51
|
Hwang YS, Bishop AE, Polak JM, Mantalaris A. EnhancedIn vitro chondrogenic differentiation of murine embryonic stem cells. BIOTECHNOL BIOPROC E 2007. [DOI: 10.1007/bf02931088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
52
|
Sritanaudomchai H, Pavasuthipaisit K, Kitiyanant Y, Kupradinun P, Mitalipov S, Kusamran T. Characterization and multilineage differentiation of embryonic stem cells derived from a buffalo parthenogenetic embryo. Mol Reprod Dev 2007; 74:1295-302. [PMID: 17290421 DOI: 10.1002/mrd.20592] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Embryonic stem (ES) cells derived from mammalian embryos have the ability to form any terminally differentiated cell of the body. We herein describe production of parthenogenetic buffalo (Bubalus Bubalis) blastocysts and subsequent isolation of an ES cell line. Established parthenogenetic ES (PGES) cells exhibited diploid karyotype and high telomerase activity. PGES cells showed remarkable long-term proliferative capacity providing the possibility for unlimited expansion in culture. Furthermore, these cells expressed key ES cell-specific markers defined for primate species including stage-specific embryonic antigen-4 (SSEA-4), tumor rejection antigen-1-81 (TRA-1-81), and octamer-binding transcription factor 4 (Oct-4). In vitro, in the absence of a feeder layer, cells readily formed embryoid bodies (EBs). When cultured for an extended period of time, EBs spontaneously differentiated into derivatives of three embryonic germ layers as detected by PCR for ectodermal (nestin, oligodendrocytes, and tubulin), mesodermal (scleraxis, alpha-skeletal actin, collagen II, and osteocalcin) and endodermal markers (insulin and alpha-fetoprotein). Differentiation of PGES cells toward chondrocyte lineage was directed by supplementing serum-containing media with ascorbic acid, beta-glycerophosphate, and dexamethasone. Moreover, when PGES cells were injected into nude mice, teratomas with derivatives representing all three embryonic germ layers were produced. Our results suggest that the cell line isolated from a parthenogenetic blastocyst holds properties of ES cells, and can be used as an in vitro model to study the effects of imprinting on cell differentiation and as an a invaluable material for extensive molecular studies on imprinted genes.
Collapse
|
53
|
Kurosawa H. Methods for inducing embryoid body formation: in vitro differentiation system of embryonic stem cells. J Biosci Bioeng 2007; 103:389-98. [PMID: 17609152 DOI: 10.1263/jbb.103.389] [Citation(s) in RCA: 349] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2007] [Accepted: 02/13/2007] [Indexed: 02/07/2023]
Abstract
When cultured in suspension without antidifferentiation factors, embryonic stem (ES) cells spontaneously differentiate and form three-dimensional multicellular aggregates called embryoid bodies (EBs). EBs recapitulate many aspects of cell differentiation during early embryogenesis, and play an important role in the differentiation of ES cells into a variety of cell types in vitro. There are several methods for inducing the formation of EBs from ES cells. The three basic methods are liquid suspension culture in bacterial-grade dishes, culture in methylcellulose semisolid media, and culture in hanging drops. Recently, the methods using a round-bottomed 96-well plate and a conical tube are adopted for forming EBs from predetermined numbers of ES cells. For the production of large numbers of EBs, stirred-suspension culture using spinner flasks and bioreactors is performed. Each of these methods has its own peculiarity; thus, the features of formed EBs depending on the method used. Therefore, we should choose an appropriate method for EB formation according to the objective to be attained. In this review, we summarize the studies on in vitro differentiation of ES cells via EB formation and highlight the EB formation methods recently developed including the techniques, devices, and procedures involved.
Collapse
Affiliation(s)
- Hiroshi Kurosawa
- Division of Medicine and Engineering Science, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 4-3-11 Takeda, Kofu, Yamanashi, Japan.
| |
Collapse
|
54
|
Koay EJ, Hoben GMB, Athanasiou KA. Tissue engineering with chondrogenically differentiated human embryonic stem cells. Stem Cells 2007; 25:2183-90. [PMID: 17540854 DOI: 10.1634/stemcells.2007-0105] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This study describes the development and application of a novel strategy to tissue engineer musculoskeletal cartilages with human embryonic stem cells (hESCs). This work expands the presently limited understanding of how to chondrogenically differentiate hESCs through the use of chondrogenic medium alone (CM) or CM with two growth factor regimens: transforming growth factor (TGF)-beta3 followed by TGF-beta1 plus insulin-like growth factor (IGF)-I or TGF-beta3 followed by bone morphogenic protein (BMP)-2. It also extends the use of the resulting chondrogenically differentiated cells for cartilage tissue engineering through a scaffoldless approach called self-assembly, which was conducted in two modes: with (a) embryoid bodies (EBs) or (b) a suspension of cells enzymatically dissociated from the EBs. Cells from two of the differentiation conditions (CM alone and TGF-beta3 followed by BMP-2) produced fibrocartilage-like constructs with high collagen I content, low collagen II content, relatively high total collagen content (up to 24% by dry weight), low sulfated glycosaminoglycan content (approximately 4% by dry weight), and tensile properties on the order of megapascals. In contrast, hESCs treated with TGF-beta3 followed by TGF-beta1 + IGF-I produced constructs with no collagen I. Results demonstrated significant differences among the differentiation conditions in terms of other biochemical and biomechanical properties of the self-assembled constructs, suggesting that distinct growth factor regimens differentially modulate the potential of the cells to produce cartilage. Furthermore, this work shows that self-assembly of cells obtained by enzymatic dissociation of EBs is superior to self-assembly of EBs. Overall, the results of this study raise the possibility of manipulating the characteristics of hESC-generated tissue toward specific musculoskeletal cartilage applications.
Collapse
Affiliation(s)
- Eugene J Koay
- Rice University, Department of Bioengineering, Houston, Texas 77251-1892, USA
| | | | | |
Collapse
|
55
|
Woll NL, Heaney JD, Bronson SK. Osteogenic nodule formation from single embryonic stem cell-derived progenitors. Stem Cells Dev 2007; 15:865-79. [PMID: 17253949 DOI: 10.1089/scd.2006.15.865] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The process of bone formation can be approximated in vitro in the form of a mineralized nodule. Osteoprogenitors and mesenchymal stem cells (MSCs), the immediate precursors of the osteoprogenitor, proliferate and differentiate into osteoblasts when placed into culture. These osteoblasts secrete and mineralize a matrix during a period of 3-4 weeks. The differentiation potential of embryonic stem (ES) cells suggests that ES cells should also have the ability to form osteogenic nodules in vitro. ES cells were allowed to form embryoid bodies (EBs) and were cultured in suspension for 2 days; EBs were disrupted and plated as single cells at concentrations as low as 25 cells/cm(2). We provide five lines of evidence for osteogenesis in these ES cell-derived cultures: (1) cell and colony morphology as revealed by phase-contrast microscopy, (2) mineralization of extracellular matrix as revealed by von Kossa staining, (3) quantitative real-time PCR (QRT-PCR) analysis of cDNA from entire plates and individual colonies revealing expression of genes characteristic of, and specific for, osteoblasts, (4) confocal microscopy of nodules from osteocalcin-green fluorescent protein (GFP) ES cell lines demonstrating the appropriate stage and position of osteoblasts expressing the reporter, and (5) immunostaining of nodules with a type I collagen antibody. Our method of initiating osteogenesis from ES cell-derived cultures is the only described method that allows for the observation and manipulation of the commitment stage of mesengenesis from single embryonic progenitors.
Collapse
Affiliation(s)
- Nicole L Woll
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033-0850, USA
| | | | | |
Collapse
|
56
|
Toh WS, Yang Z, Liu H, Heng BC, Lee EH, Cao T. Effects of Culture Conditions and Bone Morphogenetic Protein 2 on Extent of Chondrogenesis from Human Embryonic Stem Cells. Stem Cells 2007; 25:950-60. [PMID: 17218402 DOI: 10.1634/stemcells.2006-0326] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The study of human embryonic stem cells (hESCs) can provide invaluable insights into the development of numerous human cell and tissue types in vitro. In this study, we addressed the potential of hESCs to undergo chondrogenesis and demonstrated the potential of hESC-derived embryoid bodies (EBs) to undergo a well-defined full-span chondrogenesis from chondrogenic induction to hypertrophic maturation. We compared chondrogenic differentiation of hESCs through EB direct-plating outgrowth system and EB-derived high-density micromass systems under defined serumfree chondrogenic conditions and demonstrated that cell-to-cell contact and bone morphogenetic protein 2 (BMP2) treatment enhanced chondrocyte differentiation, resulting in the formation of cartilaginous matrix rich in collagens and proteoglycans. Provision of a high-density three-dimensional (3D) microenvironment at the beginning of differentiation is critical in driving chondrogenesis because increasing EB seeding numbers in the EB-outgrowth system was unable to enhance chondrogenesis. Temporal order of chondrogenic differentiation and hypertrophic maturation indicated by the gene expression profiles of Col 1, Col 2, and Col 10, and the deposition of extracellular matrix (ECM) proteins, proteoglycans, and collagen II and X demonstrated that the in vivo progression of chondrocyte maturation is recapitulated in the hESC-derived EB model system established in this study. Furthermore, we also showed that BMP2 can influence EB differentiation to multiple cell fates, including that of extraembryonic endodermal and mesenchymal lineages in the EB-outgrowth system, but was more committed to driving the chondrogenic cell fate in the EB micromass system. Overall, our findings provide a potential 3D model system using hESCs to delineate gene function in lineage commitment and restriction of chondrogenesis during embryonic cartilage development.
Collapse
Affiliation(s)
- Wei Seong Toh
- Stem Cell Laboratory, Faculty of Dentistry, Yong Loo Lin School of Medicine, Tissue Engineering Program, National University of Singapore, Singapore
| | | | | | | | | | | |
Collapse
|
57
|
Toh WS, Yang Z, Heng BC, Cao T. Differentiation of human embryonic stem cells toward the chondrogenic lineage. Methods Mol Biol 2007; 407:333-349. [PMID: 18453266 DOI: 10.1007/978-1-59745-536-7_23] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Human embryonic stem cells (hESCs) have the ability to self-replicate and differentiate into cells from all three embryonic germ layers, thereby holding great promise for tissue regeneration applications. However, controlling the differentiation of hESCs and obtaining homogenous differentiated cell populations still remain a challenge. We present a highly efficient and reproducible experimental system that mimics the three-dimensional (3-D) environment of in vivo chondrogenesis and that supports the directed differentiation of human embryoid body (EB)-derived cells toward the chondrogenic lineage under serum-free chondrogenic culture conditions in the presence of bone morphogenetic protein-2 (BMP-2).
Collapse
Affiliation(s)
- Wei Seong Toh
- Stem Cell Laboratory, Faculty of Dentistry, National University of Singapore, Singapore
| | | | | | | |
Collapse
|
58
|
Abstract
PURPOSE OF REVIEW The most commonly used therapies in nephrology target the reduction of acute injury, reduction of the rate of progression, or renal replacement therapy. The purpose of this review is to examine new evidence that renal progenitors can be used for therapeutic purposes. Stem cells possess two characteristics, self-renewal and the capacity for multilineage differentiation. They are typically classified as derived from embryos or from the adult. RECENT FINDINGS New studies on embryonic stem cells show that they can be use to enrich for specific renal progenitors, which integrate into mature structures. Studies on adult stem cells show that almost all kidney cell types can be renewed by adult stem cells originating in bone marrow. Moreover, some animal studies demonstrate that a phenotype such as the aging and diabetic phenotype can be transferred from progenitors residing in the bone marrow, suggesting that the bone marrow contains renal progenitors that may be useful for therapeutic purposes. SUMMARY Stem cell therapy opens the door to regenerative nephrology. Embryonic stem cells are a useful tool to determine the pathways to convert a pluripotent stem cell into renal progenitors. Adult stem cells in the bone marrow or in a specific kidney niche may provide a source of stem cells with a therapeutic potential.
Collapse
Affiliation(s)
- Cecile Vigneau
- Mount Sinai School of Medicine, Department of Medicine, New York, New York 10029, USA
| | | | | | | | | |
Collapse
|
59
|
Hwang NS, Varghese S, Zhang Z, Elisseeff J. Chondrogenic differentiation of human embryonic stem cell-derived cells in arginine-glycine-aspartate-modified hydrogels. ACTA ACUST UNITED AC 2006; 12:2695-706. [PMID: 16995803 DOI: 10.1089/ten.2006.12.2695] [Citation(s) in RCA: 190] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Human embryonic stem cells (hESCs) have the potential to self-renew and generate multiple cell types, producing critical building blocks for tissue engineering and regenerative medicine applications. Here, we describe the efficient derivation and chondrogenic differentiation of mesenchymal-like cells from hESCs. These cells exhibit mesenchymal stem cell (MSC) surface markers, including CD29, CD44, CD105, and platelet-derived growth factor receptor-alpha. Under appropriate growth conditions, the hESC-derived cells proliferated without phenotypic changes and maintained MSC surface markers. The chondrogenic capacity of the cells was studied in pellet culture and after encapsulation in poly(ethylene glycol)-diacrylate (PEGDA) hydrogels with exogenous extracellular proteins or arginineglycine- aspartate (RGD)-modified PEGDA hydrogels. The hESC-derived cells exhibited growth factor- dependent matrix production in pellet culture but did not produce tissue characteristic of cartilage morphology. In PEGDA hydrogels containing exogenous hyaluronic acid or type I collagen, no significant cell growth or matrix production was observed. In contrast, when these cells were encapsulated in RGDmodified poly(ethylene glycol)hydrogels, neocartilage with basophilic extracellular matrix deposition was observed within 3 weeks of culture, producing cartilage-specific gene up-regulation and extracellular matrix production. Our results indicate that precursor cells characteristic of a MSC population can be cultured from differentiating hESCs through embryoid bodies, thus holding great promise for a potentially unlimited source of cells for cartilage tissue engineering.
Collapse
Affiliation(s)
- Nathaniel S Hwang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, USA
| | | | | | | |
Collapse
|
60
|
Hwang YS, Randle WL, Bielby RC, Polak JM, Mantalaris A. Enhanced derivation of osteogenic cells from murine embryonic stem cells after treatment with HepG2-conditioned medium and modulation of the embryoid body formation period: application to skeletal tissue engineering. ACTA ACUST UNITED AC 2006; 12:1381-92. [PMID: 16846337 DOI: 10.1089/ten.2006.12.1381] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Despite the considerable progress made in directing embryonic stem cell (ESC) differentiation to therapeutically useful lineages, several issues remain to be resolved before ESCs can be used for cell therapy: 1) increasing the efficiency of specific lineage generation, and 2) developing time- and cost-effective culture systems for controlling ESC differentiation. Our study aimed to develop efficient methods to enhance mesodermal differentiation and thereby upregulate osteogenic differentiation of ESCs. Specifically, murine ESCs (mESCs) were cultured in the presence of 50% conditioned medium (CM) from the human hepatocarcinoma cell line HepG2, which resulted in enhanced mesoderm formation during embryoid body (EB) formation in the CM-treated mESCs (CM-mESCs). By varying the length of EB culture time, we achieved the selective control and stimulation of osteogenic differentiation and suppression of cardiogenic differentiation. Hence, reducing the EB culture of the CM-mESCs to 1 day resulted in 5-10-fold enhancement of osteogenic differentiation, as determined by bone nodule formation, higher alkaline phosphatase activity, the presence of well-organized osteoblast-cadherin in the bone nodules, and increased cbfa-1/runx2 gene expression. In contrast, increasing the EB culture of the CM-mESCs to 5 days resulted in three- to four-fold enhanced cardiogenic differentiation. These findings for development of highly efficient culture systems and protocols for mESC differentiation into osteogenic lineage that are time- and cost-effective can be used in skeletal tissue engineering applications.
Collapse
Affiliation(s)
- Yu-Shik Hwang
- Department of Chemical Engineering, Imperial College London, London, UK
| | | | | | | | | |
Collapse
|
61
|
Abstract
Injuries to the knee meniscus, particularly those in the avascular region, pose a complex problem and a possible solution is tissue engineering of a replacement tissue. Tissue engineering of the meniscus involves scaffold selection, addition of cells, and stimulation of the construct to synthesize, maintain, or enhance matrix production. An acellular collagen implant is currently in clinical trials and there are promising results with other scaffolds, composed of both polymeric and natural materials. The addition of cells to these constructs may promote good matrix production in vitro, but has been studied in a limited manner in animal studies. Cell sources ranging from fibroblasts to stem cells could be used to overcome challenges in cell procurement, expansion, and synthetic capacity currently encountered in studies with fibrochondrocytes. Manipulation of construct culture with exogenous growth factors and mechanical stimulation will also likely play a role in these strategies.
Collapse
Affiliation(s)
- Gwendolyn M Hoben
- Department of Bioengineering, Rice University, Houston, TX 77251, USA
| | | |
Collapse
|
62
|
Elisseeff J, Ferran A, Hwang S, Varghese S, Zhang Z. The role of biomaterials in stem cell differentiation: applications in the musculoskeletal system. Stem Cells Dev 2006; 15:295-303. [PMID: 16846368 DOI: 10.1089/scd.2006.15.295] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The capabilities of stem cells continue to be revealed, leading to excitement regarding potential new therapies. Regenerative medicine is an area in which stem cells hold great promise for overcoming the challenge of limited cell sources for tissue repair. Biomaterials play an important role in directing tissue growth and may provide another tool to manipulate and control stem cell behavior. Biomaterials are made from natural or synthetic polymers and can be processed into three-dimensional scaffolds designed to promote cell proliferation and/or differentiation that ultimately produces new tissue. Stem cells will have a significant impact on the fields of regenerative medicine and tissue engineering as a powerful cell source that will work, in conjunction with biomaterials, to treat tissue and organ loss. Herein, we survey our latest research on applying embryonic stem (ES) cells to hydrogel biomaterials for engineering musculoskeletal tissues, emphasizing the unique biomaterial requirements of ES cells for differentiation and tissue development.
Collapse
Affiliation(s)
- J Elisseeff
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.
| | | | | | | | | |
Collapse
|
63
|
Abstract
The regenerative capacity of cartilage is limited. Transplantation methods used to treat cartilage lesions are based mainly on primary cultures of chondrocytes, which dedifferentiate during cultivation in vitro and lose their functional properties. Stem cells are considered as an alternative source to generate cells for two reasons: first, they can almost indefinitely divide in culture, and second, they are able to differentiate into various mature cell types. Herein, we asked the question whether chondrocytes could be differentiated from mouse embryonic stem (ES) cells to a state suitable for regenerative use. When cultivated as embryoid bodies (EBs), murine ES cells differentiate into mesenchymal progenitor cells, which progressively develop into mature, hypertrophic chondrocytes and transdifferentiate into calcifying cells recapitulating all of the cellular processes of chondrogenesis. Chondrocytes isolated from EBs exhibit a high regenerative capacity. They dedifferentiate initially in culture, but later reexpress stable characteristics of mature chondrocytes. However, in cultures of chondrocytes isolated from EBs, additional mesenchymal cell types can be observed. Mesenchymal stem (MS) cells from bone marrow have already been used in tissue engineering settings. We compared the chondrogenic differentiation of MS and ES cells.
Collapse
Affiliation(s)
- J Kramer
- Department of Medical Molecular Biology, University of Lübeck, Campus Lübeck, Lübeck, Germany.
| | | | | | | |
Collapse
|
64
|
Liu H, Collins SF, Suggs LJ. Three-dimensional culture for expansion and differentiation of mouse embryonic stem cells. Biomaterials 2006; 27:6004-14. [PMID: 16860386 DOI: 10.1016/j.biomaterials.2006.06.016] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2006] [Accepted: 06/14/2006] [Indexed: 02/01/2023]
Abstract
Differentiation of embryonic stem (ES) cells typically requires cell-cell aggregation in the form of embryoid bodies (EBs). This process is not very well controlled and final cell numbers can be limited by EB agglomeration and the inability to drive differentiation towards a desired cell type. This study compares three-dimensional (3D) fibrin culture to conventional two-dimensional (2D) suspension culture and to culture in a semisolid methylcellulose medium solution. Two types of fibrin culture were evaluated, including a PEGylated fibrin gel. PEGylation with a difunctional PEG derivative retarded fibrinogen migration during through sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) as a result of crosslinking, similarly, degradation was slowed in the PEGylated gel. ES cell proliferation was higher in both the fibrin and PEGylated fibrin gels versus 2D and methylcellulose controls. FACS analysis and real-time-PCR revealed differences in patterns of differentiation for the various culture systems. Culture in PEGylated fibrin or methylcellulose culture demonstrated features characteristic of less extensive differentiation relative to fibrin and 2D culture as evidenced by the transcription factor Oct-4. Fibrin gels showed gene and protein expression similar to that in 2D culture. Both fibrin and 2D cultures demonstrated statistically greater cell numbers positive for the vascular mesoderm marker, VE-cadherin.
Collapse
Affiliation(s)
- Hui Liu
- Zimmer Inc., 12024 Vista Parke Drive, Austin, TX 78726, USA
| | | | | |
Collapse
|
65
|
Kramer J, Steinhoff J, Klinger M, Fricke L, Rohwedel J. Cells differentiated from mouse embryonic stem cells via embryoid bodies express renal marker molecules. Differentiation 2006; 74:91-104. [PMID: 16533308 DOI: 10.1111/j.1432-0436.2006.00062.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Differentiation of mouse embryonic stem (ES) cells via embryoid bodies (EB) is established as a suitable model to study cellular processes of development in vitro. ES cells are known to be pluripotent because of their capability to differentiate into cell types of all three germ layers including germ cells. Here, we show that ES cells differentiate into renal cell types in vitro. We found that genes were expressed during EB cultivation, which have been previously described to be involved in renal development. Marker molecules characteristic for terminally differentiated renal cell types were found to be expressed predominantly during late stages of EB cultivation, while marker molecules involved in the initiation of nephrogenesis were already expressed during early steps of EB development. On the cellular level--using immunostaining--we detected cells expressing podocin, nephrin and wt-1, characteristic for differentiated podocytes and other cells, which expressed Tamm-Horsfall protein, a marker for distal tubule epithelial cells of kidney tissue. Furthermore, the proximal tubule marker molecules renal-specific oxido reductase, kidney androgen-related protein and 25-hydroxyvitamin D3alpha-hydroxylase were found to be expressed in EBs. In particular, we could demonstrate that cells expressing podocyte marker molecules assemble to distinct ring-like structures within the EBs. Because the differentiation efficiency into these cell types is still relatively low, application of fibroblast growth factor (FGF)-2 in combination with leukaemia inhibitory factor was tested for induction, but did not enhance ES cell-derived renal differentiation in vitro.
Collapse
Affiliation(s)
- Jan Kramer
- Department of Medical Molecular Biology, University of Lübeck, D-23538 Lübeck, Germany.
| | | | | | | | | |
Collapse
|
66
|
Hwang NS, Kim MS, Sampattavanich S, Baek JH, Zhang Z, Elisseeff J. Effects of Three-Dimensional Culture and Growth Factors on the Chondrogenic Differentiation of Murine Embryonic Stem Cells. Stem Cells 2006; 24:284-91. [PMID: 16109760 DOI: 10.1634/stemcells.2005-0024] [Citation(s) in RCA: 173] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Embryonic stem (ES) cells have the ability to self-replicate and differentiate into cells from all three germ layers, holding great promise for tissue regeneration applications. However, controlling the differentiation of ES cells and obtaining homogenous cell populations still remains a challenge. We hypothesize that a supportive three-dimensional (3D) environment provides ES cell-derived cells an environment that more closely mimics chondrogenesis in vivo. In the present study, the chondrogenic differentiation capability of ES cell-derived embryoid bodies (EBs) encapsulated in poly(ethylene glycol)-based (PEG) hydrogels was examined and compared with the chondrogenic potential of EBs in conventional monolayer culture. PEG hydrogel-encapsulated EBs and EBs in monolayer were cultured in vitro for up to 17 days in chondrogenic differentiation medium in the presence of transforming growth factor (TGF)-beta1 or bone morphogenic protein-2. Gene expression and protein analyses indicated that EB-PEG hydrogel culture upregulated cartilage-relevant markers compared with a monolayer environment and induction of chondrocytic phenotype was stimulated with TGF-beta1. Histology of EBs in PEG hydrogel culture with TGF-beta1 demonstrated basophilic extracellular matrix deposition characteristic of neocartilage. These findings suggest that EB-PEG hydrogel culture, with an appropriate growth factor, may provide a suitable environment for chondrogenic differentiation of intact ES cell-derived EBs.
Collapse
Affiliation(s)
- Nathaniel S Hwang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Clark Hall 106, 3400 North Charles Street, Baltimore, Maryland 21218, USA
| | | | | | | | | | | |
Collapse
|
67
|
Abstract
Human embryonic stem cells (hESC) provide a potentially unlimited source of specialized cell types for regenerative medicine. Nonetheless, one of the key requirements used to fulfill this potential is the ability to direct the differentiation of hESC to selective fates in vitro. Studies have reported the development of culture strategies to derive multipotent mesenchymal precursors from hESCs in vitro. This chapter reviews the techniques that allow the selective derivation of such precursors and their differentiation toward various mesenchymal cell types. It also discusses current limitations and future perspectives on the use of hESC-derived mesenchymal tissues.
Collapse
Affiliation(s)
- Tiziano Barberi
- Division of Neurosciences, Beckham Research Institute of the City of Hope, Duarte, California, USA
| | | |
Collapse
|
68
|
Dravida S, Pal R, Khanna A, Tipnis SP, Ravindran G, Khan F. The transdifferentiation potential of limbal fibroblast-like cells. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2005; 160:239-51. [PMID: 16253345 DOI: 10.1016/j.devbrainres.2005.09.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2005] [Revised: 09/03/2005] [Accepted: 09/16/2005] [Indexed: 11/21/2022]
Abstract
We report the identification and isolation of limbal fibroblast-like cells from adult corneo-limbal tissue possessing self-renewing capacity and multilineage differentiation potential. The cells form cell aggregates or clusters, which express molecular markers, specific for ectoderm, mesoderm and endoderm lineages in vitro. Further, these cells mature into a myriad of cell types including neurons, corneal cells, osteoblasts, chondrocytes, adipocytes, cardiomyocytes, hepatocytes and pancreatic islet cells. Despite originating from a non-embryonic source, they express ESC and other stem cell markers important for maintaining an undifferentiated state. This multipotential capability, relatively easy isolation and high rate of ex vivo proliferation capacity make these cells a promising therapeutic tool.
Collapse
Affiliation(s)
- Subhadra Dravida
- Embryonic and Ocular Stem Cell Group, Reliance Life Sciences Pvt. Ltd., Dhirubhai Ambani Life Sciences Centre, R-282, TTC Area of MIDC, Thane Belapur Road, Rabale, Navi Mumbai 400 701, India.
| | | | | | | | | | | |
Collapse
|
69
|
Abstract
The inadequacy of current treatment modalities and insufficiency of donor organs for cadaveric transplantation have driven a search for improved methods of dealing with renal failure. The rising concept of cell-based therapeutics has provided a framework around which new approaches are being generated, and its combination with advances in stem cell research stands to bring both fields to clinical fruition. This budding partnership is presently in its very early stages, but an examination of the cell-based therapies currently under development clearly shows the magnitude of the role that stem cells will ultimately play. The issue over reports of unexpected plasticity in adult stem cell differentiation remains a focus of debate, and evidence for bone marrow-derived stem cell contributions to renal repair has been challenged. The search for adult renal stem cells, which could have a considerable impact on much of the work discussed here, appears to be narrowing. The use of embryonic tissue in research continues to provide valuable insights but will be the subject of intense societal scrutiny and debate before it reaches the stage of clinical application. Embryonic stem (ES) cells, with their ability to generate all, or nearly all, of the cell types in the adult body and a possible source of cells genetically identical to the donor, hold great promise but face ethical and political hurdles for human use. Immunoisolation of heterologous cells by encapsulation creates opportunities for their safe use as a component of implanted or ex vivo devices.
Collapse
Affiliation(s)
- James C Brodie
- Division of Nephrology, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA.
| | | |
Collapse
|
70
|
Kramer J, Klinger M, Kruse C, Faza M, Hargus G, Rohwedel J. Ultrastructural analysis of mouse embryonic stem cell-derived chondrocytes. ACTA ACUST UNITED AC 2005; 210:175-85. [PMID: 16211392 DOI: 10.1007/s00429-005-0020-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2005] [Indexed: 11/30/2022]
Abstract
Pluripotent embryonic stem (ES) cells cultivated as cellular aggregates, so called embryoid bodies (EBs), differentiate spontaneously into different cell types of all three germ layers in vitro resembling processes of cellular differentiation during embryonic development. Regarding chondrogenic differentiation, murine ES cells differentiate into progenitor cells, which form pre-cartilaginous condensations in the EB-outgrowths and express marker molecules characteristic for mesenchymal cell types such as Sox5 and Sox6. Later, mature chondrocytes appear which express collagen type II, and the collagen fibers show a typical morphology as demonstrated by electron-microscopical analysis. These mature chondrogenic cells are organized in cartilage nodules and produce large amounts of extracellular proteoglycans as revealed by staining with cupromeronic blue. Finally, cells organized in nodules express collagen type X, indicating the hypertrophic stage. In conclusion, differentiation of murine ES cells into chondrocytes proceeds from the undifferentiated stem cell via progenitor cells up to mature chondrogenic cells, which then undergo hypertrophy. Furthermore, because the ES-cell-derived chondrocytes did not express elastin, a marker for elastic cartilage tissue, we suggest the cartilage nodules to resemble hyaline cartilage tissue.
Collapse
Affiliation(s)
- Jan Kramer
- Department of Medical Molecular Biology, University of Lübeck, Lübeck, Germany.
| | | | | | | | | | | |
Collapse
|
71
|
Abstract
OBJECTIVES Tissue engineering has the potential to make a significant impact on improving tissue repair in the craniofacial system. The general strategy for tissue engineering includes seeding cells on a biomaterial scaffold. The number of scaffold and cell choices for tissue engineering systems is continually increasing and will be reviewed. DESIGN Multilayered hydrogel systems were developed to coculture different cell types and develop osteochondral tissues for applications including the temporomandibular joint. EXPERIMENTAL VARIABLE Hydrogels are one form of scaffold that can be applied to cartilage and bone repair using fully differentiated cells, adult and embryonic stem cells. OUTCOME MEASURE Case studies represent an overview of our laboratory's investigations. RESULTS Bilayered scaffolds to promote tissue development and the formation of more complex osteochondral tissues were developed and proved to be effective. CONCLUSION Tissue engineering provides a venue to investigate tissue development of mutant or diseased cells and potential therapeutics.
Collapse
Affiliation(s)
- J Elisseeff
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.
| | | | | | | |
Collapse
|
72
|
Huojia M, Muraoka N, Yoshizaki K, Fukumoto S, Nakashima M, Akamine A, Nonaka K, Ohishi M. TGF-beta3 induces ectopic mineralization in fetal mouse dental pulp during tooth germ development. Dev Growth Differ 2005; 47:141-52. [PMID: 15839999 DOI: 10.1111/j.1440-169x.2005.00790.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Several members of the transforming growth factor (TGF)-beta superfamily are expressed in developing teeth from the initiation stage through adulthood. Of those, TGF-beta1 regulates odontoblast differentiation and dentin extracellular matrix synthesis. However, the molecular mechanism of TGF-beta3 in dental pulp cells is not clearly understood. In the present study, beads soaked with human recombinant TGF-beta3 induced ectopic mineralization in dental pulp from fetal mouse tooth germ samples, which increased in a dose-dependent manner. Further, TGF-beta3 promoted mRNA expression, and increased protein levels of osteocalcin (OCN) and type I collagen (COL I) in dental pulp cells. We also observed that the expression of dentin sialophosphoprotein and dentin matrix protein 1 was induced by TGF-beta3 in primary cultured dental pulp cells, however, not in calvaria osteoblasts, whereas OCN, osteopontin and osteonectin expression was increased after treatment with TGF-beta3 in both dental pulp cells and calvaria osteoblasts. Dentin sialoprotein was also partially detected in the vicinity of TGF-beta3 soaked beads in vivo. These results indicate for the first time that TGF-beta3 induces ectopic mineralization through upregulation of OCN and COL I expression in dental pulp cells, and may regulate the differentiation of dental pulp stem cells to odontoblasts.
Collapse
Affiliation(s)
- Muhetaer Huojia
- Division of Maxillofacial Diagnostic and Surgical Science, Faculty of Dental Science, Kyushu University, Higashi-Ku, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | |
Collapse
|
73
|
Singla DK, Kumar D, Sun B. Transforming growth factor-beta2 enhances differentiation of cardiac myocytes from embryonic stem cells. Biochem Biophys Res Commun 2005; 332:135-41. [PMID: 15896309 DOI: 10.1016/j.bbrc.2005.04.098] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2005] [Accepted: 04/13/2005] [Indexed: 01/09/2023]
Abstract
Stem cell therapy holds great promise for the treatment of injured myocardium, but is challenged by a limited supply of appropriate cells. Three different isoforms of transforming growth factor-beta (TGF-beta) -beta1, -beta2, and -beta3 exhibit distinct regulatory effects on cell growth, differentiation, and migration during embryonic development. We compared the effects of these three different isoforms on cardiomyocyte differentiation from embryonic stem (ES) cells. In contrast to TGF-beta1, or -beta3, treatment of mouse ES cells with TGF-beta2 isoform significantly increased embryoid body (EB) proliferation as well as the extent of the EB outgrowth that beat rhythmically. At 17 days, 49% of the EBs treated with TGF-beta2 exhibited spontaneous beating compared with 15% in controls. Cardiac myocyte specific protein markers sarcomeric myosin and alpha-actin were demonstrated in beating EBs and cells isolated from EBs. In conclusion, TGF-beta2 but not TGF-beta1, or -beta3 promotes cardiac myocyte differentiation from ES cells.
Collapse
|
74
|
Heng BC, Cao T, Lee EH. Directing stem cell differentiation into the chondrogenic lineage in vitro. Stem Cells 2005; 22:1152-67. [PMID: 15579636 DOI: 10.1634/stemcells.2004-0062] [Citation(s) in RCA: 164] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A major area in regenerative medicine is the application of stem cells in cartilage tissue engineering and reconstructive surgery. This requires well-defined and efficient protocols for directing the differentiation of stem cells into the chondrogenic lineage, followed by their selective purification and proliferation in vitro. The development of such protocols would reduce the likelihood of spontaneous differentiation of stem cells into divergent lineages upon transplantation, as well as reduce the risk of teratoma formation in the case of embryonic stem cells. Additionally, such protocols could provide useful in vitro models for studying chondrogenesis and cartilaginous tissue biology. The development of pharmacokinetic and cytotoxicity/genotoxicity screening tests for cartilage-related biomaterials and drugs could also utilize protocols developed for the chondrogenic differentiation of stem cells. Hence, this review critically examines the various strategies that could be used to direct the differentiation of stem cells into the chondrogenic lineage in vitro.
Collapse
Affiliation(s)
- Boon Chin Heng
- Faculty of Dentistry, National University of Singapore, Singapore
| | | | | |
Collapse
|
75
|
Sekiya I, Larson BL, Vuoristo JT, Reger RL, Prockop DJ. Comparison of effect of BMP-2, -4, and -6 on in vitro cartilage formation of human adult stem cells from bone marrow stroma. Cell Tissue Res 2005; 320:269-76. [PMID: 15778851 DOI: 10.1007/s00441-004-1075-3] [Citation(s) in RCA: 219] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2004] [Accepted: 12/15/2004] [Indexed: 12/13/2022]
Abstract
The human adult stem cells from bone marrow stroma referred to as mesenchymal stem cells or marrow stromal cells (MSCs) are of interest because they are easily isolated and expanded and are capable of multipotential differentiation. Here, we examined the ability of recombinant human bone morphogenetic protein (BMP)-2, -4, and -6 to enhance in vitro cartilage formation of MSCs. Human MSCs were isolated from bone marrow taken from normal adult donors. The cells were pelleted and cultured for 21 days in chondrogenic medium containing transforming growth factor beta3 and dexamethasone with or without BMP-2, -4, or -6. All the BMPs tested increased chondrogenic differentiation as assayed by immunohistochemistry and by the size and weight of the cartilage synthesized. However, BMP-2 was the most effective. Microarray analyses of approximately 12,000 genes and reverse transcription-polymerase chain reaction assays established that the critical genes for cartilage synthesis were expressed in the expected time sequence in response to BMP-2. The tissue engineering of autologous cartilage derived from MSCs in vitro for transplantation will be a future alternative for patients with cartilage injuries. To obtain large amounts of cartilage rich in proteoglycans, the use of BMP-2 is recommended, instead of BMP-4 or -6.
Collapse
Affiliation(s)
- Ichiro Sekiya
- Center for Gene Therapy, Tulane University Health Sciences Center, New Orleans, LA 70112-2699, USA
| | | | | | | | | |
Collapse
|
76
|
zur Nieden NI, Kempka G, Rancourt DE, Ahr HJ. Induction of chondro-, osteo- and adipogenesis in embryonic stem cells by bone morphogenetic protein-2: effect of cofactors on differentiating lineages. BMC DEVELOPMENTAL BIOLOGY 2005; 5:1. [PMID: 15673475 PMCID: PMC548146 DOI: 10.1186/1471-213x-5-1] [Citation(s) in RCA: 167] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2004] [Accepted: 01/26/2005] [Indexed: 11/10/2022]
Abstract
Background Recently, tissue engineering has merged with stem cell technology with interest to develop new sources of transplantable material for injury or disease treatment. Eminently interesting, are bone and joint injuries/disorders because of the low self-regenerating capacity of the matrix secreting cells, particularly chondrocytes. ES cells have the unlimited capacity to self-renew and maintain their pluripotency in culture. Upon induction of various signals they will then differentiate into distinctive cell types such as neurons, cardiomyocytes and osteoblasts. Results We present here that BMP-2 can drive ES cells to the cartilage, osteoblast or adipogenic fate depending on supplementary co-factors. TGFβ1, insulin and ascorbic acid were identified as signals that together with BMP-2 induce a chondrocytic phenotype that is characterized by increased expression of cartilage marker genes in a timely co-ordinated fashion. Expression of collagen type IIB and aggrecan, indicative of a fully mature state, continuously ascend until reaching a peak at day 32 of culture to approximately 80-fold over control values. Sox9 and scleraxis, cartilage specific transcription factors, are highly expressed at very early stages and show decreased expression over the time course of EB differentiation. Some smaller proteoglycans, such as decorin and biglycan, are expressed at earlier stages. Overall, proteoglycan biosynthesis is up-regulated 7-fold in response to the supplements added. BMP-2 induced chondrocytes undergo hypertrophy and begin to alter their expression profile towards osteoblasts. Supplying mineralization factors such as β-glycerophosphate and vitamin D3 with the culture medium can facilitate this process. Moreover, gene expression studies show that adipocytes can also differentiate from BMP-2 treated ES cells. Conclusions Ultimately, we have found that ES cells can be successfully triggered to differentiate into chondrocyte-like cells, which can further alter their fate to become hypertrophic, and adipocytes. Compared with previous reports using a brief BMP-2 supplementation early in differentiation, prolonged exposure increased chondrogenic output, while supplementation with insulin and ascorbic acid prevented dedifferentiation. These results provide a foundation for the use of ES cells as a potential therapy in joint injury and disease.
Collapse
Affiliation(s)
- Nicole I zur Nieden
- Molecular & Genetic Toxicology, Bayer HealthCare AG, Wuppertal, Germany
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Canada
- Faculty of Medicine, Dept. of Biochemistry & Molecular Biology, University of Calgary, HMRB 331, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Grazyna Kempka
- Molecular & Genetic Toxicology, Bayer HealthCare AG, Wuppertal, Germany
| | - Derrick E Rancourt
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Canada
| | - Hans-Jürgen Ahr
- Molecular & Genetic Toxicology, Bayer HealthCare AG, Wuppertal, Germany
| |
Collapse
|
77
|
Kim MS, Hwang NS, Lee J, Kim TK, Leong K, Shamblott MJ, Gearhart J, Elisseeff J. Musculoskeletal Differentiation of Cells Derived from Human Embryonic Germ Cells. Stem Cells 2005; 23:113-23. [PMID: 15625128 DOI: 10.1634/stemcells.2004-0110] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Stem cells have the potential to significantly improve cell and tissue regeneration therapies, but little is understood about how to control their behavior. We investigated the potential differentiation capability of cells derived from human embryonic germ (EG) cells into musculoskeletal lineages by providing a three-dimensional environment with increased cell-cell contact and growth factors. Cells were clustered into pellets to mimic the mesenchyme condensation process during limb development. LVEC cells, an embryoid body-derived (EBD) cell culture generated from EG cells, were cultured in micromass pellets for 21 days in the presence of bone morphogenetic protein 2 (BMP2) and/or transforming growth factor beta-3 (TGFbeta3). Gene expression for cartilage-, bone-, and muscle-specific matrix proteins--including collagen types I, II, III, IX, X; aggrecan; cartilage proteoglycan link protein; cartilage oligomeric protein; chondroitin sulfate-4-S; and myf5--was upregulated in the pellets treated with TGFbeta3, while mRNAs for neurofilament heavy (NFH), a neuron marker, and flk-1, a hematopoietic marker, decreased. Total collagen and proteoglycan production exhibited a time-dependent increase in the pellets treated with TGFbeta3, further confirming the expression of characteristic musculoskeletal markers. Furthermore, our results indicate the ability to select or differentiate stem cells toward a musculoskeletal lineage from a heterogenous EBD cell line.
Collapse
Affiliation(s)
- Myoung Sook Kim
- Department of Biomedical Engineering, Johns Hopkins University, 3400N. Charles Street, Clark 106, Baltimore, Maryland 21218, USA
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Pellizzer C, Bello E, Adler S, Hartung T, Bremer S. Detection of tissue-specific effects by methotrexate on differentiating mouse embryonic stem cells. ACTA ACUST UNITED AC 2004; 71:331-41. [PMID: 15505807 DOI: 10.1002/bdrb.20019] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Pluripotent embryonic stem (ES) cells offer a unique possibility to monitor the differentiation of several cell types in vitro. This study attempts to identify marker genes during in vitro cell differentiation of murine ES cells and allow a prediction of chemical effects on cell differentiation of specific target tissues. The study focused on the expression pattern of key genes involved in cardiomyocyte and osteoblast differentiation: Oct-4, Brachyury, Nkx2.5, alpha myosin heavy chain, Cbfa1, and Osteocalcin. METHODS Methotrexate was selected due to its well-characterized teratogenic effects. Several in vivo studies have demonstrated the specific interactions of methotrexate with bone formation whereas the cardiovascular system is not specifically affected after exposure to low concentration. The capability of murine ES cells to differentiate in vitro into cardiomyocytes as well as into osteoblasts have been used to demonstrate the target cell specificity in vitro, at non-cytotoxic concentration. RESULTS Exposure of differentiating ES cells did not result in any gene profile modification of the selected cardiomyocyte specific genes, whereas the expression of osteoblast specific key genes, Cbfa1 and Osteocalcin, decreased. At the latter stages of skeletal differentiation we observed a 30% decrease in gene expression for Cbfa1 and a 60% decrease for Osteocalcin, with reference to the control. Early marker genes for undifferentiated cells and mesodermal cells were not modified after methotrexate treatment. CONCLUSIONS These results show the possibility to integrate specific in vitro tests for teratogenicity in a test strategy for developmental toxicity.
Collapse
Affiliation(s)
- Cristian Pellizzer
- European Centre for the Validation of Alternative Methods, Institute for Health and Consumer Protection, Joint Research Centre, Ispra, Italy
| | | | | | | | | |
Collapse
|
79
|
Krejci P, Bryja V, Pachernik J, Hampl A, Pogue R, Mekikian P, Wilcox WR. FGF2 inhibits proliferation and alters the cartilage-like phenotype of RCS cells. Exp Cell Res 2004; 297:152-64. [PMID: 15194433 DOI: 10.1016/j.yexcr.2004.03.011] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2004] [Revised: 03/04/2004] [Indexed: 11/22/2022]
Abstract
Several forms of human dwarfism are due to activating mutations in FGFR3 highlighting the role of FGF signaling in the growth attenuation of cartilage. Here, we studied the effects of FGF2 on RCS chondrocytes. Treatment with FGF2 induced growth arrest in the G1 phase of the cell cycle and partial de-differentiation of cells manifested by changes in cell morphology, loss of the cartilage-like extracellular matrix, and down-regulation of aggrecan expression. FGF2 activated phospholipase Cgamma, protein kinase B, and Erk and p38 MAP kinases. Chemical inhibition of FGFR3 and MEK1/2 antagonized FGF2-mediated growth arrest. Expression of a dominant-negative Ras mutant resulted in a partial reversal of growth inhibition while expression of constitutively activated Ras led to Erk-dependent growth arrest, further demonstrating the role of the Ras/Erk pathway in this phenotype. At the molecular level, FGF2-induced growth arrest was initiated by disintegration of cyclin D3-cdk6 complex followed by increased association of p21(WAF1) and p27(Kip1) with the cyclin-cdk2 and cyclin-cdk4 complexes leading to inhibition of their kinase activities and ultimately to underphosphorylation of the p107 and p130 pocket proteins. Both p21(WAF1) and p27(Kip1) accumulated upon FGF2 treatment, but this accumulation occurred at the protein level at least partially due to interaction with transcriptionally induced cyclin D1.
Collapse
Affiliation(s)
- Pavel Krejci
- Steven Spielberg Pediatric Research Center, Burns and Allen Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | | | | | | | | | | |
Collapse
|
80
|
de la Fuente R, Abad JL, García-Castro J, Fernández-Miguel G, Petriz J, Rubio D, Vicario-Abejón C, Guillén P, González MA, Bernad A. Dedifferentiated adult articular chondrocytes: a population of human multipotent primitive cells. Exp Cell Res 2004; 297:313-28. [PMID: 15212937 DOI: 10.1016/j.yexcr.2004.02.026] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2003] [Revised: 02/10/2004] [Indexed: 01/13/2023]
Abstract
OBJECTIVE To test the hypothesis that dedifferentiated adult human cartilage chondrocytes (HAC) are a true multipotent primitive population. METHODS Studies to characterize dedifferentiated HAC included cell cycle and quiescence analysis, cell fusion, flow-FISH telomere length assays, and ABC transporter analysis. Dedifferentiated HAC were characterized by flow cytometry, in parallel with bone marrow mesenchymal stem cells (MSC) and processed lipoaspirate (PLA) cells. The in vitro differentiation potential of dedifferentiated HAC was studied by cell culture under several inducing conditions, in multiclonal and clonal cell populations. RESULTS Long-term HAC cultures were chromosomically stable and maintained cell cycle dynamics while showing telomere shortening. The phenotype of dedifferentiated HAC was quite similar to that of human bone marrow MSC. In addition, this population expressed human embryonic stem cell markers. Multiclonal populations of dedifferentiated HAC differentiated to chondrogenic, osteogenic, adipogenic, myogenic, and neurogenic lineages. Following VEGF induction, dedifferentiated HAC expressed characteristics of endothelial cells, including AcLDL uptake. A total of 53 clonal populations of dedifferentiated HAC were efficiently expanded; 17 were able to differentiate to chondrogenic, osteogenic, and adipogenic lineages. No correlation was observed between telomere length or quiescent population and differentiation potential in the clones assayed. CONCLUSION Dedifferentiated HAC should be considered a human multipotent primitive population.
Collapse
Affiliation(s)
- Ricardo de la Fuente
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Kramer J, Hegert C, Rohwedel J. In vitro differentiation of mouse ES cells: bone and cartilage. Methods Enzymol 2004; 365:251-68. [PMID: 14696351 DOI: 10.1016/s0076-6879(03)65018-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Affiliation(s)
- Jan Kramer
- Department of Internal Medicine I, University of Lübeck, Lübeck, D-23538, Germany
| | | | | |
Collapse
|
82
|
Affiliation(s)
- John McNeish
- Genetic Technologies, Pfizer Global Research and Development, Groton 06340, Connecticut, USA.
| |
Collapse
|
83
|
Tanaka H, Murphy CL, Murphy C, Kimura M, Kawai S, Polak JM. Chondrogenic differentiation of murine embryonic stem cells: Effects of culture conditions and dexamethasone. J Cell Biochem 2004; 93:454-62. [PMID: 15372628 DOI: 10.1002/jcb.20171] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Pluripotent embryonic stem (ES) cells have the capability to differentiate to various cell types and may represent an alternative cell source for the treatment of cartilage defects. Here, we show that differentiation of ES cells toward the chondrogenic lineage can be enhanced by altering the culture conditions. Chondrogenesis was observed in intact embryoid body (EB) cultures, as detected by an increase in mRNA levels for aggrecan and Sox9 genes. Collagen IIB mRNA, the mature chondrocyte-specific splice variant, was absent at day 5, but appeared at later time points. Dexamethasone treatment of alginate-encapsulated EB cultures did not have a strong chondrogenic effect. Nor was chondrogenesis enhanced by alginate encapsulation compared to simple plating of EBs. However, disruption of day 5 EBs and culture as a micromass or pelleted mass, significantly enhanced the expression of the cartilage marker gene collagen type II and the transcription factor Sox9 compared to all other treatments. Histological and immunohistochemical analysis of pellet cultures revealed cartilage-like tissue characterized by metachromatically stained extracellular matrix and type II collagen immunoreactivity, indicative of chondrogenesis. These findings have potentially important implications for cartilage tissue engineering, since they may enable the increase in differentiated cell numbers needed for the in vitro development of functional cartilaginous tissue suitable for implantation.
Collapse
Affiliation(s)
- Hiroshi Tanaka
- Department of Orthopedic Surgery, Yamaguchi University School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-8505, Japan.
| | | | | | | | | | | |
Collapse
|
84
|
Affiliation(s)
- Shulamit Levenberg
- Massachusetts Institute of Technology, Chemical Engineering Department, Cambridge, Massachusetts 02139, USA
| | | |
Collapse
|
85
|
Barbero A, Ploegert S, Heberer M, Martin I. Plasticity of clonal populations of dedifferentiated adult human articular chondrocytes. ARTHRITIS AND RHEUMATISM 2003; 48:1315-25. [PMID: 12746904 DOI: 10.1002/art.10950] [Citation(s) in RCA: 297] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVE To investigate whether adult human articular chondrocytes (AHACs), dedifferentiated by monolayer expansion, can differentiate toward diverse mesenchymal lineages and, if so, whether this ability is regulated by growth factors during monolayer expansion. METHODS AHACs were expanded as multiclonal or clonal populations in medium without (control) or with factors enhancing cell dedifferentiation (transforming growth factor beta1, fibroblast growth factor 2, and platelet-derived growth factor type BB [TFP]). Cells were then cultured under conditions promoting chondrogenic, osteogenic, or adipogenic differentiation, and the acquired phenotypes were assessed histologically, biochemically, and by real-time reverse transcriptase-polymerase chain reaction. RESULTS Multiclonal populations of both control- and TFP-expanded AHACs differentiated toward the chondrogenic, osteogenic, and adipogenic lineages. Compared with control-expanded AHACs, TFP-expanded cells displayed enhanced chondrogenic differentiation capacity (2.4-fold higher glycosaminoglycan/DNA content and 2,500-fold higher up-regulation of type II collagen) and osteogenic differentiation capacity (9.4-fold higher increase in alkaline phosphatase activity and 12.4-fold higher up-regulation of bone sialoprotein), but reduced formation of adipocytes (5.2-fold lower oil red O-positive cells/area). Clonal populations of AHACs could be efficiently expanded in TFP, but not in control medium. Most TFP-expanded clones were able to redifferentiate only into chondrocytes (7 of 20) or were unable to differentiate (6 of 20). However, some clones (2 of 20) differentiated toward all of the lineages investigated, thus displaying characteristics of mesenchymal progenitor cells. CONCLUSION Dedifferentiated AHACs exhibit differentiation plasticity, which is modulated by growth factors used during monolayer expansion and is highly heterogeneous across different clones. Clonal culture of AHACs in the presence of regulatory molecules could lead to the identification of AHAC subpopulations with enhanced cartilage repair capacity.
Collapse
|