51
|
Hilkens P, Meschi N, Lambrechts P, Bronckaers A, Lambrichts I. Dental Stem Cells in Pulp Regeneration: Near Future or Long Road Ahead? Stem Cells Dev 2015; 24:1610-22. [PMID: 25869156 DOI: 10.1089/scd.2014.0510] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although regenerative endodontic procedures have yielded an impressive body of favorable outcomes, the treatment of necrotic immature permanent teeth in particular remains to be a challenge. Recent advances in dental stem cell (DSC) research have gained increasing insight in their regenerative potential and prospective use in the formation of viable dental tissues. Numerous studies have already reported successful dental pulp regeneration following application of dental pulp stem cells, stem cells from the apical papilla, or dental follicle precursor cells in different in vivo models. Next to responsive cells, dental tissue engineering also requires the support of an appropriate scaffold material, ranging from naturally occurring polymers to treated dentin matrix components. However, the routine use and banking of DSCs still holds some major challenges, such as culture-associated differences, patient-related variability, and the effects of culture medium additives. Only in-depth evaluation of these problems and the implementation of standardized models and protocols will effectively lead to better alternatives for patients who no longer benefit from current treatment protocols.
Collapse
Affiliation(s)
- Petra Hilkens
- 1 Laboratory of Morphology, Biomedical Research Institute (BIOMED), Hasselt University , Diepenbeek, Belgium
| | - Nastaran Meschi
- 2 Department of Oral Health Sciences, KU Leuven and Dentistry, University Hospitals Leuven , Leuven, Belgium
| | - Paul Lambrechts
- 2 Department of Oral Health Sciences, KU Leuven and Dentistry, University Hospitals Leuven , Leuven, Belgium
| | - Annelies Bronckaers
- 1 Laboratory of Morphology, Biomedical Research Institute (BIOMED), Hasselt University , Diepenbeek, Belgium
| | - Ivo Lambrichts
- 1 Laboratory of Morphology, Biomedical Research Institute (BIOMED), Hasselt University , Diepenbeek, Belgium
| |
Collapse
|
52
|
Reitinger S, Schimke M, Klepsch S, de Sneeuw S, Yani SL, Gaßner R, Ertl P, Lepperdinger G. Systemic impact molds mesenchymal stromal/stem cell aging. Transfus Apher Sci 2015; 52:285-9. [PMID: 25910539 DOI: 10.1016/j.transci.2015.04.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Aging is associated with an accruing emergence of non-functional tissues. Mesenchymal stem cells (MSC) bring forth progenitors with multi-lineage differentiation potential, yet, they also exhibit anti-inflammatory and tissue-protective properties. Due to aging, altered tissue microenvironments constrict controlled stem cell proliferation and progenitor differentiation, thus diminishing the fitness of MSC. Therefore, deepening our understanding of metabolic, molecular and environmental factors impacting on MSC during human aging as well as providing new vistas on their role in promoting healthy aging and preventing age-associated disease is pivot. It is anticipated that integrative quantification of systemic parameters dominantly impacting on MSC will also enable effective personalized prognosis and provision of effective early medical interventions. Working along this line, it can be envisaged that standards in medical therapies can be individually adjusted by accounting not solely for the patient's chronological age or other physical parameters rather than specific physiological parameters which are believed to functionally shape stem cell niches within the bone marrow.
Collapse
Affiliation(s)
- Stephan Reitinger
- Institute for Biomedical Aging Research, University Innsbruck, Rennweg 10, Innsbruck A-6020, Austria
| | - Magdalena Schimke
- Institute for Biomedical Aging Research, University Innsbruck, Rennweg 10, Innsbruck A-6020, Austria
| | - Sebastian Klepsch
- Institute for Biomedical Aging Research, University Innsbruck, Rennweg 10, Innsbruck A-6020, Austria
| | - Snezana de Sneeuw
- Institute for Biomedical Aging Research, University Innsbruck, Rennweg 10, Innsbruck A-6020, Austria
| | - Stella Lukas Yani
- Institute for Biomedical Aging Research, University Innsbruck, Rennweg 10, Innsbruck A-6020, Austria
| | - Robert Gaßner
- University Clinic for Maxillofacial Surgery Innsbruck, Anichstr. 35, Innsbruck A-6020, Austria
| | - Peter Ertl
- Austrian Institute for Technology, Muthgasse 11, Wien A-1190, Austria
| | - Günter Lepperdinger
- Institute for Biomedical Aging Research, University Innsbruck, Rennweg 10, Innsbruck A-6020, Austria; Department of Cell Biology and Genetics, University Salzburg, Hellbrunnerstr. 34, Salzburg 5020, Austria.
| |
Collapse
|
53
|
Kumar P, Satyam A, Fan X, Rochev Y, Rodriguez BJ, Gorelov A, Joshi L, Raghunath M, Pandit A, Zeugolis DI. Accelerated Development of Supramolecular Corneal Stromal-Like Assemblies from Corneal Fibroblasts in the Presence of Macromolecular Crowders. Tissue Eng Part C Methods 2015; 21:660-70. [PMID: 25535812 DOI: 10.1089/ten.tec.2014.0387] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Tissue engineering by self-assembly uses the cells' secretome as a regeneration template and biological factory of trophic factors. Despite the several advantages that have been witnessed in preclinical and clinical settings, the major obstacle for wide acceptance of this technology remains the tardy extracellular matrix formation. In this study, we assessed the influence of macromolecular crowding (MMC)/excluding volume effect, a biophysical phenomenon that accelerates thermodynamic activities and biological processes by several orders of magnitude, in human corneal fibroblast (HCF) culture. Our data indicate that the addition of negatively charged galactose derivative (carrageenan) in HCF culture, even at 0.5% serum, increases by 12-fold tissue-specific matrix deposition, while maintaining physiological cell morphology and protein/gene expression. Gene analysis indicates that a glucose derivative (dextran sulfate) may drive corneal fibroblasts toward a myofibroblast lineage. Collectively, these results indicate that MMC may be suitable not only for clinical translation and commercialization of tissue engineering by self-assembly therapies, but also for the development of in vitro pathophysiology models.
Collapse
Affiliation(s)
- Pramod Kumar
- 1 Network of Excellence for Functional Biomaterials (NFB), Bioscience Research Building, National University of Ireland Galway (NUI Galway) , Galway, Ireland
| | - Abhigyan Satyam
- 1 Network of Excellence for Functional Biomaterials (NFB), Bioscience Research Building, National University of Ireland Galway (NUI Galway) , Galway, Ireland
| | - Xingliang Fan
- 1 Network of Excellence for Functional Biomaterials (NFB), Bioscience Research Building, National University of Ireland Galway (NUI Galway) , Galway, Ireland
| | - Yury Rochev
- 1 Network of Excellence for Functional Biomaterials (NFB), Bioscience Research Building, National University of Ireland Galway (NUI Galway) , Galway, Ireland
| | - Brian J Rodriguez
- 2 Conway Institute of Biomolecular & Biomedical Research, University College Dublin , Dublin, Ireland
| | - Alexander Gorelov
- 3 School of Chemistry & Chemical Biology, University College Dublin , Dublin, Ireland
| | - Lokesh Joshi
- 4 Alimentary Glycoscience Research Cluster, NUI Galway , Galway, Ireland
| | - Michael Raghunath
- 5 Department of Bioengineering, Faculty of Engineering, National University of Singapore , Singapore, Singapore .,6 Tissue Engineering Programme, Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Abhay Pandit
- 1 Network of Excellence for Functional Biomaterials (NFB), Bioscience Research Building, National University of Ireland Galway (NUI Galway) , Galway, Ireland
| | - Dimitrios I Zeugolis
- 1 Network of Excellence for Functional Biomaterials (NFB), Bioscience Research Building, National University of Ireland Galway (NUI Galway) , Galway, Ireland
| |
Collapse
|
54
|
Macromolecularly crowded in vitro microenvironments accelerate the production of extracellular matrix-rich supramolecular assemblies. Sci Rep 2015; 5:8729. [PMID: 25736020 PMCID: PMC4348624 DOI: 10.1038/srep08729] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 01/27/2015] [Indexed: 12/13/2022] Open
Abstract
Therapeutic strategies based on the principles of tissue engineering by self-assembly put forward the notion that functional regeneration can be achieved by utilising the inherent capacity of cells to create highly sophisticated supramolecular assemblies. However, in dilute ex vivo microenvironments, prolonged culture time is required to develop an extracellular matrix-rich implantable device. Herein, we assessed the influence of macromolecular crowding, a biophysical phenomenon that regulates intra- and extra-cellular activities in multicellular organisms, in human corneal fibroblast culture. In the presence of macromolecules, abundant extracellular matrix deposition was evidenced as fast as 48 h in culture, even at low serum concentration. Temperature responsive copolymers allowed the detachment of dense and cohesive supramolecularly assembled living substitutes within 6 days in culture. Morphological, histological, gene and protein analysis assays demonstrated maintenance of tissue-specific function. Macromolecular crowding opens new avenues for a more rational design in engineering of clinically relevant tissue modules in vitro.
Collapse
|
55
|
Haque N, Kasim NHA, Rahman MT. Optimization of pre-transplantation conditions to enhance the efficacy of mesenchymal stem cells. Int J Biol Sci 2015; 11:324-34. [PMID: 25678851 PMCID: PMC4323372 DOI: 10.7150/ijbs.10567] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 12/20/2014] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are considered a potential tool for cell based regenerative therapy due to their immunomodulatory property, differentiation potentials, trophic activity as well as large donor pool. Poor engraftment and short term survival of transplanted MSCs are recognized as major limitations which were linked to early cellular ageing, loss of chemokine markers during ex vivo expansion, and hyper-immunogenicity to xeno-contaminated MSCs. These problems can be minimized by ex vivo expansion of MSCs in hypoxic culture condition using well defined or xeno-free media i.e., media supplemented with growth factors, human serum or platelet lysate. In addition to ex vivo expansion in hypoxic culture condition using well defined media, this review article describes the potentials of transient adaptation of expanded MSCs in autologous serum supplemented medium prior to transplantation for long term regenerative benefits. Such transient adaptation in autologous serum supplemented medium may help to increase chemokine receptor expression and tissue specific differentiation of ex vivo expanded MSCs, thus would provide long term regenerative benefits.
Collapse
Affiliation(s)
- Nazmul Haque
- 1. Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia. ; 2. Regenerative Dentistry Research Group, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Noor Hayaty Abu Kasim
- 1. Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia. ; 2. Regenerative Dentistry Research Group, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Mohammad Tariqur Rahman
- 3. Department of Biotechnology, Faculty of Science, International Islamic University Malaysia, Kuantan, Malaysia
| |
Collapse
|
56
|
Li J, Dong J, Zhang ZH, Zhang DC, You XY, Zhong Y, Chen MS, Liu SM. miR-10a restores human mesenchymal stem cell differentiation by repressing KLF4. J Cell Physiol 2014; 228:2324-36. [PMID: 23696417 PMCID: PMC4285942 DOI: 10.1002/jcp.24402] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 05/07/2013] [Indexed: 12/20/2022]
Abstract
miRNAs have recently been shown to play a significant role in human aging. However, data demonstrating the effects of aging-related miRNAs in human mesenchymal stem cells (hMSCs) are limited. We observed that hMSC differentiation decreased with aging. We also identified that miR-10a expression was significantly decreased with age by comparing the miRNA expression of hMSCs derived from young and aged individuals. Therefore, we hypothesized that the downregulation of miR-10a may be associated with the decreased differentiation capability of hMSCs from aged individuals. Lentiviral constructs were used to up- or downregulate miR-10a in young and old hMSCs. Upregulation of miR-10a resulted in increased differentiation to adipogenic, osteogenic, and chondrogenic lineages and in reduced cell senescence. Conversely, downregulation of miR-10a resulted in decreased cell differentiation and increased cell senescence. A chimeric luciferase reporter system was generated, tagged with the full-length 3′-UTR region of KLF4 harboring the seed-matched sequence with or without four nucleotide mutations. These constructs were cotransfected with the miR-10a mimic into cells. The luciferase activity was significantly repressed by the miR-10a mimic, proving the direct binding of miR-10a to the 3′-UTR of KLF4. Direct suppression of KLF4 in aged hMSCs increased cell differentiation and decreased cell senescence. In conclusion, miR-10a restores the differentiation capability of aged hMSCs through repression of KLF4. Aging-related miRNAs may have broad applications in the restoration of cell dysfunction caused by aging. J. Cell. Physiol. 228: 2324–2336, 2013. © The Authors. Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jiao Li
- Department of Cardiology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Cardiovascular Disease, Guangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
57
|
Gobbi A, Karnatzikos G, Sankineani SR. One-step surgery with multipotent stem cells for the treatment of large full-thickness chondral defects of the knee. Am J Sports Med 2014; 42:648-57. [PMID: 24458240 DOI: 10.1177/0363546513518007] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Chondral lesions in athletically active patients cause considerable morbidity, and treatment with existing cell-based therapies can be challenging. Bone marrow has been shown as a possible source of multipotent stem cells (MSCs) with chondrogenic potential and is easy to harvest during the same surgical procedure. PURPOSE To investigate the clinical outcome in a group of active patients with large full-thickness chondral defects of the knee treated with 1-step surgery using bone marrow-derived MSCs and a second-generation matrix. STUDY DESIGN Case series; Level of evidence, 4. METHODS From January 2007 to February 2010, 25 patients (average age, 46.5 years) with symptomatic large chondral defects of the knee (International Cartilage Repair Society grade 4) who underwent cartilage transplantation with MSCs and a collagen type I/III matrix were followed up for a minimum of 3 years. The average lesion size was 8.3 cm(2). Coexisting injuries were treated during the same surgical procedure in 18 patients. All patients underwent a standard postoperative rehabilitation program. Preoperative and postoperative evaluations at 1-year, 2-year, and final follow-up included radiographs, magnetic resonance imaging (MRI), and visual analog scale (VAS) for pain, International Knee Documentation Committee (IKDC), Knee injury and Osteoarthritis Outcome Score (KOOS), Lysholm, Marx, and Tegner scores. Seven patients underwent second-look arthroscopic surgery, with 4 consenting to a tissue biopsy. RESULTS No patients were lost at final follow-up. The average preoperative values for the evaluated scores were significantly improved at final follow-up (P < .001): VAS, 5.4 ± 0.37 to 0.48 ± 0.19; IKDC subjective, 37.92 ± 4.52 to 81.73 ± 2.42; KOOS pain, 61.04 ± 3.95 to 93.32 ± 1.92; KOOS symptoms, 55.64 ± 3.23 to 89.32 ± 2.32; KOOS activities of daily living, 63.96 ± 4.48 to 91.20 ± 2.74; KOOS sports, 34.20 ± 5.04 to 80.00 ± 3.92; KOOS quality of life, 32.20 ± 4.43 to 83.04 ± 3.37; Lysholm, 46.36 ± 2.25 to 86.52 ± 2.73; Marx, 3.00 ± 0.79 to 9.04 ± 0.79; and Tegner, 2.12 ± 0.32 to 5.64 ± 0.26. Patients younger than 45 years of age and those with smaller or single lesions showed better outcomes. The MRI scans showed good stability of the implant and complete filling of the defect in 80% of patients, and hyaline-like cartilage was found in the histological analysis of the biopsied tissue. No adverse reactions or postoperative complications were noted. CONCLUSION The treatment of large chondral defects with MSCs is an effective procedure and can be performed routinely in clinical practice. Moreover, it can be achieved with 1-step surgery, avoiding a previous surgical procedure to harvest cartilage and subsequent chondrocyte cultivation.
Collapse
Affiliation(s)
- Alberto Gobbi
- Alberto Gobbi, Orthopaedic Arthroscopic Surgery International (OASIBioresearch Foundation, G.A. Amadeo 24, 20133 Milan, Italy. )
| | | | | |
Collapse
|
58
|
Mesenchymal stem cells for regenerative therapy: optimization of cell preparation protocols. BIOMED RESEARCH INTERNATIONAL 2014; 2014:951512. [PMID: 24511552 PMCID: PMC3912818 DOI: 10.1155/2014/951512] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 12/08/2013] [Indexed: 12/14/2022]
Abstract
Administration of bone marrow-derived mesenchymal stem cells (MSCs) is an innovative approach for the treatment of a range of diseases that are not curable by current therapies including heart failure. A number of clinical trials have been completed and many others are ongoing; more than 2,000 patients worldwide have been administered with culture-expanded allogeneic or autologous MSCs for the treatment of various diseases, showing feasibility and safety (and some efficacy) of this approach. However, protocols for isolation and expansion of donor MSCs vary widely between these trials, which could affect the efficacy of the therapy. It is therefore important to develop international standards of MSC production, which should be evidence-based, regulatory authority-compliant, of good medical practice grade, cost-effective, and clinically practical, so that this innovative approach becomes an established widely adopted treatment. This review article summarizes protocols to isolate and expand bone marrow-derived MSCs in 47 recent clinical trials of MSC-based therapy, which were published after 2007 onwards and provided sufficient methodological information. Identified issues and possible solutions associated with the MSC production methods, including materials and protocols for isolation and expansion, are discussed with reference to relevant experimental evidence with aim of future clinical success of MSC-based therapy.
Collapse
|
59
|
Yang H, Lim YH, Yun S, Yoon AY, Kim H. A role of cell adhesion molecules and gelatinases in human serum-induced aggregation of human eyelid-derived stem cells in vitro. Dev Reprod 2013; 17:409-20. [PMID: 25949157 PMCID: PMC4382947 DOI: 10.12717/dr.2013.17.4.409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 12/15/2013] [Accepted: 12/20/2013] [Indexed: 01/18/2023]
Abstract
Human serum (HS) has been reported to induce aggregation of human eyelid adipose-derived stem cells (HEACs) during high-density culture in vitro. The present study focused on the role of cell adhesion molecules and gelatinases during HS-induced aggregation of HEACs. HS-induced aggregation occurred between 9-15 days of culture. Cells aggregated by HS medium (HS-agg) showed stronger expression of α2, α2B, αX, and CEACAM1 genes compared to non-aggregated cells in HS medium (HS-ex) or in control FBS-cultured cells. HS-agg were distinctly labeled with antibodies against α2, α2B, and αX proteins. Western blot results demonstrated that the two integrin proteins were greatly expressed in HS-agg compared to HS-ex and control FBS-cultured cells. Treatment of HEACs with anti-integrin α2 antibody during culture in HS medium delayed aggregation formation. HS-agg exhibited strong expression of MMP1 and MMP9 compared to HS-ex or FBS-cultured cells. Conditioned media from HS-culture showed remarkable increase of MMP9 gelatinolytic activity in comparison to those from FBS-culture. However, there was no change of TIMP mRNA expression in relation to the HS-induced aggregation. Based on these results, it is suggested that integrin α2, α2B, and αX, and MMP9 might play an important role in the HS-induced aggregation of HEACs.
Collapse
Affiliation(s)
- Hyejin Yang
- Department of Biotechnology, Seoul Women's University, Seoul 139-774, Republic of Korea
| | - Yoon Hwa Lim
- Department of Biotechnology, Seoul Women's University, Seoul 139-774, Republic of Korea
| | - Sujin Yun
- Department of Biotechnology, Seoul Women's University, Seoul 139-774, Republic of Korea
| | - A Young Yoon
- Department of Biotechnology, Seoul Women's University, Seoul 139-774, Republic of Korea
| | - Haekwon Kim
- Department of Biotechnology, Seoul Women's University, Seoul 139-774, Republic of Korea
| |
Collapse
|
60
|
Hui JHP, Goyal D, Nakamura N, Ochi M. Cartilage repair: 2013 Asian update. Arthroscopy 2013; 29:1992-2000. [PMID: 24286798 DOI: 10.1016/j.arthro.2013.06.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 06/11/2013] [Indexed: 02/02/2023]
Abstract
Despite financial and regulatory hurdles, Asian scientists and clinicians have made important contributions in the area of cartilage repair. Because it is impossible to include observations on all the published articles in one review, our attempt is to highlight Asian progress in this area during recent years (2005 to the present), reviewing research development and clinical studies. In the former, our discussion of in vitro studies focuses on (1) potential sources of stem cells--such as mesenchymal stem cells (MSCs) from marrow, cord blood, synovium, and mobilized peripheral blood--which are capable of enhancing cartilage repair and (2) the use of growth factors and scaffolds with and without cells. Our discussion of animal studies attempts to summarize activities in evaluating surgical procedures and determining the route of cell administration, as well as studies on matrices and scaffolds. It ranges from the use of small animals such as rats and rabbits to larger animals like pigs and dogs. The local adherent technique, enhancement of microfracture with poly(l-lactic-co-glycolic acid) scaffold, adenovirus-mediated bone morphogenic protein (BMP) genes, and MSCs--whether they are magnetically labeled, suspended in hyaluronic acid, or immobilized with transforming growth factor-β (TGF-β)--have all been able to engineer a repair of the osteochondral defect. Although published Asian reports of clinical studies on cartilage repair are few, the findings of relevant trials are summarized in our discussion of these investigations. There has been a long history of use of laboratory-derived MSCs for cartilage repair. Recent progress has suggested the potential utility of cord blood and mobilized peripheral blood in this area, as well as more injectable bone marrow (BM)-derived stem cells. Finally, we make a few suggestions on the direction of research and development activities and the need for collaborative approaches by regulatory agencies.
Collapse
Affiliation(s)
- James H P Hui
- Cartilage Repair Program, Therapeutic Tissue Engineering Laboratory, Department of Orthopaedic Surgery, National University Health System, National University of Singapore, Singapore.
| | | | | | | | | |
Collapse
|
61
|
Jung S, Panchalingam KM, Wuerth RD, Rosenberg L, Behie LA. Large-scale production of human mesenchymal stem cells for clinical applications. Biotechnol Appl Biochem 2013; 59:106-20. [PMID: 23586791 DOI: 10.1002/bab.1006] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 01/30/2012] [Indexed: 02/06/2023]
Abstract
Human mesenchymal stem cells (hMSCs) have many potential applications in tissue engineering and regenerative medicine. Currently, hMSCs are generated through conventional static adherent cultures in the presence of fetal bovine serum (FBS) for clinical applications (e.g., multiple sclerosis). However, these methods are not appropriate to meet the expected future demand for quality-assured hMSCs for human therapeutic use. Hence, it is imperative to develop an effective hMSC production system, which should be controllable, reproducible, and scalable. To this end, efforts have been made by several international research groups to develop (i) alternative media either by replacing FBS with human-sourced supplements (such as human serum or platelet lysate) or by identifying defined serum-free formulations consisting of key growth/attachment factors, and (ii) controlled bioreactor protocols. In this regard, we review here current hMSC production technologies and future perspectives toward efficient methods for the generation of clinically relevant numbers of hMSC therapeutics.
Collapse
Affiliation(s)
- Sunghoon Jung
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | |
Collapse
|
62
|
Gottipamula S, Muttigi MS, Kolkundkar U, Seetharam RN. Serum-free media for the production of human mesenchymal stromal cells: a review. Cell Prolif 2013; 46:608-27. [PMID: 24118248 DOI: 10.1111/cpr.12063] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 07/24/2013] [Indexed: 12/29/2022] Open
Abstract
The regenerative potential of mesenchymal stromal cells (MSC) holds great promise in using them for treatment of a wide range of debilitating diseases. Several types of culture media and systems have been used for large-scale expansion of MSCs in vitro; however, the majority of them rely heavily on using foetal bovine serum (FBS)-supplement for optimal cell proliferation. FBS-based cultures pose the potential threat of spread of transmissible spongiform encephalopathy and bovine spongiform encephalopathy to MSCs and then to their recipients. A recent trend in cell culture is to change from serum-use to serum-free media (SFM). In this context, the current review focuses specifically on employment of various SFM for MSCs and discusses existences of various options with which to substitute FBS. In addition, we analyse MSC population growth kinetic patterns using various SFM for large-scale production of MSCs.
Collapse
Affiliation(s)
- S Gottipamula
- Stempeutics Research Pvt. Ltd, Shirdi Sai Baba Cancer Hospital, Manipal, 576104, India
| | | | | | | |
Collapse
|
63
|
The Current State of Clinical Cell Transplantation Trials in Iran: A Survey in 2011. ARCHIVES OF NEUROSCIENCE 2013. [DOI: 10.5812/archneurosci.9080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
64
|
Abstract
INTRODUCTION Mesenchymal stem cells (MSC) and MSC-like cells hold great promise and offer many advantages for developing effective cellular therapeutics. Current trends indicate that the clinical application of MSC will continue to increase markedly. For clinical applications, large numbers of MSC are usually required, ideally in an off-the-shelf format, thus requiring extensive MSC expansion ex vivo and subsequent cryopreservation and banking. AREAS COVERED To exploit the full potential of MSC for cell-based therapies requires overcoming significant cell-manufacturing, banking and regulatory challenges. The current review will focus on the identification of optimal cell source for MSC, the techniques for production scale-up, cryopreservation and banking and the regulatory challenges involved. EXPERT OPINION There has been considerable success manufacturing and cryopreserving MSC at laboratory scale. Surprisingly little attention, however, has been given to translate these technologies to an industrial scale. The development of cost-effective advanced technologies for producing and cryopreserving commercial-scale MSC is important for successful clinical cell therapy.
Collapse
|
65
|
Isolation of functional human endothelial cells from small volumes of umbilical cord blood. Ann Biomed Eng 2013; 41:2181-92. [PMID: 23604849 DOI: 10.1007/s10439-013-0807-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 04/04/2013] [Indexed: 12/11/2022]
Abstract
Endothelial cells (ECs) isolated from endothelial progenitor cells in blood have great potential as a therapeutic tool to promote vasculogenesis and angiogenesis and treat cardiovascular diseases. However, current methods to isolate ECs are limited by a low yield with few colonies appearing during isolation. In order to utilize blood-derived ECs for therapeutic applications, a simple method is needed that can produce a high yield of ECs from small volumes of blood without the addition of animal-derived products. For the first time, we show that human ECs can be isolated without the prior separation of blood components through the technique of diluted whole blood incubation (DWBI) utilizing commercially available human serum. We isolated ECs from small volumes of blood (~10 mL) via DWBI and characterized them with flow cytometry, immunohistochemistry, and uptake of DiI-labeled acetylated low density lipoprotein (DiI-Ac-LDL). These ECs are functional as demonstrated by their ability to form tubular networks in Matrigel, adhere and align with flow under physiological fluid shear stress, and produce increased nitric oxide under fluid flow. An average of 7.0 ± 2.5 EC colonies that passed all functional tests described above were obtained per 10 mL of blood as compared to only 0.3 ± 0.1 colonies with the traditional method based on density centrifugation. The time until first colony appearance was 8.3 ± 1.2 days for ECs isolated with the DWBI method and 12 ± 1.4 days for ECs isolated with the traditional isolation method. A simplified method, such as DWBI, in combination with advances in isolation yield could enable the use of blood-derived ECs in clinical practice.
Collapse
|
66
|
Cheng MT, Liu CL, Chen TH, Lee OK. Optimization of culture conditions for stem cells derived from human anterior cruciate ligament and bone marrow. Cell Transplant 2013; 23:791-803. [PMID: 23582177 DOI: 10.3727/096368912x666430] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Tissue engineering with stem cells is a fascinating approach for treating anterior cruciate ligament (ACL) injuries. In our previous study, stem cells isolated from the human anterior cruciate ligament were shown to possess extensive proliferation and differentiation capabilities when treated with specific growth factors. However, optimal culture conditions and the usefulness of fetal bovine serum (FBS) as a growth factor in in vitro culture systems are yet to be determined. In this study, we compared the effects of different culture media containing combinations of various concentrations of FBS and the growth factors basic fibroblastic growth factor (bFGF) and transforming growth factor-β1 (TGF-β1) on the proliferation and differentiation of ligament-derived stem cells (LSCs) and bone marrow mesenchymal stem cells (BMSCs). We found that α-MEM plus 10% FBS and bFGF was able to maintain both LSCs and BMSCs in a relatively undifferentiated state but with lower major extracellular matrix (ECM) component gene expression and protein production, which is beneficial for stem cell expansion. However, the differentiation and proliferation potentials of LSCs and BMSCs were increased when cultured in MesenPRO, a commercially available stem cell medium containing 2% FBS. MesenPRO in conjunction with TGF-β1 had the greatest ability to induce the differentiation of BMSCs and LSCs to ligament fibroblasts, which was evidenced by the highest ligamentous ECM gene expression and protein production. These results indicate that culture media and growth factors play a very important role in the success of tissue engineering. With α-MEM plus 10% FBS and bFGF, rapid proliferation of stem cells can be achieved. In this study, MesenPRO was able to promote differentiation of both LSCs and BMSCs to ligament fibroblasts. Differentiation was further increased by TGF-β1. With increasing understanding of the effects of different culture media and growth factors, manipulation of stem cells in the desired direction for ligament tissue engineering can be achieved.
Collapse
Affiliation(s)
- Ming-Te Cheng
- Department of Surgery, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | | | | | | |
Collapse
|
67
|
Patrikoski M, Juntunen M, Boucher S, Campbell A, Vemuri MC, Mannerström B, Miettinen S. Development of fully defined xeno-free culture system for the preparation and propagation of cell therapy-compliant human adipose stem cells. Stem Cell Res Ther 2013; 4:27. [PMID: 23497764 PMCID: PMC3707027 DOI: 10.1186/scrt175] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Accepted: 03/04/2013] [Indexed: 01/29/2023] Open
Abstract
Introduction Adipose tissue is an attractive and abundant source of multipotent stem cells. Human adipose stem cells (ASCs) have shown to have therapeutic relevancy in diverse clinical applications. Nevertheless, expansion of ASCs is often necessary before performing clinical studies. Standard in vitro cell-culture techniques use animal-derived reagents that should be avoided in clinical use because of safety issues. Therefore, xeno- and serum-free (XF/SF) reagents are highly desirable for enhancing the safety and quality of the transplanted ASCs. Methods In the current study, animal component-free isolation and cell-expansion protocols were developed for ASCs. StemPro MSC SFM XF medium with either CELLstart™ CTS™ coating or Coating Matrix Kit were tested for their ability to support XF/SF growth. Basic stem-cell characteristics such as immunophenotype (CD3, CD11a, CD14, CD19, CD34, CD45RO, CD54, CD73, CD80, CD86, CD90, CD105, HLA-DR), proliferation, and differentiation potential were assessed in XF/SF conditions and compared with human serum (HS) or traditionally used fetal bovine serum (FBS) cultures. Results ASCs cultured in XF/SF conditions had significantly higher proliferation rates compared with HS/FBS cultures. Characteristic immunophenotypes of ASCs were maintained in every condition; however, cells expanded in XF/SF conditions showed significantly lower expression of CD54 (intercellular adhesion molecule 1, ICAM-1) at low passage number. Further, multilineage differentiation potential of ASCs was maintained in every culture condition. Conclusions Our findings demonstrated that the novel XF/SF conditions maintained the basic stem cell features of ASCs and the animal-free workflow followed in this study has great potential in clinical cell therapies.
Collapse
|
68
|
Xia YC, Redhu NS, Moir LM, Koziol-White C, Ammit AJ, Al-Alwan L, Camoretti-Mercado B, Clifford RL. Pro-inflammatory and immunomodulatory functions of airway smooth muscle: Emerging concepts. Pulm Pharmacol Ther 2013; 26:64-74. [DOI: 10.1016/j.pupt.2012.05.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 05/08/2012] [Accepted: 05/09/2012] [Indexed: 12/22/2022]
|
69
|
Song Y, Yun S, Yang HJ, Yoon AY, Kim H. Aggregation of Human Eyelid Adipose-derived Stem Cells by Human Body Fluids. Dev Reprod 2012; 16:339-51. [PMID: 25949109 PMCID: PMC4282241 DOI: 10.12717/dr.2012.16.4.339] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 12/21/2012] [Accepted: 12/24/2012] [Indexed: 01/22/2023]
Abstract
Fetal bovine serum (FBS) is the most frequently used serum for the cultivation of mammalian cells. However, since animal-derived materials might not be appropriate due to safety issues, allogeneic human serum (HS) has been used to replace FBS, particularly for the culture of human cells. While there has been a debate about the advantages of HS, its precise effect on human adult stem cells have not been clarified. The present study aimed to investigate the effect of HS on the human eyelid adipose stem cells (HEACs) in vitro. When HEACs were cultivated in a medium containing 10% HS, many cells moved into several spots and aggregated there. The phenomenon was observed as early as 9 days following 10% HS treatment, and 12 days following 5% HS plus 5% FBS treatment. However, the aggregation was never observed when the same cells were cultivated with 10% FBS or bovine serum albumin. To examine whether cell density might affect the aggregation, cells were seeded with different densities on 12-well dish. Until the beginning of aggregation, cells seeded at low densities exhibited the longest culture period of 16 days whereas cells seeded at high densities showed the shortest period of 9 days to form aggregation. The number of cells was 15.1±0.2×104 as the least for the low density group, and 29.3±2.8×104 as the greatest for the high density group. When human cord blood serum or normal bovine serum was examined for the same effect on HEACs, interestingly, cord blood serum induced the aggregation of cells whereas bovine serum treatment has never induced. When cells were cultivated with 10% HS for 9 days, they were obtained and analyzed by RT-PCR. Compared to FBS-cultivated HEACs, HS-cultivated HEACs did not express VIM, and less expressed GATA4, PALLD. On the other hand, HS-cultivated HEACs expressed MAP2 more than FBS-cultivated HEACs. In conclusion, human adult stem cells could move and form aggregates by the treatment with human body fluids.
Collapse
Affiliation(s)
- Yeonhwa Song
- Dept. of Biotechnology, Seoul Women's University, Seoul 139-774, Korea
| | - Sujin Yun
- Dept. of Biotechnology, Seoul Women's University, Seoul 139-774, Korea
| | - Hye Jin Yang
- Dept. of Biotechnology, Seoul Women's University, Seoul 139-774, Korea
| | - A Young Yoon
- Dept. of Biotechnology, Seoul Women's University, Seoul 139-774, Korea
| | - Haekwon Kim
- Dept. of Biotechnology, Seoul Women's University, Seoul 139-774, Korea
| |
Collapse
|
70
|
Pisciotta A, Riccio M, Carnevale G, Beretti F, Gibellini L, Maraldi T, Cavallini GM, Ferrari A, Bruzzesi G, De Pol A. Human serum promotes osteogenic differentiation of human dental pulp stem cells in vitro and in vivo. PLoS One 2012; 7:e50542. [PMID: 23209773 PMCID: PMC3510089 DOI: 10.1371/journal.pone.0050542] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 10/24/2012] [Indexed: 12/23/2022] Open
Abstract
Human dental pulp is a promising alternative source of stem cells for cell-based tissue engineering in regenerative medicine, for the easily recruitment with low invasivity for the patient and for the self-renewal and differentiation potential of cells. So far, in vitro culture of mesenchymal stem cells is usually based on supplementing culture and differentiation media with foetal calf serum (FCS). FCS is known to contain a great quantity of growth factors, and thus to promote cell attachment on plastic surface as well as expansion and differentiation. Nevertheless, FCS as an animal origin supplement may represent a potential means for disease transmission besides leading to a xenogenic immune response. Therefore, a significant interest is focused on investigating alternative supplements, in order to obtain a sufficient cell number for clinical application, avoiding the inconvenients of FCS use. In our study we have demonstrated that human serum (HS) is a suitable alternative to FCS, indeed its addition to culture medium induces a high hDPSCs proliferation rate and improves the in vitro osteogenic differentiation. Furthermore, hDPSCs-collagen constructs, pre-differentiated with HS-medium in vitro for 10 days, when implanted in immunocompromised rats, are able to restore critical size parietal bone defects. Therefore these data indicate that HS is a valid substitute for FCS to culture and differentiate in vitro hDPSCs in order to obtain a successful bone regeneration in vivo.
Collapse
Affiliation(s)
- Alessandra Pisciotta
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Riccio
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- * E-mail:
| | - Gianluca Carnevale
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesca Beretti
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Lara Gibellini
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Tullia Maraldi
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Gian Maria Cavallini
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Adriano Ferrari
- Department of Biomedical, Metabolic and Neuroscience, University of Modena and Reggio Emilia, Children Rehabilitation Special Unit, IRCCS Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
| | | | - Anto De Pol
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
71
|
Ho EA, Osooly M, Strutt D, Masin D, Yang Y, Yan H, Bally M. Characterization of long-circulating cationic nanoparticle formulations consisting of a two-stage PEGylation step for the delivery of siRNA in a breast cancer tumor model. J Pharm Sci 2012; 102:227-36. [PMID: 23132529 DOI: 10.1002/jps.23351] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 10/03/2012] [Accepted: 10/09/2012] [Indexed: 11/06/2022]
Abstract
Polyethylene glycol (PEG) has been used widely in liposomal formulations as a strategy to inhibit opsonization by plasma proteins and to prolong liposome plasma circulation time. PEG can be incorporated onto the surface of liposomes either during the spontaneous self-assembling process or inserted after vesicle formation. The advantages of employing the PEG postinsertion method include improved drug encapsulation efficiency and the ability to incorporate PEG conjugates for enhanced cell binding and uptake. In this study, we propose to evaluate a cationic lipid nanoparticle formulation containing two PEGylation steps: pre- and post-siRNA insertion. Our results indicate that formulations consisting of the extra PEG post-insertion step significantly increased siRNA circulation in the plasma by two-folds in comparison with the formulations consisting of only the single PEGylation step. Moreover, this formulation was able to efficiently carry siRNA to the tumor site, increase siRNA stability and significantly downregulate luciferase mRNA expression by >50% when compared with the controls in an intraperitoneal and subcutaneous breast cancer tumor model. Overall, our cationic lipid nanoparticle formulation displayed enhanced plasma circulation, reduced liver accumulation, enhanced tumor targeting, and effective gene knockdown--demonstrating excellent utility for the delivery of siRNA.
Collapse
Affiliation(s)
- Emmanuel A Ho
- University of Manitoba, Winnipeg, Manitoba, R3E 0T5, Canada.
| | | | | | | | | | | | | |
Collapse
|
72
|
Bornstein R, Macias MI, de la Torre P, Grande J, Flores AI. Human decidua-derived mesenchymal stromal cells differentiate into hepatic-like cells and form functional three-dimensional structures. Cytotherapy 2012; 14:1182-1192. [PMID: 22900961 DOI: 10.3109/14653249.2012.706706] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND AIMS Previously, we have shown that human decidua-derived mesenchymal stromal cells (DMSC) are mesenchymal stromal cells (MSC) with a clonal differentiation capacity for the three embryonic layers. The endodermal capacity of DMSC was revealed by differentiation into pulmonary cells. In this study, we examined the hepatic differentiation of DMSC. METHODS DMSC were cultured in hepatic differentiation media or co-cultured with murine liver homogenate and analyzed with phenotypic, molecular and functional tests. RESULTS AND CONCLUSIONS DMSC in hepatic differentiation media changed their fibroblast morphology to a hepatocyte-like morphology and later formed a 3-dimensional (3-D) structure or hepatosphere. Moreover, the hepatocyte-like cells and the hepatospheres expressed liver-specific markers such as synthesis of albumin (ALB), hepatocyte growth factor receptor (HGFR), α-fetoprotein (AFP) and cytokeratin-18 (CK-18), and exhibited hepatic functions including glycogen storage capacity and indocyanine green (ICG) uptake/secretion. Human DMSC co-cultured with murine liver tissue homogenate in a non-contact in vitro system showed hepatic differentiation, as evidenced by expression of AFP and ALB genes. The switch in the expression of these two genes resembled liver development. Indeed, the decrease in AFP and increase in ALB expression throughout the co-culture were consistent with the expression pattern observed during normal liver organogenesis in the embryo. Interestingly, AFP and ALB expression was significantly higher when DMSC were co-cultured with injured liver tissue, indicating that DMSC respond differently under normal and pathologic micro-environmental conditions. In conclusion, DMSC-derived hepatospheres and DMSC co-cultured with liver homogenate could be suitable in vitro models for toxicologic, developmental and pre-clinical hepatic regeneration studies.
Collapse
Affiliation(s)
- Rafael Bornstein
- Madrid Cord Blood Bank, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | | | | | | |
Collapse
|
73
|
Dave LYH, Nyland J, McKee PB, Caborn DNM. Mesenchymal stem cell therapy in the sports knee: where are we in 2011? Sports Health 2012; 4:252-7. [PMID: 23016095 PMCID: PMC3435924 DOI: 10.1177/1941738111427250] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Background: The relationship between biological tissue healing following knee injury or surgery and long-term clinical outcome has come to the forefront of sports medicine practice. This has led many knee surgeons to incorporate biologically mediated healing factors into the management of knee injuries. In particular, the clinical use of mesenchymal stem cells has opened new horizons. Evidence Acquisition: Relevant studies were identified through a search of PubMed from January 2000 to April 2011, combining the term mesenchymal stem cells with articular cartilage, anterior cruciate ligament, and meniscus. Relevant citations from the reference lists of selected studies were also reviewed. Results: Knee injury treatment with mesenchymal stem cells shows potential. Most reports represent animal model studies; few advances have been translated to human clinical applications. Conclusion: Mesenchymal stem cell use to promote healing following knee injury is likely to increase. There are scientific methodological concerns and ethical and legal issues regarding mesenchymal stem cell use for treating knee injuries.
Collapse
Affiliation(s)
- Lee Yee Han Dave
- Department of Orthopaedic Surgery, University of Louisville, Louisville, Kentucky
| | | | | | | |
Collapse
|
74
|
Redhu NS, Gounni AS. The high affinity IgE receptor (FcεRI) expression and function in airway smooth muscle. Pulm Pharmacol Ther 2012; 26:86-94. [PMID: 22580035 DOI: 10.1016/j.pupt.2012.04.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 04/24/2012] [Accepted: 04/27/2012] [Indexed: 12/27/2022]
Abstract
The airway smooth muscle (ASM) is no longer considered as merely a contractile apparatus and passive recipient of growth factors, neurotransmitters and inflammatory mediators signal but a critical player in the perpetuation and modulation of airway inflammation and remodeling. In recent years, a molecular link between ASM and IgE has been established through Fc epsilon receptors (FcεRs) in modulating the phenotype and function of these cells. Particularly, the expression of high affinity IgE receptor (FcεRI) has been noted in primary human ASM cells in vitro and in vivo within bronchial biopsies of allergic asthmatic subjects. The activation of FcεRI on ASM cells suggests a critical yet almost completely ignored network which may modulate ASM cell function in allergic asthma. This review is intended to provide a historical perspective of IgE effects on ASM and highlights the recent updates in the expression and function of FcεRI, and to present future perspectives of activation of this pathway in ASM cells.
Collapse
Affiliation(s)
- Naresh Singh Redhu
- Department of Immunology, Faculty of Medicine, University of Manitoba, 419 Apotex Centre, 750 McDermot Ave, Winnipeg, Manitoba, Canada R3E 0T5
| | | |
Collapse
|
75
|
Jung S, Panchalingam KM, Rosenberg L, Behie LA. Ex vivo expansion of human mesenchymal stem cells in defined serum-free media. Stem Cells Int 2012; 2012:123030. [PMID: 22645619 PMCID: PMC3356989 DOI: 10.1155/2012/123030] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 01/31/2012] [Indexed: 02/06/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) are presently being evaluated for their therapeutic potential in clinical studies to treat various diseases, disorders, and injuries. To date, early-phase studies have indicated that the use of both autologous and allogeneic hMSCs appear to be safe; however, efficacy has not been demonstrated in recent late-stage clinical trials. Optimized cell bioprocessing protocols may enhance the efficacy as well as safety of hMSC therapeutics. Classical media used for generating hMSCs are typically supplemented with ill-defined supplements such as fetal bovine serum (FBS) or human-sourced alternatives. Ideally, culture media are desired to have well-defined serum-free formulations that support the efficient production of hMSCs while maintaining their therapeutic and differentiation capacity. Towards this objective, we review here current cell culture media for hMSCs and discuss medium development strategies.
Collapse
Affiliation(s)
- Sunghoon Jung
- Pharmaceutical Production Research Facility (PPRF), Schulich School of Engineering, University of Calgary, Calgary, AB, Canada T2N 1N4
| | - Krishna M. Panchalingam
- Pharmaceutical Production Research Facility (PPRF), Schulich School of Engineering, University of Calgary, Calgary, AB, Canada T2N 1N4
| | | | - Leo A. Behie
- Pharmaceutical Production Research Facility (PPRF), Schulich School of Engineering, University of Calgary, Calgary, AB, Canada T2N 1N4
| |
Collapse
|
76
|
Characterization and modulation of fibroblast/endothelial cell co-cultures for the in vitro preformation of three-dimensional tubular networks. Cell Biol Int 2012; 35:1097-110. [PMID: 21418038 DOI: 10.1042/cbi20100718] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Various assays of different complexity are used in research on angiogenesis in health and disease. The results of these assays increasingly impact the field of tissue engineering because preformed microvascular networks may connect and conduct to the vascular system of the host, thereby helping us to support the survival of implanted cells and tissue constructs. An interesting model that supports the formation of EC (endothelial cells) tubular structures in vitro is based on co-culturing them with fibroblasts. Our initial multilayer approach was recently transferred into a three-dimensional spheroid model using HUVEC (human umbilical vein endothelial cells) as model cells. The aim of the present study is to further characterize, extend and validate this fibroblast/EC spheroid co-culture system. We have evaluated the model with a maximum size of 600-650 μm attained on day 3 from inoculation of 4×104 fibroblasts with 1×104 EC. Cell count and spheroid diameter significantly decreased as a function of time, but the EC network that developed over a period of 14 days in culture was clearly visible and viable, and central cell death was excluded. We successfully included HMVEC (human microvascular endothelial cells) of dermal origin in the system and replaced FBS (fetal bovine serum) with human AB serum, which positively impacted the EC network formation at optimized concentrations. The need for exogenous growth factors [VEGF (vascular endothelial growth factor), EGF (epithelial growth factor), bFGF (basic fibroblast growth factor) and IGF-1 (insulin-like growth factor-1)] routinely added to classical EC media was also assessed. The behaviour of both fibroblasts and EC in response to a combination of these exogenous growth factors differed critically in fibroblast/EC spheroid co-cultures compared with the same cells in the multilayer approach. VEGF was the most relevant exogenous factor for EC network formation in fibroblast/EC multilayers, but was ineffective in the spheroid system. IGF-1 was found, in general, to be dispensable; however, while it had a negative impact on EC networking in the presence of bFGF and EGF in the multilayer, it did not in the spheroid approach. We conclude that the critical determinants of EC network formation and cell survival are not universal, but have to be specifically optimized for each culture model.
Collapse
|
77
|
Brown PM, Hutchison JD, Crockett JC. Absence of glutamine supplementation prevents differentiation of murine calvarial osteoblasts to a mineralizing phenotype. Calcif Tissue Int 2011; 89:472-82. [PMID: 21972050 DOI: 10.1007/s00223-011-9537-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 09/11/2011] [Indexed: 10/17/2022]
Abstract
Osteoblasts in vitro differentiate from a proliferating to a mineralizing phenotype upon transfer to a medium rich in beta-glycerophosphate and ascorbic acid. The nutritional requirements of the cells at different stages of this differentiation process are not known. In other cell types, nutritional supplementation during surgery can improve the outcome in terms of speed of patient recovery and prognosis. There is therefore the potential for supplementation at the site of fracture repair or bone grafting with critical osteoblast nutritional factors to potentially accelerate healing. In this study we investigate which common cell nutrients are required for the proliferating and mineralizing stages of osteoblast differentiation. Medium containing 5.5 mM glucose was sufficient to achieve maximal proliferation of primary calvarial osteoblasts and human osteoblast cell lines, with some added benefit of additional glutamine supplementation. However, when cells were stimulated to mineralize, glucose was insufficient to support their energetic requirements. Only when cells were supplemented with glucose together with glutamine were high levels of osteocalcin expression observed together with mineralized nodules in culture, suggesting that this would be a useful combination to assess in cultures of primary human osteoblasts to determine whether it may have beneficial effects during fracture surgery, bone grafting, and fixation of uncemented arthroplasty implants.
Collapse
Affiliation(s)
- Philip M Brown
- Musculoskeletal Research Programme, Division of Applied Medicine, Institute of Medical Sciences, University of Aberdeen, UK
| | | | | |
Collapse
|
78
|
Abstract
BACKGROUND Avascular meniscal injuries are largely incapable of healing; the most common treatment remains partial meniscectomy despite the risk of subsequent osteoarthritis. Meniscal responses to injury are partially mediated through synovial activity and strategies have been investigated to encourage healing through stimulating or transplanting adjacent synovial lining. However, with their potential for chondrogenesis, synovial fibroblast-like stem cells hold promise for meniscal cartilage tissue engineering. QUESTIONS/PURPOSES Thus, specific purposes of this review were to (1) examine how the synovial intima and synoviomeniscal junction affect current meniscal treatment modalities; and (2) examine the components of tissue engineering (cells, scaffolds, bioactive agents, and bioreactors) in the specific context of how cells of synovial origin may be used for meniscal healing or regeneration. METHODS An online bibliographic search through PubMed was performed in March 2010. Studies were subjectively evaluated and reviewed if they addressed the question posed. Fifty-four resources were initially retrieved, which offered information on the chondrogenic potential of synovial-based cells that could prove valuable for meniscal fibrocartilage engineering. RESULTS Based on the positive effects of adjoining synovium on meniscal healing as used in some current treatment modalities, the chondrogenic potential of fibroblast-like stem cells of synovial origin make this cell source a promising candidate for cell-based tissue engineering strategies. CONCLUSIONS The abundance of autologous synovial lining, its ability to regenerate, and the potential of synovial-derived stem cells to produce a wide spectrum of chondral matrix components make it an ideal candidate for future meniscal engineering investigations.
Collapse
Affiliation(s)
- Derek B. Fox
- University of Missouri, Comparative Orthopaedic Laboratory, Columbia, MO USA ,University of Missouri, Veterinary Medical Teaching Hospital, 900 East Campus Drive, Columbia, MO USA
| | | |
Collapse
|
79
|
Jung S, Sen A, Rosenberg L, Behie LA. Human mesenchymal stem cell culture: rapid and efficient isolation and expansion in a defined serum-free medium. J Tissue Eng Regen Med 2011; 6:391-403. [PMID: 21744510 DOI: 10.1002/term.441] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2010] [Accepted: 05/12/2011] [Indexed: 11/11/2022]
Abstract
Human mesenchymal stem cells (hMSCs) are typically obtained for research or therapeutic applications by isolating and subculturing adherent cells from bone marrow on tissue-culture substrates using growth media. The purity and properties of the resulting populations can be affected greatly by the conditions under which they are cultured. Fetal bovine serum (FBS), although ill-defined, has been widely used as a critical requirement for conventional hMSC culture. However, a defined serum-free medium would greatly facilitate the development of robust, clinically acceptable bioprocesses for reproducibly generating quality-assured cells. The present study provides evidence demonstrating that a defined serum-free medium (PPRF-msc6) shows several beneficial features over a conventional FBS-containing medium for the production of hMSCs. When compared to control FBS-based cultures, PPRF-msc6 medium supported the derivation of hMSCs from primary cultures of bone marrow cells in a more rapid and consistent manner. Furthermore, hMSCs cultured in PPRF-msc6 exhibited: (a) a greater colony-forming capacity in primary as well as passaged cultures; (b) negligible lag phase and explicit exponential growth; (c) lower population doubling times (21-26 h vs 35-38 h between passage levels 1 and 10); (d) a greater number of population doublings (62 ± 4 vs 43 ± 2; over a 2 month period); and (e) a higher degree of homogeneity in size. Our data demonstrate that PPRF-msc6 is an important development which opens the door for the rapid, efficient and reproducible production of hMSCs in clinical settings.
Collapse
Affiliation(s)
- Sunghoon Jung
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta, Canada
| | | | | | | |
Collapse
|
80
|
Fan M, Chen W, Liu W, Du GQ, Jiang SL, Tian WC, Sun L, Li RK, Tian H. The effect of age on the efficacy of human mesenchymal stem cell transplantation after a myocardial infarction. Rejuvenation Res 2010; 13:429-38. [PMID: 20583954 DOI: 10.1089/rej.2009.0986] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Clinical trials of cardiac cell therapy have indicated limited benefits in aging patients, even though preclinical studies using young animals consistently reported significant improvements. Animal studies have demonstrated reduced efficacy of donor cells isolated from older individuals. Here, we evaluated the effects of donor age on the function of human mesenchymal stem cells (hMSCs) in the context of cell therapy for ischemic cardiomyopathy. METHODS In vitro, we compared the growth and clonogenic potential of hMSCs isolated from young or old patients (1-5 vs. 50-70 years old). In vivo, we injected young or old hMSCs (2.0 x 10(6)) (or medium) into the infarcted myocardia of immunosuppressed rats immediately after coronary artery ligation (myocardial infarction [MI]). We assessed cardiac function (echocardiography) at 1, 2, and 4 weeks after MI, and myocardial matrix metalloproteinase-2 (MMP-2), MMP-9, and tissue inhibitor of matrix metalloproteinase-3 (TIMP-3) levels at 1 week. RESULTS In vitro, growth and colony-forming unit fibroblast (CFU-F) formation were markedly diminished in old hMSCs (p < 0.001 and p < 0.05, respectively, vs. young). In vivo, compared with old hMSCs or medium, young hMSCs best preserved ejection fraction, fractional shortening (p < 0.05), and left ventricular end-diastolic and end-systolic volumes (p < 0.01). Recipients of young hMSCs also exhibited increases in vascular density and TIMP-3 protein levels and activity (p < 0.05), and decreases in MMP protein levels and activity (p < 0.05). CONCLUSIONS The regenerative capacity of hMSCs was significantly influenced by age. Transplanting young hMSCs improved functional outcomes after an MI by preventing matrix degradation and promoting angiogenesis. The clinical implication is that aged patients require an optimized source of stem cells for treatment.
Collapse
Affiliation(s)
- Ming Fan
- Division of Cardiovascular Surgery, Second Affiliated Hospital, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Harbin, China
| | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Eve DJ, Fillmore RW, Borlongan CV, Sanberg PR. Stem cell research in cell transplantation: sources, geopolitical influence, and transplantation. Cell Transplant 2010; 19:1493-509. [PMID: 21054954 DOI: 10.3727/096368910x540612] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
If the rapidly progressing field of stem cell research reaches its full potential, successful treatments and enhanced understanding of many diseases are the likely results. However, the full potential of stem cell science will only be reached if all possible avenues can be explored and on a worldwide scale. Until 2009, the US had a highly restrictive policy on obtaining cells from human embryos and fetal tissue, a policy that pushed research toward the use of adult-derived cells. Currently, US policy is still in flux, and retrospective analysis does show the US lagging behind the rest of the world in the proportional increase in embryonic/fetal stem cell research. The majority of US studies being on either a limited number of cell lines, or on cells derived elsewhere (or funded by other sources than Federal) rather than on freshly isolated embryonic or fetal material. Neural, mesenchymal, and the mixed stem cell mononuclear fraction are the most commonly investigated types, which can generally be classified as adult-derived stem cells, although roughly half of the neural stem cells are fetal derived. Other types, such as embryonic and fat-derived stem cells, are increasing in their prominence, suggesting that new types of stem cells are still being pursued. Sixty percent of the reported stem cell studies involved transplantation, of which over three quarters were allogeneic transplants. A high proportion of the cardiovascular systems articles were on allogeneic transplants in a number of different species, including several autologous studies. A number of pharmaceutical grade stem cell products have also recently been tested and reported on. Stem cell research shows considerable promise for the treatment of a number of disorders, some of which have entered clinical trials; over the next few years it will be interesting to see how these treatments progress in the clinic.
Collapse
Affiliation(s)
- David J Eve
- Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL 33612, USA.
| | | | | | | |
Collapse
|
82
|
Shin KS, Lee HJ, Jung J, Cha DH, Kim GJ. Culture and in vitro hepatogenic differentiation of placenta-derived stem cells, using placental extract as an alternative to serum. Cell Prolif 2010; 43:435-44. [PMID: 20887550 DOI: 10.1111/j.1365-2184.2010.00693.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Translational research using adult stem cells derived from various tissues has been highlighted in cell-based therapy. However, there are many limitations to using conventional culture systems of adult stem cells for clinically applicability, including limited combinations of cytokines and use of nutrients derived from animals. Here, we have investigated the effects of placental extract (PE) for culture of placenta-derived stem cells (PDSCs) as well as their potential for hepatogenic differentiation. MATERIALS AND METHODS Placental extract, extracted using water-soluble methods, was used as a supplement for culture of PDSCs. Cell viability was determined using the MTT assay, and cytokine assay was performed using Luminex assay kit. Gene expression, indocyanine green (ICG) up-take, PAS (Periodic Acid-Schiff) staining and urea production were also analysed. RESULTS The placental extract contained several types of cytokine and chemokine essential for maintenance and differentiation of stem cells. Expression of stemness markers in PDSCs cultured with PE is no different from that of PDSCs cultured with foetal bovine serum (FBS). After hepatogenic differentiation, expression patterns for hepatocyte-specific markers in PDSCs cultured with PE were consistent and potential for hepatogenic differentiation of PDSCs cultured with PE was similar to that of PDSCs cultured with FBS, as shown by PAS staining and urea production assays. CONCLUSIONS Our findings revealed that placental extract could be used as a new component for culture of adult stem cells, as well as for development of human-based medium, in translational research for regenerative medicine.
Collapse
Affiliation(s)
- K S Shin
- Department of Biomedical Science, CHA University, Seoul, South Korea
| | | | | | | | | |
Collapse
|
83
|
Han HS, Lee S, Kim JH, Seong SC, Lee MC. Changes in chondrogenic phenotype and gene expression profiles associated with the in vitro expansion of human synovium-derived cells. J Orthop Res 2010; 28:1283-91. [PMID: 20225285 DOI: 10.1002/jor.21129] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We undertook this study to characterize changes in the proliferative capacities, chondrogenic phenotypes, and gene expression profiles of human synovium-derived progenitor cells from osteoarthritic patients during in vitro expansion. Cells isolated from osteoarthritic synovia were cultured, and growth rates during serial passages were evaluated. Surface molecule expressions were determined by flow cytometry and cytogenetic analyses were performed. After chondrogenic differentiation in cell pellets, we evaluated type II collagen and glycosaminoglycan (GAG) synthesis. To assess whether the in vitro expansion of synovium-derived cells affects gene expression, we performed microarray analyses on cells at passage 0, 1, 2, 4, 6, and 8. Synovium-derived cells were rapidly expanded in vitro through passage 8 (about 130 days), and after passage 6, the proliferation rates decreased slightly with a wide range of individual variations. The expressions of CD166, CD49a, and CD106 decreased, whereas those of CD10, CD29, CD44, CD73, CD90, and CD105 showed no significant change. Karyotype analysis revealed no evidence of chromosome abnormalities. The staining of type II collagen and GAG in differentiated cell pellets showed rapid weakening. Genome-wide microarray analysis showed that synovium-derived cells from late passages over-expressed genes associated with cell cycle prolongation and cell aging, and less-expressed genes associated with cell growth stimulation. The in vitro expansion of synovium-derived cells was accompanied with decreased proliferative capacity and the chondrogenic phenotype, which might be modulated by change in gene expression patterns.
Collapse
Affiliation(s)
- Hyuk-Soo Han
- Department of Orthopaedic Surgery, Seoul National University, College of Medicine, 28 Yongondong, Chongnogu, 110-744 Seoul, South Korea
| | | | | | | | | |
Collapse
|
84
|
Apoptosis in chondrogenesis of human mesenchymal stem cells: effect of serum and medium supplements. Apoptosis 2010; 15:439-49. [PMID: 19949977 DOI: 10.1007/s10495-009-0431-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Apoptosis is an inevitable process during development and is evident in the formation of articular cartilage and endochondral ossification of growth plate. Mesenchymal stem cells (MSCs) can serve as alternative sources for cell therapy in focal chondral lesions or diffuse osteoarthritis. But there are few, if any, studies investigating apoptosis during chondrogenesis by MSCs. The aim of this study was to find the better condition to prevent apoptosis during chondrogenesis by MSCs. Apoptosis were evaluated in MSCs induced in different chondrogenic media by the use of Annexin V, TUNEL staining, lysosomal labeling with lysotracker and immunostaining of apoptotic markers. We found apparent apoptosis was demonstrated by Annexin V, TUNEL staining and lysosomal labeling during chondrogenesis. Meanwhile, the degree of apoptosis was related to the reagents of the defined chondrogenic medium. Adding serum in medium increased apoptosis, however, TGF-beta1 inhibited apoptosis. The apoptosis was associated with the activation of caspase-3, the increase in the Bax/Bcl-2 ratio, the loss of lysosomal integrity, and the increase of PARP-cleavage. Pro-inflammatory cytokines, IL-1alpha, IL-1beta and TNFalpha did not induce any increase in apoptosis. Interestingly, the inhibition of apoptosis by serum free medium supplemented with ITS was also associated with an increase in the expression of type II collagen, and a decrease in the expression of type X collagen, Runx2, and other osteogenic genes, while TGF-beta1 increased the expression of Sox9, type II and type X collagen and decreased the expression of osteogenic genes. These data suggest apoptosis occurs during chondrogenesis by MSCs by cell death intrinsic pathway activation and this process may be modulated by culture conditions.
Collapse
|
85
|
Hess R, Douglas T, Myers KA, Rentsch B, Rentsch C, Worch H, Shrive NG, Hart DA, Scharnweber D. Hydrostatic pressure stimulation of human mesenchymal stem cells seeded on collagen-based artificial extracellular matrices. J Biomech Eng 2010; 132:021001. [PMID: 20370238 DOI: 10.1115/1.4000194] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Human mesenchymal stem cells (hMSCs) from bone marrow are considered a promising cell source for bone tissue engineering applications because of their ability to differentiate into cells of the osteoblastic lineage. Mechanical stimulation is able to promote osteogenic differentiation of hMSC; however, the use of hydrostatic pressure (HP) has not been well studied. Artificial extracellular matrices containing collagen and chondroitin sulfate (CS) have promoted the expression of an osteoblastic phenotype by hMSCs. However, there has been little research into the combined effects of biochemical stimulation by matrices and simultaneous mechanical stimulation. In this study, artificial extracellular matrices generated from collagen and/or CS were coated onto polycaprolactone-co-lactide substrates, seeded with hMSCs and subjected to cyclic HP at various time points during 21 days after cell seeding to investigate the effects of biochemical, mechanical, and combined biochemical and mechanical stimulations. Cell differentiation was assessed by analyzing the expression of alkaline phosphatase (ALP) at the protein- and mRNA levels, as well as for calcium accumulation. The timing of HP stimulation affected hMSC proliferation and expression of ALP activity. HP stimulation after 6 days was most effective at promoting ALP activity. CS-containing matrices promoted the osteogenic differentiation of hMSCs. A combination of both CS-containing matrices and cyclic HP yields optimal effects on osteogenic differentiation of hMSCs on scaffolds compared with individual responses.
Collapse
Affiliation(s)
- Ricarda Hess
- Institute of Material Science, Max Bergmann Center of Biomaterials, Technische Universitat Dresden, 01069 Dresden, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Furlani D, Li W, Pittermann E, Klopsch C, Wang L, Knopp A, Jungebluth P, Thedinga E, Havenstein C, Westien I, Ugurlucan M, Li RK, Ma N, Steinhoff G. A transformed cell population derived from cultured mesenchymal stem cells has no functional effect after transplantation into the injured heart. Cell Transplant 2009; 18:319-31. [PMID: 19558780 DOI: 10.3727/096368909788534906] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) are multipotent cells characterized by their self-renewal and differentiation potential. Accumulating clinical and preclinical evidence indicate MSCs are a promising cell source for regenerative medical therapies. However, undesirable immortalization, spontaneous transformation, and tumorigenic potential from long-term cultured MSCs have been reported in human and mouse. We report rat MSCs isolated from young donors could undergo transformation in early passage culture. We aimed to characterize the transformed population and determine their therapeutic effects after intracardiac transplantation in the infarcted myocardium. MSCs were isolated from bone marrow of Lewis rats according to standard protocols and cultured under standard conditions. Phenotype of growing cells was assessed by flow cytometry. Following acute myocardial infarction in rats, cells were delivered by intracardiac injection. Cardiac functions were assessed by pressure-volume loops. Infarction size and pathologic effects were evaluated after 6 weeks. The abnormal colonies were detected in culture as early at passage 3. They were noted to appear as distinctly different morphology from typical MSCs, which changed from a normal elongated spindle shape to a compact abnormal morphology. They exhibited rapid cell proliferation. Some subclones lost contact inhibition of cell division and formed multilayer aggregates. Chromosomal instability was detected. They were devoid of surface markers CD29, CD44, CD90, and CD117. Furthermore, there was no significant improvement on infarction size and cardiac function 6 weeks after cell transplantation. Our study highlights the need for establishment of biosafety criteria in regulating culture- expanded MSCs to achieve the full clinical therapeutic benefits.
Collapse
Affiliation(s)
- Dario Furlani
- Department of Cardiac Surgery, University Rostock, Rostock, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Zheng C, Yang S, Guo Z, Liao W, Zhang L, Yang R, Han ZC. Human multipotent mesenchymal stromal cells from fetal lung expressing pluripotent markers and differentiating into cell types of three germ layers. Cell Transplant 2009; 18:1093-109. [PMID: 19650974 DOI: 10.3727/096368909x12483162197042] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) are a promising cell type for cell transplantation; however, their utilization remains limited until the availability of adequate alternative sources of MSCs and the thorough understanding of the biology of MSCs isolated from various sources are realized. Fetal lung has been identified as a rich source of MSCs. To explore the therapeutic potential of passaged fetal lung MSCs (FLMSCs), the present study evaluated their growth kinetics, telomere length, karyotype, immunophenotype, and the differentiation potential during in vitro expansion. FLMSCs could be easily amplified in vitro with no significant shorting of telomere length and had a normal karyotype. No significant differences between passage 5 or passage 25 were observed in the immunophenotype analysis using flow cytometry. Moreover, flow cytometry results provided the first demonstration, to our knowledge, that FLMSCs stably expressed pluripotent markers including Oct4, Nanog, Sox2, TRA-1-60, c-Myc, and SSEA-4 through 25 passages. In vitro differentiation studies as identified by confocal microscopy, flow cytometry, RT-PCR, and immunohistochemistry showed that FLMSCs had extended capacity of differentiating into mesodermal, ectodermal, and endodermal lineages, and that their potential for adipogenic, osteogenic, and chondrogenic differentiation may be maintained over 25 passages. Furthermore, osteogenic and chondrogenic differentiation was used as an indicator of their differentiation capability in vivo, as evidenced by ectopic bone and cartilage formation. In summary, these results suggest that FLMSCs are a primitive population and that their extensive in vitro expansion does not involve significant functional modification of the cells, including morphology, growth, karyotype, immunophenotype, and mesodermal differentiation potential. Hence, FLMSCs might constitute an attractive cell resource for cell transplantation to induce regeneration of damaged tissues/organs.
Collapse
Affiliation(s)
- Cuiling Zheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, P. R. China
| | | | | | | | | | | | | |
Collapse
|
88
|
Fan J, Varshney RR, Ren L, Cai D, Wang DA. Synovium-derived mesenchymal stem cells: a new cell source for musculoskeletal regeneration. TISSUE ENGINEERING PART B-REVIEWS 2009; 15:75-86. [PMID: 19196118 DOI: 10.1089/ten.teb.2008.0586] [Citation(s) in RCA: 163] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Ever since synovium-derived mesenchymal stem cells (SMSCs) were first identified and successfully isolated in 2001, as a brand new member in MSC families, they have been increasingly regarded as a promising therapeutic cell species for musculoskeletal regeneration, particularly for reconstructions of cartilage, bones, tendons, and muscles. Besides the general multipotency in common among the MSC community, SMSCs excel other sourced MSCs in higher ability of proliferation and superiority in chondrogenesis. This review summarizes the latest advances in SMSC-related studies covering their specific isolation methodologies, biological insights, and practical applications in musculoskeletal therapeutics of which an emphasis is cast on engineered chondrogenesis.
Collapse
Affiliation(s)
- Jiabing Fan
- Center for Stem Cell Biology and Tissue Engineering, Sun Yat-sen University, Guangzhou, PR China
| | | | | | | | | |
Collapse
|