1
|
Moro LG, Guarnier LP, Azevedo MF, Fracasso JAR, Lucio MA, de Castro MV, Dias ML, Lívero FADR, Ribeiro-Paes JT. A Brief History of Cell Culture: From Harrison to Organs-on-a-Chip. Cells 2024; 13:2068. [PMID: 39768159 PMCID: PMC11674496 DOI: 10.3390/cells13242068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/11/2024] [Accepted: 10/20/2024] [Indexed: 01/11/2025] Open
Abstract
This comprehensive overview of the historical milestones in cell culture underscores key breakthroughs that have shaped the field over time. It begins with Wilhelm Roux's seminal experiments in the 1880s, followed by the pioneering efforts of Ross Granville Harrison, who initiated groundbreaking experiments that fundamentally shaped the landscape of cell culture in the early 20th century. Carrel's influential contributions, notably the immortalization of chicken heart cells, have marked a significant advancement in cell culture techniques. Subsequently, Johannes Holtfreter, Aron Moscona, and Joseph Leighton introduced methodological innovations in three-dimensional (3D) cell culture, initiated by Alexis Carrel, laying the groundwork for future consolidation and expansion of the use of 3D cell culture in different areas of biomedical sciences. The advent of induced pluripotent stem cells by Takahashi and Yamanaka in 2006 was revolutionary, enabling the reprogramming of differentiated cells into a pluripotent state. Since then, recent innovations have included spheroids, organoids, and organ-on-a-chip technologies, aiming to mimic the structure and function of tissues and organs in vitro, pushing the boundaries of biological modeling and disease understanding. In this review, we overview the history of cell culture shedding light on the main discoveries, pitfalls and hurdles that were overcome during the transition from 2D to 3D cell culture techniques. Finally, we discussed the future directions for cell culture research that may accelerate the development of more effective and personalized treatments.
Collapse
Affiliation(s)
- Lincoln Gozzi Moro
- Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo—USP, São Paulo 01246-904, Brazil; (L.G.M.); (M.V.d.C.)
| | - Lucas Pires Guarnier
- Department of Genetic, Ribeirão Preto Medical School, University of São Paulo—USP, Ribeirão Preto 14040-904, Brazil;
| | | | | | - Marco Aurélio Lucio
- Graduate Program in Environment and Regional Development, University of Western São Paulo, Presidente Prudente 19050-920, Brazil;
| | - Mateus Vidigal de Castro
- Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo—USP, São Paulo 01246-904, Brazil; (L.G.M.); (M.V.d.C.)
| | - Marlon Lemos Dias
- Precision Medicine Research Center, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro—UFRJ, Rio de Janeiro 21941-630, Brazil;
| | | | - João Tadeu Ribeiro-Paes
- Department of Genetic, Ribeirão Preto Medical School, University of São Paulo—USP, Ribeirão Preto 14040-904, Brazil;
- Laboratory of Genetics and Cell Therapy (GenTe Cel), Department of Biotechnology, São Paulo State University—UNESP, Assis 19806-900, Brazil
| |
Collapse
|
2
|
Marson RF, Regner AP, da Silva Meirelles L. Mesenchymal "stem" cells, or facilitators for the development of regenerative macrophages? Pericytes at the interface of wound healing. Front Cell Dev Biol 2023; 11:1148121. [PMID: 36936686 PMCID: PMC10017474 DOI: 10.3389/fcell.2023.1148121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Cultured mesenchymal stromal cells are among the most used cells in clinical trials. Currently, their potential benefits include provision of mature cell types through differentiation, and secretion of various types of paracrine signaling molecules. Even though research on these cells has spanned some decades now, surprisingly, their therapeutic potential has not been fully translated into clinical practice yet, which calls for further understanding of their intrinsic nature and modes of action. In this review, after discussing pieces of evidence that suggest that some perivascular cells may exhibit mesenchymal stem cell characteristics in vivo, we examine the possibility that subpopulations of perivascular and/or adventitial cells activated after tissue injury behave as MSCs and contribute to the resolution of tissue injury by providing cues for the development of regenerative macrophages at injured sites. Under this perspective, an important contribution of cultured MSCs (or their acellular products, such as extracellular vesicles) used in cell therapies would be to instigate the development of M2-like macrophages that support the tissue repair process.
Collapse
Affiliation(s)
- Renan Fava Marson
- Graduate Program in Cellular and Molecular Biology Applied to Health—PPGBioSaúde, Lutheran University of Brazil, Canoas, Brazil
| | - Andrea Pereira Regner
- Graduate Program in Cellular and Molecular Biology Applied to Health—PPGBioSaúde, Lutheran University of Brazil, Canoas, Brazil
- School of Medicine, Lutheran University of Brazil, Canoas, Brazil
| | - Lindolfo da Silva Meirelles
- Graduate Program in Cellular and Molecular Biology Applied to Health—PPGBioSaúde, Lutheran University of Brazil, Canoas, Brazil
- School of Medicine, Lutheran University of Brazil, Canoas, Brazil
- *Correspondence: Lindolfo da Silva Meirelles, ,
| |
Collapse
|
3
|
|
4
|
Vašíček J, Kováč M, Baláži A, Kulíková B, Tomková M, Olexiková L, Čurlej J, Bauer M, Schnabl S, Hilgarth M, Hubmann R, Shehata M, Makarevich AV, Chrenek P. Combined approach for characterization and quality assessment of rabbit bone marrow-derived mesenchymal stem cells intended for gene banking. N Biotechnol 2019; 54:1-12. [PMID: 31400479 DOI: 10.1016/j.nbt.2019.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 07/16/2019] [Accepted: 08/01/2019] [Indexed: 01/05/2023]
Abstract
Rabbit mesenchymal stem cells (rMSCs) are promising agents for the preservation of genetic biodiversity in domestic rabbit breeds. However, rMSCs must meet certain requirements to be used for cryopreservation in animal gene banks. Currently, there are numerous discrepancies in the published data regarding the rMSC phenotype, which may complicate efforts to evaluate their purity and suitability for reuse after cryopreservation in gene and tissue banks. We propose a combined approach (flow cytometry, PCR, differentiation and ultrastructure studies) for the characterization and recovery of rMSCs after cryopreservation. Flow cytometric analyses of rMSCs confirmed the expression of CD29, CD44, vimentin, desmin and α-SMA. RT-PCR revealed the expression of other markers at the mRNA level (SSEA-4, CD73, CD90, CD105, CD146 and CD166). rMSCs showed efficient multilineage differentiation into adipo-, chondro- and osteogenic lineages, SOX2 expression (pluripotency) and typical MSC morphology and ultrastructure. The confirmed rMSCs were subsequently used for cryopreservation. Efficient recovery of rMSCs after cryogenic freezing was demonstrated by high cell viability, normal ultrastructure of reseeded rMSCs, high expression of CD29 and CD44 and lineage differentiation capacity. The proposed combined approach could be used for characterization, cryopreservation and recovery of rMSCs as genetic resources for native rabbit breeds.
Collapse
Affiliation(s)
- Jaromír Vašíček
- NPPC - Research Institute for Animal Production in Nitra, Hlohovecká 2, 951 41 Lužianky, Slovak Republic; Research Centre AgroBioTech, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovak Republic; Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovak Republic.
| | - Michal Kováč
- Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovak Republic
| | - Andrej Baláži
- NPPC - Research Institute for Animal Production in Nitra, Hlohovecká 2, 951 41 Lužianky, Slovak Republic
| | - Barbora Kulíková
- NPPC - Research Institute for Animal Production in Nitra, Hlohovecká 2, 951 41 Lužianky, Slovak Republic
| | - Mária Tomková
- Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovak Republic
| | - Lucia Olexiková
- NPPC - Research Institute for Animal Production in Nitra, Hlohovecká 2, 951 41 Lužianky, Slovak Republic
| | - Jozef Čurlej
- Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovak Republic
| | - Miroslav Bauer
- NPPC - Research Institute for Animal Production in Nitra, Hlohovecká 2, 951 41 Lužianky, Slovak Republic; Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, Nábrežie mládeže 91, 949 74 Nitra, Slovak Republic
| | - Susanne Schnabl
- Department, of Internal Medicine I, Division of Haematology and Haemostaseology, Comprehensive Cancer Centre Vienna, Drug and Target Screening Unit DTSU, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
| | - Martin Hilgarth
- Department, of Internal Medicine I, Division of Haematology and Haemostaseology, Comprehensive Cancer Centre Vienna, Drug and Target Screening Unit DTSU, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
| | - Rainer Hubmann
- Department, of Internal Medicine I, Division of Haematology and Haemostaseology, Comprehensive Cancer Centre Vienna, Drug and Target Screening Unit DTSU, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
| | - Medhat Shehata
- Department, of Internal Medicine I, Division of Haematology and Haemostaseology, Comprehensive Cancer Centre Vienna, Drug and Target Screening Unit DTSU, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
| | - Alexander V Makarevich
- NPPC - Research Institute for Animal Production in Nitra, Hlohovecká 2, 951 41 Lužianky, Slovak Republic
| | - Peter Chrenek
- NPPC - Research Institute for Animal Production in Nitra, Hlohovecká 2, 951 41 Lužianky, Slovak Republic; Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovak Republic; Faculty of Animal Breeding and Biology, UTP University of Science and Technology, Mazowiecka 28, 85-084 Bydgoszcz, Poland
| |
Collapse
|
5
|
The Influence of Aspiration Volume on the Number of Osteoblastic Progenitors Obtained From Bone Marrow in Children. J Pediatr Orthop 2019; 39:382-386. [PMID: 31305383 DOI: 10.1097/bpo.0000000000000949] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Autologous bone marrow aspirates are utilized to treat various conditions in children. The biological value of bone marrow aspirate depends on the concentration of competent osteoblastic progenitors present in the aspirate. It has been shown in adults that increasing bone marrow aspiration volume beyond 2 mL decreases the concentration of osteoblast progenitor cells because of dilution of the sample with peripheral blood. The effect of varying bone marrow aspiration volumes on the osteoblast cell content has not been determined in children. METHODS In total, 21 children (3 male and 18 female patients, age range 8 mo to 14 y) scheduled for pelvic osteotomy were included in the study. Three separate bone marrow aspirates of 1, 5, and 10 mL were obtained from the anterior superior iliac crest. Total number of nucleated cells was counted per aspirate and the prevalence of alkaline phosphatase-positive colony-forming units was determined per million nucleated cells. RESULTS We measured a significant, proportional increase in the total number of nucleated bone marrow precursor cells between the 1 and 5 mL samples (mean±SD, 27±13 and 152±78 million nucleated cells, respectively; P<0.0001). When the aspiration volume doubled from 5 to 10 mL the total number of nucleated cells was 178±76 million (P=0.17). A proportional increase from 2214 alkaline phosphatase-positive colony-forming units in the 1 mL sample to 14,100 alkaline phosphatase-positive colony-forming units in the 5 mL sample was observed. However, the number of colony-forming units per aspirate decreased to 11,880 in the 10 mL sample. CONCLUSIONS These data demonstrate that in children aspiration up to 5 mL bone marrow from the iliac crest yields a proportional increase in osteoblastic progenitor cells per aspirate. Increasing the aspiration volume beyond 5 mL results in hemodilution, rather than further selection of osteoblastic material. CLINICAL RELEVANCE These data provide clinicians with a guideline for optimizing aspiration volume of bone marrow in children. LEVEL OF EVIDENCE Level II-development of diagnostic criteria on basis of consecutive patients.
Collapse
|
6
|
Wilson A, Webster A, Genever P. Nomenclature and heterogeneity: consequences for the use of mesenchymal stem cells in regenerative medicine. Regen Med 2019; 14:595-611. [PMID: 31115266 PMCID: PMC7132560 DOI: 10.2217/rme-2018-0145] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are in development for many clinical indications, based both on ‘stem’ properties (tissue repair or regeneration) and on signaling repertoire (immunomodulatory and anti-inflammatory effects). Potential conflation of MSC properties with those of tissue-derived stromal cells presents difficulties in comparing study outcomes and represents a source of confusion in cell therapy development. Cultured MSCs demonstrate significant heterogeneity in clonogenicity and multi-lineage differentiation potential. However in vivo biology of MSCs includes native functions unrelated to regenerative medicine applications, so do nomenclature and heterogeneity matter? In this perspective we examine some consequences of the nomenclature debate and heterogeneity of MSCs. Regulatory expectations are considered, emphasizing that product development should prioritize detailed characterization of therapeutic cell populations for specific indications.
Collapse
Affiliation(s)
- Alison Wilson
- Department of Biology, University of York, York YO10 5DD, UK
| | - Andrew Webster
- Science & Technology Studies Unit, Department of Sociology, University of York, York YO10 5DD, UK
| | - Paul Genever
- Department of Biology, University of York, York YO10 5DD, UK
| |
Collapse
|
7
|
Hausherr TC, Nuss K, Thein E, Krähenbühl S, Applegate LA, Pioletti DP. Effect of temporal onsets of mechanical loading on bone formation inside a tissue engineering scaffold combined with cell therapy. Bone Rep 2018; 8:173-179. [PMID: 29955636 PMCID: PMC6020271 DOI: 10.1016/j.bonr.2018.04.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 12/28/2022] Open
Abstract
Several approaches to combine bone substitutes with biomolecules, cells or mechanical loading have been explored as an alternative to the limitation and risk-related bone auto- and allo-grafts. In particular, human bone progenitor cells seeded in porous poly(L-lactic acid)/tricalcium phosphate scaffolds have shown promising results. Furthermore, the application of mechanical loading has long been known to be a key player in the regulation of bone architecture and mechanical properties. Several in vivo studies have pointed out the importance of its temporal offset. When an early mechanical loading was applied a few days after scaffold implantation, it was ineffective on bone formation, whereas a delayed mechanical loading of several weeks was beneficial for bone tissue regeneration. No information is reported to date on the effectiveness of applying a mechanical loading in vivo on cell-seeded scaffold with respect to bone formation in a bone site. In our study, we were interested in human bone progenitor cells due to their low immunogenicity, sensitivity to mechanical loading and capacity to differentiate into osteogenic human bone progenitor cells. The latest capacity allowed us to test two different bone cell fates originating from the same cell type. Therefore, the general aim of this study was to assess the outcome on bone formation when human bone progenitor cells or pre-differentiated osteogenic human bone progenitor cells are combined with early and delayed mechanical loading inside bone tissue engineering scaffolds. Scaffolds without cells, named cell-free scaffold, were used as control. Surprisingly, we found that (1) the optimal solution for bone formation is the combination of cell-free scaffolds and delayed mechanical loading and that (2) the timing of the mechanical application is crucial and dependent on the cell type inside the implanted scaffolds. Bone substitutes can contain osteogenic cells or be mechanically stimulated. Both approaches are simultaneously tested in vivo. The combination of cell-free scaffolds and delayed mechanical loading was optimal. The timing of the mechanical application was crucial and dependent on the seeded cell type.
Collapse
Affiliation(s)
- T C Hausherr
- Laboratory of Biomechanical Orthopedics, Ecole Polytechnique Fédérale de Lausanne, Switzerland
| | - K Nuss
- Musculoskeletal Research Unit, Vetsuisse Faculty, University of Zürich, Switzerland
| | - E Thein
- Orthopedic and Traumatology Department, University Hospital of Lausanne (CHUV), Switzerland
| | - S Krähenbühl
- Regenerative Therapy Unit, Plastic and Reconstructive Surgery, University Hospital of Lausanne (CHUV), Switzerland
| | - L A Applegate
- Regenerative Therapy Unit, Plastic and Reconstructive Surgery, University Hospital of Lausanne (CHUV), Switzerland
| | - D P Pioletti
- Laboratory of Biomechanical Orthopedics, Ecole Polytechnique Fédérale de Lausanne, Switzerland
| |
Collapse
|
8
|
Qin W, Zhao W, Li X, Peng Y, Harlow LM, Li J, Qin Y, Pan J, Wu Y, Ran L, Ke HZ, Cardozo CP, Bauman WA. Mice with sclerostin gene deletion are resistant to the severe sublesional bone loss induced by spinal cord injury. Osteoporos Int 2016; 27:3627-3636. [PMID: 27436301 DOI: 10.1007/s00198-016-3700-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 07/01/2016] [Indexed: 12/11/2022]
Abstract
UNLABELLED Bone loss after spinal cord injury (SCI) is rapid, severe, and refractory to interventions studied to date. Mice with sclerostin gene deletion are resistant to the severe sublesional bone loss induced by SCI, further indicating pharmacological inhibition of sclerostin may represent a promising novel approach to this challenging medical problem. INTRODUCTION The bone loss secondary to spinal cord injury (SCI) is associated with several unique pathological features, including the permanent immobilization, neurological dysfunction, and systemic hormonal alternations. It remains unclear how these complex pathophysiological changes are linked to molecular alterations that influence bone metabolism in SCI. Sclerostin is a key negative regulator of bone formation and bone mass. We hypothesized that sclerostin could function as a major mediator of bone loss following SCI. METHODS To test this hypothesis, 10-week-old female sclerostin knockout (SOST KO) and wild type (WT) mice underwent complete spinal cord transection or laminectomy (Sham). RESULTS At 8 weeks after SCI, substantial loss of bone mineral density was observed at the distal femur and proximal tibia in WT mice but not in SOST KO mice. By μCT, trabecular bone volume of the distal femur was markedly decreased by 64 % in WT mice after SCI. In striking contrast, there was no significant reduction of bone volume in SOST KO/SCI mice compared with SOST KO/sham. Histomorphometric analysis of trabecular bone revealed that the significant reduction in bone formation rate following SCI was observed in WT mice but not in SOST KO mice. Moreover, SCI did not alter osteoblastogenesis of marrow stromal cells in SOST KO mice. CONCLUSION Our findings demonstrate that SOST KO mice were protected from the major sublesional bone loss that invariably follows SCI. The evidence indicates that sclerostin is an important mediator of the marked sublesional bone loss after SCI, and that pharmacological inhibition of sclerostin may represent a promising novel approach to this challenging clinical problem.
Collapse
Affiliation(s)
- W Qin
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Roa, Bronx, NY, 10468, USA.
- Departments of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - W Zhao
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Roa, Bronx, NY, 10468, USA
| | - X Li
- Amgen Inc, Thousand Oaks, CA, USA
| | - Y Peng
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Roa, Bronx, NY, 10468, USA
| | - L M Harlow
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Roa, Bronx, NY, 10468, USA
| | - J Li
- Indiana University Purdue University, Indianapolis, IN, USA
| | - Y Qin
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Roa, Bronx, NY, 10468, USA
| | - J Pan
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Roa, Bronx, NY, 10468, USA
| | - Y Wu
- Departments of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Institute of Gene Engineering Animal Models for Human Diseases, Dalian Medical University, Dalian, China
| | - L Ran
- Institute of Gene Engineering Animal Models for Human Diseases, Dalian Medical University, Dalian, China
| | | | - C P Cardozo
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Roa, Bronx, NY, 10468, USA
- Departments of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - W A Bauman
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Roa, Bronx, NY, 10468, USA
- Departments of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
9
|
Wise JK, Alford AI, Goldstein SA, Stegemann JP. Synergistic enhancement of ectopic bone formation by supplementation of freshly isolated marrow cells with purified MSC in collagen-chitosan hydrogel microbeads. Connect Tissue Res 2016; 57:516-525. [PMID: 26337827 PMCID: PMC4864208 DOI: 10.3109/03008207.2015.1072519] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Bone marrow-derived mesenchymal stem cells (MSC) can differentiate osteogenic lineages, but their tissue regeneration ability is inconsistent. The bone marrow mononuclear cell (BMMC) fraction of adult bone marrow contains a variety of progenitor cells that may potentiate tissue regeneration. This study examined the utility of BMMC, both alone and in combination with purified MSC, as a cell source for bone regeneration. METHODS Fresh BMMC, culture-expanded MSC, and a combination of BMMC and MSC were encapsulated in collagen-chitosan hydrogel microbeads for pre-culture and minimally invasive delivery. Microbeads were cultured in growth medium for 3 days, and then in either growth or osteogenic medium for 17 days prior to subcutaneous injection in the rat dorsum. RESULTS MSC remained viable in microbeads over 17 days in pre-culture, while some of the BMMC fraction were nonviable. After 5 weeks of implantation, microCT and histology showed that supplementation of BMMC with MSC produced a strong synergistic effect on the volume of ectopic bone formation, compared to either cell source alone. Microbeads containing only fresh BMMC or only cultured MSC maintained in osteogenic medium resulted in more bone formation than their counterparts cultured in growth medium. Histological staining showed evidence of residual microbead matrix in undifferentiated samples and indications of more advanced tissue remodeling in differentiated samples. CONCLUSIONS These data suggest that components of the BMMC fraction can act synergistically with predifferentiated MSC to potentiate ectopic bone formation. The microbead system may have utility in delivering desired cell populations in bone regeneration applications.
Collapse
Affiliation(s)
- Joel K. Wise
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Andrea I. Alford
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Steven A. Goldstein
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA,Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
10
|
Benayahu D. The Osteogenic Compartment of Bone Marrow: Cell Biology and Clinical Application. Hematology 2016; 4:427-35. [DOI: 10.1080/10245332.1999.11746469] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Dafna Benayahu
- Department of Cell Biology and Histology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 66978, Israel
| |
Collapse
|
11
|
Bone transplantation and tissue engineering, part IV. Mesenchymal stem cells: history in orthopedic surgery from Cohnheim and Goujon to the Nobel Prize of Yamanaka. INTERNATIONAL ORTHOPAEDICS 2015; 39:807-17. [PMID: 25750132 DOI: 10.1007/s00264-015-2716-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 02/16/2015] [Indexed: 12/13/2022]
Abstract
In 1867 the German pathologist Cohnheim hypothesized that non-hematopoietic, bone marrow-derived cells could migrate through the blood stream to distant sites of injury and participate in tissue regeneration. In 1868, the French physiologist Goujon studied the osteogenic potential of bone marrow on rabbits. Friedenstein demonstrated the existence of a nonhematopoietic stem cell within bone marrow more than a hundred years later. Since this discovery, the research on mesenchymal stem cell (MSC) has explored their therapeutic potential. The prevalent view during the second century was that mature cells were permanently locked into the differentiated state and could not return to a fully immature, pluripotent stem-cell state. Recently, Japanese scientist (first orthopaedist) Shinya Yamanaka proved that introduction of a small set of transcription factors into a differentiated cell was sufficient to revert the cell to a pluripotent state. Yamanaka shared the Nobel Prize in Physiology or Medicine and opened a new door for potential applications of MSCs. This manuscript describes the concept of MSCs from the period when it was relegated to the imagination to the beginning of the twenty-first century and their application in orthopaedic surgery.
Collapse
|
12
|
Nagai H, Kobayashi-Fujioka M, Fujisawa K, Ohe G, Takamaru N, Hara K, Uchida D, Tamatani T, Ishikawa K, Miyamoto Y. Effects of low crystalline carbonate apatite on proliferation and osteoblastic differentiation of human bone marrow cells. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2015; 26:99. [PMID: 25655499 DOI: 10.1007/s10856-015-5431-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 12/09/2014] [Indexed: 06/04/2023]
Abstract
Carbonated apatite (CO3Ap) is the inorganic component of bone. We have proposed a new method for the fabrication of CO3Ap blocks based on a dissolution-precipitation method using a synthetic precursor. The aim of this study is to examine the effects of low crystalline CO3Ap on initial cell attachment, proliferation and osteoblastic differentiation of human bone marrow cells (hBMCs) using sintered hydroxyapatite and tissue culture plates as controls. Initial cell attachment and proliferation were assessed with a MTT assay. Expression of osteoblastic markers was examined by reverse transcription-polymerase chain reaction. XRD and FT-IR results showed formation of B-type carbonate apatite with lower crystallinity. No difference was observed for initial cell attachment between HAp and CO3Ap discs. hBMSC attached more significantly on tissue culture plate than on HAp and CO3Ap discs. The number of cells on HAp was higher than that on CO3Ap until day 7, after which the number of cells was similar. hBMSC proliferated more significantly on tissue culture plate than on HAp and CO3Ap discs. In contrast, hBMCs incubated on CO3Ap demonstrated much higher expression of osteoblastic markers of differentiation, such as type I collagen, alkaline phosphatase, osteopontin and osteocalcin, than hBMCs on HAp. On the tissue culture plate, they were not any change throughout the culture period. These results demonstrated that low crystalline CO3Ap exhibit higher osteoinductivity than HAp.
Collapse
Affiliation(s)
- Hirokazu Nagai
- Subdivision of Molecular Oral Medicine, Division of Integrated Sciences of Translational Research, Department of Oral Surgery, Institute of Health Biosciences, The University of Tokushima Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8504, Japan,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Farrell MJ, Shin JI, Smith LJ, Mauck RL. Functional consequences of glucose and oxygen deprivation on engineered mesenchymal stem cell-based cartilage constructs. Osteoarthritis Cartilage 2015; 23:134-42. [PMID: 25241241 PMCID: PMC4275365 DOI: 10.1016/j.joca.2014.09.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 09/03/2014] [Accepted: 09/05/2014] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Tissue engineering approaches for cartilage repair have focused on the use of mesenchymal stem cells (MSCs). For clinical success, MSCs must survive and produce extracellular matrix in the physiological context of the synovial joint, where low nutrient conditions engendered by avascularity, nutrient utilization, and waste production prevail. This study sought to delineate the role of microenvironmental stressors on MSC viability and functional capacity in three dimensional (3D) culture. DESIGN We evaluated the impact of glucose and oxygen deprivation on the functional maturation of 3D MSC-laden agarose constructs. Since MSC isolation procedures result in a heterogeneous cell population, we also utilized micro-pellet culture to investigate whether clonal subpopulations respond to these microenvironmental stressors in a distinct fashion. RESULTS MSC health and the functional maturation of 3D constructs were compromised by both glucose and oxygen deprivation. Importantly, glucose deprivation severely limited viability, and so compromised the functional maturation of 3D constructs to the greatest extent. The observation that not all cells died suggested there exists heterogeneity in the response of MSC populations to metabolic stressors. Population heterogeneity was confirmed through a series of studies utilizing clonally derived subpopulations, with a spectrum of matrix production and cell survival observed under conditions of metabolic stress. CONCLUSIONS Our findings show that glucose deprivation has a significant impact on functional maturation, and that some MSC subpopulations are more resilient to metabolic challenge than others. These findings suggest that pre-selection of subpopulations that are resilient to metabolic challenge may improve in vivo outcomes.
Collapse
Affiliation(s)
- M J Farrell
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA; Translational Musculoskeletal Research Center, Philadelphia VA Medical Center, Department of Veterans Affairs, Philadelphia, PA 19104, USA
| | - J I Shin
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - L J Smith
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Translational Musculoskeletal Research Center, Philadelphia VA Medical Center, Department of Veterans Affairs, Philadelphia, PA 19104, USA; Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - R L Mauck
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA; Translational Musculoskeletal Research Center, Philadelphia VA Medical Center, Department of Veterans Affairs, Philadelphia, PA 19104, USA.
| |
Collapse
|
14
|
Recovery of neurological function of ischemic stroke by application of conditioned medium of bone marrow mesenchymal stem cells derived from normal and cerebral ischemia rats. J Biomed Sci 2014; 21:5. [PMID: 24447306 PMCID: PMC3922747 DOI: 10.1186/1423-0127-21-5] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 12/23/2013] [Indexed: 01/01/2023] Open
Abstract
Background Several lines of evidence have demonstrated that bone marrow-derived mesenchymal stem cells (BM-MSC) release bioactive factors and provide neuroprotection for CNS injury. However, it remains elusive whether BM-MSC derived from healthy donors or stroke patients provides equal therapeutic potential. The present work aims to characterize BM-MSC prepared from normal healthy rats (NormBM-MSC) and cerebral ischemia rats (IschBM-MSC), and examine the effects of their conditioned medium (Cm) on ischemic stroke animal model. Results Isolated NormBM-MSC or IschBM-MSC formed fibroblastic like morphology and expressed CD29, CD90 and CD44 but failed to express the hematopoietic marker CD34. The number of colony formation of BM-MSC was more abundant in IschBM-MSC than in NormBM-MSC. This is in contrast to the amount of Ficoll-fractionated mononuclear cells from normal donor and ischemic rats. The effect of cm of BM-MSC was further examined in cultures and in middle cerebral artery occlusion (MCAo) animal model. Both NormBM-MSC Cm and IschBM-MSC Cm effectively increased neuronal connection and survival in mixed neuron-glial cultures. In vivo, intravenous infusion of NormBM-MSC Cm and IschBM-MSC Cm after stroke onset remarkably improved functional recovery. Furthermore, NormBM-MSC Cm and IschBM-MSC Cm increased neurogenesis and attenuated microglia/ macrophage infiltration in MCAo rat brains. Conclusions Our data suggest equal effectiveness of BM-MSC Cm derived from ischemic animals or from a normal population. Our results thus revealed the potential of BM-MSC Cm on treatment of ischemic stroke.
Collapse
|
15
|
Kuroda Y, Dezawa M. Mesenchymal stem cells and their subpopulation, pluripotent muse cells, in basic research and regenerative medicine. Anat Rec (Hoboken) 2013; 297:98-110. [PMID: 24293378 DOI: 10.1002/ar.22798] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 09/13/2013] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) have gained a great deal of attention for regenerative medicine because they can be obtained from easy accessible mesenchymal tissues, such as bone marrow, adipose tissue, and the umbilical cord, and have trophic and immunosuppressive effects to protect tissues. The most outstanding property of MSCs is their potential for differentiation into cells of all three germ layers. MSCs belong to the mesodermal lineage, but they are known to cross boundaries from mesodermal to ectodermal and endodermal lineages, and differentiate into a variety of cell types both in vitro and in vivo. Such behavior is exceptional for tissue stem cells. As observed with hematopoietic and neural stem cells, tissue stem cells usually generate cells that belong to the tissue in which they reside, and do not show triploblastic differentiation. However, the scientific basis for the broad multipotent differentiation of MSCs still remains an enigma. This review summarizes the properties of MSCs from representative mesenchymal tissues, including bone marrow, adipose tissue, and the umbilical cord, to demonstrate their similarities and differences. Finally, we introduce a novel type of pluripotent stem cell, multilineage-differentiating stress-enduring (Muse) cells, a small subpopulation of MSCs, which can explain the broad spectrum of differentiation ability in MSCs.
Collapse
Affiliation(s)
- Yasumasa Kuroda
- Department of Anatomy and Anthropology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | |
Collapse
|
16
|
Wise JK, Alford AI, Goldstein SA, Stegemann JP. Comparison of uncultured marrow mononuclear cells and culture-expanded mesenchymal stem cells in 3D collagen-chitosan microbeads for orthopedic tissue engineering. Tissue Eng Part A 2013; 20:210-24. [PMID: 23879621 DOI: 10.1089/ten.tea.2013.0151] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Stem cell-based therapies have shown promise in enhancing repair of bone and cartilage. Marrow-derived mesenchymal stem cells (MSC) are typically expanded in vitro to increase cell number, but this process is lengthy, costly, and there is a risk of contamination and altered cellular properties. Potential advantages of using fresh uncultured bone marrow mononuclear cells (BMMC) include heterotypic cell and paracrine interactions between MSC and other marrow-derived cells including hematopoietic, endothelial, and other progenitor cells. In the present study, we compared the osteogenic and chondrogenic potential of freshly isolated BMMC to that of cultured-expanded MSC, when encapsulated in three-dimensional (3D) collagen-chitosan microbeads. The effect of low and high oxygen tension on cell function and differentiation into orthopedic lineages was also examined. Freshly isolated rat BMMC (25 × 10(6) cells/mL, containing an estimated 5 × 10(4) MSC/mL) or purified and culture-expanded rat bone marrow-derived MSC (2 × 10(5) cells/mL) were added to a 65-35 wt% collagen-chitosan hydrogel mixture and fabricated into 3D microbeads by emulsification and thermal gelation. Microbeads were cultured in control MSC growth media in either 20% O2 (normoxia) or 5% O2 (hypoxia) for an initial 3 days, and then in control, osteogenic, or chondrogenic media for an additional 21 days. Microbead preparations were evaluated for viability, total DNA content, calcium deposition, and osteocalcin and sulfated glycosaminoglycan expression, and they were examined histologically. Hypoxia enhanced initial progenitor cell survival in fresh BMMC-microbeads, but it did not enhance osteogenic potential. Fresh uncultured BMMC-microbeads showed a similar degree of osteogenesis as culture-expanded MSC-microbeads, even though they initially contained only 1/10th the number of MSC. Chondrogenic differentiation was not strongly supported in any of the microbead formulations. This study demonstrates the microbead-based approach to culturing and delivering cells for tissue regeneration, and suggests that fresh BMMC may be an alternative to using culture-expanded MSC for bone tissue engineering.
Collapse
Affiliation(s)
- Joel K Wise
- 1 Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan
| | | | | | | |
Collapse
|
17
|
Han J, Menicanin D, Gronthos S, Bartold PM. Stem cells, tissue engineering and periodontal regeneration. Aust Dent J 2013; 59 Suppl 1:117-30. [PMID: 24111843 DOI: 10.1111/adj.12100] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The aim of this review is to discuss the clinical utility of stem cells in periodontal regeneration by reviewing relevant literature that assesses the periodontal-regenerative potential of stem cells. We consider and describe the main stem cell populations that have been utilized with regard to periodontal regeneration, including bone marrow-derived mesenchymal stem cells and the main dental-derived mesenchymal stem cell populations: periodontal ligament stem cells, dental pulp stem cells, stem cells from human exfoliated deciduous teeth, stem cells from apical papilla and dental follicle precursor cells. Research into the use of stem cells for tissue regeneration has the potential to significantly influence periodontal treatment strategies in the future.
Collapse
Affiliation(s)
- J Han
- Colgate Australian Clinical Dental Research Centre, School of Dentistry, The University of Adelaide, South Australia
| | | | | | | |
Collapse
|
18
|
Tan SL, Ahmad TS, Selvaratnam L, Kamarul T. Isolation, characterization and the multi-lineage differentiation potential of rabbit bone marrow-derived mesenchymal stem cells. J Anat 2013; 222:437-50. [PMID: 23510053 DOI: 10.1111/joa.12032] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2013] [Indexed: 12/23/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are recognized by their plastic adherent ability, fibroblastic-like appearance, expression of specific surface protein markers, and are defined by their ability to undergo multi-lineage differentiation. Although rabbit bone marrow-derived MSCs (rbMSCs) have been used extensively in previous studies especially in translational research, these cells have neither been defined morphologically and ultrastructurally, nor been compared with their counterparts in humans in their multi-lineage differentiation ability. A study was therefore conducted to define the morphology, surface marker proteins, ultrastructure and multi-lineage differentiation ability of rbMSCs. Herein, the primary rbMSC cultures of three adult New Zealand white rabbits (at least 4 months old) were used for three independent experiments. rbMSCs were isolated using the gradient-centrifugation method, an established technique for human MSCs (hMSCs) isolation. Cells were characterized by phase contrast microscopy observation, transmission electron microscopy analysis, reverse transcriptase-polymerase chain reaction (PCR) analysis, immunocytochemistry staining, flow cytometry, alamarBlue(®) assay, histological staining and quantitative (q)PCR analysis. The isolated plastic adherent cells were in fibroblastic spindle-shape and possessed eccentric, irregular-shaped nuclei as well as rich inner cytoplasmic zones similar to that of hMSCs. The rbMSCs expressed CD29, CD44, CD73, CD81, CD90 and CD166, but were negative (or dim positive) for CD34, CD45, CD117 and HLD-DR. Despite having similar morphology and phenotypic expression, rbMSCs possessed significantly larger cell size but had a lower proliferation rate as compared with hMSCs. Using established protocols to differentiate hMSCs, rbMSCs underwent osteogenic, adipogenic and chondrogenic differentiation. Interestingly, differentiated rbMSCs demonstrated higher levels of osteogenic (Runx2) and chondrogenic (Sox9) gene expressions than that of hMSCs (P < 0.05). There was, however, no difference in the adipogenic (Pparγ) expressions between these cell types (P > 0.05). rbMSCs possess similar morphological characteristics to hMSCs, but have a higher potential for osteogenic and chondrogenic differentiation, despite having a lower cell proliferation rate than hMSCs. The characteristics reported here may be used as a comprehensive set of criteria to define or characterize rbMSCs.
Collapse
Affiliation(s)
- Sik-Loo Tan
- Tissue Engineering Group, National Orthopaedics Centre of Excellence in Research & Learning, Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | | | | | | |
Collapse
|
19
|
Bone marrow concentrate for autologous transplantation in minipigs. Characterization and osteogenic potential of mesenchymal stem cells. Vet Comp Orthop Traumatol 2012; 26:34-41. [PMID: 23171924 DOI: 10.3415/vcot-11-11-0165] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 07/16/2012] [Indexed: 12/31/2022]
Abstract
Autologous bone marrow plays an increasing role in the treatment of bone, cartilage and tendon healing disorders. Cell-based therapies display promising results in the support of local regeneration, especially therapies using intra-operative one-step treatments with autologous progenitor cells. In the present study, bone marrow-derived cells were concentrated in a point-of-care device and investigated for their mesenchymal stem cell (MSC) characteristics and their osteogenic potential. Bone marrow was harvested from the iliac crest of 16 minipigs. The mononucleated cells (MNC) were concentrated by gradient density centrifugation, cultivated, characterized by flow cytometry and stimulated into osteoblasts, adipocytes, and chondrocytes. Cell differentiation was investigated by histological and immunohistological staining of relevant lineage markers. The proliferation capacity was determined via colony forming units of fibroblast and of osteogenic alkaline-phosphatase-positive-cells. The MNC could be enriched 3.5-fold in nucleated cell concentrate in comparison to bone marrow. Flow cytometry analysis revealed a positive signal for the MSC markers. Cells could be differentiated into the three lines confirming the MSC character. The cellular osteogenic potential correlated significantly with the percentage of newly formed bone in vivo in a porcine metaphyseal long-bone defect model. This study demonstrates that bone marrow concentrate from minipigs display cells with MSC character and their osteogenic differentiation potential can be used for osseous defect repair in autologous transplantations.
Collapse
|
20
|
Mifune Y, Matsumoto T, Murasawa S, Kawamoto A, Kuroda R, Shoji T, Kuroda T, Fukui T, Kawakami Y, Kurosaka M, Asahara T. Therapeutic superiority for cartilage repair by CD271-positive marrow stromal cell transplantation. Cell Transplant 2012; 22:1201-11. [PMID: 23044363 DOI: 10.3727/096368912x657378] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Recent reports indicated that human isolated CD271+ bone marrow mesenchymal stromal cells (BM-MSCs) have a greater expansion and potential for multipotent differentiation including chondrogenesis than classical plastic adherent (PA) BM-MSCs in vitro. Therefore, we set up a hypothesis that CD271+ MSCs may have a greater chondrogenic potential than PA-MSCs in vitro and in vivo. We investigated the superiority of CD271+ MSCs on chondrogenesis using in vitro expansion and pellet culture system and in vivo rat model of cartilage defect when compared to PA-MSCs. In the in vitro study, CD271+ MSCs showed higher expansion potential and produced larger pellets with higher expressions of chondrogenic genes when compared to the control groups. During the culture, CD271 expression decreased, which resulted in decreased chondrogenesis. In the in vivo study, immunohistochemical staining demonstrated differentiated human chondrocytes identified as double-stained cells with human-specific collagen type 2 and human leukocyte antigen-ABC in CD271+ and PA groups. The number of double-stained cells was significantly higher in the CD271+ group than PA group. Real-time RT-PCR analysis of tissue RNA isolated from the chondral defect site for human-specific chondrogenic markers demonstrated a significantly higher expression in CD271+ group than PA group. Macroscopic examination of chondral defect sites at week 8 revealed glossy white and well-integrated repaired tissues in the CD271+ and PA groups, but not in the PBS group. The average histological score in the CD271+ group was significantly greater than in the other groups. Apoptosis analysis at the cell transplanted site with TUNEL staining showed that the CD271+ group had significantly fewer apoptotic chondrocytes compared with the PA group. These results indicate that CD271+ MSCs have a greater chondrogenic potential than PA-MSCs in both in vitro and in vivo conditions.
Collapse
Affiliation(s)
- Yutaka Mifune
- Stem Cell Translational Research, Kobe Institute of Biomedical Research and Innovation, Kobe, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Muscari C, Bonafè F, Fiumana E, Oranges CM, Pinto V, Caldarera CM, Guarnieri C, Morselli PG. Comparison between stem cells harvested from wet and dry lipoaspirates. Connect Tissue Res 2012; 54:34-40. [PMID: 22853627 DOI: 10.3109/03008207.2012.717130] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Adipose-derived stem cells (ASC) are usually isolated from lipoaspirates, but it is not known if the anesthetic solution injected into adipose tissue affects cell yield and functions. Two different samples were drawn from the abdominal region of female subjects. In the first, a physiological solution containing lidocaine/adrenaline was injected (wet liposuction, WL), while in the contralateral area, the sample was collected without injecting any solution (dry liposuction, DL). The aspirates were processed to investigate the yield of the stromal-vascular fraction (SVF) cells and ASC frequency, growth rate, apoptosis, and differentiation potential. The solid dried mass of fresh WL isolates was lower than that of DL isolates (p < 0.01) due to the presence, in the former, of a liquid solution. As a consequence, the amount of WL-SVF cells was 18.7% lower than those obtained from DL (p < 0.01); this difference was also observed under culture conditions. In addition, the number of colony-forming unit-fibroblasts (CFU-Fs) obtained from 1 × 10(3) SVF cells was 25.5% lower in WL-aspirates than DL-aspirates (p < 0.05) owing, at least in part, to the observed presence of ASC [corrected] in the liquid solution of the WL isolates. After WL and DL, no differences were observed in ASC growth rate, apoptosis, or differentiation potential toward adipogenic, osteogenic, and endothelial cell lineages. In conclusion, WL yields about 40% fewer ASC than DL due to the combined effect of tissue dilution and the reduced frequency of ASC in the SVF. The main biological features of ASC are suitable for cell-based therapies.
Collapse
Affiliation(s)
- Claudio Muscari
- Department of Biochemistry, University of Bologna, Bologna, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Wise JK, Sumner DR, Virdi AS. Modulation of stromal cell-derived factor-1/CXC chemokine receptor 4 axis enhances rhBMP-2-induced ectopic bone formation. Tissue Eng Part A 2012; 18:860-9. [PMID: 22035136 DOI: 10.1089/ten.tea.2011.0187] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Enhancement of in vivo mobilization and homing of endogenous mesenchymal stem cells (MSCs) to an injury site is an innovative strategy for improvement of bone tissue engineering and repair. The present study was designed to determine whether mobilization by AMD3100 and/or local homing by delivery of stromal cell-derived factor-1 (SDF-1) enhances recombinant human bone morphogenetic protein-2 (rhBMP-2) induced ectopic bone formation in an established rat model. Rats received an injection of either saline or AMD3100 treatment 1 h before harvesting of bone marrow for in vitro colony-forming unit-fibroblasts (CFU-F) culture or the in vivo subcutaneous implantation of absorbable collagen sponges (ACSs) loaded with saline, recombinant human bone morphogenetic protein-2 (rhBMP-2), SDF-1, or the combination of SDF-1 and rhBMP-2. AMD3100 treatment resulted in a significant decrease in CFU-F number, compared with saline, which confirmed that a single systemic AMD3100 treatment rapidly mobilized MSCs from the bone marrow. At 28 and 56 days, bone formation in the explanted ACS was assessed by microcomputed tomography (μCT) and histology. At 28 days, AMD3100 and/or SDF-1 had no statistically significant effect on bone volume (BV) or bone mineral content (BMC), but histology revealed more active bone formation with treatment of AMD3100, loading of SDF-1, or the combination of both AMD3100 and SDF-1, compared with saline-treated rhBMP-2 loaded ACS. At 56 days, the addition of AMD3100 treatment, loading of SDF-1, or the combination of both resulted in a statistically significant stimulatory effect on BV and BMC, compared with the saline-treated rhBMP-2 loaded ACS. Histology of the 56-day ACS were consistent with the μCT analysis, exhibiting more mature and mineralized bone formation with AMD3100 treatment, SDF-1 loading, or the combination of both, compared with the saline-treated rhBMP-2 loaded ACS. The present study is the first that provides evidence of the efficacy of AMD3100 and SDF-1 treatment to stimulate trafficking of MSCs to an ectopic implant site, in order to ultimately enhance rhBMP-2 induced long-term bone formation.
Collapse
Affiliation(s)
- Joel K Wise
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, Illinois 60612, USA
| | | | | |
Collapse
|
23
|
Otto WR, Sarraf CE. Culturing and differentiating human mesenchymal stem cells for biocompatible scaffolds in regenerative medicine. Methods Mol Biol 2012; 806:407-426. [PMID: 22057467 DOI: 10.1007/978-1-61779-367-7_27] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Mesenchymal stem cells from a variety of sites are a natural resource that using appropriate skills can be cultured in the laboratory, in scaffolds, to provide differentiated-cell replacement tissues, for clinical application. To perform such work with human cells, strict ethical integrity must be observed at all stages. Adipocytes, osteocytes and chrondrocytes are amongst the most desirable end-point cells. Hydrolytic degradable scaffolds allow implanted cells to synthesise their own extracellular matrix in situ after implantation, degeneration of the foreign scaffold to temporally match creation of the new innate one. For preliminary in vitro stem cell differentiation protocols, initial investigation is commonly performed with stem cells in commercially available porous collagen sponges or cell-free small intestinal submucosa. Differentiation of stem cells to a specific phenotype is achieved by culturing them in apposite culture media under precise conditions. Once the cells have differentiated, they are checked and characterised in a wide variety of systems. This chapter describes differentiation media for adipocytes, osteocytes, chondrocytes, myocytes and neural precursors and methods of observing their characteristics by microscopy using phase contrast microscopy, standard light microscopy and electron microscopy with tinctorial, immunocytochemical and electron dense stains, respectively. Cell sorting techniques are not dealt with here. Immunocytochemistry/microscopy staining for specific differentiated-cell antigens is an invaluable procedure, and the range of commercially available antibodies is wide. Precautions need to be considered for using actively proliferating cells in vivo, so that implanted cells remain controlled by the body's molecular signals and avoid development of malignancy.
Collapse
Affiliation(s)
- William R Otto
- Histopathology Laboratory, Cancer Research UK, London, UK
| | | |
Collapse
|
24
|
Wenceslau CV, Miglino MA, Martins DS, Ambrósio CE, Lizier NF, Pignatari GC, Kerkis I. Mesenchymal Progenitor Cells from Canine Fetal Tissues: Yolk Sac, Liver, and Bone Marrow. Tissue Eng Part A 2011; 17:2165-76. [DOI: 10.1089/ten.tea.2010.0678] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Cristiane Valverde Wenceslau
- Departments of Surgery and Pathology, University of São Paulo, São Paulo, Brazil
- National Institute of Science and Technology in Stem Cell and Cell Therapy (INCTC), Ribeirão Preto, Brazil
| | - Maria Angélica Miglino
- Departments of Surgery and Pathology, University of São Paulo, São Paulo, Brazil
- National Institute of Science and Technology in Stem Cell and Cell Therapy (INCTC), Ribeirão Preto, Brazil
| | - Daniele Santos Martins
- Faculty of Animal Sciences and Food Engineering, University of São Paulo, São Paulo, Brazil
| | - Carlos Eduardo Ambrósio
- National Institute of Science and Technology in Stem Cell and Cell Therapy (INCTC), Ribeirão Preto, Brazil
- Faculty of Animal Sciences and Food Engineering, University of São Paulo, São Paulo, Brazil
| | | | | | - Irina Kerkis
- Faculty of Animal Sciences and Food Engineering, University of São Paulo, São Paulo, Brazil
- Laboratory of Genetics, Butantan Institute, São Paulo, Brazil
| |
Collapse
|
25
|
Kuroda Y, Kitada M, Wakao S, Dezawa M. Bone marrow mesenchymal cells: how do they contribute to tissue repair and are they really stem cells? Arch Immunol Ther Exp (Warsz) 2011; 59:369-78. [PMID: 21789625 DOI: 10.1007/s00005-011-0139-9] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Accepted: 03/07/2011] [Indexed: 01/01/2023]
Abstract
Adult stem cells typically generate the cell types of the tissue in which they reside, and thus the range of their differentiation is considered limited. Bone marrow mesenchymal stem cells (MSCs) are different from other somatic stem cells in that they differentiate not only into the same mesodermal-lineage such as bone, cartilage, and adipocytes but also into other lineages of ectodermal and endodermal cells. Thus, MSCs are a unique type of adult stem cells. In addition, MSCs home to damaged sites, differentiate into cells specific to the tissue and contribute to tissue repair. Therefore, application of MSCs in the treatment of various diseases, including liver dysfunction, myocardial infarction, and central nervous system repair, has been initiated. Because MSCs are generally harvested as adherent cells from bone marrow aspirates, however, they comprise heterogeneous cell populations and their wide-ranging differentiation ability and repair functions are not yet clear. Recent evidence suggests that a very small subpopulation of cells that assume a repair function with the ability to differentiate into trilineage cells resides among human MSCs and effective utilization of such cells is expected to improve the repair effect of MSCs. This review summarizes recent advances in the clarification of MSC properties and discusses future perspectives.
Collapse
Affiliation(s)
- Yasumasa Kuroda
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Seiryo-machi, Aoba-ku, Sendai, Japan.
| | | | | | | |
Collapse
|
26
|
Dai LJ, Moniri MR, Zeng ZR, Zhou JX, Rayat J, Warnock GL. Potential implications of mesenchymal stem cells in cancer therapy. Cancer Lett 2011; 305:8-20. [PMID: 21396770 DOI: 10.1016/j.canlet.2011.02.012] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 02/13/2011] [Accepted: 02/15/2011] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cells (MSCs) are the first type of stem cells to be utilized in clinical regenerative medicine, mainly owing to their capacity for multipotent differentiation and the feasibility of autologous transplantation. More recently, the specific tumor-oriented migration and incorporation of MSCs have been demonstrated in various pre-clinical models, highlighting the potential for MSCs to be used as an ideal carrier for anticancer gene delivery. Engineered with specific anticancer genes, MSCs possess the ability of dual-targeting tumor cells. This contrasts with non-engineered native MSCs which have intrinsic pro- and anti-tumorigenic properties. Engineered MSCs are capable of producing specific anticancer agents locally and constantly. Astute investigation on engineered MSCs may lead to a new avenue toward an efficient therapy for patients with cancer.
Collapse
Affiliation(s)
- Long-Jun Dai
- Department of Surgery, University of British Columbia, Vancouver, Canada.
| | | | | | | | | | | |
Collapse
|
27
|
Peister A, Woodruff MA, Prince JJ, Gray DP, Hutmacher DW, Guldberg RE. Cell sourcing for bone tissue engineering: amniotic fluid stem cells have a delayed, robust differentiation compared to mesenchymal stem cells. Stem Cell Res 2011; 7:17-27. [PMID: 21531647 DOI: 10.1016/j.scr.2011.03.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 03/08/2011] [Accepted: 03/08/2011] [Indexed: 12/16/2022] Open
Abstract
Cell based therapies for bone regeneration are an exciting emerging technology, but the availability of osteogenic cells is limited and an ideal cell source has not been identified. Amniotic fluid-derived stem cells (AFS) and bone-marrow derived mesenchymal stem cells (MSCs) were compared to determine their osteogenic differentiation capacity in both 2D and 3D environments. In 2D culture, the AFS cells produced more mineralized matrix but delayed peaks in osteogenic markers. Cells were also cultured on 3D scaffolds constructed of poly-ε-caprolactone for 15 weeks. MSCs differentiated more quickly than AFS cells on 3D scaffolds, but mineralized matrix production slowed considerably after 5 weeks. In contrast, the rate of AFS cell mineralization continued to increase out to 15 weeks, at which time AFS constructs contained 5-fold more mineralized matrix than MSC constructs. Therefore, cell source should be taken into consideration when used for cell therapy, as the MSCs would be a good choice for immediate matrix production, but the AFS cells would continue robust mineralization for an extended period of time. This study demonstrates that stem cell source can dramatically influence the magnitude and rate of osteogenic differentiation in vitro.
Collapse
Affiliation(s)
- Alexandra Peister
- Department of Biology, Morehouse College, 830 Westview Dr. SW, Atlanta, GA 30314, USA.
| | | | | | | | | | | |
Collapse
|
28
|
Jäger M, Herten M, Fochtmann U, Fischer J, Hernigou P, Zilkens C, Hendrich C, Krauspe R. Bridging the gap: bone marrow aspiration concentrate reduces autologous bone grafting in osseous defects. J Orthop Res 2011; 29:173-80. [PMID: 20740672 DOI: 10.1002/jor.21230] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Accepted: 07/01/2010] [Indexed: 02/04/2023]
Abstract
Although autologous bone grafting represents an effective tool to induce osteogenic regeneration in local bone defects or pseudarthroses, it is associated with significant donor site morbidity and limited by the amount available for grafting. We investigate the potency of bone marrow aspiration concentrate (BMAC) to augment bone grafting and support bone healing. The functional and radiographic outcome of 39 patients with volumetric bone deficiencies treated with BMAC are presented and evaluated in a prospective clinical trial. A collagen sponge (Col) served as scaffold in 12 patients and a bovine hydroxyapatite (HA) was applied in the other 27 individuals. The minimal follow-up was 6 months. Clinical and radiographic findings were completed by in vitro data. All patients showed new bone formation in radiographs during follow-up. However, two patients underwent revision surgery due to a lack in bone healing. In contrast to the Col group, the postoperative bone formation appeared earlier in the HA group (HA group: 6.8 weeks vs. Col group 13.6 weeks). Complete bone healing was achieved in the HA group after 17.3 weeks compared to 22.4 weeks in the Col group. The average concentration factor of BMAC was 5.2 (SD 1.3). Flow cytometry confirmed the mesenchymal nature of the cells. Cells from BMAC created earlier and larger colonies of forming units fibroblasts (CFU-F) compared to cells from bone marrow aspirate. BMAC combined with HA can reduce the time needed for healing of bone defects when compared to BMAC in combination with collagen sponge.
Collapse
Affiliation(s)
- Marcus Jäger
- Research Laboratory for Regenerative Medicine and Biomaterials, Department of Orthopaedics, Heinrich-Heine University Medical School, Moorenstr. 5, D-40225 Duesseldorf, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Habold C, Momken I, Ouadi A, Bekaert V, Brasse D. Effect of prior treatment with resveratrol on density and structure of rat long bones under tail-suspension. J Bone Miner Metab 2011; 29:15-22. [PMID: 20458604 DOI: 10.1007/s00774-010-0187-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Accepted: 04/01/2010] [Indexed: 01/26/2023]
Abstract
Physical inactivity during space flight or prolonged bed rest causes rapid and marked loss of bone mass in humans. Resveratrol, a red wine polyphenol that is currently under study for its therapeutic antioxidant properties, has been shown to significantly modulate biomarkers of bone metabolism, i.e., to promote osteoblast differentiation and to prevent bone loss induced by estrogen deficiency. However, there is no direct evidence supporting its inhibitory effect toward bone loss during physical inactivity. In the present study, effects of resveratrol on bone mineral density (BMD), bone mineral content, and bone structure were examined in the femora and tibiae of tail-suspended and unsuspended rats using X-ray micro-computed tomography (micro-CT). Rats were treated with 400 mg/kg/day of resveratrol for 45 days and half of them were suspended during the last 2 weeks of treatment. Suspension caused a decrease in tibial and femoral BMD and deterioration of trabecular and cortical bone. Bone deterioration during suspension was paralleled by increased bone marrow area, which could be caused by an increase in stromal cells with osteoclastogenic potential or in adipocytes. Resveratrol had a preventive effect against bone loss induced by hindlimb immobilization. In particular, trabecular bone in the proximal tibial metaphysis was totally preserved in rats treated with resveratrol before tail-suspension.
Collapse
Affiliation(s)
- Caroline Habold
- Institut Pluridisciplinaire Hubert Curien, CNRS UMR 7178, Université de Strasbourg, 23 rue du Loess, Strasbourg Cedex 2, France.
| | | | | | | | | |
Collapse
|
30
|
Ji Y, Christopherson GT, Kluk MW, Amrani O, Jackson WM, Nesti LJ. Heterotopic Ossification Following Musculoskeletal Trauma: Modeling Stem and Progenitor Cells in Their Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 720:39-50. [DOI: 10.1007/978-1-4614-0254-1_4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
31
|
Charbord P. Bone marrow mesenchymal stem cells: historical overview and concepts. Hum Gene Ther 2010; 21:1045-56. [PMID: 20565251 DOI: 10.1089/hum.2010.115] [Citation(s) in RCA: 303] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
This review describes the historical emergence of the concept of bone marrow mesenchymal stem cells (MSCs), summarizing data on Wolf and Trentin's hematopoietic inductive microenvironment; Dexter's hematopoiesis-supportive stromal cells; Friedenstein's osteogenic cells; and Pittenger's trilineal osteoblastic, chondrocytic, and adipocytic precursors; to finally introduce the specific bone marrow mesenchymal stem cells with differentiation potential to four lineages (mesenchymal and vascular smooth muscle lineages), and stromal and immunomodulatory capacities. Two points are the object of detailed discussion. The first point envisions the stem cell attributes (multipotentiality, self-renewal, tissue regeneration, population heterogeneity, plasticity, and lineage priming) compared with that of the paradigmatic hematopoietic stem cell. In the second point, we discuss the possible existence of bone marrow cells with greater differentiation potential, eventually pluripotential cells. The latter point raises the issues of cell fusion, reprogramming, or selection under nonstandardized conditions of rare populations of neuroectodermal origin, or of cells that had undergone mesenchymal-to-epithelial transition. In the last section, we review data on MSC senescence and possible malignant transformation secondary to extensive culture, gene transfer of telomerase, or mutations such as leading to Ewing's sarcoma. The set of data leads to the conclusion that bone marrow MSCs constitute a specific adult tissue stem cell population. The multiple characteristics of this stem cell type account for the versatility of the mechanisms of injured tissue repair. Although MSC administration may be extremely useful in a number of clinical applications, their transplantation is not without risks that must not be overlooked when developing cell therapy protocols.
Collapse
Affiliation(s)
- Pierre Charbord
- Institut National de la Recherche et Santé Médicale U, Université Paris XI, Kremlin Bicêtre, France.
| |
Collapse
|
32
|
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that are being clinically explored as a new therapeutic for treating a variety of immune-mediated diseases. First heralded as a regenerative therapy for skeletal tissue repair, MSCs have recently been shown to modulate endogenous tissue and immune cells. Preclinical studies of the mechanism of action suggest that the therapeutic effects afforded by MSC transplantation are short-lived and related to dynamic, paracrine interactions between MSCs and host cells. Therefore, representations of MSCs as drug-loaded particles may allow for pharmacokinetic models to predict the therapeutic activity of MSC transplants as a function of drug delivery mode. By integrating principles of MSC biology, therapy, and engineering, the field is armed to usher in the next generation of stem cell therapeutics.
Collapse
Affiliation(s)
- Biju Parekkadan
- Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Shriners Hospital for Children, and Harvard Medical School, Boston, Massachusetts 02114, USA.
| | | |
Collapse
|
33
|
Teixeira CC, Liu Y, Thant LM, Pang J, Palmer G, Alikhani M. Foxo1, a novel regulator of osteoblast differentiation and skeletogenesis. J Biol Chem 2010; 285:31055-65. [PMID: 20650891 DOI: 10.1074/jbc.m109.079962] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Skeletogenesis depends on the activity of bone-forming cells derived from mesenchymal cells. The pathways that control mesenchymal cell differentiation are not well understood. We propose that Foxo1 is an early molecular regulator during mesenchymal cell differentiation into osteoblasts. In mouse embryos, Foxo1 expression is higher in skeletal tissues, while Foxo1 silencing has a drastic impact on skeletogenesis and craniofacial development, specially affecting pre-maxilla, nasal bone, mandible, tibia, and clavicle. Similarly, Foxo1 activity and expression increase in mouse mesenchymal cells under the influence of osteogenic stimulants. In addition, silencing Foxo1 blocks the expression of osteogenic markers such as Runx2, alkaline phosphatase, and osteocalcin and results in decreased culture calcification even in the presence of strong osteogenic stimulants. Conversely, the expression of these markers increases significantly in response to Foxo1 overexpression. One mechanism through which Foxo1 affects mesenchymal cell differentiation into osteoblasts is through regulation of a key osteogenic transcription factor, Runx2. Indeed, our results show that Foxo1 directly interacts with the promoter of Runx2 and regulates its expression. Using a tibia organ culture model, we confirmed that silencing Foxo1 decreases the expression of Runx2 and impairs bone formation. Furthermore, our data reveals that Runx2 and Foxo1 interact with each other and cooperate in the transcriptional regulation of osteoblast markers. In conclusion, our in vitro, ex vivo, and in vivo results strongly support the notion that Foxo1 is an early molecular regulator in the differentiation of mesenchymal cells into osteoblast.
Collapse
Affiliation(s)
- Cristina C Teixeira
- Department of Orthodontics, New York University College of Dentistry, New York, New York 10010, USA
| | | | | | | | | | | |
Collapse
|
34
|
Jin HJ, Nam HY, Bae YK, Kim SY, Im IR, Oh W, Yang YS, Choi SJ, Kim SW. GD2 expression is closely associated with neuronal differentiation of human umbilical cord blood-derived mesenchymal stem cells. Cell Mol Life Sci 2010; 67:1845-58. [PMID: 20165901 PMCID: PMC11115935 DOI: 10.1007/s00018-010-0292-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2009] [Revised: 01/08/2010] [Accepted: 01/26/2010] [Indexed: 01/09/2023]
Abstract
GD2 ganglioside has been identified as a key determinant of bone marrow-derived mesenchymal stem cells (BM-MSCs). Here, we characterized GD2 ganglioside expression and its implications in umbilical cord blood-derived MSCs (UCB-MSCs). Using immune-selection analysis, we showed that both GD2-positive and GD2-negative UCB-MSCs expressed general stem cell markers and possessed mesodermal lineage differentiation potential. Although the fraction of GD2-expressing cells was lower in UCB-MSC than in BM-MSC populations, inhibition of GD2 synthesis in UCB-MSCs suppressed neuronal differentiation and down-regulated basic helix-loop-helix (bHLH) transcription factors, which are involved in early stage neuronal differentiation. In addition, the levels of bHLH factors in neuronally induced UCB-MSCs were significantly higher in GD2-positive than GD2-negative cells. Our data demonstrate that GD2 ganglioside expression is associated with regulation of bHLH factors and identify neurogenic-capable UCB-MSCs, providing new insights into the potential clinical applications of MSC-based therapy.
Collapse
Affiliation(s)
- Hye Jin Jin
- Biomedical Research Institute, Medipost Co., Ltd, 1571-17 Seocho-3 dong, Seocho-Gu, Seoul, 136-736 Korea
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, 388-1 Pungnap-2 dong, Songpa-Gu, Seoul, 136-736 Korea
| | - Hae Yun Nam
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, 388-1 Pungnap-2 dong, Songpa-Gu, Seoul, 136-736 Korea
| | - Yun Kyong Bae
- Biomedical Research Institute, Medipost Co., Ltd, 1571-17 Seocho-3 dong, Seocho-Gu, Seoul, 136-736 Korea
| | - Soo Yeon Kim
- Mercersburg Academy, 300 East Seminary Street, Mercersburg, PA 17236 USA
| | - I. Rang Im
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, 388-1 Pungnap-2 dong, Songpa-Gu, Seoul, 136-736 Korea
| | - Wonil Oh
- Biomedical Research Institute, Medipost Co., Ltd, 1571-17 Seocho-3 dong, Seocho-Gu, Seoul, 136-736 Korea
| | - Yoon Sun Yang
- Biomedical Research Institute, Medipost Co., Ltd, 1571-17 Seocho-3 dong, Seocho-Gu, Seoul, 136-736 Korea
| | - Soo Jin Choi
- Biomedical Research Institute, Medipost Co., Ltd, 1571-17 Seocho-3 dong, Seocho-Gu, Seoul, 136-736 Korea
| | - Seong Who Kim
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, 388-1 Pungnap-2 dong, Songpa-Gu, Seoul, 136-736 Korea
| |
Collapse
|
35
|
Isenmann S, Arthur A, Zannettino ACW, Turner JL, Shi S, Glackin CA, Gronthos S. TWIST family of basic helix-loop-helix transcription factors mediate human mesenchymal stem cell growth and commitment. Stem Cells 2010; 27:2457-68. [PMID: 19609939 DOI: 10.1002/stem.181] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The TWIST family of basic helix-loop-helix transcription factors, Twist-1 and Dermo-1 are known mediators of mesodermal tissue development and contribute to correct patterning of the skeleton. In this study, we demonstrate that freshly purified human bone marrow-derived mesenchymal stromal/stem cells (MSC) express high levels of Twist-1 and Dermo-1 which are downregulated following ex vivo expansion. Enforced expression of Twist-1 or Dermo-1 in human MSC cultures increased expression of the MSC marker, STRO-1, and the early osteogenic transcription factors, Runx2 and Msx2. Conversely, overexpression of Twist-1 and Dermo-1 was associated with a decrease in the gene expression of osteoblast-associated markers, bone morphogenic protein-2, bone sialoprotein, osteopontin, alkaline phosphatase and osteocalcin. High expressing Twist-1 or Dermo-1 MSC lines exhibited an enhanced proliferative potential of approximately 2.5-fold compared with control MSC populations that were associated with elevated levels of Id-1 and Id-2 gene expression. Functional studies demonstrated that high expressing Twist-1 and Dermo-1 MSC displayed a decreased capacity for osteo/chondrogenic differentiation and an enhanced capacity to undergo adipogenesis. These findings implicate the TWIST gene family members as potential mediators of MSC self-renewal and lineage commitment in postnatal skeletal tissues by exerting their effects on genes involved in the early stages of bone development.
Collapse
Affiliation(s)
- Sandra Isenmann
- Mesenchymal Stem Cell Group, Division of Haematology, Institute of Medical and Veterinary Science/Hanson Institute/ CSCR, University of Adelaide, SA, Australia
| | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Osteoporosis and diabetes affect a large proportion of the elderly population. The prevalence of diabetes and osteoporosis is increasing. Compared with individuals without diabetes, both men and women with diabetes have a higher risk of fractures, particularly at the hip, with consequent significant morbidity and mortality. Type 1 diabetes is associated with decreased bone mass and although bone mass data for Type 2 diabetes may or may not be decreased, there is evidence of altered bone quality in diabetes. The mechanisms involved include effects of insulin, insulin-like growth factor 1, cytokines, advanced glycation end products, and altered calcium homeostasis. In addition, a drug-induced increase in the incidence of fractures has been noted with the use of thiazolidinediones (TZDs). TZDs improve insulin sensitivity and have multitude other beneficial effects. Osteoblasts and adipocytes are derived from a common multipotential mesenchymal stem cell progenitor, with activation of peroxisome proliferator-activated receptor γ2 by both currently available TZDs (i.e. rosiglitazone and pioglitazone) stimulating adipogenesis and inhibiting osteoblastogenesis. The use of both rosiglitazone and pioglitazone is associated with an increased fracture risk, with changes in bone turnover markers and decreased bone mineral density.
Collapse
Affiliation(s)
- Subhashini Yaturu
- Section of Endocrinology and Metabolism, Overton Brooks VA Medical Center/Louisiana State Health Sciences Center, Shreveport, Louisiana 71101-4295, USA.
| |
Collapse
|
37
|
Salgado CJ, Raju A, Licata L, Patel M, Rojavin Y, Wasielewski S, Diarra C, Gordon A, Norcross A, Kent KA. Effects of hyperbaric oxygen therapy on an accelerated rate of mandibular distraction osteogenesis. J Plast Reconstr Aesthet Surg 2009; 62:1568-72. [DOI: 10.1016/j.bjps.2008.06.071] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Revised: 04/16/2008] [Accepted: 06/11/2008] [Indexed: 10/21/2022]
|
38
|
Jackson WM, Aragon AB, Bulken-Hoover JD, Nesti LJ, Tuan RS. Putative heterotopic ossification progenitor cells derived from traumatized muscle. J Orthop Res 2009; 27:1645-51. [PMID: 19517576 PMCID: PMC3014572 DOI: 10.1002/jor.20924] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Heterotopic ossification (HO) is a frequent complication following combat-related trauma, but the pathogenesis of traumatic HO is poorly understood. Building on our recent identification of mesenchymal progenitor cells (MPCs) in traumatically injured muscle, the goal of this study was to evaluate the osteogenic potential of the MPCs in order to assess the role of these cells in HO formation. Compared to bone marrow-derived mesenchymal stem cells (MSCs), a well-characterized population of osteoprogenitor cells, the MPCs exhibited several significant differences during osteogenic differentiation and in the expression of genes related to osteogenesis. Upon osteogenic induction, MPCs showed increased alkaline phosphatase activity, production of a mineralized matrix, and up-regulated expression of the osteoblast-associated genes CBFA1 and alkaline phosphatase. However, MPCs did not appear to reach terminal differentiation as the expression of osteocalcin was not substantially up-regulated. With the exception of a few genes, the osteogenic gene expression profile of traumatized muscle-derived MPCs was comparable to that of the MSCs after osteogenic induction. These findings indicate that traumatized muscle-derived MPCs have the potential to function as osteoprogenitor cells when exposed to the appropriate biochemical environment and are the putative osteoprogenitor cells that initiate ectopic bone formation in HO.
Collapse
Affiliation(s)
- Wesley M. Jackson
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Amber B. Aragon
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland,Department of Orthopaedics and Rehabilitation, Walter Reed Army Medical Center, Washington, District of Columbia
| | - Jamie D. Bulken-Hoover
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland,Department of Orthopaedics and Rehabilitation, Walter Reed Army Medical Center, Washington, District of Columbia
| | - Leon J. Nesti
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland,Department of Orthopaedics and Rehabilitation, Walter Reed Army Medical Center, Washington, District of Columbia,Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Rocky S. Tuan
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| |
Collapse
|
39
|
Marcantonio NA, Boehm CA, Rozic R, Au A, Wells A, Muschler GF, Griffith LG. The influence of tethered epidermal growth factor on connective tissue progenitor colony formation. Biomaterials 2009; 30:4629-38. [PMID: 19540579 PMCID: PMC3119364 DOI: 10.1016/j.biomaterials.2009.05.061] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2009] [Accepted: 05/11/2009] [Indexed: 01/14/2023]
Abstract
Strategies to combine aspirated marrow cells with scaffolds to treat connective tissue defects are gaining increasing clinical attention and use. In situations such as large defects where initial survival and proliferation of transplanted connective tissue progenitors (CTPs) are limiting, therapeutic outcomes might be improved by using the scaffold to deliver growth factors that promote the early stages of cell function in the graft. Signaling by the epidermal growth factor receptor (EGFR) plays a role in cell survival and has been implicated in bone development and homeostasis. Providing epidermal growth factor (EGF) in a scaffold-tethered format may sustain local delivery and shift EGFR signaling to pro-survival modes compared to soluble ligand. We therefore examined the effect of tethered EGF on osteogenic colony formation from human bone marrow aspirates in the context of three different adhesion environments using a total of 39 donors. We found that tethered EGF, but not soluble EGF, increased the numbers of colonies formed regardless of adhesion background, and that tethered EGF did not impair early stages of osteogenic differentiation.
Collapse
Affiliation(s)
- Nicholas A. Marcantonio
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Cynthia A. Boehm
- Department of Orthopaedic Surgery and Biomedical Engineering, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | - Richard Rozic
- Department of Orthopaedic Surgery and Biomedical Engineering, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | - Ada Au
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - George F. Muschler
- Department of Orthopaedic Surgery and Biomedical Engineering, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | - Linda G. Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
40
|
Zhu M, Kohan E, Bradley J, Hedrick M, Benhaim P, Zuk P. The effect of age on osteogenic, adipogenic and proliferative potential of female adipose-derived stem cells. J Tissue Eng Regen Med 2009; 3:290-301. [PMID: 19309766 DOI: 10.1002/term.165] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Human adipose tissue is an ideal source of autologous cells that is both plentiful and easily obtainable in large quantities through the simple surgical procedure of liposuction. The stromal vascular fraction of adipose tissue contains a stem cell population, adipose-derived stem cells (ASCs), capable of adipogenic, osteogenic, myogenic and chondrogenic differentiation. These cells have already been recognized to possess great therapeutic potential in tissue engineering and regeneration. In this study, we sought to determine the effect of donor age on the growth kinetics and differentiation potential of ASCs. For this, ASCs were isolated from liposuctioned adipose tissue obtained from female patients in the age range 20-58 years. Population doubling time was calculated over 2 weeks and differentiation potential was determined by assaying for adipogenesis and osteogenesis. ASCs obtained from older donors appeared to have a slower rate of proliferation, but this relationship was not significant. While adipogenic potential was unrelated to donor age, a distinct relationship between donor age and osteogenic potential was observed. The aetiology of this age-dependent change in osteogenic potential was not due to any changes in the number of precursors with osteogenic capacity in the adipose sample. These findings have important implications for emerging cell-based therapeutic strategies, such as tissue engineering, in addition to treatment of various metabolic bone disorders including osteoporosis.
Collapse
Affiliation(s)
- Min Zhu
- Cytori Therapeutics, La Jolla, CA, USA
| | | | | | | | | | | |
Collapse
|
41
|
Kim HK, Cho SG, Kim JH, Doan TKP, Hu QS, Ulhaq R, Song EK, Yoon TR. Mevinolin enhances osteogenic genes (ALP, type I collagen and osteocalcin), CD44, CD47 and CD51 expression during osteogenic differentiation. Life Sci 2009; 84:290-5. [DOI: 10.1016/j.lfs.2008.12.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2008] [Revised: 12/02/2008] [Accepted: 12/11/2008] [Indexed: 10/21/2022]
|
42
|
Ackema KB, Charité J. Mesenchymal stem cells from different organs are characterized by distinct topographic Hox codes. Stem Cells Dev 2008; 17:979-91. [PMID: 18533811 DOI: 10.1089/scd.2007.0220] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Mesenchymal stem cells (MSC) are multipotent cells found as part of the stromal compartment of the bone marrow and in many other organs. They can be identified in vitro as CFU-F (colony forming unit-fibroblast) based on their ability to form adherent colonies of fibroblast-like cells in culture. MSC expanded in vitro retain characteristics appropriate to their tissue of origin. This is reflected in their propensity for differentiating towards specific lineages, and their capacity to generate, upon retransplantation in vivo, a stroma supporting typical lineages of hematopoietic cells. Hox genes encode master regulators of regional specification and organ development in the embryo and are widely expressed in the adult. We investigated whether they could be involved in determining tissue-specific properties of MSC. Hox gene expression profiles of individual CFU-F colonies derived from various organs and anatomical locations were generated, and the relatedness between these profiles was determined using hierarchical cluster analysis. This revealed that CFU-F have characteristic Hox expression signatures that are heterogeneous but highly specific for their anatomical origin. The topographic specificity of these Hox codes is maintained during differentiation, suggesting that they are an intrinsic property of MSC. Analysis of Hox codes of CFU-F from vertebral bone marrow suggests that MSC originate over a large part of the anterioposterior axis, but may not originate from prevertebral mesenchyme. These data are consistent with a role for Hox proteins in specifying cellular identity of MSC.
Collapse
Affiliation(s)
- Karin B Ackema
- Department of Cell Biology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | |
Collapse
|
43
|
Molchanova EA, Payushina OV, Starostin VI. Effects of growth factors on multipotent bone marrow mesenchymal stromal cells. BIOL BULL+ 2008. [DOI: 10.1134/s1062359008060010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
44
|
Post S, Abdallah BM, Bentzon JF, Kassem M. Demonstration of the presence of independent pre-osteoblastic and pre-adipocytic cell populations in bone marrow-derived mesenchymal stem cells. Bone 2008; 43:32-39. [PMID: 18456590 DOI: 10.1016/j.bone.2008.03.011] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2007] [Revised: 03/04/2008] [Accepted: 03/06/2008] [Indexed: 10/22/2022]
Abstract
Mesenchymal stem cells (MSC) are defined as plastic-adherent, clonal cells that are common progenitors for osteoblasts and adipocytes. An inverse relationship between bone and fat has been observed in several clinical conditions and has been suggested to be caused by re-directing MSC differentiation into one particular lineage. However, this inverse relationship between bone and fat is not consistent and under certain in vivo conditions, bone and fat can change independently suggesting separate precursor cell populations. In order to test for this hypothesis, we extensively characterized two plastic-adherent clonal MSC lines (mMSC1 and mMSC2) derived from murine bone marrow. The two cell lines grew readily in culture and have undergone more than 100 population doublings with no apparent differences in their growth rates. Both cell lines were positive for the murine MSC marker Sca-1 and mMSC1 was also positive for CD13. Both cell lines were exposed to in vitro culture induction of osteogenesis and adipogenesis. mMSC1 and not mMSC2 were only able to differentiate to adipocytes evidenced by the expression of adipocyte markers (aP2, adiponectin, adipsin, PPARgamma2 and C/EBPa) and the presence of mature adipocytes visualized by Oil Red O staining. On the other hand, mMSC2 and not mMSC1 differentiated to osteoblast lineage as demonstrated by up-regulation of osteoblastic makers (CBFA1/RUNX2, Osterix, alkaline phosphatase, bone sialoprotein and osteopontin) and formation of alizarin red stained mineralized matrix in vitro. Consistent with the in vitro results, mMSC2 and not mMSC1, were able to form bone in vivo after subcutaneous implantation in immune-deficient (NOD/SCID) mice. Our data suggest that contrary to the current belief, bone marrow contains clonal subpopulations of cells that are committed to either osteoblast or adipocyte lineage. These cell populations may undergo independent changes during aging and in bone diseases and thus represent important targets for therapy.
Collapse
Affiliation(s)
- S Post
- Laboratory for Molecular Endocrinology (KMEB), Department of Endocrinology and Metabolism, University Hospital of Odense, DK-5000 Odense C, Denmark
| | - B M Abdallah
- Laboratory for Molecular Endocrinology (KMEB), Department of Endocrinology and Metabolism, University Hospital of Odense, DK-5000 Odense C, Denmark
| | - J F Bentzon
- Department of Cardiology Research Unit, Clinical Institute, Skejby University Hospital, Aarhus, Denmark
| | - M Kassem
- Laboratory for Molecular Endocrinology (KMEB), Department of Endocrinology and Metabolism, University Hospital of Odense, DK-5000 Odense C, Denmark.
| |
Collapse
|
45
|
|
46
|
Gimble JM, Guilak F, Nuttall ME, Sathishkumar S, Vidal M, Bunnell BA. In vitro Differentiation Potential of Mesenchymal Stem Cells. ACTA ACUST UNITED AC 2008; 35:228-238. [PMID: 21547120 DOI: 10.1159/000124281] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2007] [Accepted: 03/07/2008] [Indexed: 12/19/2022]
Abstract
SUMMARY: Mesenchymal stem cells (MSCs) represent a class of multipotent progenitor cells that have been isolated from multiple tissue sites. Of these, adipose tissue and bone marrow offer advantages in terms of access, abundance, and the extent of their documentation in the literature. This review focuses on the in vitro differentiation capability of cells derived from adult human tissue. Multiple, independent studies have demonstrated that MSCs can commit to mesodermal (adipocyte, chondrocyte, hematopoietic support, myocyte, osteoblast, tenocyte), ectodermal (epithelial, glial, neural), and endodermal (hepatocyte, islet cell) lineages. The limitations and promises of these studies in the context of tissue engineering are discussed.
Collapse
Affiliation(s)
- Jeffrey M Gimble
- Stem Cell Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | | | | | | | | | | |
Collapse
|
47
|
Culture of mesenchymal stem/progenitor cells in adhesion-independent conditions. Methods Cell Biol 2008. [PMID: 18442652 DOI: 10.1016/s0091-679x(08)00012-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
48
|
Kim HK, Kim JH, Abbas AA, Kim DO, Park SJ, Chung JY, Song EK, Yoon TR. Red light of 647 nm enhances osteogenic differentiation in mesenchymal stem cells. Lasers Med Sci 2008; 24:214-22. [DOI: 10.1007/s10103-008-0550-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2007] [Accepted: 02/05/2008] [Indexed: 11/30/2022]
|
49
|
Specific plasma membrane protein phenotype of culture-amplified and native human bone marrow mesenchymal stem cells. Blood 2007; 111:2631-5. [PMID: 18086871 DOI: 10.1182/blood-2007-07-099622] [Citation(s) in RCA: 195] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We have studied the plasma membrane protein phenotype of human culture-amplified and native bone marrow mesenchymal stem cells (BM MSCs). We have found, using microarrays and flow cytometry, that cultured cells express specifically 113 transcripts and 17 proteins that were not detected in hematopoietic cells. These antigens define a lineage-homogenous cell population of mesenchymal cells, clearly distinct from the hematopoietic lineages, and distinguishable from other cultured skeletal mesenchymal cells (periosteal cells and synovial fibroblasts). Among the specific membrane proteins present on cultured MSCs, 9 allowed the isolation from BM mononuclear cells of a minute population of native MSCs. The enrichment in colony-forming units-fibroblasts was low for CD49b, CD90, and CD105, but high for CD73, CD130, CD146, CD200, and integrin alphaV/beta5. In addition, the expression of CD73, CD146, and CD200 was down-regulated in differentiated cells. The new marker CD200, because of its specificity and immunomodulatory properties, deserves further in-depth studies.
Collapse
|
50
|
Thibault MM, Hoemann CD, Buschmann MD. Fibronectin, vitronectin, and collagen I induce chemotaxis and haptotaxis of human and rabbit mesenchymal stem cells in a standardized transmembrane assay. Stem Cells Dev 2007; 16:489-502. [PMID: 17610379 DOI: 10.1089/scd.2006.0100] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The mesenchymal stem cell (MSC) is a critical element in tissue repair and regeneration. Its ability to differentiate into multiple connective tissue cell types and to self-renew has made it a prime candidate in regenerative medicine strategies. Currently, the environmental cues responsible for in situ recruitment and control of MSC distribution at repair sites are not entirely revealed and in particular the role of extracellular matrix (ECM) proteins as motogenic factors has not been studied. Here we have used a standardized transmembrane chemotaxis assay to assess the chemotactic and haptotactic potential of fibronectin, vitronectin, and collagen type 1 on MSCs from both rabbit and human origin. The use of both cell types was based in part on the widespread use of rabbit models for musculoskeletal-related tissue engineering and repair models and their unknown correspondence to human in terms of MSC migration. The optimized assay yielded a greatly increased chemotactic response toward known factors such as platelet-derived growth factor-BB (PDGF)-BB compared to previous studies. Our primary finding was that all three ECM proteins tested (fibronectin, vitronectin, and collagen I) induced significant motogenic activity, in both soluble and insoluble forms, for both rabbit and human MSCs. These results suggest that ECM proteins could play roles as significant as cytokines in the recruitment of pluripotential repair cells wound and tissue repair sites. Furthermore, designed ECM coatings of scaffolds or implants could provide a new tool to control both cell influx and outflux from the scaffold post-implantation. Finally, the similarity of motogenic behavior of both rabbit and human cells suggests the rabbit is a reliable model for assessing MSC recruitment in repair and regeneration strategies.
Collapse
Affiliation(s)
- Marc M Thibault
- Department of Chemical Engineering, Institute of Biomedical Engineering, Ecole Polytechnique, Montreal, Quebec, Canada
| | | | | |
Collapse
|