201
|
Pak J, Chang JJ, Lee JH, Lee SH. Safety reporting on implantation of autologous adipose tissue-derived stem cells with platelet-rich plasma into human articular joints. BMC Musculoskelet Disord 2013; 14:337. [PMID: 24289766 PMCID: PMC4219585 DOI: 10.1186/1471-2474-14-337] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Accepted: 11/27/2013] [Indexed: 02/06/2023] Open
Abstract
Background Adipose tissue-derived stem cells (ADSCs), a type of mesenchymal stem cells (MSCs), have great potential as therapeutic agents in regenerative medicine. Numerous animal studies have documented the multipotency of ADSCs, showing their capabilities to differentiate into tissues such as muscle, bone, cartilage, and tendon. However, the safety of autologous ADSC injections into human joints is only beginning to be understood and the data are lacking. Methods Between 2009 and 2010, 91 patients were treated with autologous ADSCs with platelet-rich plasma (PRP) for various orthopedic conditions. Stem cells in the form of stromal vascular fraction (SVF) were injected with PRP into various joints (n = 100). All patients were followed for symptom improvement with visual analog score (VAS) at one month and three months. Approximately one third of the patients were followed up with third month magnetic resonance imaging (MRI) of the injected sites. All patients were followed up by telephone questionnaires every six months for up to 30 months. Results The mean follow-up time for all patients was 26.62 ± 0.32 months. The follow-up time for patients who were treated in 2009 and early 2010 was close to three years. The relative mean VAS of patients at the end of one month follow-up was 6.55 ± 0.32, and at the end of three months follow-up was 4.43 ± 0.41. Post-procedure MRIs performed on one third of the patients at three months failed to demonstrate any tumor formation at the implant sites. Further, no tumor formation was reported in telephone long-term follow-ups. However, swelling of injected joints was common and was thought to be associated with death of stem cells. Also, tenosinovitis and tendonitis in elderly patients, all of which were either self-limited or were remedied with simple therapeutic measures, were common as well. Conclusions Using both MRI tracking and telephone follow ups in 100 joints in 91 patients treated, no neoplastic complications were detected at any ADSC implantation sites. Based on our longitudinal cohort, the autologous and uncultured ADSCs/PRP therapy in the form of SVF could be considered to be safe when used as percutaneous local injections.
Collapse
Affiliation(s)
- Jaewoo Pak
- National Leading Research Laboratory, Department of Biological Sciences, Myongji University, 116 Myongjiro, Yongin, Gyeonggido 449-728, Republic of Korea.
| | | | | | | |
Collapse
|
202
|
Sousa BR, Parreira RC, Fonseca EA, Amaya MJ, Tonelli FMP, Lacerda SMSN, Lalwani P, Santos AK, Gomes KN, Ulrich H, Kihara AH, Resende RR. Human adult stem cells from diverse origins: An overview from multiparametric immunophenotyping to clinical applications. Cytometry A 2013; 85:43-77. [DOI: 10.1002/cyto.a.22402] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 09/27/2013] [Accepted: 10/01/2013] [Indexed: 02/06/2023]
Affiliation(s)
- Bruna R. Sousa
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Ricardo C. Parreira
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Emerson A Fonseca
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Maria J. Amaya
- Department of Internal Medicine, Section of Digestive Diseases; Yale University School of Medicine; New Haven Connecticut
| | - Fernanda M. P. Tonelli
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Samyra M. S. N. Lacerda
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Pritesh Lalwani
- Faculdade de Ciências Farmacêuticas; Universidade Federal do Amazonas; Manaus AM Brazil
| | - Anderson K. Santos
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Katia N. Gomes
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Henning Ulrich
- Departamento de Bioquímica; Instituto de Química, Universidade de São Paulo; São Paulo SP Brazil
| | - Alexandre H. Kihara
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição; Universidade Federal do ABC; Santo André SP Brazil
| | - Rodrigo R. Resende
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| |
Collapse
|
203
|
Ying X, Chen X, Cheng S, Guo X, Chen H, Xu HZ. Phosphoserine promotes osteogenic differentiation of human adipose stromal cells through bone morphogenetic protein signalling. Cell Biol Int 2013; 38:309-17. [PMID: 24155130 DOI: 10.1002/cbin.10203] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Accepted: 10/04/2013] [Indexed: 01/03/2023]
Abstract
Phosphoserine has potential effectiveness as a simple substrate in preparing bone replacement materials, which could enhance bone forming ability. However, there is a need to investigate the independent effect of phosphoserine on osteogenic differentiation of human adipose stem cells (hADSCs). hADSCs were cultured in an osteogenic medium with phosphoserine. Cell proliferation was analysed by CCK8 and osteogenic differentiation was measured by alkaline phosphatase (ALP) activity, von Kossa staining and real time-polymerase chain reaction (RT-PCR). No stimulatory effect of phosphoserine on cell proliferation was noted at Days 1, 4 and 7. Deposition of calcium increased after the addition of phosphoserine. mRNA expression of type I collagen (COL-I), alkaline phosphatase (ALP), osteocalcin (OCN), Osterix, bone morphogenetic protein-2 (BMP-2) and RUNX2 increased markedly with phosphoserine treatment. The BMP-2 antagonist, noggin, and its receptor kinase inhibitors, dorsomorphin and LDN-193189, attenuated phosphoserine-promoted ALP activity. BMP-responsive and Runx2-responsive reporters were activated by phosphoserine treatment. Thus phosphoserine can promote osteogenic differentiation of hADSCs, probably by activating BMP and Runx2 pathways, which could be a promising approach for enhancing osteogenic capacity of cell-based construction in bone tissue engineering.
Collapse
Affiliation(s)
- Xiaozhou Ying
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Wenzhou Medical College, Wenzhou, 325000, China
| | | | | | | | | | | |
Collapse
|
204
|
Stem cells in plastic surgery: a review of current clinical and translational applications. Arch Plast Surg 2013; 40:666-75. [PMID: 24286038 PMCID: PMC3840172 DOI: 10.5999/aps.2013.40.6.666] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Revised: 09/24/2013] [Accepted: 09/25/2013] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Stem cells are a unique cell population characterized by self-renewal and cellular differentiation capabilities. These characteristics, among other traits, make them an attractive option for regenerative treatments of tissues defects and for aesthetic procedures in plastic surgery. As research regarding the isolation, culture and behavior of stem cells has progressed, stem cells, particularly adult stem cells, have shown promising results in both translational and clinical applications. METHODS The purpose of this review is to evaluate the applications of stem cells in the plastic surgery literature, with particular focus on the advances and limitations of current stem cell therapies. Different key areas amenable to stem cell therapy are addressed in the literature review; these include regeneration of soft tissue, bone, cartilage, and peripheral nerves, as well as wound healing and skin aging. RESULTS The reviewed studies demonstrate promising results, with favorable outcomes and minimal complications in the cited cases. In particular, adipose tissue derived stem cell (ADSC) transplants appear to provide effective treatment options for bony and soft tissue defects, and non-healing wounds. ADSCs have also been shown to be useful in aesthetic surgery. CONCLUSIONS Further studies involving both the basic and clinical science aspects of stem cell therapies are warranted. In particular, the mechanism of action of stem cells, their interactions with the surrounding microenvironment and their long-term fate require further elucidation. Larger randomized trials are also necessary to demonstrate the continued safety of transplanted stem cells as well as the efficacy of cellular therapies in comparison to the current standards of care.
Collapse
|
205
|
Wolff J, Sándor GK, Miettinen A, Tuovinen VJ, Mannerström B, Patrikoski M, Miettinen S. GMP-level adipose stem cells combined with computer-aided manufacturing to reconstruct mandibular ameloblastoma resection defects: Experience with three cases. Ann Maxillofac Surg 2013; 3:114-25. [PMID: 24205470 PMCID: PMC3814659 DOI: 10.4103/2231-0746.119216] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Background: The current management of large mandibular resection defects involves harvesting of autogenous bone grafts and repeated bending of generic reconstruction plates. However, the major disadvantage of harvesting large autogenous bone grafts is donor site morbidity and the major drawback of repeated reconstruction plate bending is plate fracture and difficulty in reproducing complex facial contours. The aim of this study was to describe reconstruction of three mandibular ameloblastoma resection defects using tissue engineered constructs of beta-tricalcium phosphate (β-TCP) granules, recombinant human bone morphogenetic protein-2 (rhBMP-2), and Good Manufacturing Practice (GMP) level autologous adipose stem cells (ASCs) with progressively increasing usage of computer-aided manufacturing (CAM) technology. Materials and Methods: Patients’ three-dimensional (3D) images were used in three consecutive patients to plan and reverse-engineer patient-specific saw guides and reconstruction plates using computer-aided additive manufacturing. Adipose tissue was harvested from the anterior abdominal walls of three patients before resection. ASCs were expanded ex vivo over 3 weeks and seeded onto a β-TCP scaffold with rhBMP-2. Constructs were implanted into patient resection defects together with rapid prototyped reconstruction plates. Results: All three cases used one step in situ bone formation without the need for an ectopic bone formation step or vascularized flaps. In two of the three patients, dental implants were placed 10 and 14 months following reconstruction, allowing harvesting of bone cores from the regenerated mandibular defects. Histological examination and in vitro analysis of cell viability and cell surface markers were performed and prosthodontic rehabilitation was completed. Discussion: Constructs with ASCs, β-TCP scaffolds, and rhBMP-2 can be used to reconstruct a variety of large mandibular defects, together with rapid prototyped reconstruction hardware which supports placement of dental implants.
Collapse
Affiliation(s)
- Jan Wolff
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland ; Department of Eye, Ear and Oral Diseases, Tampere University Hospital, Tampere, Finland
| | | | | | | | | | | | | |
Collapse
|
206
|
Adipose-derived mesenchymal cells for bone regereneration: state of the art. BIOMED RESEARCH INTERNATIONAL 2013; 2013:416391. [PMID: 24307997 PMCID: PMC3838853 DOI: 10.1155/2013/416391] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 09/25/2013] [Indexed: 12/21/2022]
Abstract
Adipose tissue represents a hot topic in regenerative medicine because of the tissue source abundance, the relatively easy retrieval, and the inherent biological properties of mesenchymal stem cells residing in its stroma. Adipose-derived mesenchymal stem cells (ASCs) are indeed multipotent somatic stem cells exhibiting growth kinetics and plasticity, proved to induce efficient tissue regeneration in several biomedical applications. A defined consensus for their isolation, classification, and characterization has been very recently achieved. In particular, bone tissue reconstruction and regeneration based on ASCs has emerged as a promising approach to restore structure and function of bone compromised by injury or disease. ASCs have been used in combination with osteoinductive biomaterial and/or osteogenic molecules, in either static or dynamic culture systems, to improve bone regeneration in several animal models. To date, few clinical trials on ASC-based bone reconstruction have been concluded and proved effective. The aim of this review is to dissect the state of the art on ASC use in bone regenerative applications in the attempt to provide a comprehensive coverage of the topics, from the basic laboratory to recent clinical applications.
Collapse
|
207
|
Abstract
Orthopedic injuries are common and a source of much misery and economic stress. Several relevant tissues, such as cartilage, meniscus, and intra-articular ligaments, do not heal. And even bone, which normally regenerates spontaneously, can fail to mend. The regeneration of orthopedic tissues requires 4 key components: cells, morphogenetic signals, scaffolds, and an appropriate mechanical environment. Although differentiated cells from the tissue in question can be used, most cellular research focuses on the use of mesenchymal stem cells. These can be retrieved from many different tissues, and one unresolved question is the degree to which the origin of the cells matters. Embryonic and induced pluripotent stem cells are also under investigation. Morphogenetic signals are most frequently supplied by individual recombinant growth factors or native mixtures provided by, for example, platelet-rich plasma; mesenchymal stem cells are also a rich source of trophic factors. Obstacles to the sustained delivery of individual growth factors can be addressed by gene transfer or smart scaffolds, but we still lack detailed, necessary information on which delivery profiles are needed. Scaffolds may be based on natural products, synthetic materials, or devitalized extracellular matrix. Strategies to combine these components to regenerate tissue can follow traditional tissue engineering practices, but these are costly, cumbersome, and not well suited to treating large numbers of individuals. More expeditious approaches make full use of intrinsic biological processes in vivo to avoid the need for ex vivo expansion of autologous cells and multiple procedures. Clinical translation remains a bottleneck.
Collapse
Affiliation(s)
- Christopher H Evans
- Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Collaborative Research Center, AO Foundation, Davos, Switzerland.
| |
Collapse
|
208
|
Bellotti C, Stanco D, Ragazzini S, Romagnoli L, Martella E, Lazzati S, Marchetti C, Donati D, Lucarelli E. Analysis of the karyotype of expanded human adipose-derived stem cells for bone reconstruction of the maxillo-facial region. Int J Immunopathol Pharmacol 2013; 26:3-9. [PMID: 24046944 DOI: 10.1177/03946320130260s102] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Mesenchymal stem cells (MSC) and adipose-derived stem cells (ASC) were recently proposed for bone maxillofacial reconstruction in association with biomaterials. For this application MSC must be ex-vivo expanded in order to obtain, for a given volume of implanted biomaterial, a relevant number of bone forming cells. Previously conducted pre-clinical studies suggested that a concentration of 6 x 10(8) ASC associated with 900 mg of anorganic bovine bone (ABB) could be effective for human maxillary sinus floor elevation. A keystone issue to guarantee the quality and safety of Advanced Therapy Medicinal Products containing expanded MSC and ASC is their chromosome stability in culture: this topic has been widely investigated and conflicting results have been published. Abnormal karyotype of human ex-vivo expanded MSC and ASC was found by some authors, while, at the same time, several other studies showed the MSC and ASC karyotype to be normal. It is therefore important that all the results obtained on MSC and ASC karyotype analysis be published. Given this context, the aim of this manuscript, aim of this manuscript is to verify the karyotype stability of ASC in view of their applications in clinical trials. ASC obtained from the adipose tissue of 4 donors were expanded over extended culture time. Based on previous ASC expansions we hypothesized to be able to obtain 6 x 10(8) cells by passage 7. Karyotype analysis of 30 metaphases was planned to be investigated at passage 2, 7, and 15 in all the cultures. No abnormalities were found in the karyotype of two donors at all the passages tested, while a translocation was found in 2 metaphases of a donor at passage 7, but not at passage 15, and in the fourth donor in 5 metaphases a trisomy was found at passage 15. Chromosomal abnormalities were detected only after extended ASC expansion. Whether these anomalies can be related to risk for the patient's safety will have to be demonstrated by in-vivo studies.
Collapse
Affiliation(s)
- C Bellotti
- Osteoarticular Regeneration Laboratory, Rizzoli Orthopaedic Institute, 40136 Bologna, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
209
|
Kyllönen L, Haimi S, Säkkinen J, Kuokkanen H, Mannerström B, Sándor GKB, Miettinen S. Exogenously added BMP-6, BMP-7 and VEGF may not enhance the osteogenic differentiation of human adipose stem cells. Growth Factors 2013; 31:141-53. [PMID: 23879371 DOI: 10.3109/08977194.2013.817404] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In the present study bone morphogenetic protein (BMP)-6 alone or in synergy with BMP-7 and vascular endothelial growth factor (VEGF) were tested with human adipose stem cells (hASCs) seeded on cell culture plastic or 3D bioactive glass. Osteogenic medium (OM) was used as a positive control for osteogenic differentiation. The same growth factor groups were also tested combined with OM. None of the growth factor treatments could enhance the osteogenic differentiation of hASCs in 3D- or 2D-culture compared to control or OM. In 3D-culture OM promoted significantly total collagen production, whereas in 2D-culture OM induced high total ALP activity and mineralization compared to control and growth factors groups, but also high cell proliferation. In this study, hASCs did not respond to exogenously added growth although various parameters of the study set-up may have affected these findings contradictory to the previous literature.
Collapse
Affiliation(s)
- Laura Kyllönen
- Adult Stem Cells, Institute of Biomedical Technology, Biokatu, University of Tampereo , Finland
| | | | | | | | | | | | | |
Collapse
|
210
|
Mizuno H. Adipose-derived stem cells for regenerative medicine in the field of plastic and reconstructive surgery. J Oral Biosci 2013. [DOI: 10.1016/j.job.2013.04.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
211
|
Mesenchymal stem cells in the treatment of pediatric diseases. World J Pediatr 2013; 9:197-211. [PMID: 23929252 DOI: 10.1007/s12519-013-0425-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Accepted: 06/04/2013] [Indexed: 02/06/2023]
Abstract
BACKGROUND In recent years, the incredible interests in mesenchymal stem cells have boosted the expectations of both patients and physicians. Unlike embryonic stem cells, neither their procurement nor their use is deemed controversial. Moreover, their immunomodulatory capacity coupled with low immunogenicity has opened up their allogenic use, consequently broadening the possibilities for their application. In May 2012, Canadian health regulators approved Prochymal, the first mesenchymal stem cells-based drug, for acute graft-versus-host diseases in children who have failed to respond to steroid treatment. The aim of this article is to review the recent advances in mesenchymal stem cells for pediatric diseases. DATA SOURCES A literature review was performed on PubMed from 1966 to 2013 using the MeSH terms "mesenchymal stem cells", "clinical trials" and "children". Additional articles were identified by a hand search of the references list in the initial search. RESULTS The following categories are described: general properties, mechanisms of action, graft-versus-host diseases, cardiovascular diseases, liver diseases, inflammatory bowel diseases, osteoarticular diseases, autoimmune diseases, type 1 diabetes, and lung diseases. CONCLUSIONS Mesenchymal stem cells, owing to their availability, immunomodulatory properties, low immunogenicity, and therapeutic potential, have become one of the most attractive options for the treatment of a wide range of diseases. It is expected to see more and more clinical trials and applications of mesenchymal stem cells for pediatric diseases in the near future.
Collapse
|
212
|
|
213
|
McIntosh KR, Frazier T, Rowan BG, Gimble JM. Evolution and future prospects of adipose-derived immunomodulatory cell therapeutics. Expert Rev Clin Immunol 2013; 9:175-84. [PMID: 23390948 DOI: 10.1586/eci.12.96] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Over the past two decades, tissue engineering and regenerative medicine have evolved from what many considered a theoretical science to what is now a clinical reality. Tissue engineering combines biomaterial scaffolds, growth factors and stem or progenitor cells to repair damaged tissues. Adipose tissue, an abundant and easily accessed tissue, is a potential source of stromal/stem cells for regenerative therapeutic applications. Like bone marrow-derived mesenchymal stem cells, adipose-derived stromal/stem cells display both immunomodulatory and immunosuppressive properties. The adipose cells exert these actions, in part, through their secretion of paracrine growth factors. This review highlights recent developments in the isolation, characterization and preclinical application of adipose-derived cells and the challenges facing their translation into clinical practice.
Collapse
|
214
|
Jakobsen C, Sørensen JA, Kassem M, Thygesen TH. Mesenchymal stem cells in oral reconstructive surgery: a systematic review of the literature. J Oral Rehabil 2013; 40:693-706. [DOI: 10.1111/joor.12079] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2013] [Indexed: 11/30/2022]
Affiliation(s)
- C. Jakobsen
- Department of Oral and Maxillofacial Surgery; Odense University Hospital (OUH); Odense Denmark
| | - J. A. Sørensen
- Department of Plastic and Reconstructive Surgery; Odense University Hospital (OUH); Odense Denmark
| | - M. Kassem
- Endocrinology Research Unit; Odense University Hospital (OUH); Odense Denmark
| | - T. H. Thygesen
- Department of Oral and Maxillofacial Surgery; Odense University Hospital (OUH); Odense Denmark
| |
Collapse
|
215
|
Froelich K, Mickler J, Steusloff G, Technau A, Ramos Tirado M, Scherzed A, Hackenberg S, Radeloff A, Hagen R, Kleinsasser N. Chromosomal aberrations and deoxyribonucleic acid single-strand breaks in adipose-derived stem cells during long-term expansion in vitro. Cytotherapy 2013; 15:767-81. [DOI: 10.1016/j.jcyt.2012.12.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2012] [Revised: 12/21/2012] [Accepted: 12/22/2012] [Indexed: 01/01/2023]
|
216
|
Grottkau BE, Lin Y. Osteogenesis of Adipose-Derived Stem Cells. Bone Res 2013; 1:133-45. [PMID: 26273498 DOI: 10.4248/br201302003] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 04/12/2013] [Indexed: 01/01/2023] Open
Abstract
Current treatment options for skeletal repair, including immobilization, rigid fixation, alloplastic materials and bone grafts, have significant limitations. Bone tissue engineering offers a promising method for the repair of bone deficieny caused by fractures, bone loss and tumors. The use of adipose derived stem cells (ASCs) has received attention because of the self-renewal ability, high proliferative capacity and potential of osteogenic differentiation in vitro and in vivo studies of bone regeneration. Although cell therapies using ASCs are widely promising in various clinical fields, no large human clinical trials exist for bone tissue engineering. The aim of this review is to introduce how they are harvested, examine the characterization of ASCs, to review the mechanisms of osteogenic differentiation, to analyze the effect of mechanical and chemical stimuli on ASC osteodifferentiation, to summarize the current knowledge about usage of ASC in vivo studies and clinical trials, and finally to conclude with a general summary of the field and comments on its future direction.
Collapse
Affiliation(s)
- Brian E Grottkau
- Department of Orthopaedic Surgery, MassGeneral Hospital for Children and the Pediatric Orthopaedic Laboratory for Tissue Engineering and Regenerative Medicine, Harvard Medical School , Boston, Massachusetts, USA
| | - Yunfeng Lin
- Department of Orthopaedic Surgery, MassGeneral Hospital for Children and the Pediatric Orthopaedic Laboratory for Tissue Engineering and Regenerative Medicine, Harvard Medical School , Boston, Massachusetts, USA
| |
Collapse
|
217
|
Ahtiainen K, Mauno J, Ellä V, Hagström J, Lindqvist C, Miettinen S, Ylikomi T, Kellomäki M, Seppänen R. Autologous adipose stem cells and polylactide discs in the replacement of the rabbit temporomandibular joint disc. J R Soc Interface 2013; 10:20130287. [PMID: 23720535 DOI: 10.1098/rsif.2013.0287] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The temporomandibular joint (TMJ) disc lacks functional replacement after discectomy. We investigated tissue-engineered bilayer polylactide (PLA) discs and autologous adipose stem cells (ASCs) as a potential replacement for the TMJ disc. These ASC discs were pre-cultured either in control or in differentiation medium, including transforming growth factor (TGF)-β1 for one week. Prior to implantation, expression of fibrocartilaginous genes was measured by qRT-PCR. The control and differentiated ASC discs were implanted, respectively, in the right and left TMJs of rabbits for six (n = 5) and 12 months (n = 5). Thereafter, the excised TMJ areas were examined with cone beam computed tomography (CBCT) and histology. No signs of infection, inflammation or foreign body reactions were detected at histology, whereas chronic arthrosis and considerable condylar hypertrophy were observed in all operated joints at CBCT. The left condyle treated with the differentiated ASC discs appeared consistently smoother and more sclerotic than the right condyle. The ASC disc replacement resulted in dislocation and morphological changes in the rabbit TMJ. The ASC discs pre-treated with TGF-β1 enhanced the condylar integrity. While adverse tissue reactions were not shown, the authors suggest that with improved attachment and design, the PLA disc and biomaterial itself would hold potential for TMJ disc replacement.
Collapse
Affiliation(s)
- Katja Ahtiainen
- Department of Cell Biology, School of Medicine, University of Tampere, 33014 Tampere, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
218
|
Leng B, Jin X, Lin Q, Chen L, Wang Y, Du Z, Lin K, Chang J, Gu X, Wang C. A comparative study of proliferation and osteogenic differentiation of rat adipose-derived stem cells in β-tricalcium phosphate (β-TCP), forsterite (Mg2SiO4) and clinoenstatite (MgSiO3). ACTA ACUST UNITED AC 2013. [DOI: 10.1007/s11434-013-5874-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
219
|
Fang GW, Liu XS, Lv TZ, Shu HS, Shao H. A new choice for the treatment of segmental tibial bone defects: Intramedullary nail internal fixation combined adipose-derived stem cells technique. Med Hypotheses 2013; 80:422-4. [DOI: 10.1016/j.mehy.2012.12.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 10/16/2012] [Accepted: 12/31/2012] [Indexed: 01/22/2023]
|
220
|
Schubert T, Poilvache H, Galli C, Gianello P, Dufrane D. Galactosyl-knock-out engineered pig as a xenogenic donor source of adipose MSCs for bone regeneration. Biomaterials 2013; 34:3279-89. [DOI: 10.1016/j.biomaterials.2013.01.057] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 01/10/2013] [Indexed: 12/25/2022]
|
221
|
Gimble J, Rad MR, Yao S. Adipose Tissue–Derived Stem Cells and Their Regeneration Potential. STEM CELLS IN CRANIOFACIAL DEVELOPMENT AND REGENERATION 2013:241-258. [DOI: 10.1002/9781118498026.ch12] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
222
|
Sándor GKB. Tissue engineering of bone: Clinical observations with adipose-derived stem cells, resorbable scaffolds, and growth factors. Ann Maxillofac Surg 2013; 2:8-11. [PMID: 23483030 PMCID: PMC3591085 DOI: 10.4103/2231-0746.95308] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Introduction: Tissue engineering offers a simple, nonallergenic, and viable solution for the reconstruction of human tissues such as bone. With deeper understanding of the stem cell's pathobiology, the unique properties of these tissues can be effectively harnessed for the benefit of the patients. A primary source of mesenchymal stem cells (MSCs) for bone regeneration is from adipose tissue to provide adipose-derived stem cells (ASCs). The interdependency between adipogenesis and osteogenesis has been well established. The objective of this article is to present the preliminary clinical observation with reconstruction of craniofacial osseous defects larger than critical size with ASC. Materials and Methods: Patients with large craniofacial osseous defects only were included in this study. Autogenous fat from the anterior abdominal wall of the patients was harvested from 23 patients, taken to a central tissue banking laboratory and prepared. All patients were reconstructed with ASCs, resorbable scaffolds, and growth factor as required. Vascularized soft tissue beds were prepared for ectopic bone formation and later microvascular translocation as indicated. Results: 23 ASC seeded resorbable scaffolds have been combined with rhBMP-2 and successfully implanted into humans to reconstruct their jaws except for three failures. The failures included one infection and two cases of inadequate bone formation. Discussion: The technique of ASC-aided reconstruction of large defects still remains extremely sensitive as it takes longer duration and is costlier than the conventional standard immediate reconstruction. Preliminary results and clinical observations of these cases are extremely encouraging. In future, probably with evolving technological advances, ASC-aided reconstruction will be regularly used in clinical practise.
Collapse
Affiliation(s)
- George K B Sándor
- Professor of Tissue Engineering, Regea Institute for Regenerative Medicine, University of Tampere, Tampere, Finland
| |
Collapse
|
223
|
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
|
224
|
Kyllönen L, Haimi S, Mannerström B, Huhtala H, Rajala KM, Skottman H, Sándor GK, Miettinen S. Effects of different serum conditions on osteogenic differentiation of human adipose stem cells in vitro. Stem Cell Res Ther 2013; 4:17. [PMID: 23415114 PMCID: PMC3706769 DOI: 10.1186/scrt165] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Accepted: 02/04/2013] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Currently, human adipose stem cells (hASCs) are differentiated towards osteogenic lineages using culture medium supplemented with L-ascorbic acid 2-phosphate (AsA2-P), dexamethasone (Dex) and beta-glycerophosphate (β-GP). Because this osteogenic medium (OM1) was initially generated for the differentiation of bone marrow-derived mesenchymal stem cells, the component concentrations may not be optimal for the differentiation of hASCs. After preliminary screening, two efficient osteogenic media (OM2 and OM3) were chosen to be compared with the commonly used osteogenic medium (OM1). To further develop the culture conditions towards clinical usage, the osteo-inductive efficiencies of OM1, OM2 and OM3 were compared using human serum (HS)-based medium and a defined, xeno-free medium (RegES), with fetal bovine serum (FBS)-based medium serving as a control. METHODS To compare the osteo-inductive efficiency of OM1, OM2 and OM3 in FBS-, HS- and RegES-based medium, the osteogenic differentiation was assessed by alkaline phosphatase (ALP) activity, mineralization, and expression of osteogenic marker genes (runx2A, DLX5, collagen type I, osteocalcin, and ALP). RESULTS In HS-based medium, the ALP activity increased significantly by OM3, and mineralization was enhanced by both OM2 and OM3, which have high AsA2-P and low Dex concentrations. ALP activity and mineralization of hASCs was the weakest in FBS-based medium, with no significant differences between the OM compositions due to donor variation. However, the qRT-PCR data demonstrated significant upregulation of runx2A mRNA under osteogenic differentiation in FBS- and HS-based medium, particularly by OM3 under FBS conditions. Further, the expression of DLX5 was greatly stimulated by OM1 to 3 on day 7 when compared to control. The regulation of collagen type I, ALP, and osteocalcin mRNA was modest under induction by OM1 to 3. The RegES medium was found to support the proliferation and osteogenic differentiation of hASCs, but the composition of the RegES medium hindered the comparison of OM1, OM2 and OM3. CONCLUSIONS Serum conditions affect hASC proliferation and differentiation significantly. The ALP activity and mineralization was the weakest in FBS-based medium, although osteogenic markers were upregulated on mRNA level. When comparing the OM composition, the commonly used OM1 was least effective. Accordingly, higher concentration of AsA2-P and lower concentration of Dex, as in OM2 and OM3, should be used for the osteogenic differentiation of hASCs in vitro.
Collapse
|
225
|
Farré-Guasch E, Prins HJ, Overman JR, ten Bruggenkate CM, Schulten EA, Helder MN, Klein-Nulend J. Human Maxillary Sinus Floor Elevation as a Model for Bone Regeneration Enabling the Application of One-Step Surgical Procedures. TISSUE ENGINEERING PART B-REVIEWS 2013; 19:69-82. [DOI: 10.1089/ten.teb.2012.0404] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Elisabet Farré-Guasch
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam, Research Institute MOVE, University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Henk-Jan Prins
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam, Research Institute MOVE, University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
- Department of Oral and Maxillofacial Surgery, Research Institute MOVE, VU University Medical Center/Academic Centre for Dentistry Amsterdam, Amsterdam, The Netherlands
| | - Janice R. Overman
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam, Research Institute MOVE, University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
- Department of Oral and Maxillofacial Surgery, Research Institute MOVE, VU University Medical Center/Academic Centre for Dentistry Amsterdam, Amsterdam, The Netherlands
| | - Christiaan M. ten Bruggenkate
- Department of Oral and Maxillofacial Surgery, Research Institute MOVE, VU University Medical Center/Academic Centre for Dentistry Amsterdam, Amsterdam, The Netherlands
| | - Engelbert A.J.M. Schulten
- Department of Oral and Maxillofacial Surgery, Research Institute MOVE, VU University Medical Center/Academic Centre for Dentistry Amsterdam, Amsterdam, The Netherlands
| | - Marco N. Helder
- Department of Orthopaedics, Research Institute MOVE, VU University Medical Center, Amsterdam, The Netherlands
| | - Jenneke Klein-Nulend
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam, Research Institute MOVE, University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
226
|
Sándor GK, Tuovinen VJ, Wolff J, Patrikoski M, Jokinen J, Nieminen E, Mannerström B, Lappalainen OP, Seppänen R, Miettinen S. Adipose stem cell tissue-engineered construct used to treat large anterior mandibular defect: a case report and review of the clinical application of good manufacturing practice-level adipose stem cells for bone regeneration. J Oral Maxillofac Surg 2013; 71:938-50. [PMID: 23375899 DOI: 10.1016/j.joms.2012.11.014] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 09/18/2012] [Accepted: 11/15/2012] [Indexed: 02/08/2023]
Abstract
PURPOSE Large mandibular resection defects historically have been treated using autogenous bone grafts and reconstruction plates. However, a major drawback of large autogenous bone grafts is donor-site morbidity. PATIENTS AND METHODS This report describes the replacement of a 10-cm anterior mandibular ameloblastoma resection defect, reproducing the original anatomy of the chin, using a tissue-engineered construct consisting of β-tricalcium phosphate (β-TCP) granules, recombinant human bone morphogenetic protein-2 (BMP-2), and Good Manufacturing Practice-level autologous adipose stem cells (ASCs). Unlike prior reports, 1-step in situ bone formation was used without the need for an ectopic bone-formation step. The reconstructed defect was rehabilitated with a dental implant-supported overdenture. An additive manufactured medical skull model was used preoperatively to guide the prebending of patient-specific hardware, including a reconstruction plate and titanium mesh. A subcutaneous adipose tissue sample was harvested from the anterior abdominal wall of the patient before resection and simultaneous reconstruction of the parasymphysis. ASCs were isolated and expanded ex vivo over the next 3 weeks. The cell surface marker expression profile of ASCs was similar to previously reported results and ASCs were analyzed for osteogenic differentiation potential in vitro. The expanded cells were seeded onto a scaffold consisting of β-TCP and BMP-2 and the cell viability was evaluated. The construct was implanted into the parasymphyseal defect. RESULTS Ten months after reconstruction, dental implants were inserted into the grafted site, allowing harvesting of bone cores. Histologic examination and in vitro analysis of cell viability and cell surface markers were performed and prosthodontic rehabilitation was completed. CONCLUSION ASCs in combination with β-TCP and BMP-2 offer a promising construct for the treatment of large, challenging mandibular defects without the need for ectopic bone formation and allowing rehabilitation with dental implants.
Collapse
Affiliation(s)
- George K Sándor
- Department of Oral and Maxillofacial Surgery, University of Oulu, Oulu, Finland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
227
|
Carvalho PP, Gimble JM, Dias IR, Gomes ME, Reis RL. Xenofree enzymatic products for the isolation of human adipose-derived stromal/stem cells. Tissue Eng Part C Methods 2013; 19:473-8. [PMID: 23126465 DOI: 10.1089/ten.tec.2012.0465] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Human adipose-derived stromal/stem cells (ASCs) are an abundant, readily available population of adult stem cells that reside in adipose tissue and that have a great potential utility for tissue engineering and regenerative medicine therapeutic applications. Several preclinical studies have shown that ASCs have therapeutic applicability, but a standardized isolation and expansion methodology for clinical cell therapy has yet to be established. ASC are typically isolated and expanded using reagents with xenogenic components and this may pose certain risks and safety issues, such as exposure to infectious agents and immune reactions, creating further obstacles to the translation of ASC-based therapies to clinical scenarios. The objective of this study was to determine the suitability and efficacy of various alternative enzymatic products, CLS1 (Worthington), CLSAFA (Worthington), NB4 (SERVA), and Liberase (Roche), for the digestion of adipose tissue and subsequent isolation of ASCs, assessing cell functionality concerning their proliferation and differentiation ability. Results show that there are no statistically significant differences on yield and proliferation of cells isolated after enzymatic digestion with any of the studied products. The differentiation potential of the cells was not affected, and cell surface marker expression was similar among all products. We concluded that clinical grade products can replace current research-grade products effectively in our cell isolation protocols without any negative effect in the yield or function of human ASCs.
Collapse
Affiliation(s)
- Pedro P Carvalho
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Avepark, Guimarães, Portugal
| | | | | | | | | |
Collapse
|
228
|
Liu G, Zhang Y, Liu B, Sun J, Li W, Cui L. Bone regeneration in a canine cranial model using allogeneic adipose derived stem cells and coral scaffold. Biomaterials 2013; 34:2655-64. [PMID: 23343633 DOI: 10.1016/j.biomaterials.2013.01.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 01/01/2013] [Indexed: 12/29/2022]
Abstract
Adipose tissue derived stem cells (ASCs) based therapies for the repair and regeneration of various tissues have been widely investigated recently because of their multilineage potential and self-renewal capability. Our previous study demonstrated that autologous ASCs loaded onto natural coral scaffolds could repair cranial critical-sized defects (CSDs) in a canine model. The objective of this study was to determine whether the use of allogeneic ASCs could heal the same defect without the use of immunosuppressive therapy. The pedigree mismatch, mixed lymphocyte reaction assays (MLRs) and allogeneic skin graft experiments were performed to confirm unrelated ASC donors and recipients. A total of 12 adult Beagle dogs were enrolled in this study and divided into two groups. Bilateral cranial CSDs were created in each animal. The right-side defect was treated with allogeneic ASCs delivered onto a coral scaffold, and the left defect was either filled with an autologous ASC/coral composite (Group 1, n = 5) or with one coral scaffold alone (Group 2, n = 5). The systematic immune response and bone healing were evaluated postoperatively. The results showed that allogeneic ASC transplantation did not induce a systemic immune response by the hosts, and allogeneic ASCs could repair the cranial CSDs in an analogous way to that of the autologous cells. Moreover, both the green fluorescently labeled allogeneic and autologous ASCs were detected within the lacunae of newly formed bone in the defect site at 24 weeks, illustrating that the grafted ASCs contributed directly to bone regeneration in vivo. Thus, we concluded that allogeneic ASCs have the capacity to regenerate bone within craniofacial defects, providing an alternative source of seed cells for bone tissue engineering.
Collapse
Affiliation(s)
- Guangpeng Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, PR China
| | | | | | | | | | | |
Collapse
|
229
|
|
230
|
Gimble JM, Bunnell BA, Frazier T, Rowan B, Shah F, Thomas-Porch C, Wu X. Adipose-derived stromal/stem cells: a primer. Organogenesis 2013; 9:3-10. [PMID: 23538753 DOI: 10.4161/org.24279] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Until recently, the complexity of adipose tissue and its physiological role was not well appreciated. This changed with the discovery of adipokines such as leptin. The cellular composition of adipose tissue is heterogeneous and changes as a function of diabetes and disease states such as diabetes. Tissue engineers view adipose tissue as a rich source of adult stromal/stem cells isolated by collagenase digestion. In vitro and in vivo studies have documented that adipose stromal/stem cells are multipotent, with the ability to differentiate along the adipocyte, chondrocyte, osteoblast and other lineage pathways. The adipose stromal/stem cells secrete a wide range of cytokines and growth factors with potential paracrine actions. Furthermore, adipose stromal/stem cells exert immunomodulatory functions when added to mixed lymphocyte reactions, suggesting that they can be transplanted allogeneically. This review article focuses on these mechanisms of adipose stromal/stem cell action and their potential utility as cellular therapeutics.
Collapse
Affiliation(s)
- Jeffrey M Gimble
- Stem Cell Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | | | | | | | | | | | | |
Collapse
|
231
|
Li J, Gao J, Cha P, Chang Q, Liao Y, Liu C, Li K, Lu F. Supplementing fat grafts with adipose stromal cells for cosmetic facial contouring. Dermatol Surg 2012; 39:449-56. [PMID: 23279651 DOI: 10.1111/dsu.12058] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Numerous methods have been proposed to enhance the survival of fat grafts, but no definitive treatment is available. Stromal vascular fraction (SVF)-assisted cell therapy offers new perspectives for improving fat graft survival. OBJECTIVES To determine whether SVF supplementation could improve graft retention in patients undergoing autologous fat grafting for cosmetic improvement of facial contour. METHODS We retrospectively analyzed data from 38 women who underwent fat transplantation with SVF (n = 26) or fat grafting alone (n = 12) between October 2010 and January 2012. Each patient underwent computed tomography, and photographs were taken before and 6 months after surgery. The Philips Extended Brilliance Workspace was used for analysis of volume augmentation. RESULTS All patients showed cosmetic improvements, but the degree varied. No complications were evidenced during follow-up. Fat survival was higher with SVF (64.8 ± 10.2%) than fat grafting alone (46.4 ± 9.3%) (p < .01). SVF supplementation resulted in better clinical improvement than fat grafting alone. CONCLUSION Supplementing fat grafts with SVF for cosmetic facial contouring can improve the survival of fat grafts over fat grafting alone and provides satisfactory outcomes without major complications. Autologous fat grafting has been used for various cosmetic treatments and difficult reconstructive indications such as temporal depression, wrinkles of nasolabial folds, and hemifacial atrophy, with no incisional scar or complications associated with foreign materials, although problems such as a low rate of graft survival because of early resorption remain. (Aesthet Plast Surg, 14, 1990 and 127) Despite many innovations to overcome these problems, (Dermatol Surg, 26, 2000 and 1159); (Ann Plast Surg, 60, 2008 and 594); (Dermatol Surg, 27, 2001 and 819); (Dermatol Surg, 28, 2002 and 987) we lack a definitive method of fat processing that ensures maximal graft take and viability. (Plast Reconstr Surg, 115, 2005 and 197); (Dermatol Surg, 37, 2011 and 619).
Collapse
Affiliation(s)
- Jie Li
- Department of Plastic and Reconstructive Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
232
|
Khanna-Jain R, Mannerström B, Vuorinen A, Sándor GK, Suuronen R, Miettinen S. Osteogenic differentiation of human dental pulp stem cells on β-tricalcium phosphate/poly (l-lactic acid/caprolactone) three-dimensional scaffolds. J Tissue Eng 2012; 3:2041731412467998. [PMID: 23316276 PMCID: PMC3540691 DOI: 10.1177/2041731412467998] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Functional tissue engineering for bone augmentation requires the appropriate combination of biomaterials, mesenchymal stem cells, and specific differentiation factors. Therefore, we investigated the morphology, attachment, viability, and proliferation of human dental pulp stem cells cultured in xeno-free conditions in human serum medium seeded on β-tricalcium phosphate/poly(l-lactic acid/caprolactone) three-dimensional biomaterial scaffold. Additionally, osteogenic inducers dexamethasone and vitamin D3 were compared to achieve osteogenic differentiation. Dental pulp stem cells cultured in human serum medium maintained their morphology; furthermore, cells attached, remained viable, and increased in cell number within the scaffold. Alkaline phosphatase staining showed the osteogenic potential of dental pulp stem cells under the influence of osteogenic medium containing vitamin D3 or dexamethasone within the scaffolds. Maintenance of dental pulp stem cells for 14 days in osteogenic medium containing vitamin D3 resulted in significant increase in osteogenic markers as shown at mRNA level in comparison to osteogenic medium containing dexamethasone. The results of this study show that osteogenic medium containing vitamin D3 osteo-induced dental pulp stem cells cultured in human serum medium within β-tricalcium phosphate/poly(l-lactic acid/caprolactone) three-dimensional biomaterial, which could be directly translated clinically.
Collapse
Affiliation(s)
- Rashi Khanna-Jain
- Adult Stem Cells Group, Institute of Biomedical Technology, University of Tampere, Tampere, Finland ; BioMediTech, Tampere, Finland ; Science Centre, Tampere University Hospital, Tampere, Finland
| | | | | | | | | | | |
Collapse
|
233
|
Tsuno H, Yoshida T, Nogami M, Koike C, Okabe M, Noto Z, Arai N, Noguchi M, Nikaido T. Application of human amniotic mesenchymal cells as an allogeneic transplantation cell source in bone regenerative therapy. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2012. [DOI: 10.1016/j.msec.2012.07.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
234
|
Waselau M, Patrikoski M, Juntunen M, Kujala K, Kääriäinen M, Kuokkanen H, Sándor GK, Vapaavuori O, Suuronen R, Mannerström B, von Rechenberg B, Miettinen S. Effects of bioactive glass S53P4 or beta-tricalcium phosphate and bone morphogenetic protein-2 and bone morphogenetic protein-7 on osteogenic differentiation of human adipose stem cells. J Tissue Eng 2012; 3:2041731412467789. [PMID: 23316275 PMCID: PMC3540822 DOI: 10.1177/2041731412467789] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The effects of bioactive glass S53P4 or beta-tricalcium phosphate; and bone morphogenetic proteins bone morphogenetic protein-2, bone morphogenetic protein-7, or bone morphogenetic protein-2 + 7 on osteogenic differentiation of human adipose stem cells were compared in control medium, osteogenic medium, and bone morphogenetic protein-supplemented osteogenic medium to assess suitability for bone tissue engineering. Cell amount was evaluated with qDNA measurements; osteogenic differentiation using marker gene expression, alkaline phosphate activity, and angiogenic potential was measured by vascular endothelial growth factor expression. As compared to beta-tricalcium phosphate, cell amount was significantly greater for bioactive glass in control medium after 7 days and in osteogenic medium after 14 days, and alkaline phosphate activity was always significantly greater for bioactive glass in control medium. However, alkaline phosphate activity increased for beta-tricalcium phosphate and decreased for bioactive glass granules in osteogenic medium. For both biomaterials, bone morphogenetic protein supplementation decreased cell amount and osteogenic differentiation of human adipose stem cells, and vascular endothelial growth factor expressions correlated with cell amounts. Effects of culture medium on human adipose stem cells are biomaterial dependent; bioactive glass in control medium enhanced osteogenic differentiation most effectively.
Collapse
Affiliation(s)
- Martin Waselau
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland ; Adult Stem Cell Group, Institute of Biomedical Technology, University of Tampere, Tampere, Finland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
235
|
Abstract
Chronic wounds continue to be a major challenge for the medical profession, and plastic surgeons are frequently called in to help in the management of such wounds. Apart from the obvious morbidity to the patient, these problem wounds can be a major drain on the already scarce hospital resources. Sometimes, these chronic wounds can be more taxing than the underlying disease itself. Although many newer methods are available to handle such situations, the role of stem cells in the management of such wounds is an exciting area that needs to be explored further. A review of literature has been done regarding the role of stem cells in the management of chronic wounds. The abnormal pathology in such wounds is discussed and the possible role of stem cells for optimal healing in such cases would be detailed.
Collapse
Affiliation(s)
- Ramesh Kumar Sharma
- Department of Plastic Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | |
Collapse
|
236
|
Zanetti AS, McCandless GT, Chan JY, Gimble JM, Hayes DJ. Characterization of novel akermanite:poly-ϵ-caprolactone scaffolds for human adipose-derived stem cells bone tissue engineering. J Tissue Eng Regen Med 2012; 9:389-404. [PMID: 23166107 DOI: 10.1002/term.1646] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 07/17/2012] [Accepted: 10/04/2012] [Indexed: 12/19/2022]
Abstract
In this study, three different akermanite:poly-ϵ-caprolactone (PCL) composite scaffolds (wt%: 75:25, 50:50, 25:75) were characterized in terms of structure, compression strength, degradation rate and in vitro biocompatibility to human adipose-derived stem cells (hASC). Pure ceramic scaffolds [CellCeram™, custom-made, 40:60 wt%; β-tricalcium phosphate (β-TCP):hydroxyapatite (HA); and akermanite] and PCL scaffolds served as experimental controls. Compared to ceramic scaffolds, the authors hypothesized that optimal akermanite:PCL composites would have improved compression strength and comparable biocompatibility to hASC. Electron microscopy analysis revealed that PCL-containing scaffolds had the highest porosity but CellCeram™ had the greatest pore size. In general, compression strength in PCL-containing scaffolds was greater than in ceramic scaffolds. PCL-containing scaffolds were also more stable in culture than ceramic scaffolds. Nonetheless, mass losses after 21 days were observed in all scaffold types. Reduced hASC metabolic activity and increased cell detachment were observed after acute exposure to akermanite:PCL extracts (wt%: 75:25, 50:50). Among the PCL-containing scaffolds, hASC cultured for 21 days on akermanite:PCL (wt%: 75:25) discs displayed the highest viability, increased expression of osteogenic markers (alkaline phosphatase and osteocalcin) and lowest IL-6 expression. Together, the results indicate that akermanite:PCL composites may have appropriate mechanical and biocompatibility properties for use as bone tissue scaffolds.
Collapse
Affiliation(s)
- A S Zanetti
- Department of Biological Engineering, Louisiana State University and LSU AgCenter, Baton Rouge, Louisiana, USA
| | | | | | | | | |
Collapse
|
237
|
Stem cells in dentistry--Part II: Clinical applications. J Prosthodont Res 2012; 56:229-48. [PMID: 23137671 DOI: 10.1016/j.jpor.2012.10.001] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 10/03/2012] [Indexed: 12/29/2022]
Abstract
New technologies that facilitate solid alveolar ridge augmentation are receiving considerable attention in the field of prosthodontics because of the growing requirement for esthetic and functional reconstruction by dental implant treatments. Recently, several studies have demonstrated potential advantages for stem-cell-based therapies in regenerative treatments. Mesenchymal stem/stromal cells (MSCs) are now an excellent candidate for tissue replacement therapies, and tissue engineering approaches and chair-side cellular grafting approaches using autologous MSCs represent the clinical state of the art for stem-cell-based alveolar bone regeneration. Basic studies have revealed that crosstalk between implanted donor cells and recipient immune cells plays a key role in determining clinical success that may involve the recently observed immunomodulatory properties of MSCs. Part II of this review first overviews progress in regenerative dentistry to consider the implications of the stem cell technology in dentistry and then highlights cutting-edge stem-cell-based alveolar bone regenerative therapies. Factors that affect stem-cell-based bone regeneration as related to the local immune response are then discussed. Additionally, pre-clinical stem cell studies for the regeneration of teeth and other oral organs as well as possible applications of MSC-based immunotherapy in dentistry are outlined. Finally, the marketing of stem cell technology in dental stem cell banks with a view toward future regenerative therapies is introduced.
Collapse
|
238
|
Gharibi B, Hughes FJ. Effects of medium supplements on proliferation, differentiation potential, and in vitro expansion of mesenchymal stem cells. Stem Cells Transl Med 2012. [PMID: 23197689 DOI: 10.5966/sctm.2010-0031] [Citation(s) in RCA: 211] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) possess great potential for use in regenerative medicine. However, their clinical application may be limited by the ability to expand their cell numbers in vitro while maintaining their differential potentials and stem cell properties. Thus the aim of this study was to test the effect of a range of medium supplements on MSC self-renewal and differentiation potential. Cells were cultured until confluent and subcultured continuously until reaching senescence. Medium supplementation with fibroblast growth factor (FGF)-2, platelet-derived growth factor (PDGF)-BB, ascorbic acid (AA), and epidermal growth factor (EGF) both increased proliferation rate and markedly increased number of cell doublings before reaching senescence, with a greater than 1,000-fold increase in total cell numbers for AA, FGF-2, and PDGF-BB compared with control cultures. Long-term culture was associated with loss of osteogenic/adipocytic differentiation potential, particularly with FGF-2 supplementation but also with AA, EGF, and PDGF-BB. In addition FGF-2 resulted in reduction in expression of CD146 and alkaline phosphatase, but this was partially reversible on removal of the supplement. Cells expressed surface markers including CD146, CD105, CD44, CD90, and CD71 by flow cytometry throughout, and expression of these putative stem cell markers persisted even after loss of differentiation potentials. Overall, medium supplementation with FGF-2, AA, EGF, and PDGF-BB greatly enhanced the total in vitro expansion capacity of MSC cultures, although differentiation potentials were lost prior to reaching senescence. Loss of differentiation potential was not reflected by changes in stem cell surface marker expression.
Collapse
Affiliation(s)
- Borzo Gharibi
- Department of Periodontology, King's College London, London, UK
| | | |
Collapse
|
239
|
Zanetti AS, Sabliov C, Gimble JM, Hayes DJ. Human adipose-derived stem cells and three-dimensional scaffold constructs: a review of the biomaterials and models currently used for bone regeneration. J Biomed Mater Res B Appl Biomater 2012; 101:187-99. [PMID: 22997152 DOI: 10.1002/jbm.b.32817] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 07/28/2012] [Accepted: 08/07/2012] [Indexed: 12/24/2022]
Abstract
In the past decade, substantial strides have been taken toward the use of human adipose-derived stromal/stem cells (hASC) in the regeneration of bone. Since the discovery of the hASC osteogenic potential, many models have combined hASC with biodegradable scaffold materials. In general, rats and immunodeficient (nude) mice models for nonweight bearing bone formation have led the way to assess hASC osteogenic potential in vivo. The goal of this review is to present an overview of the recent literature describing hASC osteogenesis in conjunction with three-dimensional scaffolds for bone regeneration.
Collapse
Affiliation(s)
- Andrea S Zanetti
- Department of Biological and Agricultural Engineering, Louisiana State University and LSU AgCenter, Louisiana, USA
| | | | | | | |
Collapse
|
240
|
Gimble JM, Bunnell BA, Guilak F. Human adipose-derived cells: an update on the transition to clinical translation. Regen Med 2012; 7:225-35. [PMID: 22397611 DOI: 10.2217/rme.11.119] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The pace of discovery involving adipose-derived cells continues to accelerate at both the preclinical and clinical translational levels. Adipose tissue is a source of freshly isolated, heterogeneous stromal vascular fraction cells and culture-expanded, adherent and relatively homogeneous adipose stromal/stem cells. Both populations display regenerative capacity in soft and hard tissue repair, ischemic insults and autoimmune diseases. While their major mechanism of action has been attributed to both direct lineage differentiation and/or paracrine factor release, current evidence favors a paracrine mechanism. Over 40 clinical trials using adipose-derived cells conducted in 15 countries have been registered with the NIH, the majority of which are Phase I or Phase I/II safety studies. This review focuses on the literature of the past 2 years in order to assess the status of clinical and preclinical studies on adipose-derived cell therapies for regenerative medicine.
Collapse
Affiliation(s)
- Jeffrey M Gimble
- Center for Stem Cell Research & Regenerative Medicine, Department of Pharmacology, Tulane University Health Science Center, New Orleans, LA 70112, USA.
| | | | | |
Collapse
|
241
|
Platelet-Rich Plasma Promotes Angiogenesis of Prefabricated Vascularized Bone Graft. J Oral Maxillofac Surg 2012; 70:2191-7. [DOI: 10.1016/j.joms.2011.09.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Revised: 09/18/2011] [Accepted: 09/19/2011] [Indexed: 11/22/2022]
|
242
|
Lohberger B, Payer M, Rinner B, Kaltenegger H, Wolf E, Schallmoser K, Strunk D, Rohde E, Berghold A, Pekovits K, Wildburger A, Leithner A, Windhager R, Jakse N. Tri-lineage potential of intraoral tissue-derived mesenchymal stromal cells. J Craniomaxillofac Surg 2012; 41:110-8. [PMID: 22898339 DOI: 10.1016/j.jcms.2012.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 06/01/2012] [Accepted: 06/01/2012] [Indexed: 12/24/2022] Open
Abstract
The purpose of this study was to analyse the potential of intraoral tissues as a source of mesenchymal stromal and progenitor cells (MSPCs) for usage in future cell-based therapy models. Cells were isolated from four different tissues harvested during oral surgery intervention: (1) bone explants from the posterior maxilla, (2) bone explants from the oblique line, (3) from the mandibular periosteum, and (4) from the dental pulp. Donor sites and tissues were evaluated in terms of their accessibility, donor-site morbidity and average time period until appearance of MSPC colonies. Cell characterization was performed by flow cytometry and evaluation of in vitro osteogenic, adipogenic and chondrogenic differentiation potential. Adherent cell colonies were isolated from tissues from all sites after 4-8 days. The cells showed characteristics of MSPCs, so they were expanded up to clinical scales and demonstrated multipotency. The lowest donor-site morbidity was observed in the posterior maxilla harvests, while the highest donor-site morbidity was associated with harvests from mandibular sites. All sites seem to be potential sources of mesenchymal stromal and progenitor cells for tissue engineering approaches. Therefore, harvest morbidity and patient acceptance should affect the choice of the appropriate site.
Collapse
Affiliation(s)
- Birgit Lohberger
- Department of Orthopedic Surgery, Medical University of Graz, Graz, Austria.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
243
|
Mizuno H, Tobita M, Uysal AC. Concise review: Adipose-derived stem cells as a novel tool for future regenerative medicine. Stem Cells 2012; 30:804-10. [PMID: 22415904 DOI: 10.1002/stem.1076] [Citation(s) in RCA: 486] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The potential use of stem cell-based therapies for the repair and regeneration of various tissues and organs offers a paradigm shift that may provide alternative therapeutic solutions for a number of diseases. The use of either embryonic stem cells (ESCs) or induced pluripotent stem cells in clinical situations is limited due to cell regulations and to technical and ethical considerations involved in the genetic manipulation of human ESCs, even though these cells are, theoretically, highly beneficial. Mesenchymal stem cells seem to be an ideal population of stem cells for practical regenerative medicine, because they are not subjected to the same restrictions. In particular, large number of adipose-derived stem cells (ASCs) can be easily harvested from adipose tissue. Furthermore, recent basic research and preclinical studies have revealed that the use of ASCs in regenerative medicine is not limited to mesodermal tissue but extends to both ectodermal and endodermal tissues and organs, although ASCs originate from mesodermal lineages. Based on this background knowledge, the primary purpose of this concise review is to summarize and describe the underlying biology of ASCs and their proliferation and differentiation capacities, together with current preclinical and clinical data from a variety of medical fields regarding the use of ASCs in regenerative medicine. In addition, future directions for ASCs in terms of cell-based therapies and regenerative medicine are discussed.
Collapse
Affiliation(s)
- Hiroshi Mizuno
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan.
| | | | | |
Collapse
|
244
|
|
245
|
Bassi G, Pacelli L, Carusone R, Zanoncello J, Krampera M. Adipose-derived stromal cells (ASCs). Transfus Apher Sci 2012; 47:193-8. [PMID: 22818214 DOI: 10.1016/j.transci.2012.06.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adipose-derived stromal cells (ASCs) are now emerging as a good alternative to bone marrow derived mesenchymal stromal cells (BM-MSC) for cellular therapy. Similarly to BM-MSC, ASCs can be easily isolated as adherent fibroblastoid cell population after processing lipoaspirate samples. Lipoaspiration provides a great number of cells, without extensive manipulation. ASCs express classical mesenchymal markers and only at early passages express CD34. ASCs can differentiate in cells of mesodermal lineages, such as adipocytes, osteocytes and condrocytes. ASCs share with BM-MSC the same ability to inhibit the proliferation of allogeneic, activated immune cells, thus affecting in vivo in animal models the onset and course of rheumatoid arthritis (RA), experimental autoimmune encephalomyelitis (EAE), Crohn's disease (CD), ulcerous colitis (UC) and graft-versus-host disease (GvHD). On the other hand, the main molecular pathway involved in this effect is still unclear. On the basis of this functional property, ASCs are used in different clinical trials to treat RA, CD, UC and GvHD. However, the most promising field of clinical application is represented by bone defect repair. Despite the ability to regenerate injured tissues and to block the progression of inflammatory disorders, some authors reported that ASCs can also induce, in in vivo animal models, the growth and vascularization of solid and hematological tumors. Conversely, ASCs have been shown to hamper tumor cell proliferation, reduce cell viability and induce necrosis. Thus, more accurate studies, collaborative protocols, high standardization of methods, and safety controls are required to exclude transformation of transplanted ASCs.
Collapse
Affiliation(s)
- Giulio Bassi
- Laboratorio di Ricerca sulle Cellule Staminali, Sezione di Ematologia, Dipartimento di Medicina, Università degli Studi di Verona, Italy
| | | | | | | | | |
Collapse
|
246
|
Brüning A, Mylonas I. Cbfa1/Runx2-transduced adult adipose stem cells on biodegradable scaffolds for segmental bone defect repair. J Surg Res 2012; 185:e67-8. [PMID: 22795274 DOI: 10.1016/j.jss.2012.06.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 06/19/2012] [Accepted: 06/21/2012] [Indexed: 11/17/2022]
Affiliation(s)
- Ansgar Brüning
- Department of Obstetrics and Gynecology, Campus Innenstadt, Ludwig-Maximilians-University, Munich, Germany
| | | |
Collapse
|
247
|
Egusa H, Sonoyama W, Nishimura M, Atsuta I, Akiyama K. Stem cells in dentistry--part I: stem cell sources. J Prosthodont Res 2012; 56:151-65. [PMID: 22796367 DOI: 10.1016/j.jpor.2012.06.001] [Citation(s) in RCA: 223] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 06/14/2012] [Indexed: 12/21/2022]
Abstract
Stem cells can self-renew and produce different cell types, thus providing new strategies to regenerate missing tissues and treat diseases. In the field of dentistry, adult mesenchymal stem/stromal cells (MSCs) have been identified in several oral and maxillofacial tissues, which suggests that the oral tissues are a rich source of stem cells, and oral stem and mucosal cells are expected to provide an ideal source for genetically reprogrammed cells such as induced pluripotent stem (iPS) cells. Furthermore, oral tissues are expected to be not only a source but also a therapeutic target for stem cells, as stem cell and tissue engineering therapies in dentistry continue to attract increasing clinical interest. Part I of this review outlines various types of intra- and extra-oral tissue-derived stem cells with regard to clinical availability and applications in dentistry. Additionally, appropriate sources of stem cells for regenerative dentistry are discussed with regard to differentiation capacity, accessibility and possible immunomodulatory properties.
Collapse
Affiliation(s)
- Hiroshi Egusa
- Department of Fixed Prosthodontics, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | | | | | | | | |
Collapse
|
248
|
Alge DL, Bennet J, Treasure T, Voytik-Harbin S, Goebel WS, Chu TMG. Poly(propylene fumarate) reinforced dicalcium phosphate dihydrate cement composites for bone tissue engineering. J Biomed Mater Res A 2012; 100:1792-1802. [PMID: 22489012 PMCID: PMC3360828 DOI: 10.1002/jbm.a.34130] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 12/20/2011] [Accepted: 02/16/2012] [Indexed: 01/13/2023]
Abstract
Calcium phosphate cements have many desirable properties for bone tissue engineering, including osteoconductivity, resorbability, and amenability to rapid prototyping-based methods for scaffold fabrication. In this study, we show that dicalcium phosphate dihydrate (DCPD) cements, which are highly resorbable but also inherently weak and brittle, can be reinforced with poly(propylene fumarate) (PPF) to produce strong composites with mechanical properties suitable for bone tissue engineering. Characterization of DCPD-PPF composites revealed significant improvements in mechanical properties for cements with a 1.0 powder to liquid ratio. Compared with nonreinforced controls, flexural strength improved from 1.80 ± 0.19 MPa to 16.14 ± 1.70 MPa, flexural modulus increased from 1073.01 ± 158.40 MPa to 1303.91 ± 110.41 MPa, maximum displacement during testing increased from 0.11 ± 0.04 mm to 0.51 ± 0.09 mm, and work of fracture improved from 2.74 ± 0.78 J/m(2) to 249.21 ± 81.64 J/m(2) . To demonstrate the utility of our approach for scaffold fabrication, 3D macroporous scaffolds were prepared with rapid prototyping technology. Compressive testing revealed that PPF reinforcement increased scaffold strength from 0.31 ± 0.06 MPa to 7.48 ± 0.77 MPa. Finally, 3D PPF-DCPD scaffolds were implanted into calvarial defects in rabbits for 6 weeks. Although the addition of mesenchymal stem cells to the scaffolds did not significantly improve the extent of regeneration, numerous bone nodules with active osteoblasts were observed within the scaffold pores, especially in the peripheral regions. Overall, the results of this study suggest that PPF-DCPD composites may be promising scaffold materials for bone tissue engineering.
Collapse
Affiliation(s)
- Daniel L. Alge
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47908, USA
| | - Jeffrey Bennet
- Department of Oral Surgery and Hospital Dentistry, Indiana University School of Dentistry, Indianapolis, IN 46202, USA
| | - Trevor Treasure
- Department of Oral Surgery and Hospital Dentistry, Indiana University School of Dentistry, Indianapolis, IN 46202, USA
| | - Sherry Voytik-Harbin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47908, USA
| | - W. Scott Goebel
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Tien-Min Gabriel Chu
- Department of Restorative Dentistry, Division of Dental Biomaterials, Indiana University School of Dentistry, Indianapolis, IN 46202, USA
| |
Collapse
|
249
|
|
250
|
Review of biophysical factors affecting osteogenic differentiation of human adult adipose-derived stem cells. Biophys Rev 2012; 5:11-28. [PMID: 28510177 DOI: 10.1007/s12551-012-0079-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2011] [Accepted: 03/15/2012] [Indexed: 12/11/2022] Open
Abstract
Developing bone is subject to the control of a broad variety of influences in vivo. For bone repair applications, in vitro osteogenic assays are routinely used to test the responses of bone-forming cells to drugs, hormones, and biomaterials. Results of these assays are used to predict the behavior of bone-forming cells in vivo. Stem cell research has shown promise for enhancing bone repair. In vitro osteogenic assays to test the bone-forming response of stem cells typically use chemical solutions. Stem cell in vitro osteogenic assays often neglect important biophysical cues, such as the forces associated with regular weight-bearing exercise, which promote bone formation. Incorporating more biophysical cues that promote bone formation would improve in vitro osteogenic assays for stem cells. Improved in vitro osteogenic stimulation opens opportunities for "pre-conditioning" cells to differentiate towards the desired lineage. In this review, we explore the role of select biophysical factors-growth surfaces, tensile strain, fluid flow and electromagnetic stimulation-in promoting osteogenic differentiation of stem cells from human adipose. Emphasis is placed on the potential for physical microenvironment manipulation to translate tissue engineering and stem cell research into widespread clinical usage.
Collapse
|