently, securing biological stability for a sustainable and controllable release to the target tissue has been a challenge to practical applications. This challenge has been addressed to some degree with the development of appropriate carrier materials and delivery systems. This review highlights the importance and roles of those GFs, as well as their proper administration for targeting bone regeneration. Additionally, the in vitro and in vivo performance of those GFs with or without the use of carrier systems in the repair and regeneration of bone tissue is systematically addressed. Moreover, some recent advances in the utility of the GFs, such as using fusion technology, are also reviewed.
Collapse
Affiliation(s)
- Ye-Rang Yun
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 330-714, Korea
| | - Jun Hyeog Jang
- Department of Biochemistry, Inha University School of Medicine, Incheon 400-712, Korea
| | - Eunyi Jeon
- Department of Biochemistry, Inha University School of Medicine, Incheon 400-712, Korea
| | - Wonmo Kang
- Department of Biochemistry, Inha University School of Medicine, Incheon 400-712, Korea
| | - Sujin Lee
- Department of Biochemistry, Inha University School of Medicine, Incheon 400-712, Korea
| | - Jong-Eun Won
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 330-714, Korea
- Department of Nanobiomedical Science & WCU Research Center, Dankook University Graduate School, Cheonan 330-714, Korea
| | - Hae Won Kim
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan 330-714, Korea
| | - Ivan Wall
- Department of Nanobiomedical Science & WCU Research Center, Dankook University Graduate School, Cheonan 330-714, Korea
- Department of Biochemical Engineering, University College London, Torrington Place, London WC1E 7JE, UK
| |
Collapse
|
460
|
CHIANG HONGSEN, HUANG YIYOU, JIANG CHINGCHUAN. REPAIR OF ARTICULAR CARTILAGE INJURY. BIOMEDICAL ENGINEERING-APPLICATIONS BASIS COMMUNICATIONS 2012. [DOI: 10.4015/s1016237205000366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Articular cartilage defects heal poorly and lead to consequences as osteoarthritis. Clinical experience has indicated that no existing medication would substantially promote the healing process, and the cartilage defect requires surgical replacement. Allograft decays quickly for multiple reasons including the preparation process and immune reaction, and the outcome is disappointing. The extreme shortage of sparing in articular cartilage has much discouraged the use of autograft, which requires modification. The concept that constructs a chondral or osteochondral construct for the replacement of injured native tissue introduces that of tissue engineering. Limited number of cells are expanded either in vitro or in vivo, and resided temporally on a scaffold of biomaterial, which also acts as a vehicle to transfer the cells to the recipient site. Three core elements constitute this technique: the cell, a biodegradable scaffold, and an environment suitable for cells to present their proposed activity. Modern researches have kept updating those elements for a better performance of such cultivation of living tissue.
Collapse
Affiliation(s)
- HONGSEN CHIANG
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
- National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - YI-YOU HUANG
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - CHING-CHUAN JIANG
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
- National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
461
|
Khanmohammadi M, Khanjani S, Bakhtyari MS, Zarnani AH, Edalatkhah H, Akhondi MM, Mirzadegan E, Kamali K, Alimoghadam K, Kazemnejad S. Proliferation and chondrogenic differentiation potential of menstrual blood- and bone marrow-derived stem cells in two-dimensional culture. Int J Hematol 2012; 95:484-93. [PMID: 22527849 DOI: 10.1007/s12185-012-1067-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Revised: 03/15/2012] [Accepted: 03/22/2012] [Indexed: 12/18/2022]
Abstract
Menstrual blood is easily accessible, renewable, and inexpensive source of stem cells. In this study, we investigated the chondrogenic differentiation potential of menstrual blood-derived stem cells (MenSCs) compared with that of bone marrow-derived stem cells (BMSCs) in two-dimensional culture. Following characterization of isolated cells, the potential for chondrogenic differentiation of MenSCs and BMSCs was evaluated by immunocytochemical and molecular experiments. MenSCs were strongly positive for mesenchymal stem cell markers in a manner similar to that of BMSCs. In contrast to BMSCs, MenSCs exhibited marked expression of OCT4, and higher proliferative capacity. Differentiated MenSCs showed strong immunoreactivity to a monoclonal antibody against Collagen type 2, in a pattern similar to BMSCs. Accumulation of proteoglycans in differentiated MenSCs was also comparable with that in differentiated BMSCs. However, the mRNA expression patterns as judged by RT-PCR of chondrogenic markers such as Collagen 2A1, Collagen 9A1 and SOX9 in MenSCs were different from those in BMSCs. Given these findings, MenSCs appear to be a unique stem cell population with higher proliferation than and comparable chondrogenic differentiation ability to BMSCs in two-dimensional culture. Much quantitative studies at the molecular level may elucidate the reasons for the observed differences in MenSCs and BMSCs.
Collapse
Affiliation(s)
- Manijeh Khanmohammadi
- Department of Embryology and Stem Cells, Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Evin, P.O. Box 19615-1177, Tehran, Iran
| | | | | | | | | | | | | | | | | | | |
Collapse
|
462
|
Casado JG, Gomez-Mauricio G, Alvarez V, Mijares J, Tarazona R, Bernad A, Sanchez-Margallo FM. Comparative phenotypic and molecular characterization of porcine mesenchymal stem cells from different sources for translational studies in a large animal model. Vet Immunol Immunopathol 2012; 147:104-12. [PMID: 22521281 DOI: 10.1016/j.vetimm.2012.03.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 03/23/2012] [Accepted: 03/26/2012] [Indexed: 01/30/2023]
Abstract
Mesenchymal stem cells have demonstrated their potentiality for therapeutic use in treating diseases or repairing damaged tissues. However, in some cases, the results of clinical trials have been disappointing or have not worked out as well as hoped. These disappointing results can be attributed to an inadequate or insufficient preclinical study. For medical and surgical purposes, the similarities between the anatomy of pig and human make this animal an attractive preclinical model. In this sense, for mesenchymal stem cell-based therapy, it is strongly necessary to have well characterized animal-derived mesenchymal stem cell lines to validate preclinical effectiveness of these cells. In this work, porcine mesenchymal stem cells (pMSCs) were isolated from bone marrow, adipose tissue and peripheral blood and compared in terms of differentiation potential, cell surface markers and gene expression. Our results demonstrated that the isolation and in vitro expansion protocols were feasible and effective. The data presented in this work are relevant because they provide an extensive phenotypic characterization; genetic study and differentiation behavior of the most commonly used stem cell lines for clinical practices. These pMSCs are widely available to scientists and could be a valuable tool to evaluate the safety and efficacy of adoptively transferred cells.
Collapse
Affiliation(s)
- Javier G Casado
- Stem Cell Therapy Unit, Minimally Invasive Surgery Centre Jesus Uson, Caceres, Spain.
| | | | | | | | | | | | | |
Collapse
|
463
|
Colombini A, Lanteri P, Lombardi G, Grasso D, Recordati C, Lovi A, Banfi G, Bassani R, Brayda-Bruno M. Metabolic effects of vitamin D active metabolites in monolayer and micromass cultures of nucleus pulposus and annulus fibrosus cells isolated from human intervertebral disc. Int J Biochem Cell Biol 2012; 44:1019-30. [PMID: 22481027 DOI: 10.1016/j.biocel.2012.03.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 03/21/2012] [Indexed: 11/30/2022]
Abstract
Intragenic polymorphisms in the vitamin D receptor gene are linked to disc degeneration features, suggesting that alterations in the vitamer-mediated signalling could be involved in the pathophysiology of the disc and that interaction of disc cells with vitamin D metabolites may be critical for disc health. The vitamer-mediated modulation of disc cells proliferation, metabolic activity, extracellular matrix (ECM) genes expression and proteins production was investigated. It was stated that disc cells express vitamin D receptor and are very sensitive to metabolic stimuli. In monolayer cultures, 1,25(OH)(2)D(3), but not 24,25(OH)(2)D(3), determined an inhibition of the proliferation and regulated also the ECM genes expression in nucleus pulposus and annulus fibrosus cells. Micromass cultures induced a more physiologic expression pattern of extracellular matrix genes. Cells Treatment with vitamin D metabolites did not result in relevant modifications of glycosaminoglycans production, except for annulus cells, whose production was reduced after 1,25(OH)(2)D(3) treatment. Moreover, a reduced glycosaminoglycans staining in both cell types and a significant reduced aggrecan gene expression in annulus cells treated with 1,25(OH)(2)D(3) were observed. A reduction of collagen I and II staining in annulus cells 1,25(OH)(2)D(3) treated, in accordance with a downregulation of collagen genes expression, was also registered. Finally, the vitamin D receptor gene expression did not show significant metabolite-mediated modification in monolayer or micromass cultures. These findings could enhance new insights on the biochemical mechanisms regulated by vitamin D in disc cartilage and possibly involved in the development of physiological/pathological modifications of the disc.
Collapse
Affiliation(s)
- Alessandra Colombini
- I.R.C.C.S. Istituto Ortopedico Galeazzi, Via R. Galeazzi 4, 20161 Milano, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
464
|
Cui X, Hasegawa A, Lotz M, D'Lima D. Structured three-dimensional co-culture of mesenchymal stem cells with meniscus cells promotes meniscal phenotype without hypertrophy. Biotechnol Bioeng 2012; 109:2369-80. [PMID: 22422555 DOI: 10.1002/bit.24495] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 02/22/2012] [Accepted: 02/28/2012] [Indexed: 11/11/2022]
Abstract
Menisci play a crucial role in weight distribution, load bearing, shock absorption, lubrication, and nutrition of articular cartilage within the knee joint. Damage to the meniscus typically does not heal spontaneously due to its partial avascular nature. Partial or complete meniscectomy is a common clinical treatment of the defective meniscus. However, this procedure ultimately leads to osteoarthritis due to increased mechanical stress to the articular cartilage. Meniscus tissue engineering offers a promising solution for partial or complete meniscus deficiency. Mesenchymal stem cells (MSC) have the potential to differentiate into meniscal fibrochondrocyte as well as deliver trophic effects to the differentiated cells. This study tested the feasibility of using MSC co-cultured with mature meniscal cells (MC) for meniscus tissue engineering. Structured cell pellets were created using MC and MSC at varying ratios (100:0, 75:25, 50:50, 25:75, and 0:100) and cultured with or without transforming growth factor-beta 3 supplemented chondrogenic media for 21 days. The meniscal and hypertrophic gene expression, gross appearance and structure of the pellets, meniscus extracellular matrix (ECM), histology and immunohistochemistry of proteoglycan and collagen were evaluated. Co-culture of MC with MSC at 75:25 demonstrated highest levels of collagen type I and glycosaminoglycans (GAG) production, as well as the lowest levels of hypertrophic genes, such as COL10A1 and MMP13. All co-culture conditions showed better meniscus ECM production and hypertrophic inhibition as compared to MSC culture alone. The collagen fiber bundles observed in the co-cultures are important to produce heterogenic ECM structure of meniscus. In conclusion, co-culturing MC and MSC is a feasible and efficient approach to engineer meniscus tissue with enhanced ECM production without hypertrophy.
Collapse
Affiliation(s)
- Xiaofeng Cui
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California 92037, USA.
| | | | | | | |
Collapse
|
465
|
Donor age and long-term culture affect differentiation and proliferation of human bone marrow mesenchymal stem cells. Ann Hematol 2012; 91:1175-86. [PMID: 22395436 DOI: 10.1007/s00277-012-1438-x] [Citation(s) in RCA: 212] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 02/22/2012] [Indexed: 12/20/2022]
Abstract
Bone marrow-derived human mesenchymal stem cells (BM-hMSCs) represent a promising cell-based therapy for a number of degenerative conditions. Many applications require cell expansion and involve the treatment of diseases and conditions found in an aging population. Therefore, the effects of donor age and long-term passage must be clarified. In this study, the effects of donor age and long-term passage on the morphology, proliferation potential, characteristics, mesodermal differentiation ability, and transdifferentiation potential of hMSCs towards neurogenic lineage were evaluated. Cells from child donors (0-12 years, n = 6) maintained their fibroblast-like morphology up to higher passages and proliferated in a greater number than those from adult (25-50 years, n = 6) and old (over 60 years, n = 6) donors. Adipogenic, osteogenic, and neurogenic differentiation potential decreased with age, while chondrogenic potential did not change. Long-term passage affected the morphology and proliferation of hMSCs from all ages. With increasing passage number, proliferation rate decreased and cells lost their typical morphology. Expression levels of neural markers (β III tubulin and NSE) and topo II isoforms in populations of nondifferentiated hMSCs were investigated by reverse transcription polymerase chain reaction analysis. While neural marker and topo IIβ expression levels increased due to increasing passage number in adult hMSCs compared to child hMSCs, topo IIα decreased in both. These results indicated that, even under highly standardized culture conditions, donor age and long-term passage have effects on hMSC characteristics, which should be taken into account prior to stem cell-based therapies.
Collapse
|
466
|
Kim EK, Lim S, Park JM, Seo JK, Kim JH, Kim KT, Ryu SH, Suh PG. Human mesenchymal stem cell differentiation to the osteogenic or adipogenic lineage is regulated by AMP-activated protein kinase. J Cell Physiol 2012; 227:1680-7. [PMID: 21678424 DOI: 10.1002/jcp.22892] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AMP-activated protein kinase (AMPK) is an energy-sensing kinase that has recently been shown to regulate the differentiation of preadipocytes and osteoblasts. However, the role of AMPK in stem cell differentiation is largely unknown. Using in vitro culture models, the present study demonstrates that AMPK is a critical regulatory factor for osteogenic differentiation. We observed that expression and phosphorylation of AMPK were increased during osteogenesis in human adipose tissue-derived mesenchymal stem cells (hAMSC). To elucidate the role of AMPK in osteogenic differentiation, we investigated the effect of AMPK inhibition or knockdown on mineralization of hAMSC. Compound C, an AMPK inhibitor, reduced mineralized matrix deposition and suppressed the expression of osteoblast-specific genes, including alkaline phosphatase (ALP), runt-related transcription factor 2 (RUNX2), and osteocalcin (OCN). Knockdown of AMPK by shRNA-lentivirus infection also reduced osteogenesis. In addition, inhibition or knockdown of AMPK during osteogenesis inhibited ERK phosphorylation, which is required for osteogenesis. Interestingly, inhibition of AMPK induced adipogenic differentiation of hAMSC, even in osteogenic induction medium (OIM). These results provide a potential mechanism involving AMPK activation in osteogenic differentiation of hAMSC and suggest that commitment of hAMSC to osteogenic or adipogenic lineage is governed by activation or inhibition of AMPK, respectively.
Collapse
Affiliation(s)
- Eung-Kyun Kim
- Division of Molecular and Life Sciences, Pohang University of Science and Technology, Pohang, Kyungbuk, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
467
|
Hypoxia mediated isolation and expansion enhances the chondrogenic capacity of bone marrow mesenchymal stromal cells. Stem Cell Res Ther 2012; 3:9. [PMID: 22385573 PMCID: PMC3392769 DOI: 10.1186/scrt100] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 12/18/2011] [Accepted: 03/02/2012] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION The capacity of bone marrow mesenchymal stromal cells (BMSCs) to be induced into chondrocytes has drawn much attention for cell-based cartilage repair. BMSCs represent a small proportion of cells of the bone marrow stromal compartment and, thus, culture expansion is a necessity for therapeutic use. However, there is no consensus on how BMSCs should be isolated nor expanded to maximize their chondrogenic potential. During embryonic development pluripotent stem cells differentiate into chondrocytes and form cartilage in a hypoxic microenvironment. METHODS Freshly harvested human BMSCs were isolated and expanded from the aspirates of six donors, under either hypoxic conditions (3% O2) or normoxic conditions (21% O2). A colony-forming unit fibroblastic (Cfu-f) assay was used to determine the number of cell colonies developed from each donor. BMSCs at passage 2 (P2) were characterized by flow cytometry for the phenotypic expression of cell surface markers on mesenchymal stem cells. BMSCs at P2 were subsequently cultured in vitro as three-dimensional cell pellets in a defined serum-free chondrogenic medium under normoxic and hypoxic conditions. Chondrogenic differentiation of the BMSCs was characterized by biochemical and histological methods and by quantitative gene-expression analysis. RESULTS After 14 days of culture, the number of BMSC colonies developed under hypoxia was generally higher (8% to 38% depending on donor) than under normoxia. BMSCs were positive for the cell surface markers CD13, CD29, CD44, CD73, CD90, CD105 and CD151, and negative for CD34. Regardless of the oxygen tension during pellet culture, hypoxia-expanded BMSC pellets underwent a more robust chondrogenesis than normoxia-expanded BMSC pellets after three weeks of culture, as judged by increased glycosaminoglycan synthesis and Safranin O staining, along with increased mRNA expression of aggrecan, collagen II and Sox9. Hypoxic conditions enhanced the mRNA expression of hypoxia inducible factor-2 alpha (HIF-2α) but suppressed the mRNA expression of collagen X in BMSC pellet cultures regardless of the oxygen tension during BMSC isolation and propagation. CONCLUSIONS Taken together, our data demonstrate that isolation and expansion of BMSCs under hypoxic conditions augments the chondrogenic potential of BMSCs. This suggests that hypoxia-mediated isolation and expansion of BMSCs may improve clinical applications of BMSCs for cartilage repair.
Collapse
|
468
|
Hasegawa T, Miwa M, Sakai Y, Nikura T, Lee SY, Oe K, Iwakura T, Kurosaka M, Komori T. Mandibular Hematoma Cells as a Potential Reservoir for Osteoprogenitor Cells in Fractures. J Oral Maxillofac Surg 2012; 70:599-607. [DOI: 10.1016/j.joms.2011.03.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 03/10/2011] [Accepted: 03/30/2011] [Indexed: 10/17/2022]
|
469
|
Shin SH, Yoo JJ, Kim HN, Nam J, Kim HJ. Enhanced cellular responses of human bone marrow stromal cells cultured on pretreated surface with allogenic platelet-rich plasma. Connect Tissue Res 2012; 53:318-26. [PMID: 22329757 DOI: 10.3109/03008207.2012.656859] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The principal objective of this study was to evaluate the effects of surface pretreatment with platelet-rich plasma (PRP) on the cellular functions of human bone marrow stromal cells (hBMSCs). The surfaces of tissue culture plates (TCPs) were pretreated by adding PRP followed by centrifugation to bring platelets closer to the surface, followed by incubation for 60 min at 37°C. Then, hBMSCs were seeded onto TCP and TCP pretreated with PRP (TCP-PRP), followed by culture in osteogenic medium. Cell attachment, proliferation, and osteogenic differentiation were evaluated. Field emission scanning electron microscope (FE-SEM; JSM-7401F, JEOL Ltd., Japan) observations were conducted. The attachment of hBMSCs was significantly lower on TCP-PRP than on TCP. However, when the cell numbers were normalized with those observed on day 1 of culture, cellular proliferation on 5 days was significantly higher on TCP-PRP. Alkaline phosphatase activity, an index of early phase of osteoblastic differentiation, was significantly higher on TCP-PRP on day 14. Calcium deposition amount, an index of terminal osteoblastic differentiation, was also significantly higher on TCP-PRP on days 14 and 21. The results of von Kossa staining confirmed that, on day 21, the area of mineralized nodules was significantly larger on TCP-PRP. FE-SEM observation demonstrated that activated platelets and fibrin network covered the surface after PRP treatment. An increase in the number of hBMSCs and their cellular products was evident on the FE-SEM observation, and the fibrin network remained on day 21. Our results demonstrate that a PRP-treated surface enhanced early proliferation and late osteogenic differentiation of hBMSCs.
Collapse
Affiliation(s)
- Seung Han Shin
- Department of Orthopedic Surgery, College of Medicine, Seoul National University, Jongno-gu, Seoul, Korea
| | | | | | | | | |
Collapse
|
470
|
Sato M, Uchida K, Nakajima H, Miyazaki T, Guerrero AR, Watanabe S, Roberts S, Baba H. Direct transplantation of mesenchymal stem cells into the knee joints of Hartley strain guinea pigs with spontaneous osteoarthritis. Arthritis Res Ther 2012; 14:R31. [PMID: 22314040 PMCID: PMC3392826 DOI: 10.1186/ar3735] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 12/29/2011] [Accepted: 02/07/2012] [Indexed: 01/22/2023] Open
Abstract
INTRODUCTION Mesenchymal stem cells (MSCs) can differentiate into various connective tissue cells. Several techniques have been used for the clinical application of MSCs in articular cartilage repair; however, there are many issues associated with the selection of the scaffold material, including its ability to support cell viability and differentiation and its retention and degradation in situ. The application of MSCs via a scaffold also requires a technically demanding surgical procedure. The aim of this study was to test the outcome of intra-articular transplantation of mesenchymal stem cells suspended in hyaluronic acid (HA) in the knee joints of Hartley strain guinea pigs with spontaneous osteoarthritis (OA). METHODS Commercially available human MSCs were cultured, labeled with carboxyfluorescein diacetate succinimidyl ester (CFDA-SE), suspended in either PBS or HA, and injected into the knee joints of 7-month-old animals. The control animals were injected with either PBS or HA alone. The animals were sacrificed at 1, 3, and 5 weeks post transplantation, the knee joints harvested, and fluorescent microscopic analysis was performed. Histological and immunohistochemical analysis were performed at 5 weeks post transplantation. RESULTS At 5 weeks post transplantation, partial cartilage repair was noted in the HA-MSC group but not in the other groups. Examination of CFDA-SE-labeled cells demonstrated migration, differentiation, and proliferation of MSC in the HA-MSC group. There was strong immunostaining for type II collagen around both residual chondrocytes and transplanted MSCs in the OA cartilage. CONCLUSION This scaffold-free and technically undemanding technique appears to result in the regeneration of articular cartilage in the spontaneous OA animal model. Although further examination of the long-term effects of transplantation is necessary, the findings suggest that intra-articular injection of HA-MSC mixture is potentially beneficial for OA.
Collapse
Affiliation(s)
- Mitsuhiko Sato
- Department of Orthopaedics and Rehabilitation Medicine, Faculty of Medical Sciences, University of Fukui, Matsuoka-Shimoaizuki 23, Eiheiji, Fukui 910-1193, Japan
| | - Kenzo Uchida
- Department of Orthopaedics and Rehabilitation Medicine, Faculty of Medical Sciences, University of Fukui, Matsuoka-Shimoaizuki 23, Eiheiji, Fukui 910-1193, Japan
| | - Hideaki Nakajima
- Department of Orthopaedics and Rehabilitation Medicine, Faculty of Medical Sciences, University of Fukui, Matsuoka-Shimoaizuki 23, Eiheiji, Fukui 910-1193, Japan
| | - Tsuyoshi Miyazaki
- Department of Orthopaedics and Rehabilitation Medicine, Faculty of Medical Sciences, University of Fukui, Matsuoka-Shimoaizuki 23, Eiheiji, Fukui 910-1193, Japan
| | - Alexander Rodriguez Guerrero
- Department of Orthopaedics and Rehabilitation Medicine, Faculty of Medical Sciences, University of Fukui, Matsuoka-Shimoaizuki 23, Eiheiji, Fukui 910-1193, Japan
| | - Shuji Watanabe
- Department of Orthopaedics and Rehabilitation Medicine, Faculty of Medical Sciences, University of Fukui, Matsuoka-Shimoaizuki 23, Eiheiji, Fukui 910-1193, Japan
| | - Sally Roberts
- Institute for Science & Technology in Medicine, Keele University at the RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Hisatoshi Baba
- Department of Orthopaedics and Rehabilitation Medicine, Faculty of Medical Sciences, University of Fukui, Matsuoka-Shimoaizuki 23, Eiheiji, Fukui 910-1193, Japan
| |
Collapse
|
471
|
Isolation of the stromal-vascular fraction of mouse bone marrow markedly enhances the yield of clonogenic stromal progenitors. Blood 2012; 119:e86-95. [PMID: 22262767 DOI: 10.1182/blood-2011-08-372334] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The low incidence of CFU-F significantly complicates the isolation of homogeneous populations of mouse bone marrow stromal cells (BMSCs), a common problem being contamination with hematopoietic cells. Taking advantage of burgeoning evidence demonstrating the perivascular location of stromal cell stem/progenitors, we hypothesized that a potential reason for the low yield of mouse BMSCs is the flushing of the marrow used to remove single-cell suspensions and the consequent destruction of the marrow vasculature, which may adversely affect recovery of BMSCs physically associated with the abluminal surface of blood vessels. Herein, we describe a simple methodology based on preparation and enzymatic disaggregation of intact marrow plugs, which yields distinct populations of both stromal and endothelial cells. The recovery of CFU-F obtained by pooling the product of each digestion (1631.8 + 199) reproducibly exceeds that obtained using the standard BM flushing technique (14.32 + 1.9) by at least 2 orders of magnitude (P < .001; N = 8) with an accompanying 113.95-fold enrichment of CFU-F frequency when plated at low oxygen (5%). Purified BMSC populations devoid of hematopoietic contamination are readily obtained by FACS at P0 and from freshly prepared single-cell suspensions. Furthermore, this population demonstrates robust multilineage differentiation using standard in vivo and in vitro bioassays.
Collapse
|
472
|
Naveena N, Venugopal J, Rajeswari R, Sundarrajan S, Sridhar R, Shayanti M, Narayanan S, Ramakrishna S. Biomimetic composites and stem cells interaction for bone and cartilage tissue regeneration. ACTA ACUST UNITED AC 2012. [DOI: 10.1039/c1jm14401d] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
473
|
Mantovani C, Terenghi G, Shawcross SG. Isolation of adult stem cells and their differentiation to Schwann cells. Methods Mol Biol 2012; 916:47-57. [PMID: 22914932 DOI: 10.1007/978-1-61779-980-8_5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Peripheral nerve injuries are an economic burden for society in general and despite advanced microsurgical reconstruction of the damaged nerves the functional result is unsatisfactory with poor sensory recovery and reduced motor functions (Wiberg and Terenghi, Surg Technol Int 11:303-310, 2003). In the treatment of nerve injuries transplantation of a nerve graft is often necessary, especially in nerve gap injuries.Schwann cells (SC) are the key facilitators of peripheral nerve regeneration and are responsible for the formation and maintenance of the myelin sheath around axons in peripheral nerve fibers. They are essential for nerve regeneration after nerve injuries as they produce extracellular matrix molecules, integrins, and trophic factors providing guidance and trophic support for regenerating axons (Wiberg and Terenghi, Surg Technol Int 11:303-310, 2003; Bunge, J Neurol 242:S19-21, 1994; Ide, Neurosci Res 25:101-121, 1996; Mahanthappa et al. J Neurosci 16:4673-4683, 1996). However, the use of ex vivo cultured SC within conduits is limited in its clinical application because of the concomitant donor site morbidity and the slow growth of these cells in vitro (Tohill et al. Tissue Eng 10:1359-1367, 2004).Mesenchymal stem cells (MSC or bone marrow stromal cells) and adipose-derived stem cells (ASC) are easily accessible non-hematopoietic stem cells that have proven essential for research purposes due to their plasticity and ability to differentiate into several functional cell types. This alternative source of cells is relatively simple to isolate and expand in culture. We have demonstrated that MSC and ASC can trans-differentiate along a SC lineage with functional properties and growth factor synthesis activities similar to those of native SC and could provide nerve fiber support and guidance during nerve regeneration.
Collapse
Affiliation(s)
- Cristina Mantovani
- Blond McIndoe Laboratories, School of Biomedicine, University of Manchester, Manchester, UK
| | | | | |
Collapse
|
474
|
Lavrentieva A, Hatlapatka T, Neumann A, Weyand B, Kasper C. Potential for osteogenic and chondrogenic differentiation of MSC. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2012; 129:73-88. [PMID: 22457052 DOI: 10.1007/10_2012_133] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The introduction of mesenchymal stem cells (MSC) into the field of tissue engineering for bone and cartilage repair is a promising development, since these cells can be expanded ex vivo to clinically relevant numbers and, after expansion, retain their ability to differentiate into different cell lineages. Mesenchymal stem cells isolated from various tissues have been intensively studied and characterized by many research groups. To obtain functionally active differentiated tissue, tissue engineered constructs are cultivated in vitro statically or dynamically in bioreactors under controlled conditions. These conditions include special cell culture media, addition of signalling molecules, various physical and chemical factors and the application of different mechanical stimuli. Oxygen concentration in the culture environment is also a significant factor which influences MSC proliferation, stemness and differentiation capacity. Knowledge of the different aspects which affect MSC differentiation in vivo and in vitro will help researchers to achieve directed cell fate without the addition of differentiation agents in concentrations above the physiological range.
Collapse
Affiliation(s)
- Antonina Lavrentieva
- Institut für Technische Chemie, Leibniz Universität Hannover, Callinstrasse 5, 30167, Hannover, Germany,
| | | | | | | | | |
Collapse
|
475
|
Sitasuwan P, Andrew Lee L, Bo P, Davis EN, Lin Y, Wang Q. A plant virus substrate induces early upregulation of BMP2 for rapid bone formation. Integr Biol (Camb) 2012; 4:651-60. [DOI: 10.1039/c2ib20041d] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
476
|
Sasaki JI, Matsumoto T, Egusa H, Matsusaki M, Nishiguchi A, Nakano T, Akashi M, Imazato S, Yatani H. In vitro reproduction of endochondral ossification using a 3D mesenchymal stem cell construct. Integr Biol (Camb) 2012; 4:1207-14. [DOI: 10.1039/c2ib20027a] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
477
|
Kanematsu D, Shofuda T, Yamamoto A, Ban C, Ueda T, Yamasaki M, Kanemura Y. Isolation and cellular properties of mesenchymal cells derived from the decidua of human term placenta. Differentiation 2011; 82:77-88. [PMID: 21684674 DOI: 10.1016/j.diff.2011.05.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 05/06/2011] [Accepted: 05/26/2011] [Indexed: 12/22/2022]
Abstract
The clinical promise of cell-based therapies is generally recognized, and has driven an intense search for good cell sources. In this study, we isolated plastic-adherent cells from human term decidua vera, called decidua-derived-mesenchymal cells (DMCs), and compared their properties with those of bone marrow-derived-mesenchymal stem cells (BM-MSCs). The DMCs strongly expressed the mesenchymal cell marker vimentin, but not cytokeratin 19 or HLA-G, and had a high proliferative potential. That is, they exhibited a typical fibroblast-like morphology for over 30 population doublings. Cells phenotypically identical to the DMCs were identified in the decidua vera, and genotyping confirmed that the DMCs were derived from the maternal components of the fetal adnexa. Flow cytometry analysis showed that the expression pattern of CD antigens on the DMCs was almost identical to that on BM-MSCs, but some DMCs expressed the CD45 antigen, and over 50% of them also expressed anti-fibroblast antigen. In vitro, the DMCs showed good differentiation into chondrocytes and moderate differentiation into adipocytes, but scant evidence of osteogenesis, compared with the BM-MSCs. Gene expression analysis showed that, compared with BM-MSCs, the DMCs expressed higher levels of TWIST2 and RUNX2 (which are associated with early mesenchymal development and/or proliferative capacity), several matrix metalloproteinases (MMP1, 3, 10, and 12), and cytokines (BMP2 and TGFB2), and lower levels of MSX2, interleukin 26, and HGF. Although DMCs did not show the full multipotency of BM-MSCs, their higher proliferative ability indicates that their cultivation would require less maintenance. Furthermore, the use of DMCs avoids the ethical concerns associated with the use of embryonic tissues, because they are derived from the maternal portion of the placenta, which is otherwise discarded. Thus, the unique properties of DMCs give them several advantages for clinical use, making them an interesting and attractive alternative to MSCs for regenerative medicine.
Collapse
Affiliation(s)
- Daisuke Kanematsu
- Department of Regenerative Medicine, Institute for Clinical Research, Osaka National Hospital, National Hospital Organization, 2-1-14 Hoenzaka, Chuo-ku, Osaka 540-0006, Japan
| | | | | | | | | | | | | |
Collapse
|
478
|
Hammouda GA, Abd El Rahman GM, Abou Golayel MK, Galhoom RA. Chondrogenic differentiation of cultured rat mesenchymal stem cells from bone marrow. THE EGYPTIAN JOURNAL OF HISTOLOGY 2011; 34:772-779. [DOI: 10.1097/01.ehx.0000407208.88972.9d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
479
|
Gupta MS, Cooper ES, Nicoll SB. Transforming Growth Factor-Beta 3 Stimulates Cartilage Matrix Elaboration by Human Marrow-Derived Stromal Cells Encapsulated in Photocrosslinked Carboxymethylcellulose Hydrogels: Potential for Nucleus Pulposus Replacement. Tissue Eng Part A 2011; 17:2903-10. [DOI: 10.1089/ten.tea.2011.0152] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Michelle S. Gupta
- Department of Biomedical Engineering, The City College of New York, New York, New York
| | - Elana S. Cooper
- Department of Biomedical Engineering, The City College of New York, New York, New York
| | - Steven B. Nicoll
- Department of Biomedical Engineering, The City College of New York, New York, New York
| |
Collapse
|
480
|
Willers C, Wood DJ, Zheng MH. A CURRENT REVIEW ON THE BIOLOGY AND TREATMENT OF ARTICULAR CARTILAGE DEFECTS (PART I & PART II). ACTA ACUST UNITED AC 2011. [DOI: 10.1142/s0218957703001125] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Osteochondral injury occurs predominantly in physically active young adult males. Injury to the articular cartilage and/or subchondral bone may not only cause acute joint disease resulting in osseous intracapsular (synovitis) or extracapsular pain, but may also act to spawn arthritic conditions in later life. Since the 18th century, such injury has proven difficult to treat clinically, and much therapy has been essentially palliative. Past treatments such as abrasion arthroplasty, drilling, microfracture and arthroscopic lavage have been useful in removing articular debris and promoting the formation of the fibrin clot used in most native repair mechanisms. However, the limitation of these techniques is their inability to restore the damaged cartilage and subchondral bone to their normal tissue architecture. Recent developments in tissue engineering have concentrated on the utilization of autologous chondrocyte implantation, biomaterials and growth factors to promote the regeneration of biomechanically superior hyaline articular cartilage. This paper reviews the etiology, repair biology and therapeutic techniques of cartilage and/or osteochondral injury over the previous decades, and attempts to provide insight into interesting new research directions which offer much potential for improved treatment of these troublesome lesions.
Collapse
Affiliation(s)
- Craig Willers
- Department of Orthopaedic Surgery, School of Surgery and Pathology, University of Western Australia, Nedlands, 6009, W.A., Australia
| | - David J. Wood
- Department of Orthopaedic Surgery, School of Surgery and Pathology, University of Western Australia, Nedlands, 6009, W.A., Australia
| | - Ming H. Zheng
- Department of Orthopaedic Surgery, School of Surgery and Pathology, University of Western Australia, Nedlands, 6009, W.A., Australia
| |
Collapse
|
481
|
FUKAGATA KOJI, FURUKAWA KATSUKOS, USHIDA TAKASHI. ANALYSIS OF CELL ACCUMULATION MECHANISM IN A ROTATIONAL CULTURE SYSTEM. J MECH MED BIOL 2011. [DOI: 10.1142/s0219519410003745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The accumulation mechanism of cells in a rotational culture device is investigated from the viewpoint of fluid mechanics. For simplicity, the deformation of the water surface is neglected and the cells are treated as spherical solid particles. From the numerical simulation of flow field with typical parameters used in the previous experiments, it is confirmed that the relative velocity of fluid induced by the rotational shaking is much smaller than the speed of rotation. From the analysis of particle equation of motion, it is found that the accumulation of cells toward the central region is found to be due to the interaction between the acceleration by rotational shaking and the drag force acting on the cells. The integral time scale for cell accumulation was estimated to be about 10 min for typical cases. The accumulation speed increases quadratically with the diameter of cell and the angular velocity of rotational shaking, which qualitatively support the previous experimental observation.
Collapse
Affiliation(s)
- KOJI FUKAGATA
- Department of Mechanical Engineering, Keio University, Hiyoshi 3-14-1, Kohoku-ku, Yokohama 223-8522, Japan
| | - KATSUKO S. FURUKAWA
- Department of Bioengineering, Department of Mechanical Engineering, School of Engineering, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8656, Japan
| | - TAKASHI USHIDA
- Division of Biomedical Materials and Systems, Center for Disease Biology and Integrative Medicine, School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
482
|
Eini F, Foroutan T, Bidadkosh A, Barin A, Dehghan MM, Tajik P. The effects of freeze/thawing process on cryopreserved equine umbilical cord blood-derived mesenchymal stem cells. ACTA ACUST UNITED AC 2011. [DOI: 10.1007/s00580-011-1355-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
483
|
Hwang Y, Phadke A, Varghese S. Engineered microenvironments for self-renewal and musculoskeletal differentiation of stem cells. Regen Med 2011; 6:505-24. [PMID: 21749208 DOI: 10.2217/rme.11.38] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Stem cells hold great promise for therapies aimed at regenerating damaged tissue, drug screening and studying in vitro models of human disease. However, many challenges remain before these applications can become a reality. One such challenge is developing chemically defined and scalable culture conditions for derivation and expansion of clinically viable human pluripotent stem cells, as well as controlling their differentiation with high specificity. Interaction of stem cells with their extracellular microenvironment plays an important role in determining their differentiation commitment and functions. Regenerative medicine approaches integrating cell-matrix and cell-cell interactions, and soluble factors could lead to development of robust microenvironments to control various cellular responses. Indeed, several of these recent developments have provided significant insight into the design of microenvironments that can elicit the targeted cellular response. In this article, we will focus on some of these developments with an emphasis on matrix-mediated expansion of human pluripotent stem cells while maintaining their pluripotency. We will also discuss the role of matrix-based cues and cell-cell interactions in the form of soluble signals in directing stem cell differentiation into musculoskeletal lineages.
Collapse
Affiliation(s)
- Yongsung Hwang
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0412, USA
| | | | | |
Collapse
|
484
|
Williams AR, Hare JM. Mesenchymal stem cells: biology, pathophysiology, translational findings, and therapeutic implications for cardiac disease. Circ Res 2011; 109:923-40. [PMID: 21960725 DOI: 10.1161/circresaha.111.243147] [Citation(s) in RCA: 651] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Mesenchymal stem cells (MSCs) are a prototypical adult stem cell with capacity for self-renewal and differentiation with a broad tissue distribution. Initially described in bone marrow, MSCs have the capacity to differentiate into mesoderm- and nonmesoderm-derived tissues. The endogenous role for MSCs is maintenance of stem cell niches (classically the hematopoietic), and as such, MSCs participate in organ homeostasis, wound healing, and successful aging. From a therapeutic perspective, and facilitated by the ease of preparation and immunologic privilege, MSCs are emerging as an extremely promising therapeutic agent for tissue regeneration. Studies in animal models of myocardial infarction have demonstrated the ability of transplanted MSCs to engraft and differentiate into cardiomyocytes and vasculature cells, recruit endogenous cardiac stem cells, and secrete a wide array of paracrine factors. Together, these properties can be harnessed to both prevent and reverse remodeling in the ischemically injured ventricle. In proof-of-concept and phase I clinical trials, MSC therapy improved left ventricular function, induced reverse remodeling, and decreased scar size. This article reviews the current understanding of MSC biology, mechanism of action in cardiac repair, translational findings, and early clinical trial data of MSC therapy for cardiac disease.
Collapse
Affiliation(s)
- Adam R Williams
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33101, USA.
| | | |
Collapse
|
485
|
Solorio LD, Vieregge EL, Dhami CD, Dang PN, Alsberg E. Engineered cartilage via self-assembled hMSC sheets with incorporated biodegradable gelatin microspheres releasing transforming growth factor-β1. J Control Release 2011; 158:224-32. [PMID: 22100386 DOI: 10.1016/j.jconrel.2011.11.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Revised: 11/01/2011] [Accepted: 11/04/2011] [Indexed: 10/15/2022]
Abstract
Self-assembling cell sheets have shown great potential for use in cartilage tissue engineering applications, as they provide an advantageous environment for the chondrogenic induction of human mesenchymal stem cells (hMSCs). We have engineered a system of self-assembled, microsphere-incorporated hMSC sheets capable of forming cartilage in the presence of exogenous transforming growth factor β1 (TGF-β1) or with TGF-β1 released from incorporated microspheres. Gelatin microspheres with two different degrees of crosslinking were used to enable different cell-mediated microsphere degradation rates. Biochemical assays, histological and immunohistochemical analyses, and biomechanical testing were performed to determine biochemical composition, structure, and equilibrium modulus in unconfined compression after 3 weeks of culture. The inclusion of microspheres with or without loaded TGF-β1 significantly increased sheet thickness and compressive equilibrium modulus, and enabled more uniform matrix deposition by comparison to control sheets without microspheres. Sheets incorporated with fast-degrading microspheres containing TGF-β1 produced significantly more GAG and GAG per DNA than all other groups tested and stained more intensely for type II collagen. These findings demonstrate improved cartilage formation in microsphere-incorporated cell sheets, and describe a tailorable system for the chondrogenic induction of hMSCs without necessitating culture in growth factor-containing medium.
Collapse
Affiliation(s)
- Loran D Solorio
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | | | | | | | | |
Collapse
|
486
|
Reddy BY, Xu DS, Hantash BM. Mesenchymal stem cells as immunomodulator therapies for immune-mediated systemic dermatoses. Stem Cells Dev 2011; 21:352-62. [PMID: 21864110 DOI: 10.1089/scd.2011.0404] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stems cells (MSCs) are a population of multipotent cells residing in several readily available adult tissue compartments, thus allowing for their ex vivo expansion. To date, therapeutic applications of MSCs have focused on their ability to hone to and support the repair of damaged tissues. More recent evidence suggests that MSCs possess low immunogenicity and a diverse array of immunosuppressive properties. In thisarticle, we will review the basic biology of bone marrow-derived MSCs including their immunoregulatory effects and the putative mechanisms underlying them. We will then present some of the recent clinical applications of MSCs that have leveraged these effects for the treatment of immune-mediated dermatoses such as graft-versus-host disease and systemic lupus erythematosus. Although MSCs offer great therapeutic promise, we will also highlight a number of pertinent challenges that should be overcome before their successful clinical translation.
Collapse
Affiliation(s)
- Bobby Y Reddy
- Department of Dermatology, UMDNJ-New Jersey Medical School, Newark, New Jersey, USA
| | | | | |
Collapse
|
487
|
Multipotent mesenchymal stromal stem cell expansion by plating whole bone marrow at a low cellular density: a more advantageous method for clinical use. Stem Cells Int 2011; 2012:920581. [PMID: 23715383 PMCID: PMC3195433 DOI: 10.1155/2012/920581] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 08/01/2011] [Indexed: 01/14/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a promising source for cell therapy due to their pluripotency and immunomodulant proprieties. As the identification of "optimal" conditions is important to identify a standard procedure for clinical use. Percoll, Ficoll and whole bone marrow directly plated were tested from the same sample as separation methods. The cells were seeded at the following densities: 100 000, 10 000, 1000, 100, 10 cells/cm(2). After reaching confluence, the cells were detached, pooled and re-plated at 1000, 500, 100, and 10 cells/cm(2). Statistical analyses were performed. Cumulative Population Doublings (PD) did not show significant differences for the separation methods and seeding densities but only for the plating density. Some small quantity samples plated in T25 flasks at plating densities of 10 and 100 cells/cm(2) did not produce any expansion. However, directly plated whole bone marrow resulted in a more advantageous method in terms of CFU-F number, cellular growth and minimal manipulation. No differences were observed in terms of gross morphology, differentiation potential or immunophenotype. These data suggest that plating whole bone marrow at a low cellular density may represent a good procedure for MSC expansion for clinical use.
Collapse
|
488
|
Abstract
The use of adult stem cells for therapeutic purposes has met with great success in recent years. Among several types of adult stem cells, mesenchymal stem cells (MSCs) derived from bone marrow (BM) and other sources have gained popularity for basic research and clinical applications because of their therapeutic potential in treating a variety of diseases. Because of their tissue regeneration potential and immune modulation effect, MSCs were recently used as cell-based therapy to promote revascularization, increase pancreatic β-cell proliferation, and avoid allograft rejection in islet transplantation. Taking advantage of the recent progress in gene therapy, genetically modified MSCs can further enhance and expand the therapeutic benefit of primary MSCs while retaining their stem-cell-like properties. This review aims to gain a thorough understanding of the current obstacles to successful islet transplantation and discusses the potential role of primary MSCs before or after genetic modification in islet transplantation.
Collapse
Affiliation(s)
- Hao Wu
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Zhaoyang Ye
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
489
|
Rampersad S, Ruiz JC, Petit A, Lerouge S, Antoniou J, Wertheimer MR, Mwale F. Stem Cells, Nitrogen-Rich Plasma-Polymerized Culture Surfaces, and Type X Collagen Suppression. Tissue Eng Part A 2011; 17:2551-60. [DOI: 10.1089/ten.tea.2010.0723] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Affiliation(s)
- Sonia Rampersad
- Lady Davis Institute for Medical Research, SMBD-Jewish General Hospital, Montreal, Québec, Canada
- Division of Orthopaedic Surgery, McGill University, Montreal, Québec, Canada
| | - Juan-Carlos Ruiz
- Department of Engineering Physics, École Polytechnique, Montreal, Québec, Canada
| | - Alain Petit
- Lady Davis Institute for Medical Research, SMBD-Jewish General Hospital, Montreal, Québec, Canada
| | - Sophie Lerouge
- Laboratory of Endovascular Biomaterials, Research Centre of the University of Montreal Hospital Centre (CRCHUM), Montreal, Québec, Canada
| | - John Antoniou
- Lady Davis Institute for Medical Research, SMBD-Jewish General Hospital, Montreal, Québec, Canada
- Division of Orthopaedic Surgery, McGill University, Montreal, Québec, Canada
| | | | - Fackson Mwale
- Lady Davis Institute for Medical Research, SMBD-Jewish General Hospital, Montreal, Québec, Canada
- Division of Orthopaedic Surgery, McGill University, Montreal, Québec, Canada
| |
Collapse
|
490
|
Xu B, Song G, Ju Y. Effect of focal adhesion kinase on the regulation of realignment and tenogenic differentiation of human mesenchymal stem cells by mechanical stretch. Connect Tissue Res 2011; 52:373-9. [PMID: 21401419 DOI: 10.3109/03008207.2010.541961] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Focal adhesion kinase (FAK) is a focal adhesion-associated protein kinase involved in cell adhesion and spreading. It is recruited as a participant in focal adhesion dynamics between cells and has a role in cell motility, differentiation, and survival. The role of FAK in the differentiation of human mesenchymal stem cells (hMSCs), however, is not well understood, particularly in terms of tenogenic differentiation. In this study, we reported that FAK regulates the mechanical stretch-induced realignment of hMSCs. We showed that FAK can be activated by mechanical stretch and, with a 10 μM PF 573228 (a novel small molecule inhibitor of FAK) treatment, FAK autophosphorylation at Tyr397 is significantly decreased. Moreover, our findings demonstrated that this decrease in FAK autophosphorylation at Tyr397 leads to the attenuation of upregulation of mechanical stretch-induced mRNA expression of tendon-related genes, including type I collagen, type III collagen, tenascin-C, and scleraxis. These results indicate that the FAK signaling molecule plays an important role in regulating cell realignment and tenogenic differentiation of hMSCs when induced by mechanical stretch. Collectively, our findings provide novel insight into the role of FAK in the realignment and mechanotransduction of hMSCs during the process of tenogenic differentiation induced by mechanical stretch.
Collapse
Affiliation(s)
- Baiyao Xu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, China
| | | | | |
Collapse
|
491
|
Makris EA, Hadidi P, Athanasiou KA. The knee meniscus: structure-function, pathophysiology, current repair techniques, and prospects for regeneration. Biomaterials 2011; 32:7411-31. [PMID: 21764438 PMCID: PMC3161498 DOI: 10.1016/j.biomaterials.2011.06.037] [Citation(s) in RCA: 696] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 06/17/2011] [Indexed: 02/07/2023]
Abstract
Extensive scientific investigations in recent decades have established the anatomical, biomechanical, and functional importance that the meniscus holds within the knee joint. As a vital part of the joint, it acts to prevent the deterioration and degeneration of articular cartilage, and the onset and development of osteoarthritis. For this reason, research into meniscus repair has been the recipient of particular interest from the orthopedic and bioengineering communities. Current repair techniques are only effective in treating lesions located in the peripheral vascularized region of the meniscus. Healing lesions found in the inner avascular region, which functions under a highly demanding mechanical environment, is considered to be a significant challenge. An adequate treatment approach has yet to be established, though many attempts have been undertaken. The current primary method for treatment is partial meniscectomy, which commonly results in the progressive development of osteoarthritis. This drawback has shifted research interest toward the fields of biomaterials and bioengineering, where it is hoped that meniscal deterioration can be tackled with the help of tissue engineering. So far, different approaches and strategies have contributed to the in vitro generation of meniscus constructs, which are capable of restoring meniscal lesions to some extent, both functionally as well as anatomically. The selection of the appropriate cell source (autologous, allogeneic, or xenogeneic cells, or stem cells) is undoubtedly regarded as key to successful meniscal tissue engineering. Furthermore, a large variation of scaffolds for tissue engineering have been proposed and produced in experimental and clinical studies, although a few problems with these (e.g., byproducts of degradation, stress shielding) have shifted research interest toward new strategies (e.g., scaffoldless approaches, self-assembly). A large number of different chemical (e.g., TGF-β1, C-ABC) and mechanical stimuli (e.g., direct compression, hydrostatic pressure) have also been investigated, both in terms of encouraging functional tissue formation, as well as in differentiating stem cells. Even though the problems accompanying meniscus tissue engineering research are considerable, we are undoubtedly in the dawn of a new era, whereby recent advances in biology, engineering, and medicine are leading to the successful treatment of meniscal lesions.
Collapse
Affiliation(s)
- Eleftherios A Makris
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, USA
| | | | | |
Collapse
|
492
|
Proulx-Bonneau S, Annabi B. The primary cilium as a biomarker in the hypoxic adaptation of bone marrow-derived mesenchymal stromal cells: a role for the secreted frizzled-related proteins. Biomark Insights 2011; 6:107-118. [PMID: 22084569 PMCID: PMC3201088 DOI: 10.4137/bmi.s8247] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A pivotal role in guiding mesenchymal stem cell (MSC) differentiation has recently been attributed to the primary cilium. This solitary, non-motile microtubule-based organelle emerging from the cell surface acts as a sensorial membrane structure reflecting developmental and adaptive processes associated with pathologies including human cystic kidney disease, skeletal malformations, obesity and cancer. Given that the intrinsic hypoxic adaptation of MSC remains poorly understood within ischemic tissues or hypoxic tumours, we questioned whether the hypoxia inducible factor-1α (HIF-1α) might be a downstream effector regulating cilium maintenance. We show that murine bone marrow-derived MSC cultured under hypoxic conditions (1.2% O(2)) lose their primary cilia in a time-dependent manner. Gene silencing of HIF-1α prevented cilia loss in hypoxic cultures, and generation of MSC expressing a constitutively active HIF-1α (MSC-HIF) was found to decrease primary cilium formation. A Wnt pathway-related gene expression array was also performed on MSC-HIF and indicated that the secreted Frizzled-related proteins (sFRP)-1, -3 and -4 were down-regulated, while sFRP-2 was up-regulated. Overexpression of recombinant sFRP-2 or gene silencing of sFRP-1, -3 and -4 in MSC led to primary cilium disruption. These results indicate a molecular signalling mechanism for the hypoxic disruption of the primary cilium in MSC involving an HIF-1α/sFRP axis. This mechanism contributes to our understanding of the adaptive processes possibly involved in the oncogenic transformation and tumour-supporting potential of MSC. Our current observations also open up the possibility for the primary cilia to serve as a biomarker in MSC adaptation to low oxygen tension within (patho)physiological microenvironments.
Collapse
Affiliation(s)
- Sébastien Proulx-Bonneau
- Laboratoire d’Oncologie Moléculaire, Centre de Recherche BioMED, Département de Chimie, Université du Québec à Montréal, Quebec, Canada
| | - Borhane Annabi
- Laboratoire d’Oncologie Moléculaire, Centre de Recherche BioMED, Département de Chimie, Université du Québec à Montréal, Quebec, Canada
| |
Collapse
|
493
|
Acosta FL, Metz L, Adkisson HD, Liu J, Carruthers-Liebenberg E, Milliman C, Maloney M, Lotz JC. Porcine intervertebral disc repair using allogeneic juvenile articular chondrocytes or mesenchymal stem cells. Tissue Eng Part A 2011; 17:3045-55. [PMID: 21910592 DOI: 10.1089/ten.tea.2011.0229] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Tissue engineering strategies for intervertebral disc repair have focused on the use of autologous disc-derived chondrocytes. Difficulties with graft procurement, harvest site morbidity, and functionality, however, may limit the utility of this cell source. We used an in vivo porcine model to investigate allogeneic non-disc-derived chondrocytes and allogeneic mesenchymal stem cells (MSCs) for disc repair. After denucleation, lumbar discs were injected with either fibrin carrier alone, allogeneic juvenile chondrocytes (JCs), or allogeneic MSCs. Discs were harvested at 3, 6, and 12 months, and cell viability and functionality were assessed qualitatively and quantitatively. JC-treated discs demonstrated abundant cartilage formation at 3 months, and to a lesser extent at 6 and 12 months. For the carrier and MSC-treated groups, however, there was little evidence of proteoglycan matrix or residual notochordal/chondrocyte cells, but rather a type I/II collagen-enriched scar tissue. By contrast, JCs produced a type II collagen-rich matrix that was largely absent of type I collagen. Viable JCs were observed at all time points, whereas no evidence of viable MSCs was found. These data support the premise that committed chondrocytes are more appropriate for use in disc repair, as they are uniquely suited for survival in the ischemic disc microenvironment.
Collapse
Affiliation(s)
- Frank L Acosta
- Orthopaedic Bioengineering Laboratory, University of California, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | |
Collapse
|
494
|
Dahl JP, Caballero M, Pappa AK, Madan G, Shockley WW, van Aalst JA. Analysis of human auricular cartilage to guide tissue-engineered nanofiber-based chondrogenesis: implications for microtia reconstruction. Otolaryngol Head Neck Surg 2011; 145:915-23. [PMID: 21908800 DOI: 10.1177/0194599811419092] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Nanofiber-supported, in vitro-generated cartilage may represent an optimal starting material for the development of a cartilage implant for use in microtia reconstruction. To do so, the authors aim to first characterize the molecular composition of endogenous auricular cartilage and determine if human umbilical cord mesenchymal stem cells (hUCMSCs) can be differentiated into cartilage in vitro. STUDY DESIGN Prospective, controlled. SETTING Academic research laboratory. SUBJECTS AND METHODS Human ear cartilage from normal adults, pediatric patients with microtia, and pediatric patients with preauricular appendages (n = 2) was analyzed for collagens I, II, and X and elastin expression. In parallel, hUCMSCs were cultured on either polycaprolactone (PCL) or D, L-lactide-co-glycolic acid (PLGA) nanofiber scaffolds for 21 days under chondrogenic conditions. Cells were harvested for histologic, biochemical, and quantitative polymerase chain reaction analysis. Control cells were grown under both chondrogenic and nonchondrogenic conditions in the absence of nanofiber scaffolds. RESULTS Histological analysis of human ear cartilage revealed similar levels and distribution of collagens I and X and elastin. Collagen II was not highly expressed in the microtia samples. hUCMSC cultures stained positively for glycosaminosglycans (GAG) and sulfated proteoglycans. Compared to control cells, hUCMSCs grown on PLGA nanofiber scaffolds had a higher differentiation index (P ≤ .012) and higher levels of collagen X mRNA expression (P ≤ .006). CONCLUSION These data provide information regarding the composition of endogenous ear cartilage and suggest that hUCMSCs grown on PLGA nanofiber scaffolds may represent an optimal starting material for the development of a cartilage implant for use in microtia reconstruction.
Collapse
Affiliation(s)
- John P Dahl
- Department of Otolaryngology-Head & Neck Surgery, University of North Carolina, Chapel Hill, North Carolina 27599-7195, USA
| | | | | | | | | | | |
Collapse
|
495
|
Wu H, Lu W, Mahato RI. Mesenchymal stem cells as a gene delivery vehicle for successful islet transplantation. Pharm Res 2011; 28:2098-109. [PMID: 21499838 PMCID: PMC3152657 DOI: 10.1007/s11095-011-0434-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Accepted: 03/14/2011] [Indexed: 01/01/2023]
Abstract
PURPOSE To evaluate the efficacy of human bone marrow-derived mesenchymal stem cells (hBMSCs) as gene delivery vehicles to simultaneously express human hepatocyte growth factor (HGF) and interleukin 1 receptor antagonist (IL-1Ra) to improve the outcome of islet transplantation. METHODS Morphology and islet-binding affinity of hBMSCs were checked by microscope. The expression of target genes and endogenous genes was determined by ELISA. Protection of islets by hBMSCs was evaluated in vitro by Calcein-AM/Propidium Iodide staining and in vivo by allogeneic islet transplantation study. Function and revascularization of islets was evaluated by immune fluorescence study. RESULTS Non-donor-specific hBMSCs showed strong binding affinity to human islets and protected viability and function. Transduction of hBMSCs with adenovirus encoding human HGF and human IL-1Ra (Adv-hHGF-hIL-1Ra) prior to co-culturing with islets further protected from apoptotic cell death, helped maintain 3D structures and morphology, and enhanced insulin secretion. Transplantation of human islets reconstituted with Adv-hHGF-hIL-1Ra transduced hBMSCs under the kidney capsule of streptozotocin-induced diabetic non-obese diabetic/severe combined immunodeficient (NOD-SCID) mice reversed diabetes by reducing blood glucose levels to ≤ 200 mg/dL for up to 15 weeks and reduced the number of islets required to achieving normoglycemia. Blood glucose levels of mice transplanted with islets alone reversed to ≥ 500 mg/dL 4 weeks post-transplantation. CONCLUSIONS Results indentified hBMSCs as effective gene delivery vehicles to improve the outcome of islet transplantation.
Collapse
Affiliation(s)
- Hao Wu
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 19 S. Manassas, RM 224, Memphis, Tennessee 38103-3308, USA
| | | | | |
Collapse
|
496
|
Matsumoto T, Ingham SM, Mifune Y, Osawa A, Logar A, Usas A, Kuroda R, Kurosaka M, Fu FH, Huard J. Isolation and characterization of human anterior cruciate ligament-derived vascular stem cells. Stem Cells Dev 2011; 21:859-72. [PMID: 21732814 DOI: 10.1089/scd.2010.0528] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The anterior cruciate ligament (ACL) usually fails to heal after rupture mainly due to the inability of the cells within the ACL tissue to establish an adequate healing process, making graft reconstruction surgery a necessity. However, some reports have shown that there is a healing potential of ACL with primary suture repair. Although some reports showed the existence of mesenchymal stem cell-like cells in human ACL tissues, their origin still remains unclear. Recently, blood vessels have been reported to represent a rich supply of stem/progenitor cells with a characteristic expression of CD34 and CD146. In this study, we attempted to validate the hypothesis that CD34- and CD146-expressing vascular cells exist in hACL tissues, have a potential for multi-lineage differentiation, and are recruited to the rupture site to participate in the intrinsic healing of injured ACL. Immunohistochemistry and flow cytometry analysis of hACL tissues demonstrated that it contains significantly more CD34 and CD146-positive cells in the ACL ruptured site compared with the noninjured midsubstance. CD34+CD45- cells isolated from ACL ruptured site showed higher expansionary potentials than CD146+CD45- and CD34-CD146-CD45- cells, and displayed higher differentiation potentials into osteogenic, adipogenic, and angiogenic lineages than the other cell populations. Immunohistochemistry of fetal and adult hACL tissues demonstrated a higher number of CD34 and CD146-positive cells in the ACL septum region compared with the midsubstance. In conclusion, our findings suggest that the ACL septum region contains a population of vascular-derived stem cells that may contribute to ligament regeneration and repair at the site of rupture.
Collapse
Affiliation(s)
- Tomoyuki Matsumoto
- Stem Cell Research Center, Children's Hospital of Pittsburgh, and Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
497
|
Regenerative phenotype in mice with a point mutation in transforming growth factor beta type I receptor (TGFBR1). Proc Natl Acad Sci U S A 2011; 108:14560-5. [PMID: 21841138 DOI: 10.1073/pnas.1111056108] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Regeneration of peripheral differentiated tissue in mammals is rare, and regulators of this process are largely unknown. We carried out a forward genetic screen in mice using N-ethyl-N-nitrosourea mutagenesis to identify genetic mutations that affect regenerative healing in vivo. More than 400 pedigrees were screened for closure of a through-and-through punch wound in the mouse ear. This led to the identification of a single pedigree with a heritable, fast, and regenerative wound-healing phenotype. Within 5 wk after ear-punch, a threefold decrease in the diameter of the wound was observed in the mutant mice compared with the wild-type mice. At 22 wk, new cartilage, hair follicles, and sebaceous glands were observed in the newly generated tissue. This trait was mapped to a point mutation in a receptor for TGF-β, TGFBR1. Mouse embryonic fibroblasts from the affected mice had increased expression of a subset of TGF-β target genes, suggesting that the mutation caused partial activation of the receptor. Further, bone marrow stromal cells from the mutant mice more readily differentiated to chondrogenic precursors, providing a plausible explanation for the enhanced development of cartilage islands in the regenerated ears. This mutant mouse strain provides a unique model to further explore regeneration in mammals and, in particular, the role of TGFBR1 in chondrogenesis and regenerative wound healing.
Collapse
|
498
|
Mwale F, Rampersad S, Richard H, Guoying Y, Al Rowas S, Madiraju P, Antoniou J, Laverty S. The constitutive expression of type x collagen in mesenchymal stem cells from osteoarthritis patients is reproduced in a rabbit model of osteoarthritis. J Tissue Eng 2011; 2011:587547. [PMID: 21808721 PMCID: PMC3144696 DOI: 10.4061/2011/587547] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 06/18/2011] [Indexed: 11/20/2022] Open
Abstract
The expression of type X collagen (COL X), a late-stage chondrocyte hypertrophy marker in human mesenchymal stem cells (MSCs) from osteoarthritis (OA) patients poses a major setback to current cartilage and intervertebral disc tissue engineering efforts. However, it is not yet clear whether COL X is expressed by all human bone marrow stem cells or if it is related to age, gender, site, disease status, or drug therapy. In the current study, we report that COL X expression is upregulated in MSCs from rabbits in a surgical instability model of OA (anterior cruciate ligament transection (ACLT)) when compared to control rabbit MSCs. Thus COL X expression in OA is a common phenomenon that is due to the disease process itself and not to other environmental factors. It is, therefore, critical to understand MSC phenotype in OA patients, as these cells are essential clinically for biological repair of cartilage lesions using autologous stem cells.
Collapse
Affiliation(s)
- Fackson Mwale
- Division of Orthopaedic Surgery, McGill University, Montreal, Quebec, Canada H3H 2P2
| | | | | | | | | | | | | | | |
Collapse
|
499
|
Pountos I, Giannoudis PV, Jones E, English A, Churchman S, Field S, Ponchel F, Bird H, Emery P, McGonagle D. NSAIDS inhibit in vitro MSC chondrogenesis but not osteogenesis: implications for mechanism of bone formation inhibition in man. J Cell Mol Med 2011; 15:525-34. [PMID: 20070439 PMCID: PMC3922374 DOI: 10.1111/j.1582-4934.2010.01006.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The non-steroidal anti-inflammatory drugs (NSAIDs) are widely used for analgesia but may inhibit bone formation. We investigated whether the reported NSAID effect on bone is related to inhibition of bone marrow mesenchymal stem cell (MSC) proliferation and osteogenic and chondrogenic differentiation and evaluated both cyclooxygenase (COX)-1 and COX-2 specific drugs. The effects of seven COX-1 and COX-2 inhibitors on MSC proliferation and osteogenic and chondrogenic differentiation were tested using Vybrant, sodium 3'-[1-(phenylaminocarbonyl)- 3,4-tetrazolium]-bis (4-methoxy-6-nitro) benzene sulfonic acid hydrate (XTT), functional and quantitative assays of MSC differentiation. The MSC expression of COX-1 and COX-2 and prostaglandin E2 (PGE-2) levels were evaluated serially during lineage differentiation by quantitative PCR and ELISA. None of the NSAIDs at broad range of concentration (range 10(-3) to 100 μg/ml) significantly affected MSC proliferation. Surprisingly, MSC osteogenic differentiation inhibition was not evident. However, NSAIDs affected chondrogenic potential with a reduction in sulphated glycosaminoglycans (sGAG) content by 45% and 55% with diclofenac and ketorolac, respectively (P < 0.05 compared to controls). Parecoxib and meloxicam, more COX-2 specific reagents inhibited sGAG to a lesser degree, 22% and 27% respectively (P < 0.05 compared to controls). Cartilage pellet immunohistochemistry confirmed the above results. Pellet chondrogenesis was associated with increased COX-1 expression levels but not COX-2, and COX-1 specific drugs suppressed MSC PGE-2 more than COX-2 specific inhibitors. These findings suggest that NSAIDs may inhibit bone formation via blockage of MSC chondrogenic differentiation which is an important intermediate phase in normal endochondral bone formation.
Collapse
|
500
|
Differentiation of Human Bone Marrow Mesenchymal Stem Cells to Chondrocytes for Construction of Three-dimensional Cartilage Tissue. Cytotechnology 2011; 47:11-7. [PMID: 19003040 DOI: 10.1007/s10616-005-3751-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2004] [Accepted: 03/21/2005] [Indexed: 01/13/2023] Open
Abstract
A differentiation method of human bone marrow mesenchymal stem cells (MSCs) to chondrocytes was developed for the construction of a three-dimensional (3D) cartilage tissue. The adhesive cells, which were isolated from a human bone marrow aspirate were embedded in type I collagen in a poly-L: -lactate-glycolic acid copolymer (PLGA) mesh and cultivated for 4 week together with growth factors. The degree of cellular differentiation was estimated by quantitative RT-PCR of aggrecan and type II collagen mRNAs and by staining with Safranin O. The 3D culture showed a higher degree of differentiation even without growth factors than the conventional pellet culture with growth factors, namely, dexamethasone and transforming growth factor (TGF)-beta 3. The 3D culture for 2 week with the combined addition of dexamethasone, TGF-beta 3, and insulin-like growth factor (IGF)-I reached a 30% expression of aggrecan mRNA compared with that in primary human chondrocytes, while the aggrecan mRNA expression in the conventional pellet culture was less than 2%. The sequential two-step differentiation cultivation, during which the cells were cultivated in 3D for 1 week after the conventional two-dimensional (2D) culture for 1 week, could markedly accelerate the expression of aggrecan mRNA compared with the 3D cultivation for 2 week.
Collapse
|