5651
|
Phillips BW, Vernochet C, Dani C. Differentiation of embryonic stem cells for pharmacological studies on adipose cells. Pharmacol Res 2003; 47:263-8. [PMID: 12644382 DOI: 10.1016/s1043-6618(03)00035-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The ongoing global explosion in the incidence of obesity has focused attention on the development of adipose cells. Severe obesity is the result of an increase in fat cell size in combination with increased fat cell number. New fat cells arise from a pre-existing pool of adipose stem cells that are present irrespective of age. The development of established preadipocyte cell lines has facilitated the study of different steps leading to terminal differentiation. However, these systems are limited for studying early events of differentiation as they represent cells which are already determined for the adipogenic lineage. In vitro differentiation of mouse embryonic stem (ES) cells towards the adipogenic lineage provides an alternative source of adipocytes for study in tissue culture and offers the possibility to investigate regulation of the first steps of adipose cell development. In this review, we describe the sequential requirement of retinoic acid and PPARgamma during adipogenesis in ES cells. Stimulation of ES cells with synthetic retinoids which are selective ligands of the retinoic acid receptor isotypes allowed the investigation of the contribution of the different retinoic receptors on the RA-dependent differentiation. The effects of thiazolidinediones, a new class of pharmacological agents used for the treatment of type 2 diabetes, and of statins, drugs used in therapy for lowering cholesterol, on the differentiation of ES cells into adipocytes or osteoblasts are described. Finally, we propose a model in which PPARgamma plays a key role in the decision of stem cells to undergo differentiation into adipocytes or osteoblasts, two closely related lineages.
Collapse
Affiliation(s)
- Blaine W Phillips
- Institute of Signalling, Developmental Biology and Cancer Research, CNRS UMR 6543, Centre de Biochimie, 06108 Nice Cedex 2, France
| | | | | |
Collapse
|
5652
|
Abstract
Tissue engineering is a rather new field of science. Despite this fact, some experimental investigations have already been applied in clinical studies. Compared to other medical fields, tissue engineering in urology is well established. Tissue-engineered bulking agents and tissue-engineered bladder augments are being investigated in clinical trials. Even though the knowledge gained in recent years is promising, the results of cellular therapies need to be critically judged before being finally applied in patients. Genetic engineering and stem cell research (adult undifferentiated cells) have had major impact on the field of tissue engineering over the past 2 years. By using the technology of genetic engineering, biochemical and functional qualities of tissues may be modified. Adult stem cells may help to substitute lost tissue in an autologous fashion by isolating undifferentiated cells from the body and by differentiating them into a desired cell type. These cells may be used to form native functional tissue to replace a diseased organ or organ part.
Collapse
Affiliation(s)
- G Bartsch
- Abteilung für Urologie und Kinderurologie, Urologische Universitätsklinik und Poliklinik, Universität Ulm.
| | | |
Collapse
|
5653
|
Abstract
Trauma and disease of bones and joints, frequently involving structural damage to both the articular cartilage surface and the subchondral bone, result in severe pain and disability for millions of people worldwide and represent major challenges for the orthopedic surgeons. Therapeutic repair of skeletal tissues by tissue engineering has raised the interest of the scientific community, providing very promising results in preclinical animal models and clinical pilot studies. In this review, we discuss this approach. The choice of a proper cell type is addressed. The use of terminally differentiated cells, as in the case of autologous chondrocyte implantation, is compared with the advantages/disadvantages of using more undifferentiated cell types, such as stem cells or early mesenchymal progenitors that retain multi-lineage and self-renewal potentials. The need for proper scaffold matrices is also examined, and we provide a brief overview of their fundamental properties. A description of the natural and biosynthetic materials currently used for reconstruction purposes, either of cartilage or bone, is given. Finally, we highlight the positive aspects and the remaining problems that will drive future research in articular cartilage and bone repair.
Collapse
Affiliation(s)
- Ranieri Cancedda
- Centro Biotecnologie Avanzate, Istituto Nazionale per la Ricerca sul Cancro, Largo R. Benzi, 10, 16132 Genoa, Italy.
| | | | | | | |
Collapse
|
5654
|
Abstract
Stem cells still lack integral and exhaustive legislation in Italy and in the European Union. The use of pluripotent embryonic stem cells (ESC) for cell therapy seems to be encumbered with several disadvantages, such as the frequency of aneuploidy and the risk of tumour development (i.e. formation of teratomes). In addition, the capacity for indefinite growth of ESC, which first seems to confer them an advantage, may become potentially harmful if some ESC contaminate the transplantation of their derived differentiated cells. This is, in part, contrasted by the ease of obtaining and expanding adult or cord blood-derived stem cells in vitro, and by their transdifferentiation capacity (the so-called somatic stem cell 'plasticity'). Moreover, ethical considerations make us plead against the use of human embryos for stem cell research. First and foremost, there is the ethical position that it is never permissible to stop human life in order to prolong human life, except in self-defence. We must maintain that a human embryo certainly constitutes a new human life with the direct potential of one day becoming a human infant. Therefore, human embryo should not be considered just a 'cluster of cells' but a 'person'. Humanity is at a philosophical crossroads and we need to speak up in favour of the dignity of human life from its very inception.
Collapse
Affiliation(s)
- Salvatore Mancuso
- Department of Obstetrics and Gynaecology, Catholic University of the Sacred Heart, Rome, Italy.
| |
Collapse
|
5655
|
Rangappa S, Fen C, Lee EH, Bongso A, Sim EKW, Wei EKS. Transformation of adult mesenchymal stem cells isolated from the fatty tissue into cardiomyocytes. Ann Thorac Surg 2003; 75:775-9. [PMID: 12645692 DOI: 10.1016/s0003-4975(02)04568-x] [Citation(s) in RCA: 270] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Myocardial infarction results in the death of cardiomyocytes, which are replaced by scar tissue. Cardiomyocytes cannot regenerate because they are terminally differentiated. Mesenchymal cells are pluripotent cells, which have the potential to differentiate to specialized tissues under appropriate stimuli. The aim of this study was to direct differentiation of the adult mesenchymal stem cells isolated from fatty tissue into cardiomyocytes using 5-azacytidine. METHODS Adult mesenchymal stem cells were isolated from the fatty tissue of New Zealand White rabbits and cultured in RPMI medium. Second-passaged mesenchymal cells were treated with various concentrations of 5-azacytidine and incubated for different intervals of time. The cells were plated in six-well dishes at 500, 5,000, and 50,000 cells/well. These cells were treated with 1-, 3-, 6-, 9-, and 12-micromol/L concentrations of 5-azacytidine and incubated for 12, 24, 48, and 72 hours. Later, the medium was replaced with fresh medium and incubated in a CO2 incubator. The medium was changed once at 3 to 4 days. At 2 months, the cells were fixed with 0.4% glutaraldehyde for 2 hours and later washed with phosphate-buffered saline. The transformed cells were subjected to immunostaining for the myosin heavy chain, alpha actinin, and troponin-I. RESULTS After treatment with 5-azacytidine, the adult mesenchymal stem cells were transformed into cardiomyocytes. At 1 week, some cells showed binucleation and extended cytoplasmic processes with adjacent cells. At 2 weeks, 20% to 30% of the cells increased in size and formed a ball-like appearance. At 3 weeks, these cells began to beat spontaneously in culture when observed under phase contrast microscope. Immunostaining of the transformed cells for myosin heavy chain, alpha actinin, and troponin-I was positive. The differentiated cells maintained the phenotype and did not dedifferentiate up to 2 months after treatment with 5-azacytidine. CONCLUSIONS These observations confirm that adult mesenchymal stem cells isolated from fatty tissue can be chemically transformed into cardiomyocytes. This can potentially be a source of autologous cells for myocardial repair.
Collapse
Affiliation(s)
- Sunil Rangappa
- Division of Cardiothoracic Surgery, National University Hospital, National University of Singapore, Singapore.
| | | | | | | | | | | |
Collapse
|
5656
|
Cousin B, André M, Arnaud E, Pénicaud L, Casteilla L. Reconstitution of lethally irradiated mice by cells isolated from adipose tissue. Biochem Biophys Res Commun 2003; 301:1016-22. [PMID: 12589814 DOI: 10.1016/s0006-291x(03)00061-5] [Citation(s) in RCA: 405] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
It is suggested that hematopoietic stem cells (HSC) could be found in several tissues of mesodermic origin. Among these, adipose tissue can expand throughout adult life and its expansion is not only due to mature adipocyte hypertrophy but also to the presence of precursor cells in stroma-vascular fraction (SVF). Here we report that transplantation of cells isolated from mice adipose tissue can efficiently rescue lethally irradiated mice and results in a reconstitution of major hematopoietic lineages. Donor cells can be detected in blood and in hematopoietic tissues of recipient mice. Adipose tissue contains a significant percentage of CD34, CD45 positive cells, and SVF cells were able to give rise to hematopoietic colonies in methylcellulose. We demonstrate the presence of hematopoietic progenitors in adipose tissue by phenotypic and functional characteristics. Thus adipose tissue could be considered as an important and convenient source of cells able to support hematopoiesis.
Collapse
Affiliation(s)
- Béatrice Cousin
- UMR 5018-CNRS, CHU Rangueil, Bat L1, 1 Avenue J. Poulhès, 31403, Toulouse Cedex, France.
| | | | | | | | | |
Collapse
|
5657
|
Muschler GF, Nitto H, Matsukura Y, Boehm C, Valdevit A, Kambic H, Davros W, Powell K, Easley K. Spine fusion using cell matrix composites enriched in bone marrow-derived cells. Clin Orthop Relat Res 2003:102-18. [PMID: 12567137 PMCID: PMC1425047 DOI: 10.1097/00003086-200302000-00018] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Bone marrow-derived cells including osteoblastic progenitors can be concentrated rapidly from bone marrow aspirates using the surface of selected implantable matrices for selective cell attachment. Concentration of cells in this way to produce an enriched cellular composite graft improves graft efficacy. The current study was designed to test the hypothesis that the biologic milieu of a bone marrow clot will significantly improve the efficacy of such a graft. An established posterior spinal fusion model and cancellous bone matrix was used to compare an enriched cellular composite bone graft alone, bone matrix plus bone marrow clot, and an enriched bone matrix composite graft plus bone marrow clot. Union score, quantitative computed tomography, and mechanical testing were used to define outcome. The union score for the enriched bone matrix plus bone marrow clot composite was superior to the enriched bone matrix alone and the bone matrix plus bone marrow clot. The enriched bone matrix plus bone marrow clot composite also was superior to the enriched bone matrix alone in fusion volume and in fusion area. These data confirm that the addition of a bone marrow clot to an enriched cell-matrix composite graft results in significant improvement in graft performance. Enriched composite grafts prepared using this strategy provide a rapid, simple, safe, and inexpensive method for intraoperative concentration and delivery of bone marrow-derived cells and connective tissue progenitors that may improve the outcome of bone grafting.
Collapse
Affiliation(s)
- George F Muschler
- Department of Orthopaedic Surgery, The Cleveland Clinic Foundation, Cleveland, OH 44195, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
5658
|
Abstract
Tissue replacement traditionally requires use of autologous tissue and is associated with the attendant morbidity of donor site harvest. In the case of allograft transplantation, there are concerns, similar to those associated with organ transplantation, of rejection and immunosuppression. For these reasons, emphasis has been placed on the development of tissue-engineered substitutes that incorporate autologous stem cells into tissue-engineered scaffolds. The authors' laboratory has characterized a population of cells obtained from processed lipoaspirate (PLA), which have the capacity in vitro to differentiate into osteoblasts, chondrocytes, myocytes, adipocytes, and neuron-like cells. Adipose tissue is an abundant, expendable, and easily obtained tissue that may prove to be an ideal source of autologous stem cells for engineering tissues.
Collapse
Affiliation(s)
- Daniel A De Ugarte
- Laboratory for Regenerative Bioengineering and Repair, Department of Surgery, Division of Plastic and Reconstructive Surgery, UCLA School of Medicine, 64-140 Center for Health Sciences, 10833 LeConte Avenue, Los Angeles, CA 90095-1665, USA
| | | | | | | |
Collapse
|
5659
|
Huard J, Li Y, Peng H, Fu FH. Gene therapy and tissue engineering for sports medicine. J Gene Med 2003; 5:93-108. [PMID: 12539148 DOI: 10.1002/jgm.344] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Sports injuries usually involve tissues that display a limited capacity for healing. The treatment of sports injuries has improved over the past 10 to 20 years through sophisticated rehabilitation programs, novel operative techniques, and advances in the field of biomechanical research. Despite this considerable progress, no optimal solution has been found for treatment of various sports-related injuries, including muscle injuries, ligament and tendon ruptures, central meniscal tears, cartilage lesions, and delayed bone fracture healing. New biological approaches focus on the treatment of these injuries with growth factors to stimulate and hasten the healing process. Gene therapy using the transfer of defined genes encoding therapeutic proteins represents a promising way to efficiently deliver suitable growth factors into the injured tissue. Tissue engineering, which may eventually be combined with gene therapy, may potentially result in the creation of tissues or scaffolds for regeneration of tissue defects following trauma. In this article we will discuss why gene therapy and tissue engineering are becoming increasingly important in modern orthopaedic sports medicine practice. We then will review recent research achievements in the area of gene therapy and tissue engineering for sports-related injuries, and highlight the potential clinical applications of this technology in the treatment of patients with musculoskeletal problems following sports-related injuries.
Collapse
Affiliation(s)
- Johnny Huard
- University of Pittsburgh, Department of Orthopaedic Surgery, Growth and Development Laboratory, 4151 Rangos Research Center, Pittsburgh, PA 15213, USA. jhuard+@pitt.edu
| | | | | | | |
Collapse
|
5660
|
Winter A, Breit S, Parsch D, Benz K, Steck E, Hauner H, Weber RM, Ewerbeck V, Richter W. Cartilage-like gene expression in differentiated human stem cell spheroids: a comparison of bone marrow-derived and adipose tissue-derived stromal cells. ARTHRITIS AND RHEUMATISM 2003; 48:418-29. [PMID: 12571852 DOI: 10.1002/art.10767] [Citation(s) in RCA: 315] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE To compare the chondrogenic potential of human bone marrow-derived mesenchymal stem cells (BMSC) and adipose tissue-derived stromal cells (ATSC), because the availability of an unlimited cell source replacing human chondrocytes could be strongly beneficial for cell therapy, tissue engineering, in vitro drug screening, and development of new therapeutic options to enhance the regenerative capacity of human cartilage. METHODS Quantitative gene expression of common cartilage and cell interaction molecules was analyzed using complementary DNA array technology and reverse transcription-polymerase chain reaction during optimization of cell differentiation, in order to achieve a molecular phenotype similar to that of chondrocytes in cartilage. RESULTS The multilineage potential of BMSC and ATSC was similar according to cell morphology and histology, but minor differences in marker gene expression occurred in diverse differentiation pathways. Although chondrogenic differentiation of BMSC and ATSC was indistinguishable in monolayer and remained partial, only BMSC responded (with improved chondrogenesis) to a shift to high-density 3-dimensional cell culture, and reached a gene expression profile highly homologous to that of osteoarthritic (OA) cartilage. CONCLUSION Hypertrophy of chondrocytes and high matrix-remodeling activity in differentiated BMSC spheroids and in OA cartilage may be the basis for the strong similarities in gene expression profiles between these samples. Differentiated stem cell spheroids represent an attractive tool for use in drug development and identification of drug targets in OA cartilage-like tissue outside the human body. However, optimization of differentiation protocols to achieve the phenotype of healthy chondrocytes is desired for cell therapy and tissue engineering approaches.
Collapse
Affiliation(s)
- Anja Winter
- Orthopaedic Hospital/University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
5661
|
Abstract
Adult cardiac muscle is unable to repair itself following severe disease or injury. Because of this fundamental property of the myocardium, it was long believed that the adult myocardium is a postmitotic tissue. Yet, recent studies have indicated that new cardiac myocytes are generated throughout the life span of an adult and that extracardiac cells can contribute to the renewal of individual cells within the myocardium. In addition, investigations of the phenotypic capacity of adult stem cells have suggested that their potential is not solely restricted to the differentiated cell phenotypes of the source tissue. These observations have great implications for cardiac biology, as stem cells obtained from the bone marrow and other readily accessible adult tissues may serve as a source of replacement cardiac myocytes. In this review, we describe the evidence for these new findings and discuss their implications in context of the continuing controversy over stem cell plasticity.
Collapse
Affiliation(s)
- Leonard M Eisenberg
- Department of Cell Biology and Anatomy, Medical University of South Carolina, Charleston, South Carolina 29425, USA.
| | | |
Collapse
|
5662
|
Nakahara T, Nakamura T, Tabata Y, Eto K, Shimizu Y. Regeneration of periodontal tissues based on in situ tissue engineering. ACTA ACUST UNITED AC 2003. [DOI: 10.2492/jsir.23.116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
5663
|
Abstract
The stem cells are promising for future cell-based therapy such as tissue engineering or regenerative medicine. Although Embryonic Stem Cells (ESCs) are theoretically highly beneficial, there are various limitations on their use posed by cell regulations and ethical considerations. Therefore, adult stem cells are considered to be highly available with neither ethical nor immunoreactive considerations as long as they are of autologous tissue origin. Much of work has focused on the Mesenchymal Stem Cells (MSCs) isolated from bone marrow stroma, which have been shown to possess adipogenic, osteogenic, chondrogenic, myogenic and neurogenic potential in vitro. However bone marrow procurement is severely painful for patients and the harvested cells yields low number. Our preliminary studies have identified a putative stem cell population isolated from human adipose tissue. This cell population, termed Processed Lipoaspirate Cells (PLA Cells), is found to differentiate into adipogenic, osteogenic, chondrogenic and myogenic lineage in vitro in lineage-specific culture media. In addition to these findings, our recent data shows that PLA cells can be induced to differentiate into neural precursors, which are of an ectodermal origin. Furthermore, PLA cells express multiple CD marker antigens similar to those observed on MSCs. Finally, some of PLA clonal cells have capabilities of differentiate into adipogenic, osteogenic and chondrogenic lineage. These findings suggest that human PLA have a mesodermal stem cell population. Since human adipose tissue is ubiquitous, easily obtainable in large quantity under local anesthesia with little patient discomfort, it may be an alternative stem cell source for mesenchymal tissue regeneration and engineering.
Collapse
Affiliation(s)
- Hiroshi Mizuno
- Department of Plastic and Reconstructive Surgery, Nippon Medical School, Japan.
| |
Collapse
|
5664
|
Tuli R, Seghatoleslami MR, Tuli S, Wang ML, Hozack WJ, Manner PA, Danielson KG, Tuan RS. A simple, high-yield method for obtaining multipotential mesenchymal progenitor cells from trabecular bone. Mol Biotechnol 2003; 23:37-49. [PMID: 12611268 DOI: 10.1385/mb:23:1:37] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In vitro cultures of primary, human trabecular bone-derived cells represent a useful system for investigation of the biology of osteoblasts. Our recent discovery of the multilineage mesenchymal differentiation potential of trabecular bone-derived cells suggests the potential application of these cells as mesenchymal progenitors for tissue repair and regeneration. Such applications are crucially dependent on efficient cell-isolation protocols to yield cells that optimally proliferate and differentiate. In this study, we describe a simple, high-yield procedure, requiring minimal culture expansion, for the isolation of mesenchymal progenitor cells from human trabecular bone. Moreover, these cells retain their ability to differentiate along multiple mesenchymal lineages through successive subculturing. Cell populations isolated and cultured as described here allow the efficient acquisition of a clinically significant number of cells, which may be used as the cell source for tissue-engineering applications.
Collapse
Affiliation(s)
- Richard Tuli
- Cartilage Biology and Orthopedics Branch, 50 South Drive, Rm 1503, MSC 8022, National Institute of Arthritis, and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
5665
|
Lee DH, Kang SK, Lee RH, Ryu JM, Park HY, Choi HS, Bae YC, Suh KT, Kim YK, Jung JS. Effects of peripheral benzodiazepine receptor ligands on proliferation and differentiation of human mesenchymal stem cells. J Cell Physiol 2003; 198:91-9. [PMID: 14584048 DOI: 10.1002/jcp.10391] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The peripheral benzodiazepine receptor (PBR) has been known to have many functions such as a role in cell proliferation, cell differentiation, steroidogenesis, calcium flow, cellular respiration, cellular immunity, malignancy, and apoptosis. However, the presence of PBR has not been examined in mesenchymal stem cells. In this study, we demonstrated the expression of PBR in human bone marrow stromal cells (hBMSCs) and human adipose stromal cells (hATSCs) by RT-PCR and immunocytochemistry. To determine the roles of PBR in cellular functions of human mesenchymal stem cells (hMSCs), effects of diazepam, PK11195, and Ro5-4864 were examined. Adipose differentiation of hMSCs was decreased by high concentration of PBR ligands (50 microM), whereas it was increased by low concentrations of PBR ligands (<10 microM). PBR ligands showed a biphasic effect on glycerol-3-phosphate dehydrogenase (GPDH) activity. High concentration of PBR ligands (from 25 to 75 microM) inhibited proliferation of hMSCs. However, clonazepam, which does not have an affinity to PBR, did not affect adipose differentiation and proliferation of hMSCs. The PBR ligands did not induce cell death in hMSCs. PK11195 (50 microM) and Ro5-5864 (50 microM) induced cell cycle arrest in the G(2)/M phase. These results indicate that PBR ligands play roles in adipose differentiation and proliferation of hMSCs.
Collapse
Affiliation(s)
- D H Lee
- Department of Physiology, College of Medicine, Pusan National University, Pusan, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
5666
|
Romanov YA, Svintsitskaya VA, Smirnov VN. Searching for alternative sources of postnatal human mesenchymal stem cells: candidate MSC-like cells from umbilical cord. Stem Cells 2003; 21:105-10. [PMID: 12529557 DOI: 10.1634/stemcells.21-1-105] [Citation(s) in RCA: 611] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) have the capability for renewal and differentiation into various lineages of mesenchymal tissues. These features of MSCs attract a lot of attention from investigators in the context of cell-based therapies of several human diseases. Despite the fact that bone marrow represents the main available source of MSCs, the use of bone marrow-derived cells is not always acceptable due to the high degree of viral infection and the significant drop in cell number and proliferative/differentiation capacity with age. Thus, the search for possible alternative MSC sources remains to be validated. Umbilical cord blood is a rich source of hematopoietic stem/progenitor cells and does not contain mesenchymal progenitors. However, MSCs circulate in the blood of preterm fetuses and may be successfully isolated and expanded. Where these cells home at the end of gestation is not clear. In this investigation, we have made an attempt to isolate MSCs from the subendothelial layer of umbilical cord vein using two standard methodological approaches: the routine isolation of human umbilical vein endothelial cell protocol and culture of isolated cells under conditions appropriate for bone-marrow-derived MSCs. Our results suggest that cord vasculature contains a high number of MSC-like elements forming colonies of fibroblastoid cells that may be successfully expanded in culture. These MSC-like cells contain no endothelium- or leukocyte-specific antigens but express alpha-smooth muscle actin and several mesenchymal cell markers. Therefore, umbilical cord/placenta stroma could be regarded as an alternative source of MSCs for experimental and clinical needs.
Collapse
Affiliation(s)
- Yuri A Romanov
- Laboratory of Human Stem Cells, Institute of Experimental Cardiology, National Cardiology Research Center of the Russian Ministry of Health, Moscow, Russia.
| | | | | |
Collapse
|
5667
|
Li Y, Kniss DA, Lasky LC, Yang ST. Culturing and differentiation of murine embryonic stem cells in a three-dimensional fibrous matrix. Cytotechnology 2003; 41:23-35. [PMID: 19002959 PMCID: PMC3449760 DOI: 10.1023/a:1024283521966] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Embryonic stem (ES) cells have indefinite self-renewal ability and pluripotency, and can provide a novel cell source for tissue engineering applications. In this study, a murine CCE ES cell line was used to derive hematopoietic cells in a 3-D fibrous matrix. The 3-D matrix was found to maintain the phenotypes of undifferentiated ES cells as indicated by alkaline phosphatase (ALP) activity and stage specific embryonic antigen-1 (SSEA-1) expression. In hematopoietic differentiation, cells from 3-D culture exhibited similar cell cycle distribution and SSEA-1 expression to those in the initial cell population. The Oct-4 expression was significantly down-regulated, which indicated the occurrence of differentiation, although the level was slightly higher than that in Petri dish culture. The expression of c-kit, cell surface marker for hematopoietic progenitor, was higher in the 3-D culture, suggesting a better-directed hematopoietic differentiation. Cells in the 3-D matrix tended to form large aggregates associated with fibers. For large-scale processes, a perfusion bioreactor can be used for both maintenance and differentiation cultures. As compared to the static culture, a higher growth rate and final cell density were resulted from the perfusion bioreactor due to better control of the reactor environment. At the same time, the differentiation capacity of ES cells was preserved in the perfusion culture. The ES cell culture in the fibrous matrix thus can be used as a 3-D model system to study effects of extracellular environment and associated physico-chemical parameters on ES cell maintenance and differentiation.
Collapse
Affiliation(s)
- Yan Li
- Department of Chemical Engineering, The Ohio State University, Columbus, 43210 USA
| | - Douglas A. Kniss
- Laboratory of Perinatal Research, Department of Obstetrics and Gynecology, The Ohio State University, Columbus, 43210 USA
| | - Larry C. Lasky
- Departments of Pathology and Internal Medicine, The Ohio State University, Columbus, 43210 USA
| | - Shang-Tian Yang
- Department of Chemical Engineering, The Ohio State University, Columbus, 43210 USA
| |
Collapse
|
5668
|
Affiliation(s)
- Jeffrey M Gimble
- Pennington Biomedical Research Center, Louisiana State University, 6400 Perkins Road, Baton Rouge, Louisiana 70808, USA.
| | | |
Collapse
|
5669
|
Tuan RS, Boland G, Tuli R. Adult mesenchymal stem cells and cell-based tissue engineering. Arthritis Res Ther 2003; 5:32-45. [PMID: 12716446 PMCID: PMC154434 DOI: 10.1186/ar614] [Citation(s) in RCA: 512] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2002] [Accepted: 11/01/2002] [Indexed: 12/12/2022] Open
Abstract
The identification of multipotential mesenchymal stem cells (MSCs) derived from adult human tissues, including bone marrow stroma and a number of connective tissues, has provided exciting prospects for cell-based tissue engineering and regeneration. This review focuses on the biology of MSCs, including their differentiation potentials in vitro and in vivo, and the application of MSCs in tissue engineering. Our current understanding of MSCs lags behind that of other stem cell types, such as hematopoietic stem cells. Future research should aim to define the cellular and molecular fingerprints of MSCs and elucidate their endogenous role(s) in normal and abnormal tissue functions.
Collapse
Affiliation(s)
- Rocky S Tuan
- National Institute of Arthritis, and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | |
Collapse
|
5670
|
Morizono K, De Ugarte DA, Zhu M, Zuk P, Elbarbary A, Ashjian P, Benhaim P, Chen ISY, Hedrick MH. Multilineage cells from adipose tissue as gene delivery vehicles. Hum Gene Ther 2003; 14:59-66. [PMID: 12573059 DOI: 10.1089/10430340360464714] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
We have characterized a population of mesenchymal progenitor cells from adipose tissue, termed processed lipoaspirate (PLA) cells, which have multilineage potential similar to bone marrow-derived mesenchymal stem cells and are also easily expanded in culture. The primary benefit of using adipose tissue as a source of multilineage progenitor cells is its relative abundance and ease of procurement. We examined the infection of PLA cells with adenoviral, oncoretroviral, and lentiviral vectors. We demonstrate that PLA cells can be transduced with lentiviral vectors at high efficiency. PLA cells maintain transgene expression after differentiation into adipogenic and osteogenic lineages after lentiviral transduction. Therefore, PLA cells and lentiviral vectors may be an efficient combination for use as a therapeutic gene delivery vehicle.
Collapse
Affiliation(s)
- Kouki Morizono
- Microbiology, Immunology, and Molecular Genetics and Medicine, Department of Hematology-Oncology, UCLA AIDS Institute, UCLA School of Medicine, Los Angeles, CA 90024, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
5671
|
Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P, Hedrick MH. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell 2002; 13:4279-95. [PMID: 12475952 PMCID: PMC138633 DOI: 10.1091/mbc.e02-02-0105] [Citation(s) in RCA: 4982] [Impact Index Per Article: 216.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Much of the work conducted on adult stem cells has focused on mesenchymal stem cells (MSCs) found within the bone marrow stroma. Adipose tissue, like bone marrow, is derived from the embryonic mesenchyme and contains a stroma that is easily isolated. Preliminary studies have recently identified a putative stem cell population within the adipose stromal compartment. This cell population, termed processed lipoaspirate (PLA) cells, can be isolated from human lipoaspirates and, like MSCs, differentiate toward the osteogenic, adipogenic, myogenic, and chondrogenic lineages. To confirm whether adipose tissue contains stem cells, the PLA population and multiple clonal isolates were analyzed using several molecular and biochemical approaches. PLA cells expressed multiple CD marker antigens similar to those observed on MSCs. Mesodermal lineage induction of PLA cells and clones resulted in the expression of multiple lineage-specific genes and proteins. Furthermore, biochemical analysis also confirmed lineage-specific activity. In addition to mesodermal capacity, PLA cells and clones differentiated into putative neurogenic cells, exhibiting a neuronal-like morphology and expressing several proteins consistent with the neuronal phenotype. Finally, PLA cells exhibited unique characteristics distinct from those seen in MSCs, including differences in CD marker profile and gene expression.
Collapse
Affiliation(s)
- Patricia A Zuk
- Department of Surgery and Orthopedics, Regenerative Bioengineering and Repair Laboratory, UCLA School of Medicine, Los Angeles, California 90095, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
5672
|
Warren SM, Hedrick MH, Sylvester K, Longaker MT, Chen CM. New directions in bioabsorbable technology. J Neurosurg 2002; 97:481-9. [PMID: 12449205 DOI: 10.3171/spi.2002.97.4.0481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Generating replacement tissues requires an interdisciplinary approach that combines developmental, cell, and molecular biology with biochemistry, immunology, engineering, medicine, and the material sciences. Because basic cues for tissue engineering may be derived from endogenous models, investigators are learning how to imitate nature. Endogenous models may provide the biological blueprints for tissue restoration, but there is still much to learn. Interdisciplinary barriers must be overcome to create composite, vascularized, patient-specific tissue constructs for replacement and repair. Although multistep, multicomponent tissue fabrication requires an amalgamation of ideas, the following review is limited to the new directions in bioabsorbable technology. The review highlights novel bioabsorbable design and therapeutic (gene, protein, and cell-based) strategies currently being developed to solve common spine-related problems.
Collapse
Affiliation(s)
- Stephen M Warren
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts, USA
| | | | | | | | | |
Collapse
|
5673
|
Erices A, Conget P, Rojas C, Minguell JJ. Gp130 activation by soluble interleukin-6 receptor/interleukin-6 enhances osteoblastic differentiation of human bone marrow-derived mesenchymal stem cells. Exp Cell Res 2002; 280:24-32. [PMID: 12372336 DOI: 10.1006/excr.2002.5627] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Interleukin-6 (IL-6) promotes osteodifferentiation in bone-located progenitors; however, it is not known whether this cytokine affects the differentiation of bone marrow-located osteoprogenitors. To address this issue, we prepared human bone marrow-derived mesenchymal stem cells (MSCs), which were characterized by a cell surface phenotype and multipotential nature. It was observed that in the presence of IL-6, MSCs were not differentiated into the osteogenic lineage, as evidenced by a failure to induce alkaline phosphatase activity, an earlier marker of osteodifferentiation. The lack of effect of IL-6 correlates with the observation that MSCs do not express a membrane-bound or soluble IL-6 receptor (sIL-6R). The incompetence of IL-6 was not reversed by the addition of sIL-6R alone or the sIL-6R/IL-6 complex, as it occurs in other IL-6R-negative cells. However, after MSC osteocommittment by dexamethasone, sIL-6R or the sIL-6R/IL-6 complex enhanced alkaline phosphatase activity. The effect of sIL-6R or sIL-6R/IL-6 proved to be dependent on gp130 availability, which is expressed by MSCs, and involves stat-3 phosphorylation. These data suggest that IL-6R deficiency may represent for bone marrow-located mesenchymal progenitors a sort of protective mechanism to escape the osteogenic effect of IL-6, which is produced by the MSC itself as well as by other marrow stromal cells.
Collapse
Affiliation(s)
- Alejandro Erices
- Programa Terapias Génicas y Celulares, INTA, Universidad de Chile, Santiago, Chile.
| | | | | | | |
Collapse
|
5674
|
Kapsa RMI, Wong SHA, Bertoncello I, Quigley AF, Williams B, Sells K, Marotta R, Kita M, Simmons P, Byrne E, Kornberg AJ. CD45 fraction bone marrow cells as potential delivery vehicles for genetically corrected dystrophin loci. Neuromuscul Disord 2002; 12 Suppl 1:S61-6. [PMID: 12206798 DOI: 10.1016/s0960-8966(02)00084-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Targeted correction of mutations in muscle can be delivered by direct i.m. injection of corrective DNA to the dystrophic muscle or by autologous injection of cells that have been genetically corrected after isolation from the individual with the dystrophic muscle. The successful application of chimeraplasty and short fragment homologous replacement to correct the exon 23 nonsense mdx transition at the mouse dys locus has opened up the possibility that with further development, targeted gene correction may have some future application for the treatment of muscular dystrophies. In vitro, application of targeted gene correction at the mdx dys locus results in better correction efficiencies than when applied directly to dystrophic muscle. This suggests that at least for the time being, a strategy involving ex vivo correction may be advantageous over a direct approach for delivery of gene correction to dystrophic muscle. This, particularly in view of recent developments indicating that bone-marrow-derived cells are able to systemically remodel dystrophic muscle, whilst penetration of DNA introduced to muscle is limited to individually injected muscles. Application of targeted gene correction to Duchenne dystrophy needs to account for the fact that about 65% of Duchenne muscular dystrophy cases involve large frame-shift deletion of gene sequence at the dys locus. Traditionally, whilst targeted gene correction is able to restore point mutations entirely, it remains to be seen as to whether a strategy for the 'correction' of frame shift deletions may be engineered successfully. This communication discusses the possibility of applying targeted gene correction to dystrophic muscle in Duchenne dystrophy.
Collapse
Affiliation(s)
- R M I Kapsa
- Melbourne Neuromuscular Research Institute, Clinical Neurosciences, St Vincent's Hospital, Fitzroy, Victoria, 3065, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5675
|
Abstract
Regenerative biology has now been recognized as a new field with certain aims and goals. One direction of this new field is to understand the basic mechanisms by which tissues can be repaired and restored. The other direction examines the possibility of using this basic knowledge to apply it to medicine with the goal to clinically repair damaged tissues. Regeneration of tissues can occur by the differentiation of stem cells (local or non-local) or by the transdifferentiation of local terminally differentiated cells. While the transdifferentiation aspects are old, during the past few years many data have accumulated regarding the existence of stem cells and their participation in tissue renewal. This review will present an overview of the potential of all vertebrate organs to regenerate and of the basic mechanisms involved.
Collapse
Affiliation(s)
- Panagiotis A Tsonis
- Laboratory of Molecular Biology, Department of Biology, University of Dayton, Dayton, OH 45469-2320, USA.
| |
Collapse
|
5676
|
Abstract
Generating replacement tissues requires an interdisciplinary approach that combines developmental, cell, and molecular biology with biochemistry, immunology, engineering, medicine, and the material sciences. Since the basic cues for tissue engineering may be derived from endogenous models, investigators are learning how to imitate nature. Endogenous models may provide the biologic blueprints for tissue restoration, but there is still much to learn. Interdisciplinary barriers must be overcome to create composite, vascularized, patient-specific tissue constructs for replacement and repair. Although multistep, multicomponent tissue fabrication requires an amalgamation of ideas, the following review is limited to the new directions in bioabsorbable technology. The review highlights novel bioabsorbable design and therapeutic (gene, protein, and cell-based) strategies that are currently being developed to solve common spinal problems.
Collapse
Affiliation(s)
- Stephen M Warren
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Mass, USA
| | | | | | | | | |
Collapse
|
5677
|
Abstract
Stem cells are defined by their unique properties of self-renewal and multilineage differentiation. Several decades ago, cells with such developmental plasticity have been identified in the embryo and in the bone marrow of the adult; in other organs, such cells could not be demonstrated. Here, recent findings are briefly summarized indicating that the elementary stem cell capabilities are retained by a limited number of cells present in many organs of the adult. Other data suggest that, on response to another microenvironment, "organ-specific" stem cells are able to acquire different fates. If confirmed these findings will have considerable impact on the future of clinical stem cell therapy.
Collapse
Affiliation(s)
- Robert Keller
- Department of Pathology, Institute of Experimental Immunology, University of Zurich, CH-8091 Zurich, Switzerland
| |
Collapse
|
5678
|
Jiang Y, Vaessen B, Lenvik T, Blackstad M, Reyes M, Verfaillie CM. Multipotent progenitor cells can be isolated from postnatal murine bone marrow, muscle, and brain. Exp Hematol 2002; 30:896-904. [PMID: 12160841 DOI: 10.1016/s0301-472x(02)00869-x] [Citation(s) in RCA: 575] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Recent studies have shown that cells from bone marrow (BM), muscle, and brain may have greater plasticity than previously known. We have identified multipotent adult progenitor cells (MAPC) in postnatal human and rodent BM that copurify with mesenchymal stem cells (MSC). BM MAPC proliferate without senescence and differentiate into mesodermal, neuroectodermal, and endodermal cell types. We hypothesized that cells with characteristics similar to BM MAPC can be selected and cultured from tissues other than BM. MATERIALS AND METHODS BM, whole brain, and whole muscle tissue was obtained from mice. Cells were plated on Dulbecco modified Eagle medium supplemented with 2% fetal calf serum and 10 ng/mL epidermal growth factor (EGF), 10 ng/mL platelet-derived growth factor (PDGF-BB), and 1000 units/mL leukemia inhibitory factor (LIF) for more than 6 months. Cells were maintained between 0.5 and 1.5 x 10(3) cells/cm(2). At variable time points, we tested cell phenotype by FACS and evaluated their differentiation into endothelial cells, neuroectodermal cells, and endodermal cells in vitro. We also compared the expressed gene profile in BM, muscle, and brain MAPC by Affimetrix gene array analysis. RESULTS Cells could be cultured from BM, muscle, and brain that proliferated for more than 70 population doublings (PDs) and were negative for CD44, CD45, major histocompatibility complex class I and II, and c-kit. Cells from the three tissues differentiated to cells with morphologic and phenotypic characteristics of endothelium, neurons, glia, and hepatocytes. The expressed gene profile of cells derived from the three tissues was identical (r(2) > 0.975). CONCLUSIONS This study shows that cells with MAPC characteristics can be isolated not only from BM, but also from brain and muscle tissue. Whether MAPC originally derived from BM are circulating or all organs contain stem cells with MAPC characteristics currently is being studied. Presence of MAPC in multiple tissues may help explain the "plasticity" found in multiple adult tissues.
Collapse
Affiliation(s)
- Yuehua Jiang
- Stem Cell Institute, Department of Medicine, University of Minnesota Medical School, Minneapolis 55455, USA
| | | | | | | | | | | |
Collapse
|
5679
|
Lieberman JR, Ghivizzani SC, Evans CH. Gene transfer approaches to the healing of bone and cartilage. Mol Ther 2002; 6:141-7. [PMID: 12161179 DOI: 10.1006/mthe.2000.0663] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Jay R Lieberman
- Department of Orthopaedic Surgery, The David Geffen School of Medicine at UCLA, Los Angeles, California, 90095, USA
| | | | | |
Collapse
|
5680
|
Vats A, Tolley NS, Polak JM, Buttery LDK. Stem cells: sources and applications. CLINICAL OTOLARYNGOLOGY AND ALLIED SCIENCES 2002; 27:227-32. [PMID: 12169121 DOI: 10.1046/j.1365-2273.2002.00579.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Tissue engineering is a multidisciplinary area of research aimed at regeneration of tissues and restoration of function of organs through implantation of cells/tissues grown outside the body, or stimulating cells to grow into implanted matrix. In this short review, some of the most recent developments in the use of stem cells for tissue repair and regeneration will be discussed. There is no doubt that stem cells derived from adult and embryonic sources hold great therapeutic potential but it is clear that there is still much research required before their use is commonplace. There is much debate over adult versus embryonic stem cells and whether both are required. It is probably too early to disregard one or other of these cell sources. With regard to embryonic stem cells, the major concern relates to the ethics of their creation and the proposed practice of therapeutic cloning.
Collapse
Affiliation(s)
- A Vats
- Tissue Engineering Centre, Imperial College, Chelsea & Westminster Hospital, London, UK.
| | | | | | | |
Collapse
|
5681
|
Kimura Y, Ozeki M, Inamoto T, Tabata Y. Time course of de novo adipogenesis in matrigel by gelatin microspheres incorporating basic fibroblast growth factor. TISSUE ENGINEERING 2002; 8:603-13. [PMID: 12202000 DOI: 10.1089/107632702760240526] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Controlled release of basic fibroblast growth factor (bFGF) from gelatin microspheres achieved de novo adipogenesis at the implanted site of a basement membrane extract (Matrigel). Following subcutaneous co-implantation of Matrigel and gelatin microspheres incorporating 0.1 microg of bFGF into the back of mice, adipose tissue was formed at the implanted site after 4 weeks postoperatively although the extent increased with implantation time. Formation of adipose tissue was significantly faster than the co-implantation of Matrigel, and 0.1 microg of free bFGF while a larger volume of the adipose tissue formed was retained 15 weeks later. When measured in Matrigel co-implanted with the gelatin microspheres incorporating bFGF, the number of cells infiltrated into Matrigel increased to a significantly high extent compared with the bFGF co-implantation. Matrigel alone was much less effective in inducing formation of adipose tissue. We conclude that gelatin microspheres incorporating bFGF enable Matrigel to efficiently induce de novo adipogenesis at the implanted site in respect to the formation rate and volume of adipose tissue.
Collapse
Affiliation(s)
- Yu Kimura
- The Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | | | | | | |
Collapse
|
5682
|
Abstract
Strategies to treat atherosclerotic coronary artery disease include coronary artery bypass grafting (CABG), in which grafts are used to bypass atherosclerotic vessels and restore blood flow to the ischemic myocardium. The grafts used include healthy arteries or veins harvested from a separate site. Results with arterial grafts have been superior to venous grafts; promoting the practice of total arterial revascularization using only arterial grafts. Suitable arterial grafts, however, are scarce and harvest procedures add to morbidity and cost. Tissue engineering combines the principles of engineering with life sciences for the development of biological substitutes and restore, maintain or improve tissue function. Advances in this field have included the development of tissue-engineered blood vessels, with the potential to serve as arterial grafts, conduits or fistulae. This review describes the history of tissue engineering arteries, the techniques used, and progress to date. The source of cells and the future direction of this field are explored.
Collapse
Affiliation(s)
- Briain D MacNeill
- Division of Cardiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | | |
Collapse
|
5683
|
Abstract
Observations made in the last few years support the existence of pathways, in adult humans and rodents, that allow adult stem cells to be surprisingly flexible in their differentiation repertoires. Termed plasticity, this property allows adult stem cells, assumed, until now, to be committed to generating a fixed range of progeny, to switch, when they have been relocated, to make other specialized sets of cells appropriate to their new niche. Reprogramming of some adult stem cells can occur in vivo; the stem cells normally resident in bone marrow appear particularly flexible and are able to contribute usefully to multiple recipient organs. This process produces cells with specialized structural and metabolic adaptations commensurate with their new locations. In a few examples, the degree of support is sufficient to assist or even rescue recipient mice from genetic defects. Some studies provide evidence for the expansion of the reprogrammed cells locally, but in most it remains possible that cells arrive and redifferentiate, but are no longer stem cells. Nevertheless, the fact that appropriately differentiated cells are delivered deep within organs simply by injection of bone marrow cells should make us think differently about the way that organs regenerate and repair. Migratory pathways for stem cells in adult organisms may exist that could be exploited to effect repairs using an individual's own stem cells, perhaps after gene therapy. Logical extensions of this concept are that a transplanted organ would become affected by the genetic susceptibilities of the recipient, alleles that re-express themselves via marrow-derived stem cells, and that plasticity after bone marrow transplantation would also transfer different phenotypes, affecting important parameters such as susceptibility to long-term complications of diabetes, or the ability to metabolize drugs in the liver. This article reviews some of the evidence for stem cell plasticity in rodents and man.
Collapse
Affiliation(s)
- Richard Poulsom
- Histopathology Unit, Cancer Research UK, London Research Institute, London, UK.
| | | | | | | |
Collapse
|
5684
|
Abstract
Since its discovery four decades ago, the satellite cell of skeletal muscle has been implicated as the major source of myogenic cells involved in growth and repair of muscle fibres. This review not only looks at the role of the satellite cell in these processes but discusses how cells derived from other sources and tissues have recently been implicated in muscle formation and regeneration. Muscle itself also yields cells that contribute to other cell lineages although it is currently debated as to whether these cells originate within muscle or have migrated there from other tissues. The reality of using cells from muscle or other tissues to repair diseased muscle fibres is also addressed.
Collapse
Affiliation(s)
- Kirstin Goldring
- Department of Neuromuscular Diseases, Division of Neuroscience and Psychological Medicine, Faculty of Medicine, Imperial College of Science, Technology and Medicine, Charing Cross Campus, St Dunstan's Road, London W6 8RP, UK
| | | | | |
Collapse
|
5685
|
Safford KM, Hicok KC, Safford SD, Halvorsen YDC, Wilkison WO, Gimble JM, Rice HE. Neurogenic differentiation of murine and human adipose-derived stromal cells. Biochem Biophys Res Commun 2002; 294:371-9. [PMID: 12051722 DOI: 10.1016/s0006-291x(02)00469-2] [Citation(s) in RCA: 553] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The identification of cells capable of neuronal differentiation has great potential for cellular therapies. We examined whether murine and human adipose-derived adult stem (ADAS) cells can be induced to undergo neuronal differentiation. We isolated ADAS cells from the adipose tissue of adult BalbC mice or from human liposuction tissue and induced neuronal differentiation with valproic acid, butylated hydroxyanisole, insulin, and hydrocortisone. As early as 1-3 h after neuronal induction, the phenotype of ADAS cells changed towards neuronal morphology. Following neuronal induction, muADAS cells displayed immunocytochemical staining for GFAP, nestin and NeuN and huADAS cells displayed staining for intermediate filament M, nestin, and NeuN. Following neuronal induction of murine and human ADAS cells, Western blot analysis confirmed GFAP, nestin, and NeuN protein expression. Pretreatment with EGF and basic FGF augmented the neuronal differentiation of huADAS cells. The neuronal differentiation of stromal cells from adipose tissue has broad biological and clinical implications.
Collapse
Affiliation(s)
- Kristine M Safford
- Department of Surgery, Division of Pediatric Surgery, Box 3815, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | |
Collapse
|
5686
|
Abstract
Many exciting discoveries reported over the past 3 years have caused us to expand the paradigm for understanding somatic stem cell plasticity. Within adult organs, there are not only specific stem cells that are capable of producing functional cells of one organ system, but also cells with the flexibility to differentiate into multiple other cell types. In the bone marrow, for example, in addition to hematopoietic stem cells and supportive stromal cells, there are cells with the potential to differentiate into mature cells of the heart, liver, kidney, lungs, GI tract, skin, bone, muscle, cartilage, fat, endothelium and brain. A subpopulation of cells in the brain can differentiate into all of the major cell types in the brain and also into hematopoietic and skeletal muscle cells. In this brief overview, several of these recent findings are summarized.
Collapse
Affiliation(s)
- D S Krause
- Yale University School of Medicine, Department of Laboratory Medicine, New Haven, CT 06520-8035, USA
| |
Collapse
|
5687
|
Buzańska L, Machaj EK, Zabłocka B, Pojda Z, Domańska-Janik K. Human cord blood-derived cells attain neuronal and glial features in vitro. J Cell Sci 2002; 115:2131-8. [PMID: 11973354 DOI: 10.1242/jcs.115.10.2131] [Citation(s) in RCA: 174] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neural stem cells are clonogenic, self-renewing cells with the potential to differentiate into brain-specific cell lines. Our study demonstrates that a neural-stem-cell-like subpopulation can be selected and expanded in vitro by the use of human umbilical cord blood cells, which are a relatively easily available starting material. Through a combination of antigen-driven magnetic cell sorting and subfractionation according to cell surface adhesive properties, we have isolated a clonogenic fraction devoid of hematopoietic or angiogenetic properties but with relatively high self-renewal potency. The resulting clones express nestin, a neurofilament protein that is one of the most specific markers of multipotent neural stem cells. In the presence of selected growth factors or in the rat brain co-culture system, the progeny of these cells can be oriented towards the three main neural phenotypes: neurons,astroglia and oligodendroglia. The cells show high commitment (about 30% and 40% of the population) to neuronal and astrocytic fate, respectively. Interestingly, upon differentiation, the neural-type precursor cells of cord blood origin also give rise to a relatively high proportion of oligodendrocytes — 11% of the total population of differentiating cells.
Collapse
Affiliation(s)
- L Buzańska
- Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego St. 02-106 Warsaw, Poland
| | | | | | | | | |
Collapse
|
5688
|
|
5689
|
Abstract
Human trabecular bone-derived cells (HTBs) have been used for many years as osteoblast progenitors. In this study we tested whether HTBs have stem cell characteristics; that is, whether they are pluripotent and able to self-renew. We show that HTBs readily differentiate into osteoblasts, chondrocytes, and adipocytes if subjected to the appropriate differentiating conditions. Importantly, differentiation into these three lineages is maintained in single cell clones derived by limiting dilution, following expansion over more than 20 cumulative population doublings. We conclude that cultures of HTBs are equivalent to cultures of "mesenchymal stem cells" (MSCs) isolated from bone marrow.
Collapse
Affiliation(s)
- V Sottile
- Research, Novartis Pharma AG, CH-4002 Basel, Switzerland
| | | | | | | | | |
Collapse
|
5690
|
Liang L, Bickenbach JR. Somatic epidermal stem cells can produce multiple cell lineages during development. Stem Cells 2002; 20:21-31. [PMID: 11796919 DOI: 10.1634/stemcells.20-1-21] [Citation(s) in RCA: 123] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
It has been demonstrated that several types of somatic stem cells have the remarkable capacity to differentiate into other types of tissues. We demonstrate here that stem cells from the skin, the largest organ of the body, have the capacity to form multiple cell lineages during development. Using our recently developed sorting technique, we isolated viable homogeneous populations of somatic epidermal stem and transient amplifying cells from the skin of 3-day old transgenic mice, who carried the enhanced green fluorescent protein transgene, and injected stem, TA, or unsorted basal epidermal cells into 3.5-day C57BL/6 blastocysts. Only the stem-injected blastocysts produced mice with GFP(+) cells in their tissues. We found GFP(+) cells in ectodermal, mesenchymal, and neural-crest-derived tissues in E13.5 embryos, 13-day-old neonates, and 60-day-old adult mice, proving that epidermal stem cells survived the blastocyst injection and multiplied during development. Furthermore, the injected stem cells altered their epidermal phenotype and expressed the appropriate proteins for the tissues into which they developed, demonstrating that somatic epidermal stem cells have the ability to produce cells of different lineages during development. These data suggest that somatic epidermal stem cells may show a generalized plasticity expected only of embryonic stem cells and that environmental (extrinsic) factors may influence the lineage pathway for somatic stem cells. Thus, the skin could be a source of easily accessible stem cells that are able to be reprogrammed to form multiple cell lineages.
Collapse
Affiliation(s)
- Luchuan Liang
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa 52242, USA
| | | |
Collapse
|
5691
|
|
5692
|
Huang JI, Beanes SR, Zhu M, Lorenz HP, Hedrick MH, Benhaim P. Rat extramedullary adipose tissue as a source of osteochondrogenic progenitor cells. Plast Reconstr Surg 2002; 109:1033-41; discussion 1042-3. [PMID: 11884830 DOI: 10.1097/00006534-200203000-00037] [Citation(s) in RCA: 119] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Human liposuction aspirates contain pluripotent adipose-derived mesodermal stem cells that have previously been shown to differentiate into various mesodermal cell types, including osteoblasts and chondrocytes. To develop an autologous research model of bone and cartilage tissue engineering, the authors sought to determine whether rat inguinal fat pads contain a similar population of osteochondrogenic precursor cells. It was hypothesized that the rat inguinal fat pad contains adipose-derived multipotential cells that resemble human adipose-derived mesodermal stem cells in their osteochondrogenic capacity. To test this, the authors assessed the ability of cells isolated from the rat inguinal fat pad to differentiate into osteoblasts and chondrocytes by a variety of lineage-specific histologic stains. Rat inguinal fat pads were isolated and processed from Sprague-Dawley rats into a fibroblast-like cell population. Cell cultures were placed in pro-osteogenic media containing dexamethasone, ascorbic acid, and beta-glycerol phosphate. Osteogenic differentiation was assessed at 2, 4, and 6 weeks. Alkaline phosphatase activity and von Kossa staining were performed to assess osteoblastic differentiation and the production of a calcified extracellular matrix. Cell cultures were also placed in prochondrogenic conditions and media supplemented with transforming growth factor-beta1, insulin, transferrin, and ascorbic acid. Chondrogenic differentiation was assessed at 2, 7, and 14 days by the presence of positive Alcian blue staining and type II collagen immunohistochemistry. Cells placed in osteogenic conditions changed in structure to a more cuboidal shape, formed bone nodules, stained positively for alkaline phosphatase activity, and secreted calcified extracellular matrix by 2 weeks. Cells placed in chondrogenic conditions formed cartilaginous nodules within 48 hours that stained positively for Alcian blue and type II collagen. The authors identified the rat inguinal fat pad as a source of osteochondrogenic precursors and developed a straightforward technique to isolate osteochondrogenic precursors from a small animal source. This relatively easily obtained source of osteochondrogenic cells from the rat may be useful for study of tissue engineering strategies and the basic science of stem cell biology.
Collapse
Affiliation(s)
- Jerry I Huang
- Laboratory for Regenerative Bioengineering and Repair, Department of Surgery, School of Medicine, University of California Los Angeles, 200 UCLA Medical Plaza, Los Angeles, CA 90095-6902, USA
| | | | | | | | | | | |
Collapse
|
5693
|
Tissue-Engineered Bone Using Mesenchymal Stem Cells and a Biodegradable Scaffold. J Craniofac Surg 2002. [DOI: 10.1097/00001665-200203000-00010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
5694
|
Abstract
Increased participation by the general population in athletic activities leads to increased trauma to bones, joint surfaces, and soft tissues. Management and treatment of these injuries has significantly improved over the past few decades. The application of knowledge gained from basic science research in biology and biomechanics has continuously contributed to that. Biological advances have been made in the field of gene therapy, cell therapy, and tissue engineering. Certainly, the greatest focus is bone and cartilage research that will lead to improved fracture repair in the traumatic injured population, as well as prevention of early osteoarthritic changes in the injured athletic population. In biomechanical research, contributions have been made to further understand kinematic behavior of joints that will lead to improved ligament reconstruction techniques and rehabilitation regimens. Various fixation techniques and several different ligament reconstruction techniques have been studied and validated. In the future, improved understanding of ligament healing, graft incorporation, and revascularization will lead to improved outcome of surgical reconstruction techniques in orthopaedic sports medicine. Exciting research has been performed over the past years and will be reviewed in this article.
Collapse
Affiliation(s)
- Volker Musahl
- Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | |
Collapse
|
5695
|
Caterson EJ, Nesti LJ, Danielson KG, Tuan RS. Human marrow-derived mesenchymal progenitor cells: isolation, culture expansion, and analysis of differentiation. Mol Biotechnol 2002; 20:245-56. [PMID: 11936255 DOI: 10.1385/mb:20:3:245] [Citation(s) in RCA: 176] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
A number of adult mesenchymal tissues contain subpopulations of undifferentiated cells, which retain the capacity to differentiate along multiple lineages. These mesenchymal progenitor cells may be cultured in an undifferentiated state and, when given the appropriate signals, differentiate into an expanding list of several mesenchymal and a few ectodermal derived tissues. The maintenance and propagation of the multipotential nature of these progenitor cell populations are crucially dependent on the isolation protocol, the culture expansion conditions, particularly the properties of the fetal bovine serum supplement in the culture medium. This article describes a method for selection of the appropriate serum lot, and introduces a simplified isolation technique to optimize the yield of progenitor cells that maintain the capability of undergoing multilineage differentiation in response to appropriate cues. Cell populations isolated and culture expanded in this manner, by virtue of their multiple differentiation potential, should serve as ideal candidate cells for tissue engineering applications for the repair and regeneration of tissue damaged by disease and or trauma.
Collapse
Affiliation(s)
- Edward J Caterson
- Cartilage Biology and Orthopedics Branch, National Institute of Arthritis, and Musculoskeletal Skin Diseases, National Institutes of Health, Bethesda, MD 20992-5755, USA
| | | | | | | |
Collapse
|
5696
|
Abstract
Stem cells undergo self-renewal and differentiate into multiple lineages of mature cells. The identification of stem cells in diverse adult tissues and the findings that human embryonic stem cells can be proliferated and differentiated has kindled the imagination of both scientists and the public regarding future stem cell technology. These cells could constitute an unlimited supply of diverse cell types that can be used for cell transplantation or drug discovery. The new options raise several fundamental ethical issues. This review gives an overview of the scientific basis underlying the hope generated by stem cell research and discusses current ethical and funding regulations.
Collapse
Affiliation(s)
- Gesine Paul
- Section for Neuronal Survival, Wallenberg Neuroscience Center, Lund University, BMC A10, 221 84 Lund, Sweden.
| | | | | |
Collapse
|
5697
|
Affiliation(s)
- Linda L. Demer
- From the Departments of Medicine and Physiology, UCLA School of Medicine, Los Angeles, Calif
| |
Collapse
|
5698
|
Boyan BD, Dean DD, Lohmann CH, Niederauer GG, McMillan J, Sylvia VL, Schwartz Z. Cartilage regeneration. Oral Maxillofac Surg Clin North Am 2002; 14:105-16. [DOI: 10.1016/s1042-3699(02)00017-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
5699
|
Abstract
Tissue engineering can be defined as any effort to create or induce the formation of a specific tissue in a specific location through the selection and manipulation of cells, matrices, and biologic stimuli. The biologic concepts and the biochemical and biophysical principles on which these efforts are based have become an exciting and rapidly evolving field of biomedical research. More importantly, tissue engineering is becoming a clinical reality in the practice of orthopaedic surgery, providing patients and physicians with an expanding set of practical tools for effective therapy. New and improved matrices and bioactive factors inevitably will play important roles in the evolution of orthopaedic tissue engineering. However, tissue engineering never can stray far from fundamental biologic principles, and one of these is that cells do all the work. No new tissue forms except through the activity of living cells. No bone graft, no matrix, no growth factor, no cytokine can contribute to the generation or integration of new tissue, except through the influence it has on the behavior of cells. The efficacy of all current clinical tools depends entirely on the cells in the grafted site, particularly the small subset of stem cells and progenitor cells that are capable of generating new tissue. The current authors review a series of key biologic concepts related to the rational design and selection of composites of cells and matrices in contemporary bone grafting and tissue engineering efforts. The functional paradigms of stem cell biology are reviewed, including self renewal, asymmetric and symmetric mitosis, and lineage restriction. Several potential sources for autogenous stem cells for connective tissues are discussed. Finally, a simple mathematical model is introduced as a tool for understanding the functional demands placed on stem cells and progenitors in a graft site and to provide a conceptual framework for the rational design of cell matrix composite grafts.
Collapse
Affiliation(s)
- George F Muschler
- Department of Orthopedic Surgery, The Cleveland Clinic Foundation, OH 44195, USA
| | | |
Collapse
|
5700
|
Abstract
Melanoma research has made great advances in recent years. Particularly in the field of immunology, melanoma researchers have opened new avenues for basic and translational cancer research overall. Emerging research areas such as molecular epidemiology promise to develop a similar leadership role. On the other hand, research in biology, genetics or experimental therapy of melanoma has remained confined to few laboratories and entire research areas are not covered due to lack of researchers and resources. New developments in defining stem cells in skin and bone marrow enable us to develop new concepts for melanoma development and progression. New technologies allow rapid progress but they require close cooperation between laboratories. The field has to better bridge experimental with clinical research and increase communication. Corroboration with advocacy groups should activate the public for increased awareness and funding.
Collapse
Affiliation(s)
- M Herlyn
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA.
| |
Collapse
|