5501
|
Martin JMM, Smith M, Al-Rubeai M. Cryopreservation and in vitro expansion of chondroprogenitor cells isolated from the superficial zone of articular cartilage. Biotechnol Prog 2005; 21:168-77. [PMID: 15903255 DOI: 10.1021/bp049821o] [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: 11/28/2022]
Abstract
Understanding the proliferation mechanisms of chondroprogenitor cells and their influence on cell differentiation is crucial in order to develop large-scale expansion processes for tissue engineering applications. Proliferation control mechanisms were mainly attributed to substrate limitation and cell-cell contact inhibition. The limiting substrates were found to be components of the FCS, with an optimal proliferation rate achieved in the presence of 40% FCS. In addition, the medium supply rate was found to be essential in reducing substrate limitation. In terms of FCS, 10 microL FCS cm(-2) h(-1) was the threshold feed rate required to prevent substrate limitation. Above this rate, maximum cell densities of 5.3 x 10(5) cells/cm2 were achieved, representing a 53-fold expansion. To reduce the need for high supply rates, the effect of specific growth factors was also investigated. Cell densities of 3.3 x 10(5) cells/cm2 were achieved in batch cultures using 40% FCS and 1 ng/mL TGF-beta1. Chondroprogenitor cells were expanded in this medium up to three passages without compromising their ability to differentiate and produce cartilage-like matrix in pellet cultures. In addition to substrate limitation, cell-cell contact, even at very sparse subconfluent densities, appeared capable of exerting some degree of growth inhibition. The cells exhibited deceleratory growth kinetics, characterized by a decrease of specific growth rates over time.
Collapse
Affiliation(s)
- Juan M Melero Martin
- Department of Chemical Engineering, School of Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | | | | |
Collapse
|
5502
|
Fujimura J, Ogawa R, Mizuno H, Fukunaga Y, Suzuki H. Neural differentiation of adipose-derived stem cells isolated from GFP transgenic mice. Biochem Biophys Res Commun 2005; 333:116-21. [PMID: 15939405 DOI: 10.1016/j.bbrc.2005.05.096] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2005] [Accepted: 05/17/2005] [Indexed: 01/08/2023]
Abstract
Taking advantage of homogeneously marked cells from green fluorescent protein (GFP) transgenic mice, we have recently reported that adipose-derived stromal cells (ASCs) could differentiate into mesenchymal lineages in vitro. In this study, we performed neural induction using ASCs from GFP transgenic mice and were able to induce these ASCs into neuronal and glial cell lineages. Most of the neurally induced cells showed bipolar or multipolar appearance morphologically and expressed neuronal markers. Electron microscopy revealed their neuronal morphology. Some cells also showed glial phenotypes, as shown immunocytochemically. The present study clearly shows that ASCs derived from GFP transgenic mice differentiate into neural lineages in vitro, suggesting that these cells might provide an ideal source for further neural stem cell research with possible therapeutic application for neurological disorders.
Collapse
Affiliation(s)
- Juri Fujimura
- Department of Pharmacology, Nippon Medical School, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
5503
|
Kolokol'chikova EG, Pal'tsyn AA, Shchegolev AI, Adamyan AA, Chervonskaya NV. Proliferative Activity of Adipocytes in Adipose Tissue Tumors. Bull Exp Biol Med 2005; 140:122-6. [PMID: 16254637 DOI: 10.1007/s10517-005-0427-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Electron-autoradiographic study of normal and tumor-transformed adipose tissue (common lipoma and destructive lipoma, i.e. infiltrating and degrading lipoma) showed the capacity of adipose tissue cells in lipomas, especially in destructive lipomas, to proliferation and differentiation. In vivo synthesis of DNA in mature adipocytes not observed previously is described. The role of microvascular wall cells as mesenchymal multipotent precursors in the formation of the adipose tissue is discussed. The involvement of the bone marrow mesenchymal stem cell in this process cannot be ruled out.
Collapse
Affiliation(s)
- E G Kolokol'chikova
- A. V. Vishnevskii Institute of Surgery, Russian Academy of Medical Sciences, Moscow
| | | | | | | | | |
Collapse
|
5504
|
García-Olmo D, García-Arranz M, Herreros D, Pascual I, Peiro C, Rodríguez-Montes JA. A phase I clinical trial of the treatment of Crohn's fistula by adipose mesenchymal stem cell transplantation. Dis Colon Rectum 2005; 48:1416-23. [PMID: 15933795 DOI: 10.1007/s10350-005-0052-6] [Citation(s) in RCA: 563] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
PURPOSE The effective management of fistulas in patients with Crohn's disease presents an extremely challenging problem. Mesenchymal adult stem cells extracted from certain tissues, such as adipose tissue, can differentiate into various cell types. Therefore, we have tried to use such cells to stimulate healing of Crohn's fistulas. METHODS We designed a prospective Phase I clinical trial, involving five patients with Crohn's disease, to test the feasibility and safety of autologous stem cells transplantation in the treatment of fistulas. We also studied the expression of various cell markers and the growth rates of the lipoaspirate-derived cells that were used for transplantation. RESULTS One patient was excluded because of bacterial contamination of cultured cells. We inoculated nine fistulas in four patients with autologous adipose tissue-derived stem cells at Passage 3 or earlier. Eight inoculated fistulas were followed weekly for at least eight weeks. In six fistulas, the external opening was covered with epithelium at the end of Week 8, and, thus, these fistulas were considered healed (75 percent). In the other two fistulas, there was only incomplete closure of the external opening, with a decrease in output flow (not healed; 25 percent). No adverse effects were observed in any patient at the end of the follow-up period (minimum follow-up,12 months; maximum follow-up, 30 months; follow-up average, 22 months). CONCLUSIONS To our knowledge, this is the first report of a clinical trial of cell therapy using autologous stem cells obtained from a lipoaspirate. Our results indicate that our protocol is feasible and safe for the treatment of fistulas in Crohn's disease. The number of patients included and the uncontrolled nature of Phase I clinical trials do not allow demonstration of the effectiveness of the treatment. However, the results of the present study encourage to perform further studies in Phase II.
Collapse
Affiliation(s)
- Damián García-Olmo
- Department of General Surgery, La Paz University Hospital, Madrid, Spain.
| | | | | | | | | | | |
Collapse
|
5505
|
Rodriguez AM, Elabd C, Amri EZ, Ailhaud G, Dani C. The human adipose tissue is a source of multipotent stem cells. Biochimie 2005; 87:125-8. [PMID: 15733747 DOI: 10.1016/j.biochi.2004.11.007] [Citation(s) in RCA: 268] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2004] [Accepted: 11/02/2004] [Indexed: 12/13/2022]
Abstract
Multipotent stem cells constitute an unlimited source of differentiated cells that could be used in pharmacological studies and in medicine. Recently, several publications have reported that adipose tissue contains a population of cells able to differentiate into different cell types including adipocytes, osteoblasts, myoblasts, and chondroblasts. More recently, stem cells with a multi-lineage potential at the single cell level have been isolated from human adipose tissue. These cells, called human Multipotent Adipose-Derived Stem (hMADS) cells, have been established in culture and interestingly, maintain their characteristics with long-term passaging. The adipocyte differentiation of hMADS cells has been thoroughly studied and differentiated cells exhibit the unique feature of human adipocytes. Finally, potential applications of stem cells isolated from adipose tissue in medicine will be discussed.
Collapse
Affiliation(s)
- A-M Rodriguez
- Faculté des Sciences, Centre de Biochimie, Institut de Recherche Signalisation, Biologie du Developpement et Cancer, UMR 6543 CNRS, Parc Valrose, 06108 Nice cedex 2, France
| | | | | | | | | |
Collapse
|
5506
|
Peterson B, Zhang J, Iglesias R, Kabo M, Hedrick M, Benhaim P, Lieberman JR. Healing of critically sized femoral defects, using genetically modified mesenchymal stem cells from human adipose tissue. ACTA ACUST UNITED AC 2005; 11:120-9. [PMID: 15738667 DOI: 10.1089/ten.2005.11.120] [Citation(s) in RCA: 297] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The FDA has approved the clinical use of recombinant bone morphogenetic proteins (BMPs). However, the use of recombinant BMPs in humans has required large doses of the proteins to be effective, which suggests that the delivery method of bone morphogenetic proteins needs to be optimized. Gene therapy is an alternative method to deliver such recombinant proteins, and gene transfer techniques have been tested on a variety of cell types including bone marrow cells, skin fibroblasts, peripheral blood monocytes, and muscle-derived cells. In this study, we sought to determine the ability of BMP-2-producing human adipose-derived mesenchymal stem cells to heal a critically sized femoral defect in a nude rat model. After approval by the human subjects protection committee, human adipose tissue was obtained from healthy donors. The lipoaspirate was processed as previously described (De Ugarte, D.A., et al. Cells Tissues Organs 174, 101, 2003). Cells were grown in culture and infected with a BMP-2-carrying adenovirus. Five million cells were applied to a collagen- ceramic carrier and implanted into femoral defects as previously described (Zuk, P.A., et al. Mol. Biol. 13, 4279, 2002). All animals were killed at 8 weeks. Femora were dissected out and underwent radiographic, histologic, and biomechanical analysis. Eleven of the 12 femora in the group treated with human processed lipoaspirate (HPLA) cells genetically modified to overexpress BMP-2 had healed at 8 weeks. This was assessed by radiographs, by mechanical testing, and by histology. The one femur that did not heal had a subacute infection. All eight of the femora treated with the rhBMP-2-impregnated collagen-ceramic carrier healed. No statistically significant difference was detected between these two groups. Evaluation of the control groups: group II (collagen- ceramic carrier with HPLA cells) and group III (collagen-ceramic carrier alone) showed that none of the femora had healed by 8 weeks. Our results indicate that HPLA cells genetically modified by adenoviral gene transfer to overexpress BMP-2 can induce bone formation in vivo and heal a critically sized femoral defect in an athymic rat. The HPLA cells alone did not induce significant bone formation. However, when combined with an osteoinductive factor these cells may be an effective method for enhancing bone healing and the tissue engineering of bone.
Collapse
Affiliation(s)
- Brett Peterson
- Department of Orthopedic Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California 90095-3075, USA
| | | | | | | | | | | | | |
Collapse
|
5507
|
Tondreau T, Meuleman N, Delforge A, Dejeneffe M, Leroy R, Massy M, Mortier C, Bron D, Lagneaux L. Mesenchymal stem cells derived from CD133-positive cells in mobilized peripheral blood and cord blood: proliferation, Oct4 expression, and plasticity. Stem Cells 2005; 23:1105-12. [PMID: 15955825 DOI: 10.1634/stemcells.2004-0330] [Citation(s) in RCA: 322] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In this study, we used a common procedure to assess the potential of mobilized peripheral blood (MPB) and umbilical cord blood (UCB) as sources of mesenchymal stem cells (MSCs) in comparison with bone marrow (BM). We tested three methods: plastic adhesion supplemented with 5% of BM-MSC conditioned medium, unsupplemented plastic adhesion, and selection of CD133-positive cells. MSCs derived from MPB or UCB are identified by their positive expression of mesenchymal (SH2, SH3) and negative expression of hematopoietic markers (CD14, CD34, CD45, HLA-DR). We observed that the CD133-positive cell fraction contains more MSCs with high proliferative potential. Placed in appropriate conditions, these cells proved their capacity to differentiate into adipocytes, osteocytes, chondrocytes, and neuronal/glial cells. MPB- and UCB-MSCs express Oct4, a transcriptional binding factor present in undifferentiated cells with high proliferative capacity. The selection of CD133-positive cells enabled us to obtain a homogeneous population of MSCs from UCB and MPB. These sources may have a major clinical importance thanks to their easy accessibility.
Collapse
Affiliation(s)
- Tatiana Tondreau
- Jules Bordet Institute, 121 Bd de Waterloo, 1000 Brussels, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
5508
|
Abstract
Homeostasis of continuously renewing adult tissues, such as the epidermis of the skin, is maintained by epidermal stem cells (EpiSC), which are a small population of undifferentiated, self-renewing basal keratinocyte cells that produce daughter transit amplifying (TA) cells to make up the majority of the proliferative basal cell population in the epidermis. We have isolated EpiSC from neonatal and adult skin, and shown that these cells can regenerate an epidermis that lasts long term in vitro and in vivo, and that permanently expresses a recombinant gene in the regenerated tissue (Bickenbach and Dunnwald, 2000; Dunnwald et al., 2001). When we injected murine EpiSC into the developing blastocyst environment of the mouse, we found that both neonatal and adult EpiSC retained some ability to participate in the formation of tissues from all three germ layers (Liang and Bickenbach, 2002; Bickenbach and Chinnathambi, 2004; Liang et al., 2004). Although it appears evident that EpiSC act as pluripotent stem cells, how this reprogramming takes place is not understood. EpiSC might directly transdifferentiate into other cell types or they might first dedifferentiate into a more primitive cell type, and then proceed to develop along a cell lineage pathway. To begin to unravel this, we co-cultured EpiSC with embryonic stem (ES) cells, and found that EpiSC could alter their cell lineage protein expression to that of a more primitive cell type. We also placed EpiSC in a wounded environment and found that EpiSC interacted with the mesenchymal cells repopulating the wound bed. Our findings indicate that the population of cells that we isolate as EpiSC has a pluripotent capability. This has led us to postulate a paradigm shift for somatic stem cells. We propose that tissues maintain a sequestered population of uncommitted stem cells that retain a regenerative response which is enhanced when the cells are exposed to developmental or stress influences.
Collapse
Affiliation(s)
- Jackie R Bickenbach
- Department of Anatomy and Cell Biology, College of Medicine, The University of Iowa, Room 1-457 BSB, 51 Newton Road, Iowa City, IA 52242-1109, USA.
| | | |
Collapse
|
5509
|
Shih DTB, Lee DC, Chen SC, Tsai RY, Huang CT, Tsai CC, Shen EY, Chiu WT. Isolation and characterization of neurogenic mesenchymal stem cells in human scalp tissue. Stem Cells 2005; 23:1012-20. [PMID: 15941858 DOI: 10.1634/stemcells.2004-0125] [Citation(s) in RCA: 184] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Recent studies have shown that adult tissues contain stem/ progenitor cells capable of not only generating mature cells of their tissue of origin but also transdifferentiating themselves into other tissue cells. Murine skin-derived precursor cells, for example, have been described as unique, nonmesenchymal-like stem cells capable of mesodermal and ectodermal neurogenic differentiation. Human-derived skin precursors are less well characterized. In this study, the isolation and characterization of adherent, mesenchymal stem cell-like cells from human scalp tissue (hSCPs) are described. hSCPs initially isolated by both medium-selection (ms-hSCPs) and single-cell (c-hSCPs) methods were cultured in medium containing epidermal growth factor and fibroblast growth factor-beta. Cultured ms-hSCPs and c-hSCPs demonstrated a consistent growth rate, continuously replicated in cell culture, and displayed a stable phenotype indistinguishable from each other. Both hSCPs expressed surface antigen profile (CDw90, SH2, SH4, CD105, CD166, CD44, CD49d-e, and HLA class I) similar to that of bone marrow mesenchymal stem cells (BM-MSCs). The growth kinetics, surface epitopes, and differentiation potential of c-hSCP cells were characterized and compared with BM-MSCs. In addition to differentiation along the osteogenic, chondrogenic, and adipogenic lineages, hSCPs can effectively differentiate into neuronal precursors evident by neurogenic gene expression of glial fibrillary acid protein, NCAM, neuron filament-M, and microtubule-associated protein 2 transcripts. Therefore, hSCPs may potentially be a better alternative of BM-MSCs for neural repairing, in addition to their other mesenchymal regenerative capacity. Our study suggests that hSCPs may provide an alternative adult stem cell resource that may be useful for regenerative tissue repair and autotransplantations.
Collapse
Affiliation(s)
- Daniel Tzu-bi Shih
- Center for Stem Cell Research, Wan-Fang Hospital and Graduate Institute of Cell and Molecular Biology, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan 110.
| | | | | | | | | | | | | | | |
Collapse
|
5510
|
Barberi T, Willis LM, Socci ND, Studer L. Derivation of multipotent mesenchymal precursors from human embryonic stem cells. PLoS Med 2005; 2:e161. [PMID: 15971941 PMCID: PMC1160574 DOI: 10.1371/journal.pmed.0020161] [Citation(s) in RCA: 333] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2004] [Accepted: 04/15/2005] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Human embryonic stem cells provide access to the earliest stages of human development and may serve as a source of specialized cells for regenerative medicine. Thus, it becomes crucial to develop protocols for the directed differentiation of embryonic stem cells into tissue-restricted precursors. METHODS AND FINDINGS Here, we present culture conditions for the derivation of unlimited numbers of pure mesenchymal precursors from human embryonic stem cells and demonstrate multilineage differentiation into fat, cartilage, bone, and skeletal muscle cells. CONCLUSION Our findings will help to elucidate the mechanism of mesoderm specification during embryonic stem cell differentiation and provide a platform to efficiently generate specialized human mesenchymal cell types for future clinical applications.
Collapse
Affiliation(s)
- Tiziano Barberi
- 1Laboratory of Stem Cell and Tumor Biology, Division of Neurosurgery and Developmental Biology ProgramSloan-Kettering Institute, New York, New YorkUnited States of America
| | - Lucy M Willis
- 1Laboratory of Stem Cell and Tumor Biology, Division of Neurosurgery and Developmental Biology ProgramSloan-Kettering Institute, New York, New YorkUnited States of America
| | - Nicholas D Socci
- 2Computational Biology Center, Sloan-Kettering InstituteNew York, New YorkUnited States of America
| | - Lorenz Studer
- 1Laboratory of Stem Cell and Tumor Biology, Division of Neurosurgery and Developmental Biology ProgramSloan-Kettering Institute, New York, New YorkUnited States of America
- *To whom correspondence should be addressed. E-mail:
| |
Collapse
|
5511
|
DeLany JP, Floyd ZE, Zvonic S, Smith A, Gravois A, Reiners E, Wu X, Kilroy G, Lefevre M, Gimble JM. Proteomic Analysis of Primary Cultures of Human Adipose-derived Stem Cells. Mol Cell Proteomics 2005; 4:731-40. [PMID: 15753122 DOI: 10.1074/mcp.m400198-mcp200] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Adipogenesis plays a critical role in energy metabolism and is a contributing factor to the obesity epidemic. This study examined the proteome of primary cultures of human adipose-derived adult stem (ADAS) cells as an in vitro model of adipogenesis. Protein lysates obtained from four individual donors were compared before and after adipocyte differentiation by two-dimensional gel electrophoresis and tandem mass spectroscopy. Over 170 individual protein features in the undifferentiated adipose-derived adult stem cells were identified. Following adipogenesis, over 40 proteins were up-regulated by > or = 2-fold, whereas 13 showed a > or = 3-fold reduction. The majority of the modulated proteins belonged to the following functional categories: cytoskeleton, metabolic, redox, protein degradation, and heat shock protein/chaperones. Additional immunoblot analysis documented the induction of four individual heat shock proteins and confirmed the presence of the heat shock protein 27 phosphoserine 82 isoform, as predicted by the proteomic analysis, as well as the crystallin alpha phosphorylated isoforms. These findings suggest that the heat shock protein family proteome warrants further investigation with respect to the etiology of obesity and type 2 diabetes.
Collapse
Affiliation(s)
- James P DeLany
- Stable Isotope Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
5512
|
Tondreau T, Lagneaux L, Dejeneffe M, Massy M, Mortier C, Delforge A, Bron D. Bone marrow-derived mesenchymal stem cells already express specific neural proteins before any differentiation. Differentiation 2005; 72:319-26. [PMID: 15554943 DOI: 10.1111/j.1432-0436.2004.07207003.x] [Citation(s) in RCA: 242] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Bone marrow mesenchymal stem cells (MSC) are multipotent cells. To explain their plasticity, we postulated that undifferentiated MSC may express proteins from other tissues such as neuronal tissues. MSC are obtained by two different approaches: plastic adhesion or negative depletion (RosetteSep and magnetic beads CD45/glycophorin A). MSC are evaluated through FACS analysis using a panel of antibodies (SH2, SH3, CD14, CD33, CD34, CD45, etc.). To confirm the multipotentiality in vitro, we have differentiated MSC into adipocytes, chondrocytes, osteocytes, and neuronal/glial cells using specific induction media. We have evaluated neuronal and glial proteins (Nestin, Tuj-I, betaIII Tubulin, tyrosine hydroxylase [TH], MAP-2, and GFAP) by using flow cytometry, Western blots, and RT-PCR. We found that MSC constituently express native immature neuronal proteins such as Nestin and Tuj-1. After only five passages, MSC can already express more mature neuronal or glial proteins, such as TH, MAP-2, and GFAP, without any specific induction. We noticed an increase in the expression of more mature neuronal/glial proteins (TH, MAP-2, and GFAP) after exposure to neural induction medium, thus confirming the differentiation of MSC into neurons and astrocytes. The constitutive expression of Nestin or Tuj-1 by MSC suggests that these cells are "multidifferentiated" cells and thus can retain the ability for neuronal differentiation, enhancing their potentiality to be employed in the treatment of neurological diseases.
Collapse
Affiliation(s)
- Tatiana Tondreau
- Department of Experimental Hematology, Jules Bordet Institute-ULB, 1000 Brussels, Belgium.
| | | | | | | | | | | | | |
Collapse
|
5513
|
Kang SK, Putnam L, Dufour J, Ylostalo J, Jung JS, Bunnell BA. Expression of telomerase extends the lifespan and enhances osteogenic differentiation of adipose tissue-derived stromal cells. Stem Cells 2005; 22:1356-72. [PMID: 15579653 DOI: 10.1634/stemcells.2004-0023] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Expression of TERT, the catalytic protein subunit of the telomerase complex, can be used to generate cell lines that expand indefinitely and retain multilineage potential. We have created immortal adipose stromal cell lines (ATSCs) by stably transducing nonhuman primate-derived ATSCs with a retroviral vector expressing TERT. Transduced cells (ATSC-TERT) had an increased level of telomerase activity and increased mean telomere length in the absence of malignant cellular transformation. Long-term culture of the ATSC-TERT cells demonstrated that the cells retain the ability to undergo differentiation along multiple lineages such as adipogenic, chondrogenic, and neurogenic. Untransduced cells demonstrated markedly reduced multilineage and self-renewal potentials after 12 passages in vitro. To determine the functional role of telomerase during osteogenesis, we examined osteogenic differentiation potential of ATSC-TERT cells in vitro. Compared with naive ATSCs, which typically begin to accumulate calcium after 3-4 weeks of induction by osteogenic differentiation medium, ATSC-TERT cells were found to accumulate significant amounts of calcium after only 1 week of culture in osteogenic induction medium. The cells have increased production of osteoblastic markers, such as AP2, osteoblast-specific factor 2, chondroitin sulfate proteoglycan 4, and the tumor necrosis factor receptor superfamily, compared with control ATSCs, indicating that telomerase expression may aid in maintaining the osteogenic stem cell pool during in vitro expansion. These results show that ectopic expression of the telomerase gene in nonhuman primate ATSCs prevents senescence-associated impairment of osteoblast functions and that telomerase therapy may be a useful strategy for bone regeneration and repair.
Collapse
Affiliation(s)
- Soo Kyung Kang
- Division of Gene Therapy, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA 70433, USA
| | | | | | | | | | | |
Collapse
|
5514
|
Abstract
Increasing evidence suggests that circulating progenitor cells contribute to postnatal neovascularization. These cells home to sites of ischemia, adopt an endothelial phenotype, and contribute to new blood vessel formation. Hence, the identity of the circulating cells that contribute to neovascularization is not entirely clear. Bone-marrow-derived hematopoietic progenitor cells can give rise to endothelial cells and contribute to endothelial recovery and new capillary formation after ischemia. However, nonhematopoietic stem cells within the bone marrow and adipose-tissue-derived cells, as well as cardiac and neural progenitor cells, also differentiate to endothelial cells. Progenitor cells from the different sources may be useful to augment therapeutic vascularization. The present review article summarizes the different subtypes of (endothelial) progenitor cells that can give rise to endothelial cells, enhance neovascularization, and may be suitable for therapeutic neovascularization.
Collapse
Affiliation(s)
- Carmen Urbich
- Division of Molecular Cardiology, Department of Internal Medicine IV, University of Frankfurt, Frankfurt, Germany
| | | |
Collapse
|
5515
|
Richardson GD, Arnott EC, Whitehouse CJ, Lawrence CM, Hole N, Jahoda CAB. Cultured Cells from the Adult Human Hair Follicle Dermis can be Directed Toward Adipogenic and Osteogenic Differentiation. J Invest Dermatol 2005; 124:1090-1. [PMID: 15854060 DOI: 10.1111/j.0022-202x.2005.23734.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
5516
|
Shah K, Hsich G, Breakefield XO. Neural precursor cells and their role in neuro-oncology. Dev Neurosci 2005; 26:118-30. [PMID: 15711055 DOI: 10.1159/000082132] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2003] [Accepted: 01/07/2004] [Indexed: 01/08/2023] Open
Abstract
Neural precursor cells (NPCs) provide a new mode for delivery of genes and proteins to brain tumors. These cells exist both in the developing and the adult nervous systems of all mammalian organisms. They have the ability to self-renew, migrate to diseased areas of the brain and differentiate into neurons, astrocytes and oligodendrocytes. The migratory ability of NPCs and their capacity to differentiate into all neural phenotypes provides a powerful tool for the treatment of both diffuse and localized neurological disorders. NPCs have been used in transplantation to replace damaged cells and in cancer therapy to provide therapeutic proteins and vectors to eliminate malignant cells in the brain. This review focuses on the characteristics of NPCs and their experimental use in the therapy for brain tumors. Examples are provided of monitoring migration of NPCs by bioluminescence imaging in living animals and of using them to deliver the apoptotic protein, TRAIL, to kill tumor cells.
Collapse
Affiliation(s)
- Khalid Shah
- Department of Neurology, Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
5517
|
Rim JS, Mynatt RL, Gawronska-Kozak B. Mesenchymal stem cells from the outer ear: a novel adult stem cell model system for the study of adipogenesis. FASEB J 2005; 19:1205-7. [PMID: 15857881 DOI: 10.1096/fj.04-3204fje] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Adipocytes arise from multipotent stem cells of mesodermal origin, which also give rise to the muscle, bone, and cartilage lineages. However, signals and early molecular events that commit multipotent stem cells into the adipocyte lineage are not well established mainly due to lack of an adequate model system. We have identified a novel source of adult stem cells from the external murine ears referred to here as an ear mesenchymal stem cells (EMSC). EMSC have been isolated from several standard and mutant strains of mice. They are self-renewing, clonogenic, and multipotent, since they give rise to osteocytes, chondrocytes, and adipocytes. The in vitro characterization of EMSC indicates very facile adipogenic differentiation. Morphological, histochemical, and molecular analysis after the induction of differentiation showed that EMSC maintain adipogenic potentials up to fifth passage. A comparison of EMSC to the stromal-vascular (S-V) fraction of fat depots, under identical culture conditions (isobutyl-methylxanthine, dexamethasone, and insulin), revealed much more robust and consistent adipogenesis in EMSC than in the S-V fraction. In summary, we show that EMSC can provide a novel, easily obtainable, primary culture model for the study of adipogenesis.
Collapse
Affiliation(s)
- Jong-Seop Rim
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | | | | |
Collapse
|
5518
|
Abstract
The presence of ectopic tissue in the diseased artery wall is evidence for the presence of multipotential stem cells in the vasculature. Mesenchymal stem cells were first identified in the marrow stroma, and they differentiate along multiple lineages giving rise to cartilage, bone, fat, muscle, and vascular tissue in vitro and in vivo. Transplantation studies show that marrow-derived mesenchymal stem cells appear to enter the circulation and engraft other tissues, including the artery wall, at sites of injury. Recent evidence indicates that mesenchymal stem cells are also present in normal artery wall and microvessels and that they also may enter the circulation, contributing to the population of circulating progenitor cells and engrafting other tissues. Thus, the artery wall is not only a destination but also a source of progenitor cells that have regenerative potential. Although potential artifacts, such as fusion, need to be taken into consideration, these new developments in vascular biology open important therapeutic avenues. A greater understanding of how mesenchymal stem cells from the bone marrow or artery wall bring about vascular regeneration and repair may lead to novel cell-based treatments for cardiovascular disease.
Collapse
Affiliation(s)
- Moeen Abedin
- Department of Medicine, The David Geffen School of Medicine at University of California at Los Angeles, CA 90095-1679, USA
| | | | | |
Collapse
|
5519
|
Hattori H, Sato M, Masuoka K, Ishihara M, Kikuchi T, Matsui T, Takase B, Ishizuka T, Kikuchi M, Fujikawa K, Ishihara M. Osteogenic potential of human adipose tissue-derived stromal cells as an alternative stem cell source. Cells Tissues Organs 2005; 178:2-12. [PMID: 15550755 DOI: 10.1159/000081088] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2004] [Indexed: 12/11/2022] Open
Abstract
Adult bone marrow contains mesenchymal stem cells (bone marrow-derived mesenchymal stem cells; BMSCs) which contribute to the generation of mesenchymal tissue such as bone, cartilage, muscle and adipose. However, using bone marrow as a source of stem cells has the limitation of a low cell number. An alternate source of adult stem cells that could be obtained in large quantities, under local anesthesia, with minimal discomfort would be advantageous. Human adipose tissue obtained by liposuction was processed to obtain a fibroblast-like population of cells or adipose tissue-derived stromal cells (ATSCs). In this study, we compared the osteogenic differentiation of ATSCs with that of BMSCs. Both cell types were cultured in atelocollagen honeycomb-shaped scaffolds with a membrane seal (ACHMS scaffold) for three-dimensional culturing in a specific osteogenic induction medium. Optimal osteogenic differentiation in both cell types, as determined by alkaline phosphatase cytochemistry, secretion of osteocalcin, mineral (calcium phosphate) deposition and scanning electron microscopy, was obtained with the same three-dimensional culture. Furthermore, osteoblastic lining in vivo was examined using ATSC-seeded or BMSC-seeded scaffolds in nude mice. The present results show that ATSCs have a similar ability to differentiate into osteoblasts to that of BMSCs.
Collapse
Affiliation(s)
- Hidemi Hattori
- Division of Biomedical Engineering, Research Institute, National Defense Medical College, Saitama, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5520
|
Chen X, Mao Z, Liu S, Liu H, Wang X, Wu H, Wu Y, Zhao T, Fan W, Li Y, Yew DT, Kindler PM, Li L, He Q, Qian L, Wang X, Fan M. Dedifferentiation of adult human myoblasts induced by ciliary neurotrophic factor in vitro. Mol Biol Cell 2005; 16:3140-51. [PMID: 15843428 PMCID: PMC1165399 DOI: 10.1091/mbc.e05-03-0218] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Ciliary neurotrophic factor (CNTF) is primarily known for its important cellular effects within the nervous system. However, recent studies indicate that its receptor can be highly expressed in denervated skeletal muscle. Here, we investigated the direct effect of CNTF on skeletal myoblasts of adult human. Surprisingly, we found that CNTF induced the myogenic lineage-committed myoblasts at a clonal level to dedifferentiate into multipotent progenitor cells--they not only could proliferate for over 20 passages with the expression absence of myogenic specific factors Myf5 and MyoD, but they were also capable of differentiating into new phenotypes, mainly neurons, glial cells, smooth muscle cells, and adipocytes. These "progenitor cells" retained their myogenic memory and were capable of redifferentiating into myotubes. Furthermore, CNTF could activate the p44/p42 MAPK and down-regulate the expression of myogenic regulatory factors (MRFs). Finally, PD98059, a specific inhibitor of p44/p42 MAPK pathway, was able to abolish the effects of CNTF on both myoblast fate and MRF expression. Our results demonstrate the myogenic lineage-committed human myoblasts can dedifferentiate at a clonal level and CNTF is a novel regulator of skeletal myoblast dedifferentiation via p44/p42 MAPK pathway.
Collapse
Affiliation(s)
- Xiaoping Chen
- Department of Neurophysiology, Institute of Basic Medical Sciences, Beijing 100850, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5521
|
Rubio D, Garcia-Castro J, Martín MC, de la Fuente R, Cigudosa JC, Lloyd AC, Bernad A. Spontaneous human adult stem cell transformation. Cancer Res 2005; 65:3035-9. [PMID: 15833829 DOI: 10.1158/0008-5472.can-04-4194] [Citation(s) in RCA: 734] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Human adult stem cells are being evaluated widely for various therapeutic approaches. Several recent clinical trials have reported their safety, showing them to be highly resistant to transformation. The clear similarities between stem cell and cancer stem cell genetic programs are nonetheless the basis of a recent proposal that some cancer stem cells could derive from human adult stem cells. Here we show that although they can be managed safely during the standard ex vivo expansion period (6-8 weeks), human mesenchymal stem cells can undergo spontaneous transformation following long-term in vitro culture (4-5 months). This is the first report of spontaneous transformation of human adult stem cells, supporting the hypothesis of cancer stem cell origin. Our findings indicate the importance of biosafety studies of mesenchymal stem cell biology to efficiently exploit their full clinical therapeutic potential.
Collapse
Affiliation(s)
- Daniel Rubio
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/Consejo Superior de Investigaciones Cientificas, UAM Campus de Cantoblanco, Darwin, 3 E-28049 Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
5522
|
Kim SJ, Cho HH, Kim YJ, Seo SY, Kim HN, Lee JB, Kim JH, Chung JS, Jung JS. Human adipose stromal cells expanded in human serum promote engraftment of human peripheral blood hematopoietic stem cells in NOD/SCID mice. Biochem Biophys Res Commun 2005; 329:25-31. [PMID: 15721268 DOI: 10.1016/j.bbrc.2005.01.092] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2005] [Indexed: 01/18/2023]
Abstract
Human mesenchymal stem cells (hMSC), that have been reported to be present in bone marrow, adipose tissues, dermis, muscles, and peripheral blood, have the potential to differentiate along different lineages including those forming bone, cartilage, fat, muscle, and neuron. Therefore, hMSC are attractive candidates for cell and gene therapy. The optimal conditions for hMSC expansion require medium supplemented with fetal bovine serum (FBS). Some forms of cell therapy will involve multiple doses, raising a concern over immunological reactions caused by medium-derived FBS proteins. In this study, we cultured human adipose stromal cells (hADSC) and bone marrow stroma cells (HBMSC) in human serum (HS) during their isolation and expansion, and demonstrated that they maintain their proliferative capacity and ability for multilineage differentiation and promote engraftment of peripheral blood-derived CD34(+) cells mobilized from bone marrow in NOD/SCID mice. Our results indicate that hADSC and hBMSC cultured in HS can be used for clinical trials of cell and gene therapies, including promotion of engraftment after allogeneic HSC transplantation.
Collapse
Affiliation(s)
- Su Jin Kim
- Department of Physiology, College of Medicine, Pusan National University, Pusan 602-739, Republic of Korea.
| | | | | | | | | | | | | | | | | |
Collapse
|
5523
|
von Heimburg D, Hemmrich K, Haydarlioglu S, Staiger H, Pallua N. Comparison of viable cell yield from excised versus aspirated adipose tissue. Cells Tissues Organs 2005; 178:87-92. [PMID: 15604532 DOI: 10.1159/000081719] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2004] [Indexed: 02/01/2023] Open
Abstract
The correction of soft-tissue defects by adipose tissue transplantation often produces poor and unpredictable results. The implantation of isolated and cultured preadipocytes offers a solution to this problem since these cells differentiate into adipocytes when implanted in vivo. A field of major interest is to maximize the yield of preadipocytes isolated from adipose tissue showing only low contamination with other cell types. Aspiration and excision are two concurrent clinical ways of harvesting adipose tissue for the isolation of preadipocytes. This tissue is usually discarded after surgery. In this study, the yield of preadipocytes obtained from liposuction material was compared to that of excised adipose tissue. Furthermore, we determined the loss of precursor cells if isolation of preadipocytes was delayed for 24 h. Preadipocytes were isolated from the stromal cell fraction of human subcutaneous adipose tissue samples. Harvesting of adipose tissue by suction was performed according to the Coleman procedure (manually applied negative pressure using a 10-ml syringe with a blunt tip cannula). Isolation was either carried out within 60 min after extraction or after storage for 24 h in culture medium at 4 degrees C. Isolated preadipocytes were cultured for 24 h, trypsinized and counted in a Neubauer chamber. Our results show clearly that the yield of preadipocytes isolated from liposuction material (within 60 min after extraction and after 24 h of storage) is higher than the cell yield from excised adipose tissue. Overnight storage for 24 h leads to a significant loss of preadipocytes in excised tissue but not in liposuction material. The high yield of cells isolated from liposuction material proves that extraction by suction does not damage the stromal cell fraction in the adipose tissue. If cell isolation is not performed immediately after the operation, liposuction material is clearly the better alternative for storage.
Collapse
Affiliation(s)
- Dennis von Heimburg
- Department of Plastic Surgery and Hand Surgery, Burn Center, University Hospital, Aachen University of Technology, Aachen, Germany.
| | | | | | | | | |
Collapse
|
5524
|
Wulf GG, Viereck V, Hemmerlein B, Haase D, Vehmeyer K, Pukrop T, Glass B, Emons G, Trümper L. Mesengenic progenitor cells derived from human placenta. ACTA ACUST UNITED AC 2005; 10:1136-47. [PMID: 15363170 DOI: 10.1089/ten.2004.10.1136] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Progenitor cells with differentiation capacity along multiple mesengenic lineages are attractive tools for numerous purposes in regenerative medicine. Such mesengenic progenitor cells have been isolated from adult mammalian bone marrow, and we here report placental tissue as an alternative source for these cells. By means of dissection/proteinase digestion techniques, high numbers of viable mononuclear cells were harvested from human placenta at term, and a mesenchymal cell population with characteristic expression of CD9, CD29, and CD73 was obtained in culture. The in vitro growth behavior of such placenta-derived mesengenic cells was similar to that of human bone marrow mesengenic progenitor cells. After in vitro propagation for more than three passages the cells were exclusively of maternal origin. Differentiation experiments showed differentiation potential along osteogenic, chondrogenic, adipogenic, and myogenic lineages. In conclusion, we propose human term placenta as an easily accessible, ample source of multipotent mesengenic progenitor cells.
Collapse
Affiliation(s)
- Gerald G Wulf
- Department of Hematology and Oncology, Georg August University Goettingen, 37075 Goettingen, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
5525
|
Gawronska-Kozak B. Regeneration in the ears of immunodeficient mice: identification and lineage analysis of mesenchymal stem cells. ACTA ACUST UNITED AC 2005; 10:1251-65. [PMID: 15363180 DOI: 10.1089/ten.2004.10.1251] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Wound healing in the ears of Athymic Nude-nu mice resembles regeneration. Histological analysis of the ear-punched tissues revealed the initial formation of a blastema-like structure followed by dermal, vascular, cartilage, and muscle regrowth exclusively in Athymic Nude-nu mice but not in wild-type controls (C57BL/6J). A subset of stem cells referred to here as ear mesenchymal stem cells (EMSCs) has been isolated from the external ears of regenerative (Athymic Nude-nu) and nonregenerative strains of mice. Morphological, histochemical, and molecular analysis after the induction of EMSC differentiation revealed multiple mesenchymal cell lineages (adipocytes, chondrocytes, and osteocytes) in all murine strains independent of their ability for regeneration. Thus, the absence of regeneration in wounded ears of C57BL/6J wild-type mice is not related to the absence of mesenchymal stem cell differentiation in tissue culture. Because nude mice lack T lymphocytes, it appears that in this model the absence of T lymphocytes in the wounded ears provides a microenvironment conducive to regeneration of mesenchymal tissues. These findings provide a new model to study the influence of the immune system on tissue regeneration.
Collapse
|
5526
|
Hauner H. The new concept of adipose tissue function. Physiol Behav 2005; 83:653-8. [PMID: 15621071 DOI: 10.1016/j.physbeh.2004.09.016] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2004] [Accepted: 09/14/2004] [Indexed: 11/19/2022]
Affiliation(s)
- Hans Hauner
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin, Klinikum Rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 München, Germany.
| |
Collapse
|
5527
|
Gang EJ, Jeong JA, Hong SH, Hwang SH, Kim SW, Yang IH, Ahn C, Han H, Kim H. Skeletal myogenic differentiation of mesenchymal stem cells isolated from human umbilical cord blood. ACTA ACUST UNITED AC 2005; 22:617-24. [PMID: 15277707 DOI: 10.1634/stemcells.22-4-617] [Citation(s) in RCA: 218] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Human umbilical cord blood (UCB) has been regarded as an alternative source for cell transplantation and cell therapy because of its hematopoietic and nonhematopoietic (mesenchymal) potential. Although there has been debate about whether mesenchymal stem cells (MSCs) are invariably present in UCB, several reports showed that MSC-like cells could be consistently derived from human UCB and, moreover, could differentiate into various cells of a mesodermal origin. However, it remains unclear whether these UCB-derived MSCs are also capable of differentiating into skeletal muscle cells. In this study, we isolated MSCs from human UCB and induced them to differentiate into skeletal muscle cells. During cell culture expansion, UCB-derived mononuclear cells gave rise to adherent layers of fibroblast-like cells expressing MSC-related antigens such as SH2, SH3, alpha-smooth muscle actin, CD13, CD29, and CD49e. More important, when these UCB-derived MSCs were incubated in promyogenic conditions for up to 6 weeks, they expressed myogenic markers in accordance with myogenic differentiation pattern. Both flow cytometric and reverse transcriptase-polymerase reaction analyses showed that two early myogenic markers, MyoD and myogenin, were expressed after 3 days of incubation but not after 2 weeks. At week 6, more than half of UCB-derived MSCs expressed myosin heavy chain, a late myogenic marker. Our results demonstrate that UCB-derived MSCs possess a potential of skeletal myogenic differentiation and also imply that these cells could be a suitable source for skeletal muscle repair and a useful tool of muscle-related tissue engineering.
Collapse
Affiliation(s)
- Eun Ji Gang
- Research Institute of Biotechnology, Histostem Co. 518-4 Taijul Bldg, Doonchundong, Kangdong-gu, Seoul 134-060, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
5528
|
Bronckers ALJJ, Sasaguri K, Cavender AC, D'Souza RN, Engelse MA. Expression of Runx2/Cbfa1/Pebp2alphaA during angiogenesis in postnatal rodent and fetal human orofacial tissues. J Bone Miner Res 2005; 20:428-37. [PMID: 15746987 DOI: 10.1359/jbmr.041118] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2003] [Revised: 09/07/2004] [Accepted: 10/15/2004] [Indexed: 01/26/2023]
Abstract
UNLABELLED Transient expression of Runx2 is reported in endothelial cells and vascular smooth muscle cells during vessel formation in skin, stroma of forming bones and developing periodontal ligament, developing skeletal muscle cells, and fat tissue. The data suggest that Runx2 is expressed in a multipotential mesenchymal cell population that gives rise to various osseous and nonosseous cell lineages. INTRODUCTION Runx2/Cbfa1 is a transcription factor essential for cells of osteogenic and dentinogenic lineages. Here we examined expression of Runx2/Cbfa1 (all isotypes) in several nonskeletal cell types present in developing orofacial tissues of neonatal rodents and human fetuses with special emphasis on vessel formation. MATERIALS AND METHODS Sections obtained from heads or jaws of postnatal mice, hamster, and human fetuses were immunostained with monoclonal anti-Pebp2aA antibody. Mouse and human tissues were also examined by in situ hybridization. Sections of Runx2 null mutant mice with a LacZ reporter construct inserted in the Runx2 locus were stained for Runx2 promoter activity with anti-galactosidase. RESULTS We found transient mRNA and protein expression in endothelial cells and in vascular smooth muscle cells of forming vessels in skin, alveoli of forming bone, and forming periodontal ligament. We also noticed weak and variable expression in some fibroblasts of embryonic skin, early differentiating cross-striated muscle cells, and differentiating fat cells. CONCLUSION Runx2 is not an exclusive marker for chondrogenic, osteogenic, and dentinogenic tissues, but is much more widely present in an early multipotential mesenchymal cell population that gives rise to several other lineages.
Collapse
Affiliation(s)
- Antonius L J J Bronckers
- Department of Oral Cell Biology, ACTA, Vrije Universiteit and Universiteit van Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
5529
|
Cowan CM, Aalami OO, Shi YY, Chou YF, Mari C, Thomas R, Quarto N, Nacamuli RP, Contag CH, Wu B, Longaker MT. Bone Morphogenetic Protein 2 and Retinoic Acid Acceleratein VivoBone Formation, Osteoclast Recruitment, and Bone Turnover. ACTA ACUST UNITED AC 2005; 11:645-58. [PMID: 15869441 DOI: 10.1089/ten.2005.11.645] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Reconstruction of craniofacial defects presents a substantial biomedical burden, and requires complex surgery. Interestingly, children after age 2 years and adults are unable to heal large skull defects. This nonhealing paradigm provides an excellent model system for craniofacial skeletal tissueengineering strategies. Previous studies have documented the in vivo osteogenic potential of adipose-derived stromal (ADS) cells and bone marrow-derived stromal (BMS) cells. This study investigates the ability to accelerate in vivo osteogenesis on ex vivo recombinant human bone morphogenetic protein 2 (BMP-2) and retinoic acid stimulation. Mouse osteoblasts, ADS cells, and BMS cells were seeded onto apatite-coated PLGA scaffolds, stimulated with rhBMP-2 and retinoic acid ex vivo for 4 weeks, and subsequently implanted into critically sized (4 mm) calvarial defects. Samples were harvested after 2, 4, 8, and 12 weeks. Areas of complete bony bridging were noted as early as 2 weeks in vivo; however, osteoclasts were attracted to the scaffold as identified by calcitonin receptor staining and tartrate-resistant acid phosphatase activity staining. Although the optimal method of in vitro osteogenic priming for mesenchymal cells remains unknown, these results provide evidence that BMP-2 and retinoic acid stimulation of multipotent cells ex vivo can subsequently induce significant quantities of bone formation within a short time period in vivo.
Collapse
Affiliation(s)
- Catherine M Cowan
- Department of Surgery, Stanford University School of Medicine, CA 94305, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5530
|
Katz AJ, Tholpady A, Tholpady SS, Shang H, Ogle RC. Cell Surface and Transcriptional Characterization of Human Adipose-Derived Adherent Stromal (hADAS) Cells. Stem Cells 2005; 23:412-23. [PMID: 15749936 DOI: 10.1634/stemcells.2004-0021] [Citation(s) in RCA: 605] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Adult human subcutaneous adipose tissue contains cells with intriguing multilineage developmental plasticity, much like marrow-derived mesenchymal stem cells. Putative stem or progenitor cells from fat have been given many different names in the literature, reflecting an early and evolving consensus regarding their phenotypic characterization. The study reported here used microarrays to evaluate over 170 genes relating to angiogenesis and extracellular matrix in undifferentiated, early-passage human adipose-derived adherent stromal (hADAS) cells isolated from three separate donors. The hADAS populations unanimously transcribed 66% of the screened genes, and 83% were transcribed by at least two of the three populations. The most highly transcribed genes relate to functional groupings such as cell adhesion, matrix proteins, growth factors and receptors, and proteases. The transcriptome of hADAS cells demonstrated by this work reveals many similarities to published profiles of bone marrow mesenchymal stem cells (MSCs). In addition, flow analysis of over 24 hADAS cell surface proteins (n = 7 donors) both confirms and expands on the existing literature and reveals strong intergroup correlation, despite an inconsistent nomenclature and the lack of standardized protocols for cell isolation and culture. Finally, based on flow analysis and reverse transcription polymerase chain reaction studies, our results suggest that hADAS cells do not express several proteins that are implicated as markers of "stemness" in other stem cell populations, including telomerase, CD133, and the membrane transporter ABCG2.
Collapse
Affiliation(s)
- Adam J Katz
- Department of Plastic and Reconstructive Surgery, University of Virginia, Charlottesville, VA 22908, USA.
| | | | | | | | | |
Collapse
|
5531
|
Lendeckel S, Jödicke A, Christophis P, Heidinger K, Wolff J, Fraser JK, Hedrick MH, Berthold L, Howaldt HP. Autologous stem cells (adipose) and fibrin glue used to treat widespread traumatic calvarial defects: case report. J Craniomaxillofac Surg 2005; 32:370-3. [PMID: 15555520 DOI: 10.1016/j.jcms.2004.06.002] [Citation(s) in RCA: 435] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2003] [Accepted: 06/21/2004] [Indexed: 02/08/2023] Open
Abstract
This is a report of a 7-year-old girl suffering from widespread calvarial defects after severe head injury with multifragment calvarial fractures, decompressive craniectomy for refractory intracranial hypertension and replantation of cryopreserved skull fragments. Chronic infection resulted in an unstable skull with marked bony defects. Two years after the initial injury the calvarial defects were repaired. Due to the limited amount of autologous cancellous bone available from the iliac crest, autologous adipose derived stem cells were processed simultaneously and applied to the calvarial defects in a single operative procedure. The stem cells were kept in place using autologous fibrin glue. Mechanical fixation was achieved by two large, resorbable macroporous sheets acting as a soft tissue barrier at the same time. The postoperative course was uneventful and CT-scans showed new bone formation and near complete calvarial continuity three months after the reconstruction.
Collapse
Affiliation(s)
- Stefan Lendeckel
- Department of Maxillofacial and Facial Plastic Surgery, Justus-Liebig-University Medical School, Giessen, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
5532
|
Bost F, Aouadi M, Caron L, Even P, Belmonte N, Prot M, Dani C, Hofman P, Pagès G, Pouysségur J, Le Marchand-Brustel Y, Binétruy B. The extracellular signal-regulated kinase isoform ERK1 is specifically required for in vitro and in vivo adipogenesis. Diabetes 2005; 54:402-11. [PMID: 15677498 DOI: 10.2337/diabetes.54.2.402] [Citation(s) in RCA: 263] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Hyperplasia of adipose tissue is critical for the development of obesity, but molecular mechanisms governing normal or pathological recruitment of new adipocytes remain unclear. The extracellular signal-regulated kinase (ERK) pathway plays a pivotal role in many essential cellular functions, such as proliferation and differentiation. Using ERK1(-/-) mice, we investigated the role of this isoform in adipose tissue development. Mice lacking ERK1 have decreased adiposity and fewer adipocytes than wild-type animals. Furthermore, ERK1(-/-) mice challenged with high-fat diet are resistant to obesity, are protected from insulin resistance, and have a higher postprandial metabolic rate. To get insights into cellular mechanisms implicated in reduced adiposity in ERK1(-/-) animals, we analyzed adipocyte differentiation in ERK1(-/-) cells. Compared with wild-type control cells, mouse embryo fibroblasts and cultures of adult preadipocytes isolated from ERK1(-/-) adult animals exhibit impaired adipogenesis. An inhibitor of the ERK pathway does not affect the residual adipogenesis of the ERK1(-/-) cells, suggesting that ERK2 is not implicated in adipocyte differentiation. Our results clearly link ERK1 to the regulation of adipocyte differentiation, adiposity, and high-fat diet-induced obesity. This suggests that a therapeutic approach of obesity targeting specifically the ERK1 isoform and not ERK2 would be of particular interest.
Collapse
Affiliation(s)
- Frédéric Bost
- Institut National de la Santé et de la Recherche Médicale, Unité 568, IFR 50, Faculté de Médecine, Université de Nice- Sophia Antipolis, Avenue de Valombrose, Nice, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5533
|
Li WJWJ, Tuli R, Okafor C, Derfoul A, Danielson KGKG, Hall DJDJ, Tuan RSRS. A three-dimensional nanofibrous scaffold for cartilage tissue engineering using human mesenchymal stem cells. Biomaterials 2005; 26:599-609. [PMID: 15282138 DOI: 10.1016/j.biomaterials.2004.03.005] [Citation(s) in RCA: 613] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2003] [Accepted: 03/13/2004] [Indexed: 12/16/2022]
Abstract
The utilization of adult stem cells in tissue engineering is a promising solution to the problem of tissue or organ shortage. Adult bone marrow derived mesenchymal stem cells (MSCs) are undifferentiated, multipotential cells which are capable of giving rise to chondrocytes when maintained in a three-dimensional culture and treated with members of the transforming growth factor-beta (TGF-beta) family of growth factors. In this study, we fabricated a nanofibrous scaffold (NFS) made of a synthetic biodegradable polymer, poly(-caprolactone) (PCL), and examined its ability to support in vitro chondrogenesis of MSCs. The electrospun PCL porous scaffold was constructed of uniform, randomly oriented nanofibers with a diameter of 700 nm, and structural integrity of this scaffold was maintained over a 21-day culture period. MSCs cultured in NFSs in the presence of TGF-beta1 differentiated to a chondrocytic phenotype, as evidenced by chondrocyte-specific gene expression and synthesis of cartilage-associated extracellular matrix (ECM) proteins. The level of chondrogenesis observed in MSCs seeded within NFSs was comparable to that observed for MSCs maintained as cell aggregates or pellets, a widely used culture protocol for studying chondrogenesis of MSCs in vitro. Due to the physical nature and improved mechanical properties of NFSs, particularly in comparison to cell pellets, the findings reported here suggest that the PCL NFS is a practical carrier for MSC transplantation, and represents a candidate scaffold for cell-based tissue engineering approaches to cartilage repair.
Collapse
Affiliation(s)
- W-J Wan-Ju Li
- Department of Health and Human Services, Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis, and Musculoskeletal and Skin Diseases, National Institute of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
5534
|
Casteilla L, Charrière G, Laharrague P, Cousin B, Planat-Benard V, Péricaud L, Chavoin JP. [Adipose tissue, plastic and reconstructive surgery: come back to sources]. ANN CHIR PLAST ESTH 2005; 49:409-18. [PMID: 15518941 DOI: 10.1016/j.anplas.2004.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The adipose tissue represents a large amount of adult tissue. For long time, it was considered as a filling tissue and used in plastic and reconstructive surgery. It was always studied for its main involvement in energy metabolism and energy disorders as diabetes and obesity. More recently, its endocrine functions emerged and thus play a key role in many physiological functions as inflammation and immunity. The presence of preadipocytes throughout life was demonstrated using primary culture technology from cells derived from adipose tissue. In recent papers, cells derived from adipose tissue were used for haematopoiesis, vascularisation or skeletal muscle recovery. Differentiation into functional cardiomyocytes, osteoblasts and neural cells was obtained in vitro. These spectacular data, the fact that adipose tissue is easy to sample and the possibility to create cell or tissue banks open numerous and promising perspectives in regenerative medicine.
Collapse
Affiliation(s)
- L Casteilla
- UMR 5018 CNRS UPS, IFR31, TSA 50032, IFR31, Bat. L1, CHU de Rangueil, 1, avenue Poulhès, 31059 Toulouse 09, France.
| | | | | | | | | | | | | |
Collapse
|
5535
|
Abstract
Despite the wealth of research on stem-cell biology and therapy, there have been few advances in the ex vivo expansion of adult stem cells. Fetal bovine serum, which can elicit an immune reaction in hosts receiving cellular products, remains essential to the culture methods. Recent work by Prockop et al. suggests that Dickkopf-1 (Dkk-1) could stimulate the proliferation of mesenchymal stem cells (MSCs) while maintaining an undifferentiated phenotype. The use of Dkk-1 in cell-processing laboratories might advance cell-expansion technology and greatly increase the clinical uses of MSCs.
Collapse
Affiliation(s)
- Edwin M Horwitz
- Divisions of Stem Cell Transplantation and Experimental Hematology, Department of Hematology-Oncology, St Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
5536
|
Tominaga S, Yamaguchi T, Takahashi SI, Hirose F, Osumi T. Negative regulation of adipogenesis from human mesenchymal stem cells by Jun N-terminal kinase. Biochem Biophys Res Commun 2005; 326:499-504. [PMID: 15582605 DOI: 10.1016/j.bbrc.2004.11.056] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2004] [Indexed: 01/01/2023]
Abstract
Human mesenchymal stem cells (hMSCs) are capable of differentiating into several cell types including adipocytes, osteoblasts, and chondrocytes, under appropriate culture conditions. We found that SP600125, an inhibitor of Jun N-terminal kinase (JNK), promoted adipogenesis whereas it repressed osteogenesis from hMSCs. SP600125 increased the expression of adipogenic transcription factors, CCAAT/enhancer-binding proteins alpha and beta as well as peroxisome proliferator-activated receptor gamma2, which suggested that the chemical acted on the early steps of transcriptional regulatory cascade in adipogenesis. A gene reporter assay showed that SP600125 and a dominant negative JNK promoted a transcriptional activity dependent on the cAMP-response element (CRE). Thus, JNK represses adipogenesis from hMSCs probably by, at least in part, inhibiting the transactivating function of CRE-binding protein. Another action of JNK, phosphorylation at Ser(307) of insulin receptor substrate-1, was also predicted to contribute to the repression of adipogenesis.
Collapse
Affiliation(s)
- Sachiko Tominaga
- Graduate School of Life Science, Himeji Institute of Technology, University of Hyogo, 3-2-1 Koto, Kamigori, Hyogo 678-1297, Japan
| | | | | | | | | |
Collapse
|
5537
|
Hicok KC, Du Laney TV, Zhou YS, Halvorsen YDC, Hitt DC, Cooper LF, Gimble JM. Human adipose-derived adult stem cells produce osteoid in vivo. ACTA ACUST UNITED AC 2005; 10:371-80. [PMID: 15165454 DOI: 10.1089/107632704323061735] [Citation(s) in RCA: 220] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Adult subcutaneous fat tissue is an abundant source of multipotent cells. Previous studies from our laboratory have shown that, in vitro, adipose-derived adult stem (ADAS) cells express bone marker proteins including alkaline phosphatase, type I collagen, osteopontin, and osteocalcin and produce a mineralized matrix as shown by alizarin red staining. In the current study, the ADAS cell ability to form osteoid in vivo was determined. ADAS cells were isolated from liposuction waste of three individual donors and expanded in vitro before implantation. Equal numbers of cells (3 x 10(6)) were loaded onto either hydroxyapatite/tricalcium phosphate (HA-TCP) cubes or the collagen/HA-TCP composite matrix, Collagraft, and then implanted subcutaneously into SCID mice. After 6 weeks, implants were removed, fixed, and demineralized and sectioned for hematoxylin and eosin staining. Osteoid formation was observed in 80% of HA-TCP implants loaded with ADAS cells. Only 20% of Collagraft implants were positive for the presence of osteoid matrix. Whereas 100% of HA-TCP implants loaded with hFOB 1.19 cells formed osteoid, Collagraft loaded with hFOB 1.19 cells displayed a high degree of adipose tissue within the matrix. Immunostaining of serial sections for human nuclear antigen demonstrated that the osteoid contained human cells. Osteoid formation was not observed in control HA-TCP or Collagraft matrices implanted without cells. In summary, the data demonstrate the ability of ADAS cells to form osteoid matrix in vivo. Because of their abundance and accessibility, ADAS cells may prove to be a novel cell therapeutic for bone repair and regeneration.
Collapse
|
5538
|
Nakahara T, Nakamura T, Kobayashi E, Kuremoto KI, Matsuno T, Tabata Y, Eto K, Shimizu Y. In situ tissue engineering of periodontal tissues by seeding with periodontal ligament-derived cells. ACTA ACUST UNITED AC 2005; 10:537-44. [PMID: 15165470 DOI: 10.1089/107632704323061898] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The feasibility of an in situ tissue-engineering method employing cell-based therapy with autologous periodontal ligament-derived cells was investigated. Periodontal ligament cells were obtained from six beagle dogs. Periodontal fenestration defects (6 x 4 mm) were created bilaterally at a location 6 mm apical to the marginal alveolar crest in the maxillary canines. Alkaline phosphatase-positive periodontal ligament cells (3 x 10(5) cells) were seeded onto a collagen sponge scaffold just before implantation. One defect was filled with the cell-scaffold construct, and another was left empty as the control. All animals were killed 4 weeks after surgery, and specimens were evaluated histomorphometrically. All the histomorphometrical data were analyzed by three-way analysis of variance with the Bonferroni multiple comparisons test. Regeneration of apical tissue was faster than that of coronal and isolated tissues on the control side (apical > coronal > isolated; p < 0.0001). On the other hand, on the cell-seeded side, regeneration of the cementum was observed uniformly on the root surface. Our data suggest that the seeded cells induced cementum regeneration on the root surface, indicating the potential of in situ tissue engineering using autologous cells for the regeneration of periodontal tissues.
Collapse
Affiliation(s)
- Taka Nakahara
- Section of Molecular Craniofacial Embryology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
5539
|
Justesen J, Pedersen SB, Stenderup K, Kassem M. Subcutaneous adipocytes can differentiate into bone-forming cells in vitro and in vivo. ACTA ACUST UNITED AC 2005; 10:381-91. [PMID: 15165455 DOI: 10.1089/107632704323061744] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Interconversion of bone marrow osteoblasts and adipocytes has been reported previously. However, the osteogenic potential of extramedullary adipocytes is not known. Thus, we incubated a pure culture of human subcutaneous adipocytes in control medium for 1-2 weeks. Afterward, the cells were incubated in either osteoblast medium (OB medium) containing various combinations of calcitriol, dexamethasone, ascorbic acid, and beta-glycerophosphate or in adipocyte medium (AD medium) containing HEPES, biotin, pantothenate, insulin, triiodothyronine, dexamethasone, and isobutylmethylxanthine for 4 weeks. Expression of osteoblastic and adipocytic phenotypes was examined by determination of lineage-specific mRNA markers and in vitro adipocyte and osteoblast formation. Cells were also implanted, mixed with hydroxyapatite-tricalcium phosphate powder, in the subcutaneous tissue of immunodeficient mice in order to assess in vivo bone formation potential. One week after incubation in control medium, cells formed fusiform elongated fibroblast-like cells. In OB medium, cells stained positive for alkaline phosphatase (AP) and expressed mRNAs encoding Cbfa1/Runx2, AP, and osteocalcin. In AD medium cells reacquired adipocyte morphology with multilocular lipid-filled cells. Also, the cells expressed adipocyte-specific mRNA markers: lipoprotein lipase and peroxisome proliferator-activated receptor gamma2. Bone was formed only in the in vivo implants of cells incubated in OB medium. In conclusion, extramedullary adipocytes can transdifferentiate to bone-forming cells. Because of their ease of isolation, adipocytes may be good candidates for tissue-engineering protocols aimed at creating bone tissue for the repair of nonunion fractures and large bone defects.
Collapse
Affiliation(s)
- Jeannette Justesen
- University Department of Endocrinology and Metabolism, Aarhus Amtssygehus, DK-8000 Aarhus, Denmark
| | | | | | | |
Collapse
|
5540
|
Boquest AC, Shahdadfar A, Frønsdal K, Sigurjonsson O, Tunheim SH, Collas P, Brinchmann JE. Isolation and transcription profiling of purified uncultured human stromal stem cells: alteration of gene expression after in vitro cell culture. Mol Biol Cell 2005; 16:1131-41. [PMID: 15635089 PMCID: PMC551479 DOI: 10.1091/mbc.e04-10-0949] [Citation(s) in RCA: 263] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Stromal stem cells proliferate in vitro and may be differentiated along several lineages. Freshly isolated, these cells have been too few or insufficiently pure to be thoroughly characterized. Here, we have isolated two populations of CD45-CD34+CD105+ cells from human adipose tissue which could be separated based on expression of CD31. Compared with CD31+ cells, CD31- cells overexpressed transcripts associated with cell cycle quiescence and stemness, and transcripts involved in the biology of cartilage, bone, fat, muscle, and neural tissues. In contrast, CD31+ cells overexpressed transcripts associated with endothelium and the major histocompatibility complex class II complex. Clones of CD31- cells could be expanded in vitro and differentiated into cells with characteristics of bone, fat, and neural-like tissue. On culture, transcripts associated with cell cycle quiescence, stemness, certain cytokines and organ specific genes were down-regulated, whereas transcripts associated with signal transduction, cell adhesion, and cytoskeletal +CD105+CD31- cells from human adipose tissue have stromal stem cell properties which may make them useful for tissue engineering.
Collapse
Affiliation(s)
- Andrew C Boquest
- Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway
| | | | | | | | | | | | | |
Collapse
|
5541
|
Li X, Lee JP, Balian G, Greg Anderson D. Modulation of chondrocytic properties of fat-derived mesenchymal cells in co-cultures with nucleus pulposus. Connect Tissue Res 2005; 46:75-82. [PMID: 16019417 DOI: 10.1080/03008200590954104] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Human subcutaneous fat-derived mesenchymal cells recently have been shown to have the potential to differentiate in vitro into a variety of cell types, including adipocytes, osteoblasts, chondrocytes, and myoblasts. This effect suggests that fat tissue may serve as an abundant and easily acquired source of multipotent cells for tissue engineering. The multipotential characteristics of fat-derived mesenchymal cells from the inguinial fat pad of rabbit have not been clearly defined. In this study we have isolated a population of mesenchymal cells from inguinal fat from adult New Zealand white rabbits. The cells that were maintained under various differentiation conditions were shown to differentiate in vitro into adipocytes, osteoblasts, or chondrocytes; this differentiation was demonstrated using gene expression for tissue-specific proteins. We also co-cultured the cells with intervertebral disk tissue from the nucleus pulpous or from the annulus fibrosus. The fat-derived cells co-cultured with nucleus pulposus showed an increase in expression of type II collagen and aggrecan genes, compared with cells in alginate alone and cells co-cultured with annulus fibrosus. The data suggest that the fat-derived mesenchymal cells responded to soluble mediators from the disk. Future studies on intervertebral disk reconstruction could be based on our findings with fat-derived multipotential cells from the inguinal region of the rabbit that were co-cultured with disk tissue and may prove useful in tissue engineering strategies.
Collapse
Affiliation(s)
- Xudong Li
- Department of Orthopaedic Surgery, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA.
| | | | | | | |
Collapse
|
5542
|
Mahmoudifar N, Doran PM. Tissue engineering of human cartilage in bioreactors using single and composite cell-seeded scaffolds. Biotechnol Bioeng 2005; 91:338-55. [PMID: 15959891 DOI: 10.1002/bit.20490] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Chondrocytes isolated from human fetal epiphyseal cartilage were seeded under mixed conditions into 15-mm-diameter polyglycolic acid (PGA) scaffolds and cultured in recirculation column bioreactors to generate cartilage constructs. After seeding, the cell distributions in thick (4.75 mm) and thin (2.15 mm) PGA disks were nonuniform, with higher cell densities accumulating near the top surfaces. Composite scaffolds were developed by suturing together two thin PGA disks after seeding to manipulate the initial cell distribution before bioreactor culture. The effect of medium flow direction in the bioreactors, including periodic reversal of medium flow, was also investigated. The quality of the tissue-engineered cartilage was assessed after 5 weeks of culture in terms of the tissue wet weight, glycosaminoglycan (GAG), total collagen and collagen type II contents, histological analysis of cell, GAG and collagen distributions, and immunohistochemical analysis of collagen types I and II. Significant enhancement in construct quality was achieved using composite scaffolds compared with single PGA disks. Operation of the bioreactors with periodic medium flow reversal instead of unidirectional flow yielded further improvements in tissue weight and GAG and collagen contents with the composite scaffolds. At harvest, the constructs contained GAG concentrations similar to those measured in ex vivo human adult articular cartilage; however, total collagen and collagen type II levels were substantially lower than those in adult tissue. This study demonstrates that the location of regions of high cell density in the scaffold coupled with application of dynamic bioreactor operating conditions has a significant influence on the quality of tissue-engineered cartilage.
Collapse
Affiliation(s)
- Nastaran Mahmoudifar
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia
| | | |
Collapse
|
5543
|
Kim YI, Oh IH. Cell Biological Characteristics of Adult Stem Cells. JOURNAL OF THE KOREAN MEDICAL ASSOCIATION 2005. [DOI: 10.5124/jkma.2005.48.10.993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Yeong In Kim
- Catholic High-Performance Cell Therapy Center, The Catholic University of Korea, School of Medicine, Korea. ,
| | - Il-Hoan Oh
- Catholic High-Performance Cell Therapy Center, The Catholic University of Korea, School of Medicine, Korea. ,
| |
Collapse
|
5544
|
Little M. Wilms' tumor: starting off the kidney all over again? PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2005; 40:107-32. [PMID: 17153482 DOI: 10.1007/3-540-27671-8_5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Affiliation(s)
- M Little
- Institute for Molecular Bioscience, Level 4 North, Queensland Bioscience Precinct, The University of Queensland, St. Lucia, 4072, Qld, Australia
| |
Collapse
|
5545
|
Hattori H, Masuoka K, Sato M, Ishihara M, Asazuma T, Takase B, Kikuchi M, Nemoto K, Ishihara M. Bone formation using human adipose tissue-derived stromal cells and a biodegradable scaffold. J Biomed Mater Res B Appl Biomater 2005; 76:230-9. [PMID: 16047328 DOI: 10.1002/jbm.b.30357] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Human adipose tissue, obtained by liposuction, was processed to obtain a fibroblast-like population of cells or adipose tissue-derived stromal cells (ATSCs). The ATSCs, as well as bone marrow-derived mesenchymal stem cells (BMSCs), have the capacity for renewal and the potential to differentiate into multiple lineages of mesenchymal tissues. These cells are capable of forming bone when implanted ectopically in an appropriate scaffold. The aim of this study was to evaluate a beta-tricalcium phosphate (beta-TCP) as a scaffold and to compare the potential of osteogenic differentiation of ATSCs with BMSCs. Both cell types were loaded into beta-TCP disk and cultured in an osteogenic induction medium. Optimal osteogenic differentiation in ATSCs in vitro, as determined by secretion of osteocalcin, scanning electron microscope, and histology, were obtained in the culturing with the beta-TCP disk. Furthermore, bone formation in vivo was examined by using the ATSC- or BMSC-loaded scaffolds in nude mice. The present results show that ATSCs have a similar ability to differentiate into osteoblasts and to synthesize bone in beta-TCP disk as have BMSCs.
Collapse
Affiliation(s)
- Hidemi Hattori
- Division of Biomedical Engineering, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
5546
|
Abstract
Postnatal stem cells have been isolated from a variety of tissues and they are highly expected to have potentiality to be utilized for cell-based clinical therapies. Bone marrow stromal stem cells (BMSSCs) derived from bone marrow stromal tissue have been identified as a population of multipotent mesenchymal stem cells that are capable of differentiating into osteoblasts, adipocytes, chondrocytes, muscle cells, and neural cells. The most significant tissue regeneration trait of BMSSCs is their in vivo bone regeneration capability, which has been widely studied for understanding molecular and cellular mechanisms of osteogenesis, and, more importantly, developing into a stem-cell-based therapy. Recent studies further demonstrated that BMSSC-mediated bone regeneration is a promising approach for regenerative medicine in clinical trials. However, there are some fundamental questions that remain to be answered prior to successful utilization of BMSSCs in clinical therapy. For instance, how to maintain stemness of BMSSCs will be a critical issue for developing methodologies to propagate multi-potential stem cells in vitro, in order to allow the development of effective clinical therapies.
Collapse
Affiliation(s)
- Wataru Sonoyama
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | |
Collapse
|
5547
|
Strem BM, Hicok KC, Zhu M, Wulur I, Alfonso Z, Schreiber RE, Fraser JK, Hedrick MH. Multipotential differentiation of adipose tissue-derived stem cells. Keio J Med 2005; 54:132-41. [PMID: 16237275 DOI: 10.2302/kjm.54.132] [Citation(s) in RCA: 629] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tissue engineering offers considerable promise in the repair or replacement of diseased and/or damaged tissues. The cellular component of this regenerative approach will play a key role in bringing these tissue engineered constructs from the laboratory bench to the clinical bedside. However, the ideal source of cells still remains unclear and may differ depending upon the application. Current research for many applications is focused on the use of adult stem cells. The properties of adult stem cells that make them well-suited for regenerative medicine are (1) ease of harvest for autologous transplantation, (2) high proliferation rates for ex vivo expansion and (3) multilineage differentiation capacity. This review will highlight the use of adipose tissue as a reservoir of adult stem cells and draw conclusions based upon comparisons with bone marrow stromal cells.
Collapse
|
5548
|
Nacamuli RP, Wan DC, Lenton KA, Longaker MT. New developments in pediatric plastic surgery research. Clin Plast Surg 2005; 32:123-36, ix-x. [PMID: 15636770 DOI: 10.1016/j.cps.2004.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Pediatric plastic surgery research is a rapidly expanding field. Unique in many ways, researchers in this field stand at the union of multiple scientific specialties, including biomedical engineering, tissue engineering, polymer science, molecular biology, developmental biology, and genetics. The goal of this scientific effort is to translate research advances into improved treatments for children with congenital and acquired defects. Although the last decade has seen a dramatic acceleration in research related to pediatric plastic surgery, the next 10 years will no doubt lead to novel treatment strategies with improved clinical outcomes.
Collapse
Affiliation(s)
- Randall P Nacamuli
- Children's Surgical Research Program, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford University Medical Center, 257 Campus Drive, Stanford, CA 94305-5148, USA
| | | | | | | |
Collapse
|
5549
|
Abstract
The field of tissue engineering integrates the latest advances in molecular biology, biochemistry, engineering, material science, and medical transplantation. Researchers in the developing field of regenerative medicine have identified bone tissue engineering as an attractive translational target. Clinical problems requiring bone regeneration are diverse, and no single regeneration approach will likely resolve all defects. Recent advances in the field of tissue engineering have included the use of sophisticated biocompatible scaffolds, new postnatal multipotent cell populations, and the appropriate cellular stimulation. In particular, synthetic polymer scaffolds allow for fast and reproducible construction, while still retaining biocompatible characteristics. These criteria relate to the immediate goal of determining the ideal implant. The search is becoming a reality with widespread availability of biocompatible scaffolds; however, the desired parameters have not been clearly defined. Currently, most research focuses on the use of bone morphogenetic proteins (BMPs), specifically BMP-2 and BMP-7. These proteins induce osteogenic differentiation in vitro, as well as bone defect healing in vivo. Protein-scaffold interactions that enhance BMP binding are of the utmost importance, since prolonged BMP release creates the most osteogenic microenvironment. Transition into clinical studies has had only mild success and relies on large doses of BMPs for bone formation. Advances within the field of bone tissue engineering will likely overcome these challenges and lead to more clinically relevant therapies.
Collapse
Affiliation(s)
- Catherine M Cowan
- Department of Bioengineering, University of California Los Angeles, Los Angeles, California 90095, USA
| | | | | | | |
Collapse
|
5550
|
Mitchell PO, Pavlath GK. Skeletal muscle atrophy leads to loss and dysfunction of muscle precursor cells. Am J Physiol Cell Physiol 2004; 287:C1753-62. [PMID: 15329339 DOI: 10.1152/ajpcell.00292.2004] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Atrophy of skeletal muscle leads to decreases in myofiber size and nuclear number; however, the effects of atrophic conditions on muscle precursor cells (MPC) are largely unknown. MPC lie outside myofibers and represent the main source of additional myonuclei necessary for muscle growth and repair. In the present study, we examined the properties of MPC after hindlimb suspension (HS)-induced atrophy and subsequent recovery of the mouse hindlimb muscles. We demonstrated that the number of MPC in atrophied muscles was decreased. RT-PCR analysis of cells isolated from atrophied muscles indicated that several mRNA characteristic of the myogenic program in MPC were absent. Cells isolated from atrophied muscles failed to properly proliferate and undergo differentiation into multinucleated myotubes. Thus atrophy led to a decrease in MPC and caused dysfunction in those MPC that remained. Upon regrowth of the atrophied muscles, these deleterious effects were reversed. Our data suggest that preventing loss or dysfunction of MPC may be a new pharmacological target during muscle atrophy.
Collapse
Affiliation(s)
- Patrick O Mitchell
- Department of Pharmacology, Emory University School of Medicine, 5024 O. W. Rollins Research Center, Atlanta, GA 30322, USA
| | | |
Collapse
|