151
|
Abstract
The authors present a review of biomaterials, substances traditionally derived from human or animal tissue or, more recently, biodegradable synthetics modeled after naturally occurring resources. These constructs differ from purely synthetic materials in that they are degraded or incorporated into a host's tissue. These biomaterials include a diverse array of medical products, such as acellular dermal matrix, bone substitutes, and injectables. In this review, the authors examine various clinical applications, including burn reconstruction and wound healing, breast surgery, complex abdominal wall reconstruction, craniofacial repair, and cosmetic surgery. Biomaterials such as acellular dermal matrix have proven beneficial in difficult-to-treat applications; however, more prospective data are needed to determine their true efficacy and cost-effectiveness.
Collapse
|
152
|
Nordberg RC, Loboa EG. Our Fat Future: Translating Adipose Stem Cell Therapy. Stem Cells Transl Med 2015; 4:974-9. [PMID: 26185256 DOI: 10.5966/sctm.2015-0071] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 06/17/2015] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Human adipose stem cells (hASCs) have the potential to treat patients with a variety of clinical conditions. Recent advancements in translational research, regulatory policy, and industry have positioned hASCs on the threshold of clinical translation. We discuss the progress and challenges of bringing adipose stem cell therapy into mainstream clinical use. SIGNIFICANCE This article details the advances made in recent years that have helped move human adipose stem cell therapy toward mainstream clinical use from a translational research, regulatory policy, and industrial standpoint. Four recurrent themes in translational technology as they pertain to human adipose stem cells are discussed: automated closed-system operations, biosensors and real-time monitoring, biomimetics, and rapid manufacturing. In light of recent FDA guidance documents, regulatory concerns about adipose stem cell therapy are discussed. Finally, an update is provided on the current state of clinical trials and the emerging industry that uses human adipose stem cells. This article is expected to stimulate future studies in translational adipose stem cell research.
Collapse
Affiliation(s)
- Rachel C Nordberg
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, and University of North Carolina Chapel Hill, Chapel Hill, North Carolina, USA; Department of Materials Science and Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Elizabeth G Loboa
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, and University of North Carolina Chapel Hill, Chapel Hill, North Carolina, USA; Department of Materials Science and Engineering, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
153
|
In Vitro Effects of Strontium on Proliferation and Osteoinduction of Human Preadipocytes. Stem Cells Int 2015; 2015:871863. [PMID: 26240575 PMCID: PMC4512617 DOI: 10.1155/2015/871863] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 03/10/2015] [Indexed: 12/15/2022] Open
Abstract
Development of tools to be used for in vivo bone tissue regeneration focuses on cellular models and differentiation processes. In searching for all the optimal sources, adipose tissue-derived mesenchymal stem cells (hADSCs or preadipocytes) are able to differentiate into osteoblasts with analogous characteristics to bone marrow mesenchymal stem cells, producing alkaline phosphatase (ALP), collagen, osteocalcin, and calcified nodules, mainly composed of hydroxyapatite (HA). The possibility to influence bone differentiation of stem cells encompasses local and systemic methods, including the use of drugs administered systemically. Among the latter, strontium ranelate (SR) represents an interesting compound, acting as an uncoupling factor that stimulates bone formation and inhibits bone resorption. The aim of our study was to evaluate the in vitro effects of a wide range of strontium (Sr(2+)) concentrations on proliferation, ALP activity, and mineralization of a novel finite clonal hADSCs cell line, named PA20-h5. Sr(2+) promoted PA20-h5 cell proliferation while inducing the increase of ALP activity and gene expression as well as HA production during in vitro osteoinduction. These findings indicate a role for Sr(2+) in supporting bone regeneration during the process of skeletal repair in general, and, more specifically, when cell therapies are applied.
Collapse
|
154
|
Critical steps in the isolation and expansion of adipose-derived stem cells for translational therapy. Expert Rev Mol Med 2015; 17:e11. [PMID: 26052798 DOI: 10.1017/erm.2015.10] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Since the discovery of adipose-derived stem cells (ASCs), there have been high expectations of their putative clinical use. Recent advances support these expectations, and it is expected that the transition from pre-clinical and clinical studies to implementation as a standard treatment modality is imminent. However ASCs must be isolated and expanded according to good manufacturing practice guidelines and a basic assurance of quality, safety, and medical effectiveness is needed for authorisation by regulatory agencies, such as European Medicines Agency and US Food and Drug Administration. In this review, a collection of studies investigating the influence of different steps of the isolation and expansion protocol on the yield and functionality of ASCs has been presented in an attempt to come up with best recommendations that ensure potential beneficial clinical outcome of using ASCs in any therapeutic setting. If the findings confirm the initial observations of beneficial effects of ASCs, the path is paved for implementing these ASC-based therapies as standard treatment options.
Collapse
|
155
|
Griffin M, Kalaskar DM, Butler PE, Seifalian AM. The use of adipose stem cells in cranial facial surgery. Stem Cell Rev Rep 2015; 10:671-85. [PMID: 24913279 PMCID: PMC4167434 DOI: 10.1007/s12015-014-9522-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Craniofacial malformations, have devastating psychosocial implications for many adults and children and causes huge socioeconomic burden. Currently craniofacial defects require soft tissue transfer, bone grafting techniques or difficult procedures such as microvascular free flaps. Such tissues are often limited in quantity, their harvest causes secondary large donor site defects and they lack the capability to fully restore previous form and function. Stem cell technology is being utilised for various tissue and organs of the body and consequently surgeons are eager to transfer these principles for craniofacial surgery. Adipose derived stem cells (ADSCs) are an exciting stem cell source for craniofacial surgeons due to their easy and painless isolation, relatively large abundance and familiarity with the harvesting procedure. ADSCs also have multiple desirable properties including adipogenic, osteogenic and chondrogenic potential, enhancement of angiogenesis and immunodulatory function. Due to these advantageous characteristics, ASDCs have been explored to repair craniofacial bone, soft tissue and cartilage. The desirable characteristics of ADSCs for craniofacial surgical applications will be explained. We report the experimental and clinical studies that have explored the use of ADSCs for bone, cartilage and soft tissue craniofacial defects. We conclude by establishing the key questions that are preventing the clinical application of ADSCs for craniofacial surgery.
Collapse
Affiliation(s)
- Michelle Griffin
- UCL Centre for Nanotechnology and Regenerative Medicine, Division of Surgery & Interventional Science, University College London, London, United Kingdom
| | | | | | | |
Collapse
|
156
|
Shingyochi Y, Orbay H, Mizuno H. Adipose-derived stem cells for wound repair and regeneration. Expert Opin Biol Ther 2015; 15:1285-92. [PMID: 26037027 DOI: 10.1517/14712598.2015.1053867] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The use of undifferentiated cells for cell-based tissue repair and regeneration strategies represents a promising approach for chronic wound healing. Multipotent adult stem cells isolated from adipose tissue, termed adipose-derived stem cells (ASCs), appear to be an ideal population of stem cells because they are autologous, non-immunogenic, plentiful, and easily obtained. Both preclinical and clinical studies have revealed that ASCs have potential for wound healing due to the mechanisms described below. AREAS COVERED Both in vitro and in vivo studies demonstrated that ASCs not only differentiate into keratinocytes, fibroblasts, and endothelial cells, as evidenced by their morphology, expression of cell surface markers, and gene expression, but also secrete several soluble factors, which positively contribute to wound healing in a paracrine manner. Clinical trials have been conducted using autologous ASCs with great success. EXPERT OPINION There remain many concerns regarding the use of ASCs, including how these cells act as precursors of keratinocytes, fibroblasts, and endothelial cells, or as a secretion vehicle of soluble factors. Further studies are necessary to establish the optimal strategy for the treatment of chronic wounds in patients with different disease backgrounds.
Collapse
Affiliation(s)
- Yoshiaki Shingyochi
- Juntendo University School of Medicine, Department of Plastic and Reconstructive Surgery , 2-1-1 Hongo Bunkyo-ku, Tokyo 1138421 , Japan
| | | | | |
Collapse
|
157
|
Wei J, Xu M, Zhang X, Meng S, Wang Y, Zhou T, Ma Q, Han B, Wei Y, Deng X. Enhanced Osteogenic Behavior of ADSCs Produced by Deproteinized Antler Cancellous Bone and Evidence for Involvement of ERK Signaling Pathway. Tissue Eng Part A 2015; 21:1810-1821. [PMID: 25760375 DOI: 10.1089/ten.tea.2014.0395] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Calcinated antler cancellous bone (CACB) is useful in repair of bone defects, as its composition and architecture is analogous to natural extracellular bone matrix. The use of CACB scaffolds with adipose-derived stem cells (ADSCs) in repair of rabbit mandibular bone defects was investigated along with the underlying molecular signaling pathways involved. CACB promoted the adhesion, spreading, and viability of ADSCs. Increased extracellular matrix production and expression of osteogenic markers in ADSCs were observed when seeded in CACB. The temporal kinetics of mRNA expression of ADSCs cultured in CACB lagged in comparison with that observed in cells grown in medium with osteogenic supplements. Activation of the extracellular signal-related kinases (ERK) 1/2 and RUNX-2 in CACB-cultured ADSCs was observed, and this activation was attenuated by the MeK inhibitor U0126. Microcomputed tomography scanning analysis and histological evaluations showed that loading the CACB with ADSCs resulted in enhanced new bone formation and angiogenesis when the composites were implanted in rabbit mandibular defects. These results indicated that the osteogenic behavior of ADSCs might be driven by the microenvironment formed by CACB via the ERK signaling pathway. These CACB/ADSCs composites have promising therapeutic potential for large bone defect repairs.
Collapse
Affiliation(s)
- Jinqi Wei
- 1Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
- 2First Clinical Division, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Mingming Xu
- 1Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Xuehui Zhang
- 1Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
- 3Department of Dental Materials, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Song Meng
- 1Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Yixiang Wang
- 4Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Tuanfeng Zhou
- 2First Clinical Division, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Qi Ma
- 1Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Bing Han
- 5Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Yan Wei
- 1Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Xuliang Deng
- 1Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
- 6National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| |
Collapse
|
158
|
Fanganiello RD, Ishiy FAA, Kobayashi GS, Alvizi L, Sunaga DY, Passos-Bueno MR. Increased In Vitro Osteopotential in SHED Associated with Higher IGF2 Expression When Compared with hASCs. Stem Cell Rev Rep 2015; 11:635-44. [DOI: 10.1007/s12015-015-9592-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
159
|
Strong AL, Bowles AC, MacCrimmon CP, Frazier TP, Lee SJ, Wu X, Katz AJ, Gawronska-Kozak B, Bunnell BA, Gimble JM. Adipose stromal cells repair pressure ulcers in both young and elderly mice: potential role of adipogenesis in skin repair. Stem Cells Transl Med 2015; 4:632-42. [PMID: 25900728 DOI: 10.5966/sctm.2014-0235] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 03/05/2015] [Indexed: 01/17/2023] Open
Abstract
UNLABELLED More than 2.5 million patients in the U.S. require treatment for pressure ulcers annually, and the elderly are at particularly high risk for pressure ulcer development. Current therapy for pressure ulcers consists of conservative medical management for shallow lesions and aggressive debridement and surgery for deeper lesions. The current study uses a murine model to address the hypothesis that adipose-derived stromal/stem cell (ASC) treatment would accelerate and enhance pressure ulcer repair. The dorsal skin of both young (2 months old [mo]) and old (20 mo) C57BL/6J female mice was sandwiched between external magnets for 12 hours over 2 consecutive days to initiate a pressure ulcer. One day following the induction, mice were injected with ASCs isolated from congenic mice transgenic for the green fluorescent protein under a ubiquitous promoter. Relative to phosphate-buffered saline-treated controls, ASC-treated mice displayed a cell concentration-dependent acceleration of wound closure, improved epidermal/dermal architecture, increased adipogenesis, and reduced inflammatory cell infiltration. The ASC-induced improvements occurred in both young and elderly recipients, although the expression profile of angiogenic, immunomodulatory, and reparative mRNAs differed as a function of age. The results are consistent with clinical reports that fat grafting improved skin architecture in thermal injuries; the authors of this published study have invoked ASC-based mechanisms to account for their clinical outcomes. Thus, the current proof-of-principle study sets the stage for clinical translation of autologous and/or allogeneic ASC treatment of pressure ulcers. SIGNIFICANCE Adipose-derived stromal/stem cells (ASCs) promote the healing of pressure ulcer wounds in both young and old mice. ASCs enhance wound healing rates through adipogenic differentiation and regeneration of the underlying architecture of the skin.
Collapse
Affiliation(s)
- Amy L Strong
- Center for Stem Cell Research and Regenerative Medicine and Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA; LaCell LLC, New Orleans, Louisiana, USA; Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Florida, Gainesville, Florida, USA; Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland; Departments of Medicine, Surgery, and Structural and Cellular Biology, Tulane Health Sciences Center, New Orleans, Louisiana, USA
| | - Annie C Bowles
- Center for Stem Cell Research and Regenerative Medicine and Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA; LaCell LLC, New Orleans, Louisiana, USA; Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Florida, Gainesville, Florida, USA; Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland; Departments of Medicine, Surgery, and Structural and Cellular Biology, Tulane Health Sciences Center, New Orleans, Louisiana, USA
| | - Connor P MacCrimmon
- Center for Stem Cell Research and Regenerative Medicine and Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA; LaCell LLC, New Orleans, Louisiana, USA; Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Florida, Gainesville, Florida, USA; Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland; Departments of Medicine, Surgery, and Structural and Cellular Biology, Tulane Health Sciences Center, New Orleans, Louisiana, USA
| | - Trivia P Frazier
- Center for Stem Cell Research and Regenerative Medicine and Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA; LaCell LLC, New Orleans, Louisiana, USA; Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Florida, Gainesville, Florida, USA; Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland; Departments of Medicine, Surgery, and Structural and Cellular Biology, Tulane Health Sciences Center, New Orleans, Louisiana, USA
| | - Stephen J Lee
- Center for Stem Cell Research and Regenerative Medicine and Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA; LaCell LLC, New Orleans, Louisiana, USA; Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Florida, Gainesville, Florida, USA; Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland; Departments of Medicine, Surgery, and Structural and Cellular Biology, Tulane Health Sciences Center, New Orleans, Louisiana, USA
| | - Xiying Wu
- Center for Stem Cell Research and Regenerative Medicine and Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA; LaCell LLC, New Orleans, Louisiana, USA; Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Florida, Gainesville, Florida, USA; Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland; Departments of Medicine, Surgery, and Structural and Cellular Biology, Tulane Health Sciences Center, New Orleans, Louisiana, USA
| | - Adam J Katz
- Center for Stem Cell Research and Regenerative Medicine and Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA; LaCell LLC, New Orleans, Louisiana, USA; Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Florida, Gainesville, Florida, USA; Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland; Departments of Medicine, Surgery, and Structural and Cellular Biology, Tulane Health Sciences Center, New Orleans, Louisiana, USA
| | - Barbara Gawronska-Kozak
- Center for Stem Cell Research and Regenerative Medicine and Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA; LaCell LLC, New Orleans, Louisiana, USA; Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Florida, Gainesville, Florida, USA; Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland; Departments of Medicine, Surgery, and Structural and Cellular Biology, Tulane Health Sciences Center, New Orleans, Louisiana, USA
| | - Bruce A Bunnell
- Center for Stem Cell Research and Regenerative Medicine and Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA; LaCell LLC, New Orleans, Louisiana, USA; Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Florida, Gainesville, Florida, USA; Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland; Departments of Medicine, Surgery, and Structural and Cellular Biology, Tulane Health Sciences Center, New Orleans, Louisiana, USA
| | - Jeffrey M Gimble
- Center for Stem Cell Research and Regenerative Medicine and Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA; LaCell LLC, New Orleans, Louisiana, USA; Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Florida, Gainesville, Florida, USA; Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland; Departments of Medicine, Surgery, and Structural and Cellular Biology, Tulane Health Sciences Center, New Orleans, Louisiana, USA
| |
Collapse
|
160
|
Paula ACC, Martins TMM, Zonari A, Frade SPPJ, Angelo PC, Gomes DA, Goes AM. Human adipose tissue-derived stem cells cultured in xeno-free culture condition enhance c-MYC expression increasing proliferation but bypassing spontaneous cell transformation. Stem Cell Res Ther 2015; 6:76. [PMID: 25889298 PMCID: PMC4455683 DOI: 10.1186/s13287-015-0030-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 01/24/2015] [Accepted: 03/02/2015] [Indexed: 12/13/2022] Open
Abstract
Introduction Human adipose tissue-derived stem cells (hASCs) are attractive cells for therapeutic applications and are currently being evaluated in multiple clinical trials. Prior to their clinical application, hASCs must be expanded ex vivo to obtain the required number of cells for transplantation. Fetal bovine serum is the supplement most widely used for cell culture, but it has disadvantages and it is not safe for cell therapy due to the risks of pathogen transmission and immune reaction. Furthermore, the cell expansion poses a risk of accumulating genetic abnormalities that could lead to malignant cell transformation. In this study, our aim was to evaluate the proliferation pattern as well as the resistance to spontaneous transformation of hASCs during expansion in a xeno-free culture condition. Methods hASCs were expanded in Dulbecco’s modified Eagle’s medium supplemented with pooled allogeneic human serum or fetal bovine serum to enable a side-by-side comparison. Cell viability and differentiation capacity toward the mesenchymal lineages were assessed, along with immunophenotype. Ki-67 expression and the proliferation kinetics were investigated. The expression of the transcription factors c-FOS and c-MYC was examined with Western blot, and MYC, CDKN2A, ERBB2 and TERT gene expression was assessed with quantitative PCR. Senescence was evaluated by β-gal staining. Karyotype analysis was performed and tumorigenesis assay in vivo was also evaluated. Results The hASCs expanded in medium with pooled allogeneic human serum did not show remarkable differences in morphology, viability, differentiation capacity or immunophenotype. The main difference observed was a significantly higher proliferative effect on hASCs cultured in pooled allogeneic human serum. There was no significant difference in C-FOS expression; however, C-MYC protein expression was enhanced in pooled allogeneic human serum cultures compared to fetal bovine serum cultures. No difference was observed in MYC and TERT mRNA levels. Moreover, the hASCs presented normal karyotype undergoing senescence, and did not form in vivo tumors, eliminating the possibility that spontaneous immortalization of hASCs had occurred with pooled allogeneic human serum. Conclusions This complete characterization of hASCs cultivated in pooled allogeneic human serum, a suitable xeno-free approach, shows that pooled allogeneic human serum provides a high proliferation rate, which can be attributed for the first time to C-MYC protein expression, and showed cell stability for safe clinical applications in compliance with good manufacturing practice.
Collapse
Affiliation(s)
- Ana C C Paula
- Laboratory of Cellular and Molecular Immunology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte, Minas Gerais, 31270-910, Brazil.
| | - Thaís M M Martins
- Laboratory of Cellular and Molecular Immunology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte, Minas Gerais, 31270-910, Brazil.
| | - Alessandra Zonari
- Laboratory of Cellular and Molecular Immunology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte, Minas Gerais, 31270-910, Brazil.
| | - Soraia P P J Frade
- Instituto Hermes Pardini, Av. das Nações, 2448, Vespasiano, Minas Gerais, 33200-000, Brazil.
| | - Patrícia C Angelo
- Instituto Hermes Pardini, Av. das Nações, 2448, Vespasiano, Minas Gerais, 33200-000, Brazil.
| | - Dawidson A Gomes
- Laboratory of Cellular and Molecular Immunology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte, Minas Gerais, 31270-910, Brazil.
| | - Alfredo M Goes
- Laboratory of Cellular and Molecular Immunology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte, Minas Gerais, 31270-910, Brazil.
| |
Collapse
|
161
|
Minteer DM, Marra KG, Rubin JP. Adipose stem cells: biology, safety, regulation, and regenerative potential. Clin Plast Surg 2015; 42:169-79. [PMID: 25827561 DOI: 10.1016/j.cps.2014.12.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This article discusses adipose-derived stem cell (ASC) biology, describes the current knowledge in the literature for the safety and regulation of ASCs, and provides a brief overview of the regenerative potential of ASCs. It is not an exhaustive listing of all available clinical studies or every study applying ASCs in tissue engineering and regenerative medicine, but is an objective commentary of these topics.
Collapse
Affiliation(s)
- Danielle M Minteer
- Department of Bioengineering, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Kacey G Marra
- Department of Bioengineering, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Department of Plastic Surgery, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15213, USA
| | - J Peter Rubin
- Department of Bioengineering, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Department of Plastic Surgery, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15213, USA.
| |
Collapse
|
162
|
Iwen KA, Priewe AC, Winnefeld M, Rose C, Siemers F, Rohwedel J, Cakiroglu F, Lehnert H, Schepky A, Klein J, Kramer J. Gluteal and abdominal subcutaneous adipose tissue depots as stroma cell source: gluteal cells display increased adipogenic and osteogenic differentiation potentials. Exp Dermatol 2015; 23:395-400. [PMID: 24689514 DOI: 10.1111/exd.12406] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2014] [Indexed: 02/06/2023]
Abstract
Human adipose-derived stroma cells (ADSCs) have successfully been employed in explorative therapeutic studies. Current evidence suggests that ADSCs are unevenly distributed in subcutaneous adipose tissue; therefore, the anatomical origin of ADSCs may influence clinical outcomes. This study was designed to investigate proliferation and differentiation capacities of ADSCs from the gluteal and abdominal depot of 8 females. All had normal BMI (22.01 ± 0.39 kg/m(2) ) and waist circumference (81.13 ± 2.33 cm). Examination by physicians and analysis of 31 laboratory parameters did not reveal possibly confounding medical disorders. Gluteal and abdominal adipose tissue was sampled by en bloc resection on day 7 (±1) after the last menses. Histological examination did not reveal significant depot-specific differences. As assessed by BrdU assay, proliferation of cells from both depots was similar after 24 h and analysis of 15 cell surface markers by flow cytometry identified the isolated cells as ADSCs, again without depot-specific differences. ADSCs from both depots differentiated poorly to chondroblasts. Gluteal ADSCs displayed significantly higher adipogenic differentiation potential than abdominal cells. Osteogenic differentiation was most pronounced in gluteal cells, whereas differentiation of abdominal ADSCs was severely impaired. Our data demonstrate a depot-specific difference in ADSC differentiation potential with abdominal cells failing to meet the criteria of multipotent ADSCs. This finding should be taken into account in future explorations of ADSC-derived therapeutic strategies.
Collapse
|
163
|
Grayson WL, Bunnell BA, Martin E, Frazier T, Hung BP, Gimble JM. Stromal cells and stem cells in clinical bone regeneration. Nat Rev Endocrinol 2015; 11:140-50. [PMID: 25560703 PMCID: PMC4338988 DOI: 10.1038/nrendo.2014.234] [Citation(s) in RCA: 304] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Stem-cell-mediated bone repair has been used in clinical trials for the regeneration of large craniomaxillofacial defects, to slow the process of bone degeneration in patients with osteonecrosis of the femoral head and for prophylactic treatment of distal tibial fractures. Successful regenerative outcomes in these investigations have provided a solid foundation for wider use of stromal cells in skeletal repair therapy. However, employing stromal cells to facilitate or enhance bone repair is far from being adopted into clinical practice. Scientific, technical, practical and regulatory obstacles prevent the widespread therapeutic use of stromal cells. Ironically, one of the major challenges lies in the limited understanding of the mechanisms via which transplanted cells mediate regeneration. Animal models have been used to provide insight, but these models largely fail to reproduce the nuances of human diseases and bone defects. Consequently, the development of targeted approaches to optimize cell-mediated outcomes is difficult. In this Review, we highlight the successes and challenges reported in several clinical trials that involved the use of bone-marrow-derived mesenchymal or adipose-tissue-derived stromal cells. We identify several obstacles blocking the mainstream use of stromal cells to enhance skeletal repair and highlight technological innovations or areas in which novel techniques might be particularly fruitful in continuing to advance the field of skeletal regenerative medicine.
Collapse
Affiliation(s)
- Warren L Grayson
- Department of Biomedical Engineering, Johns Hopkins University, 400 North Broadway, Baltimore, MD 21205, USA
| | - Bruce A Bunnell
- Centre for Stem Cell Research and Regenerative Medicine, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| | - Elizabeth Martin
- Centre for Stem Cell Research and Regenerative Medicine, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| | - Trivia Frazier
- Centre for Stem Cell Research and Regenerative Medicine, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| | - Ben P Hung
- Department of Biomedical Engineering, Johns Hopkins University, 400 North Broadway, Baltimore, MD 21205, USA
| | - Jeffrey M Gimble
- Centre for Stem Cell Research and Regenerative Medicine, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| |
Collapse
|
164
|
Hassan WU, Greiser U, Wang W. Role of adipose-derived stem cells in wound healing. Wound Repair Regen 2015; 22:313-25. [PMID: 24844331 DOI: 10.1111/wrr.12173] [Citation(s) in RCA: 253] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 03/01/2014] [Indexed: 12/11/2022]
Abstract
Impaired wound healing remains a challenge to date and causes debilitating effects with tremendous suffering. Recent advances in tissue engineering approaches in the area of cell therapy have provided promising treatment options to meet the challenges of impaired skin wound healing such as diabetic foot ulcers. Over the last few years, stem cell therapy has emerged as a novel therapeutic approach for various diseases including wound repair and tissue regeneration. Several different types of stem cells have been studied in both preclinical and clinical settings such as bone marrow-derived stem cells, adipose-derived stem cells (ASCs), circulating angiogenic cells (e.g., endothelial progenitor cells), human dermal fibroblasts, and keratinocytes for wound healing. Adipose tissue is an abundant source of mesenchymal stem cells, which have shown an improved outcome in wound healing studies. ASCs are pluripotent stem cells with the ability to differentiate into different lineages and to secrete paracrine factors initiating tissue regeneration process. The abundant supply of fat tissue, ease of isolation, extensive proliferative capacities ex vivo, and their ability to secrete pro-angiogenic growth factors make them an ideal cell type to use in therapies for the treatment of nonhealing wounds. In this review, we look at the pathogenesis of chronic wounds, role of stem cells in wound healing, and more specifically look at the role of ASCs, their mechanism of action and their safety profile in wound repair and tissue regeneration.
Collapse
Affiliation(s)
- Waqar Ul Hassan
- Charles Institute of Dermatology, School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| | | | | |
Collapse
|
165
|
Patel JJ, Flanagan CL, Hollister SJ. Bone Morphogenetic Protein-2 Adsorption onto Poly-ɛ-caprolactone Better Preserves Bioactivity In Vitro and Produces More Bone In Vivo than Conjugation Under Clinically Relevant Loading Scenarios. Tissue Eng Part C Methods 2015; 21:489-98. [PMID: 25345571 DOI: 10.1089/ten.tec.2014.0377] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND One strategy to reconstruct large bone defects is to prefabricate a vascularized flap by implanting a biomaterial scaffold with associated biologics into the latissimus dorsi and then transplanting this construct to the defect site after a maturation period. This strategy, similar to all clinically and regulatory feasible biologic approaches to surgical reconstruction, requires the ability to quickly (<1 h within an operating room) and efficiently bind biologics to scaffolds. It also requires the ability to localize biologic delivery. In this study, we investigated the efficacy of binding bone morphogenetic protein-2 (BMP2) to poly-ɛ-caprolactone (PCL) using adsorption and conjugation as a function of time. METHODS BMP2 was adsorbed (Ads) or conjugated (Conj) to PCL scaffolds with the same three-dimensional printed architecture while altering exposure time (0.5, 1, 5, and 16 h), temperature (4°C, 23°C), and BMP2 concentration (1.4, 5, 20, and 65 μg/mL). The in vitro release was quantified, and C2C12 cell alkaline phosphatase (ALP) expression was used to confirm bioactivity. Scaffolds with either 65 or 20 μg/mL Ads or Conj BMP2 for 1 h at 23°C were implanted subcutaneously in mice to evaluate in vivo bone regeneration. Micro-computed tomography, compression testing, and histology were performed to characterize bone regeneration. RESULTS After 1 h exposure to 65 μg/mL BMP2 at 23°C, Conj and Ads resulted in 12.83 ± 1.78 and 10.78 ± 1.49 μg BMP2 attached, respectively. Adsorption resulted in a positive ALP response and had a small burst release; whereas conjugation provided a sustained release with negligible ALP production, indicating that the conjugated BMP2 may not be bioavailable. Adsorbed 65 μg/mL BMP2 solution resulted in the greatest regenerated bone volume (15.0 ± 3.0 mm³), elastic modulus (20.1 ± 3.0 MPa), and %bone ingrowth in the scaffold interior (17.2% ± 5.4%) when compared with conjugation. CONCLUSION Adsorption may be optimal for the clinical application of prefabricating bone flaps due to BMP2 binding in a short exposure time, retained BMP2 bioactivity, and bone growth adhering to scaffold geometry and into pores with healthy marrow development.
Collapse
Affiliation(s)
- Janki J Patel
- Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan
| | | | | |
Collapse
|
166
|
Si JW, Wang XD, Shen SGF. Perinatal stem cells: A promising cell resource for tissue engineering of craniofacial bone. World J Stem Cells 2015; 7:149-159. [PMID: 25621114 PMCID: PMC4300925 DOI: 10.4252/wjsc.v7.i1.149] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 08/28/2014] [Accepted: 09/17/2014] [Indexed: 02/06/2023] Open
Abstract
In facing the mounting clinical challenge and suboptimal techniques of craniofacial bone defects resulting from various conditions, such as congenital malformations, osteomyelitis, trauma and tumor resection, the ongoing research of regenerative medicine using stem cells and concurrent advancement in biotechnology have shifted the focus from surgical reconstruction to a novel stem cell-based tissue engineering strategy for customized and functional craniofacial bone regeneration. Given the unique ontogenetical and cell biological properties of perinatal stem cells, emerging evidence has suggested these extraembryonic tissue-derived stem cells to be a promising cell source for extensive use in regenerative medicine and tissue engineering. In this review, we summarize the current achievements and obstacles in stem cell-based craniofacial bone regeneration and subsequently we address the characteristics of various types of perinatal stem cells and their novel application in tissue engineering of craniofacial bone. We propose the promising feasibility and scope of perinatal stem cell-based craniofacial bone tissue engineering for future clinical application.
Collapse
|
167
|
Potentialities of Adipose-Derived Mesenchymal Stem Cells Collected from Liposuction for Use in Cellular Therapy. Regen Med 2015. [DOI: 10.1007/978-1-4471-6542-2_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
168
|
Human Adipose-Derived Stem Cells (ASC): Their Efficacy in Clinical Applications. Regen Med 2015. [DOI: 10.1007/978-1-4471-6542-2_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
169
|
Abstract
The bony naso-orbital-ethmoid (NOE) complex is a 3-dimensional delicate anatomic structure. Damages to this region may result in severe facial dysfunction and malformation. The management and optimal surgical treatment strategies of NOE fractures remain controversial. For a patient with NOE trauma, doctors should perform comprehensive clinical examination and radiographic analysis to assess the type and extent of fracture. The results of assessment will assist doctors to make a patientspecific program for the sake of reducing post-operation complications and restoring normal appearance and function as much as possible. This review focuses on the advancement of management of NOE fractures including symptoms, classifications, diagnosis, approaches, treatment and new techniques in this field.
Collapse
Affiliation(s)
- Jun-Jun Wei
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | | | | | | | | | | |
Collapse
|
170
|
Banyard DA, Salibian AA, Widgerow AD, Evans GRD. Implications for human adipose-derived stem cells in plastic surgery. J Cell Mol Med 2014; 19:21-30. [PMID: 25425096 PMCID: PMC4288346 DOI: 10.1111/jcmm.12425] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 08/13/2014] [Indexed: 12/18/2022] Open
Abstract
Adipose-derived stem cells (ADSCs) are a subset of mesenchymal stem cells (MSCs) that possess many of the same regenerative properties as other MSCs. However, the ubiquitous presence of ADSCs and their ease of access in human tissue have led to a burgeoning field of research. The plastic surgeon is uniquely positioned to harness this technology because of the relative frequency in which they perform procedures such as liposuction and autologous fat grafting. This review examines the current landscape of ADSC isolation and identification, summarizes the current applications of ADSCs in the field of plastic surgery, discusses the risks associated with their use, current barriers to universal clinical translatability, and surveys the latest research which may help to overcome these obstacles.
Collapse
Affiliation(s)
- Derek A Banyard
- Department of Plastic Surgery, University of California, Irvine, Orange, CA, USA
| | | | | | | |
Collapse
|
171
|
Nicoletti GF, De Francesco F, D'Andrea F, Ferraro GA. Methods and Procedures in Adipose Stem Cells: State of the Art and Perspective for Translation Medicine. J Cell Physiol 2014; 230:489-95. [DOI: 10.1002/jcp.24837] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 09/22/2014] [Indexed: 12/14/2022]
Affiliation(s)
- G. F. Nicoletti
- Multidisciplinary Department of Medical-Surgical and Dental Specialties; Second University of Naples; Naples Italy
| | - F. De Francesco
- Multidisciplinary Department of Medical-Surgical and Dental Specialties; Second University of Naples; Naples Italy
| | - F. D'Andrea
- Multidisciplinary Department of Medical-Surgical and Dental Specialties; Second University of Naples; Naples Italy
| | - G. A. Ferraro
- Multidisciplinary Department of Medical-Surgical and Dental Specialties; Second University of Naples; Naples Italy
| |
Collapse
|
172
|
Kobayashi S, Kanai N, Ohki T, Takagi R, Yamaguchi N, Isomoto H, Kasai Y, Hosoi T, Nakao K, Eguchi S, Yamamoto M, Yamato M, Okano T. Prevention of esophageal strictures after endoscopic submucosal dissection. World J Gastroenterol 2014; 20:15098-15109. [PMID: 25386058 PMCID: PMC4223243 DOI: 10.3748/wjg.v20.i41.15098] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 02/14/2014] [Accepted: 05/19/2014] [Indexed: 02/06/2023] Open
Abstract
Endoscopic mucosal resection (EMR) and endoscopic submucosal dissection (ESD) have recently been accepted as less invasive methods for treating patients with early esophageal cancers such as squamous cell carcinoma and dysplasia of Barrett’s esophagus. However, the large defects in the esophageal mucosa often cause severe esophageal strictures, which dramatically reduce the patient’s quality of life. Although preventive endoscopic balloon dilatation can reduce dysphagia and the frequency of dilatation, other approaches are necessary to prevent esophageal strictures after ESD. This review describes several strategies for preventing esophageal strictures after ESD, with a particular focus on anti-inflammatory and tissue engineering approaches. The local injection of triamcinolone acetonide and other systemic steroid therapies are frequently used to prevent esophageal strictures after ESD. Tissue engineering approaches for preventing esophageal strictures have recently been applied in basic research studies. Scaffolds with temporary stents have been applied in five cases, and this technique has been shown to be safe and is anticipated to prevent esophageal strictures. Fabricated autologous oral mucosal epithelial cell sheets to cover the defective mucosa similarly to how commercially available skin products fabricated from epidermal cells are used for skin defects or in cases of intractable ulcers. Fabricated autologous oral-mucosal-epithelial cell sheets have already been shown to be safe.
Collapse
|
173
|
Dawson JI, Kanczler J, Tare R, Kassem M, Oreffo ROC. Concise review: bridging the gap: bone regeneration using skeletal stem cell-based strategies - where are we now? Stem Cells 2014; 32:35-44. [PMID: 24115290 DOI: 10.1002/stem.1559] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 08/12/2013] [Indexed: 12/11/2022]
Abstract
Skeletal stem cells confer to bone its innate capacity for regeneration and repair. Bone regeneration strategies seek to harness and enhance this regenerative capacity for the replacement of tissue damaged or lost through congenital defects, trauma, functional/esthetic problems, and a broad range of diseases associated with an increasingly aged population. This review describes the state of the field and current steps to translate and apply skeletal stem cell biology in the clinic and the problems therein. Challenges are described along with key strategies including the isolation and ex vivo expansion of multipotential populations, the targeting/delivery of regenerative populations to sites of repair, and their differentiation toward bone lineages. Finally, preclinical models of bone repair are discussed along with their implications for clinical translation and the opportunities to harness that knowledge for musculoskeletal regeneration.
Collapse
Affiliation(s)
- Jonathan I Dawson
- Bone & Joint Research Group, Centre for Human Development, Stem Cells and Regeneration Human Development and Health, Institute of Developmental Sciences, University of Southampton, United Kingdom
| | | | | | | | | |
Collapse
|
174
|
Nowacki M, Pietkun K, Pokrywczyńska M, Rasmus M, Warda K, Kloskowski T, Jundziłł A, Gagat M, Grzanka A, Bodnar M, Marszałek A, Drewa T, Czajkowski R. Filling effects, persistence, and safety of dermal fillers formulated with stem cells in an animal model. Aesthet Surg J 2014; 34:1261-9. [PMID: 25168156 DOI: 10.1177/1090820x14548212] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Research is scarce regarding the effectiveness of dermal fillers containing autologous stem cells. OBJECTIVES The authors sought to determine the local and systemic effects of adipose-derived stem cells (ADSCs) as a component of dermal fillers in an animal model. METHODS Wistar rats were injected with 1 of the following dermal fillers: ADSCs combined with hyaluronic acid (ADSC-HA), ADSCs combined with fish collagen (ADSC-COL), HA alone (CONTROL-HA), or COL alone (CONTROL-COL). Fillers were injected into the glabella, dorsum, and chest of each animal. The ADSCs were labeled with PKH26 to assess cell migration. Filling effects (FEs) were measured immediately after injection and at 1.5 months and 3 months after injection. Skin specimens were stained with hematoxylin and eosin to assess localization and persistence of ADSCs. RESULTS Mean FEs in animals implanted with ADSCs were greater and persisted longer than those of controls. No inflammatory responses were observed in any group. Three months after injection, PKH26-positive cells comprised nearly 70% of cells at the injection site in animals treated with ADSC-HA. PKH26 fluorescence also was detected in the spleen but not in the brain, kidney, or lung. CONCLUSIONS Stem cells have the potential to improve the aesthetic effects and longevity of dermal fillers.
Collapse
Affiliation(s)
- Maciej Nowacki
- Mr Nowacki is an MD and PhD student, Dr Jundziłł is a volunteer, Dr Pokrywczyńska and Dr Kloskowski are research fellows, and Ms Rasmus and Ms Warda are MSc students in the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, PolandMr Gagat is a research fellow and Dr Grzanka is Professor and Head of the Department of Histology and Embryology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Bodnar is a research fellow in the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, Poland, and Dr Marszałek is Professor and head of the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDepartment of Oncologic Pathology, Poznan University of Medical Sciences and Greater Poland Oncology Center, Poznan, PolandMs Pietkun is an MD and PhD student in the Chair of Regenerative Medicine, Department of Tissue Engineering and Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Czajkowski is a Professor and Head of the Department of Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Drewa is Professor and Head of the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, Poland, and Head of the Urology and Oncological Urology Department, Nicolaus Copernicus Hospital, Toruń, Poland
| | - Katarzyna Pietkun
- Mr Nowacki is an MD and PhD student, Dr Jundziłł is a volunteer, Dr Pokrywczyńska and Dr Kloskowski are research fellows, and Ms Rasmus and Ms Warda are MSc students in the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, PolandMr Gagat is a research fellow and Dr Grzanka is Professor and Head of the Department of Histology and Embryology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Bodnar is a research fellow in the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, Poland, and Dr Marszałek is Professor and head of the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDepartment of Oncologic Pathology, Poznan University of Medical Sciences and Greater Poland Oncology Center, Poznan, PolandMs Pietkun is an MD and PhD student in the Chair of Regenerative Medicine, Department of Tissue Engineering and Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Czajkowski is a Professor and Head of the Department of Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Drewa is Professor and Head of the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, Poland, and Head of the Urology and Oncological Urology Department, Nicolaus Copernicus Hospital, Toruń, Poland
| | - Marta Pokrywczyńska
- Mr Nowacki is an MD and PhD student, Dr Jundziłł is a volunteer, Dr Pokrywczyńska and Dr Kloskowski are research fellows, and Ms Rasmus and Ms Warda are MSc students in the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, PolandMr Gagat is a research fellow and Dr Grzanka is Professor and Head of the Department of Histology and Embryology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Bodnar is a research fellow in the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, Poland, and Dr Marszałek is Professor and head of the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDepartment of Oncologic Pathology, Poznan University of Medical Sciences and Greater Poland Oncology Center, Poznan, PolandMs Pietkun is an MD and PhD student in the Chair of Regenerative Medicine, Department of Tissue Engineering and Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Czajkowski is a Professor and Head of the Department of Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Drewa is Professor and Head of the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, Poland, and Head of the Urology and Oncological Urology Department, Nicolaus Copernicus Hospital, Toruń, Poland
| | - Marta Rasmus
- Mr Nowacki is an MD and PhD student, Dr Jundziłł is a volunteer, Dr Pokrywczyńska and Dr Kloskowski are research fellows, and Ms Rasmus and Ms Warda are MSc students in the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, PolandMr Gagat is a research fellow and Dr Grzanka is Professor and Head of the Department of Histology and Embryology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Bodnar is a research fellow in the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, Poland, and Dr Marszałek is Professor and head of the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDepartment of Oncologic Pathology, Poznan University of Medical Sciences and Greater Poland Oncology Center, Poznan, PolandMs Pietkun is an MD and PhD student in the Chair of Regenerative Medicine, Department of Tissue Engineering and Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Czajkowski is a Professor and Head of the Department of Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Drewa is Professor and Head of the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, Poland, and Head of the Urology and Oncological Urology Department, Nicolaus Copernicus Hospital, Toruń, Poland
| | - Karolina Warda
- Mr Nowacki is an MD and PhD student, Dr Jundziłł is a volunteer, Dr Pokrywczyńska and Dr Kloskowski are research fellows, and Ms Rasmus and Ms Warda are MSc students in the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, PolandMr Gagat is a research fellow and Dr Grzanka is Professor and Head of the Department of Histology and Embryology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Bodnar is a research fellow in the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, Poland, and Dr Marszałek is Professor and head of the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDepartment of Oncologic Pathology, Poznan University of Medical Sciences and Greater Poland Oncology Center, Poznan, PolandMs Pietkun is an MD and PhD student in the Chair of Regenerative Medicine, Department of Tissue Engineering and Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Czajkowski is a Professor and Head of the Department of Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Drewa is Professor and Head of the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, Poland, and Head of the Urology and Oncological Urology Department, Nicolaus Copernicus Hospital, Toruń, Poland
| | - Tomasz Kloskowski
- Mr Nowacki is an MD and PhD student, Dr Jundziłł is a volunteer, Dr Pokrywczyńska and Dr Kloskowski are research fellows, and Ms Rasmus and Ms Warda are MSc students in the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, PolandMr Gagat is a research fellow and Dr Grzanka is Professor and Head of the Department of Histology and Embryology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Bodnar is a research fellow in the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, Poland, and Dr Marszałek is Professor and head of the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDepartment of Oncologic Pathology, Poznan University of Medical Sciences and Greater Poland Oncology Center, Poznan, PolandMs Pietkun is an MD and PhD student in the Chair of Regenerative Medicine, Department of Tissue Engineering and Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Czajkowski is a Professor and Head of the Department of Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Drewa is Professor and Head of the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, Poland, and Head of the Urology and Oncological Urology Department, Nicolaus Copernicus Hospital, Toruń, Poland
| | - Arkadiusz Jundziłł
- Mr Nowacki is an MD and PhD student, Dr Jundziłł is a volunteer, Dr Pokrywczyńska and Dr Kloskowski are research fellows, and Ms Rasmus and Ms Warda are MSc students in the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, PolandMr Gagat is a research fellow and Dr Grzanka is Professor and Head of the Department of Histology and Embryology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Bodnar is a research fellow in the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, Poland, and Dr Marszałek is Professor and head of the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDepartment of Oncologic Pathology, Poznan University of Medical Sciences and Greater Poland Oncology Center, Poznan, PolandMs Pietkun is an MD and PhD student in the Chair of Regenerative Medicine, Department of Tissue Engineering and Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Czajkowski is a Professor and Head of the Department of Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Drewa is Professor and Head of the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, Poland, and Head of the Urology and Oncological Urology Department, Nicolaus Copernicus Hospital, Toruń, Poland
| | - Maciej Gagat
- Mr Nowacki is an MD and PhD student, Dr Jundziłł is a volunteer, Dr Pokrywczyńska and Dr Kloskowski are research fellows, and Ms Rasmus and Ms Warda are MSc students in the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, PolandMr Gagat is a research fellow and Dr Grzanka is Professor and Head of the Department of Histology and Embryology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Bodnar is a research fellow in the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, Poland, and Dr Marszałek is Professor and head of the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDepartment of Oncologic Pathology, Poznan University of Medical Sciences and Greater Poland Oncology Center, Poznan, PolandMs Pietkun is an MD and PhD student in the Chair of Regenerative Medicine, Department of Tissue Engineering and Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Czajkowski is a Professor and Head of the Department of Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Drewa is Professor and Head of the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, Poland, and Head of the Urology and Oncological Urology Department, Nicolaus Copernicus Hospital, Toruń, Poland
| | - Alina Grzanka
- Mr Nowacki is an MD and PhD student, Dr Jundziłł is a volunteer, Dr Pokrywczyńska and Dr Kloskowski are research fellows, and Ms Rasmus and Ms Warda are MSc students in the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, PolandMr Gagat is a research fellow and Dr Grzanka is Professor and Head of the Department of Histology and Embryology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Bodnar is a research fellow in the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, Poland, and Dr Marszałek is Professor and head of the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDepartment of Oncologic Pathology, Poznan University of Medical Sciences and Greater Poland Oncology Center, Poznan, PolandMs Pietkun is an MD and PhD student in the Chair of Regenerative Medicine, Department of Tissue Engineering and Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Czajkowski is a Professor and Head of the Department of Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Drewa is Professor and Head of the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, Poland, and Head of the Urology and Oncological Urology Department, Nicolaus Copernicus Hospital, Toruń, Poland
| | - Magdalena Bodnar
- Mr Nowacki is an MD and PhD student, Dr Jundziłł is a volunteer, Dr Pokrywczyńska and Dr Kloskowski are research fellows, and Ms Rasmus and Ms Warda are MSc students in the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, PolandMr Gagat is a research fellow and Dr Grzanka is Professor and Head of the Department of Histology and Embryology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Bodnar is a research fellow in the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, Poland, and Dr Marszałek is Professor and head of the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDepartment of Oncologic Pathology, Poznan University of Medical Sciences and Greater Poland Oncology Center, Poznan, PolandMs Pietkun is an MD and PhD student in the Chair of Regenerative Medicine, Department of Tissue Engineering and Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Czajkowski is a Professor and Head of the Department of Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Drewa is Professor and Head of the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, Poland, and Head of the Urology and Oncological Urology Department, Nicolaus Copernicus Hospital, Toruń, Poland
| | - Andrzej Marszałek
- Mr Nowacki is an MD and PhD student, Dr Jundziłł is a volunteer, Dr Pokrywczyńska and Dr Kloskowski are research fellows, and Ms Rasmus and Ms Warda are MSc students in the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, PolandMr Gagat is a research fellow and Dr Grzanka is Professor and Head of the Department of Histology and Embryology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Bodnar is a research fellow in the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, Poland, and Dr Marszałek is Professor and head of the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDepartment of Oncologic Pathology, Poznan University of Medical Sciences and Greater Poland Oncology Center, Poznan, PolandMs Pietkun is an MD and PhD student in the Chair of Regenerative Medicine, Department of Tissue Engineering and Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Czajkowski is a Professor and Head of the Department of Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Drewa is Professor and Head of the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, Poland, and Head of the Urology and Oncological Urology Department, Nicolaus Copernicus Hospital, Toruń, Poland
| | - Tomasz Drewa
- Mr Nowacki is an MD and PhD student, Dr Jundziłł is a volunteer, Dr Pokrywczyńska and Dr Kloskowski are research fellows, and Ms Rasmus and Ms Warda are MSc students in the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, PolandMr Gagat is a research fellow and Dr Grzanka is Professor and Head of the Department of Histology and Embryology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Bodnar is a research fellow in the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, Poland, and Dr Marszałek is Professor and head of the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDepartment of Oncologic Pathology, Poznan University of Medical Sciences and Greater Poland Oncology Center, Poznan, PolandMs Pietkun is an MD and PhD student in the Chair of Regenerative Medicine, Department of Tissue Engineering and Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Czajkowski is a Professor and Head of the Department of Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Drewa is Professor and Head of the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, Poland, and Head of the Urology and Oncological Urology Department, Nicolaus Copernicus Hospital, Toruń, Poland
| | - Rafał Czajkowski
- Mr Nowacki is an MD and PhD student, Dr Jundziłł is a volunteer, Dr Pokrywczyńska and Dr Kloskowski are research fellows, and Ms Rasmus and Ms Warda are MSc students in the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, PolandMr Gagat is a research fellow and Dr Grzanka is Professor and Head of the Department of Histology and Embryology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Bodnar is a research fellow in the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, Poland, and Dr Marszałek is Professor and head of the Department of Clinical Pathomorphology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDepartment of Oncologic Pathology, Poznan University of Medical Sciences and Greater Poland Oncology Center, Poznan, PolandMs Pietkun is an MD and PhD student in the Chair of Regenerative Medicine, Department of Tissue Engineering and Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Czajkowski is a Professor and Head of the Department of Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University Collegium Medicum, Bydgoszcz, PolandDr Drewa is Professor and Head of the Chair of Regenerative Medicine, Department of Tissue Engineering, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, Poland, and Head of the Urology and Oncological Urology Department, Nicolaus Copernicus Hospital, Toruń, Poland
| |
Collapse
|
175
|
Marble HD, Sutermaster BA, Kanthilal M, Fonseca VC, Darling EM. Gene expression-based enrichment of live cells from adipose tissue produces subpopulations with improved osteogenic potential. Stem Cell Res Ther 2014; 5:145. [PMID: 25287061 PMCID: PMC4619280 DOI: 10.1186/scrt502] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 09/08/2014] [Indexed: 12/11/2022] Open
Abstract
Introduction Mesenchymal stem cells have been increasingly used for cell-based therapies. Adipose-derived stem/stromal cells (ASCs) from the stromal vascular fraction (SVF) of fat tissue are a particularly attractive option for cell based therapy given their accessibility and relative abundance. However, their application in both clinical and basic science investigations is complicated by the isolation of differentiable cells within the SVF. Current enrichment strategies, such as monolayer passaging and surface marker-based sorting, can be time-consuming or overly stringent. Ideally, a population of cells with great regenerative capacity could be isolated with high yields so that extensive in vitro manipulation is not necessary. The objective of this study was to determine whether SVF cells sorted based on expression of alkaline phosphatase liver/bone/kidney (ALPL) resulted in populations with increased osteogenic differentiation potential. Methods SVF samples were obtained from four, human donors and processed to isolate initial, heterogeneous cell populations. These SVF cells underwent a four day osteogenic priming period, after which they were treated with a fluorescent, oligodeoxynucleotide molecular beacon probe specific for ALPL mRNA. Cells were separated into positive and negative groups using fluorescence-activated cell sorting (FACS) then differentiated down the osteogenic lineage. Differentiation was assessed by measuring calcified matrix production in each sample. Results Cells positive for ALPL expression (ALPL+) represented approximately 34% of the gated population, while cells negative for ALPL expression (ALPL-) represented approximately 18%. ALPL+ cells produced 3.7-fold and 2.1-fold more calcified matrix than ALPL- and unsorted SVF cells, respectively, indicating a significant improvement in osteogenic differentiation. Further, ALPL+ cells showed increases in metabolite production for both adipogenesis and chondrogenesis, suggesting that the enrichment process yields an enhanced multipotent phenotype. Osteogenic differentiation response and cell yields for ALPL+ cells were markedly improved over surface marker-sorted samples. Conclusion This study demonstrates a novel method to enrich heterogeneous SVF cells for increased osteogenic potential. The procedure requires less time and results in higher yields of therapeutically useful cells than other existing approaches. Gene expression-based sorting of MSCs is a potentially paradigm-shifting approach that could benefit applications spanning from basic science to clinical therapy. Electronic supplementary material The online version of this article (doi:10.1186/scrt502) contains supplementary material, which is available to authorized users.
Collapse
|
176
|
Paracrine effect of mesenchymal stem cells derived from human adipose tissue in bone regeneration. PLoS One 2014; 9:e107001. [PMID: 25198551 PMCID: PMC4157844 DOI: 10.1371/journal.pone.0107001] [Citation(s) in RCA: 228] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 08/08/2014] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation has proved to be a promising strategy in cell therapy and regenerative medicine. Although their mechanism of action is not completely clear, it has been suggested that their therapeutic activity may be mediated by a paracrine effect. The main goal of this study was to evaluate by radiographic, morphometric and histological analysis the ability of mesenchymal stem cells derived from human adipose tissue (Ad-MSC) and their conditioned medium (CM), to repair surgical bone lesions using an in vivo model (rabbit mandibles). The results demonstrated that both, Ad-MSC and CM, induce bone regeneration in surgically created lesions in rabbit's jaws, suggesting that Ad-MSC improve the process of bone regeneration mainly by releasing paracrine factors. The evidence of the paracrine effect of MSC on bone regeneration has a major impact on regenerative medicine, and the use of their CM can address some issues and difficulties related to cell transplants. In particular, CM can be easily stored and transported, and is easier to handle by medical personnel during clinical procedures.
Collapse
|
177
|
|
178
|
Salamon A, Adam S, Rychly J, Peters K. Long-term tumor necrosis factor treatment induces NFκB activation and proliferation, but not osteoblastic differentiation of adipose tissue-derived mesenchymal stem cells in vitro. Int J Biochem Cell Biol 2014; 54:149-62. [PMID: 25066315 DOI: 10.1016/j.biocel.2014.07.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 07/16/2014] [Accepted: 07/17/2014] [Indexed: 01/08/2023]
Abstract
The pro-inflammatory cytokine tumor necrosis factor (TNF) is well known to induce differentiation of bone matrix-resorbing osteoclasts from hematopoietic stem cells. However, the impact of TNF on differentiation of bone matrix-forming osteoblasts from mesenchymal stem cells (MSC) was only fragmentarily studied so far. Therefore, we investigated what impact long-term TNF treatment has on osteoblastic differentiation of MSC isolated from the adipose tissue (ASC) in vitro. In summary, we found continuous TNF exposure to induce the nuclear factor of kappa B pathway in ASC as well as secretion of the pro-inflammatory chemokine interleukin 8, but not the mitogen-activated protein kinase and the apoptosis pathway in ASC. Moreover, TNF neither induced nor inhibited osteoblastic differentiation of ASC, but strongly increased their proliferation rate. In that manner, pro-inflammatory conditions in vivo may generate significantly increased numbers of progenitor cells, and ASC especially, in conjunction with external stimuli, may contribute to the events of ectopic ossification observed in chronic inflammatory diseases. The substantiation of the translation of our in vitro findings to the disease context encourages further in vivo studies.
Collapse
Affiliation(s)
- Achim Salamon
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, D-18057 Rostock, Germany.
| | - Stefanie Adam
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, D-18057 Rostock, Germany
| | - Joachim Rychly
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, D-18057 Rostock, Germany
| | - Kirsten Peters
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, D-18057 Rostock, Germany
| |
Collapse
|
179
|
Abstract
Maxillary reconstruction is still an evolving art when compared to the reconstruction of the mandible. The defects of maxilla apart from affecting the functions of the speech, swallowing and mastication also cause cosmetic disfigurement. Rehabilitation of the form and function in patients with maxillary defects is either by using an obturator prosthesis or by a surgical reconstruction. Literature is abundant with a variety of reconstructive methods. The classification systems are also varied, with no universal acceptance of any one of them. The oncologic safety of these procedures is still debated, and conclusive evidence in this regard has not emerged yet. Management of the orbit is also not yet addressed properly. Tissue engineering, that has been hyped to be one of the possible solutions for this vexing reconstructive problem, has not come out with reliable and reproducible results so far. This review article discusses the rationale and oncological safety of the reconstructing the maxillary defects, critically analyzes the classification systems, offers the different reconstructive methods and touches upon the controversies in this subject. The management of the retained and exenterated orbit associated with maxillectomy is reviewed. The surgical morbidity, complications and the recent advances in this field are also looked into. An algorithm, based on our experience, is presented.
Collapse
Affiliation(s)
- Subramania Iyer
- Department of Plastic, Reconstructive and Head and Neck Surgery, Amrita Institute of Medical Sciences, Amrita University, Kochi, Kerala, India
| | - Krishnakumar Thankappan
- Department of Plastic, Reconstructive and Head and Neck Surgery, Amrita Institute of Medical Sciences, Amrita University, Kochi, Kerala, India
| |
Collapse
|
180
|
Kuismanen K, Sartoneva R, Haimi S, Mannerström B, Tomás E, Miettinen S, Nieminen K. Autologous adipose stem cells in treatment of female stress urinary incontinence: results of a pilot study. Stem Cells Transl Med 2014; 3:936-41. [PMID: 24985079 DOI: 10.5966/sctm.2013-0197] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The purpose of our study was to find out whether transurethral injections of autologous adipose stem cells (ASCs) are an effective and a safe treatment for female stress urinary incontinence (SUI). We treated five SUI patients with ASCs combined with bovine collagen gel and saline. Prior to the treatment, the ASCs were isolated from subcutaneous fat and expanded for 3 weeks in a good manufacturing practice-level laboratory. The mixture of ASCs and collagen was injected transurethrally via cystoscope. Additionally, viability, multipotency, and surface marker profile of ASCs were analyzed in vitro. We followed up with patients 3, 6, and 12 months after the injections. The primary endpoint was a cough test to measure objectively the effect of the treatment. Validated questionnaires were used to determine the subjective cure rate. After 6 months, 1 of 5 patients displayed a negative cough test with full bladder filled with 500 ml of saline. At 1 year, the cough test was negative with three patients; two of them were satisfied with the treatment and did not wish further treatment for SUI. Validated questionnaires showed some subjective improvement in all five patients. This is the first study describing the use of autologous ASCs in combination with collagen gel for female SUI treatments. Thus far, the treatment with autologous ASCs has proven safe and well tolerated. However, the feasibility and efficacy of the treatment were not optimal; therefore, additional research is needed to develop SUI injection therapies.
Collapse
Affiliation(s)
- Kirsi Kuismanen
- Department of Obstetrics and Gynaecology and School of Medicine and
| | - Reetta Sartoneva
- School of Medicine and Adult Stem Cell Group, University of Tampere, BioMediTech, Tampere, Finland; Science Center, Tampere University Hospital, Tampere, Finland
| | - Suvi Haimi
- Adult Stem Cell Group, University of Tampere, BioMediTech, Tampere, Finland; Department of Biomaterials Science and Technology, University of Twente, Enschede, The Netherlands
| | - Bettina Mannerström
- Adult Stem Cell Group, University of Tampere, BioMediTech, Tampere, Finland; Science Center, Tampere University Hospital, Tampere, Finland
| | - Eija Tomás
- Department of Obstetrics and Gynaecology and
| | - Susanna Miettinen
- Adult Stem Cell Group, University of Tampere, BioMediTech, Tampere, Finland; Science Center, Tampere University Hospital, Tampere, Finland
| | | |
Collapse
|
181
|
Senarath-Yapa K, McArdle A, Renda A, Longaker MT, Quarto N. Adipose-derived stem cells: a review of signaling networks governing cell fate and regenerative potential in the context of craniofacial and long bone skeletal repair. Int J Mol Sci 2014; 15:9314-30. [PMID: 24865492 PMCID: PMC4100096 DOI: 10.3390/ijms15069314] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 05/16/2014] [Accepted: 05/20/2014] [Indexed: 02/07/2023] Open
Abstract
Improvements in medical care, nutrition and social care are resulting in a commendable change in world population demographics with an ever increasing skew towards an aging population. As the proportion of the world's population that is considered elderly increases, so does the incidence of osteodegenerative disease and the resultant burden on healthcare. The increasing demand coupled with the limitations of contemporary approaches, have provided the impetus to develop novel tissue regeneration therapies. The use of stem cells, with their potential for self-renewal and differentiation, is one potential solution. Adipose-derived stem cells (ASCs), which are relatively easy to harvest and readily available have emerged as an ideal candidate. In this review, we explore the potential for ASCs to provide tangible therapies for craniofacial and long bone skeletal defects, outline key signaling pathways that direct these cells and describe how the developmental signaling program may provide clues on how to guide these cells in vivo. This review also provides an overview of the importance of establishing an osteogenic microniche using appropriately customized scaffolds and delineates some of the key challenges that still need to be overcome for adult stem cell skeletal regenerative therapy to become a clinical reality.
Collapse
Affiliation(s)
- Kshemendra Senarath-Yapa
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, School of Medicine, Stanford University, Stanford, CA 94305-2200, USA; E-Mails: (K.S.-Y.); (A.M.)
| | - Adrian McArdle
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, School of Medicine, Stanford University, Stanford, CA 94305-2200, USA; E-Mails: (K.S.-Y.); (A.M.)
| | - Andrea Renda
- Dipartimento di Scienze Biomediche Avanzate, Universita’ degli Studi di Napoli Federico II, Napoli 80131, Italy; E-Mail:
| | - Michael T. Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, School of Medicine, Stanford University, Stanford, CA 94305-2200, USA; E-Mails: (K.S.-Y.); (A.M.)
| | - Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, School of Medicine, Stanford University, Stanford, CA 94305-2200, USA; E-Mails: (K.S.-Y.); (A.M.)
- Dipartimento di Scienze Biomediche Avanzate, Universita’ degli Studi di Napoli Federico II, Napoli 80131, Italy; E-Mail:
| |
Collapse
|
182
|
Adipose-derived stromal cells for osteoarticular repair: trophic function versus stem cell activity. Expert Rev Mol Med 2014; 16:e9. [PMID: 24810570 PMCID: PMC4017835 DOI: 10.1017/erm.2014.9] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The identification of multipotent adipose-derived stromal cells (ASC) has raised hope that tissue regeneration approaches established with bone-marrow-derived stromal cells (BMSC) can be reproduced with a cell-type that is far more accessible in large quantities. Recent detailed comparisons, however, revealed subtle functional differences between ASC and BMSC, stressing the concept of a common mesenchymal progenitor existing in a perivascular niche across all tissues. Focussing on bone and cartilage repair, this review summarises recent in vitro and in vivo studies aiming towards tissue regeneration with ASC. Advantages of good accessibility, high yield and superior growth properties are counterbalanced by an inferiority of ASC to form ectopic bone and stimulate long-bone healing along with their less pronounced osteogenic and angiogenic gene expression signature. Hence, particular emphasis is placed on establishing whether stem cell activity of ASC is so far proven and relevant for successful osteochondral regeneration, or whether trophic activity may largely determine therapeutic outcome.
Collapse
|
183
|
Abstract
Adipose-derived stem cells (ASCs) are considered a great alternative source of mesenchymal stem cells (MSCs). Unlike bone marrow stem cells (BMSCs), ASCs can be retrieved in high numbers from lipoaspirate, a by-product of liposuction procedures. Given that ASCs represent an easily accessible and abundant source of multipotent cells, ASCs have garnered attention and curiosity from both scientific and clinical communities for their potential in clinical applications. Furthermore, their unique immunobiology and secretome are attractive therapeutic properties. A decade since the discovery of a stem cell reservoir residing within adipose tissue, ASC-based clinical trials have grown over the years around the world along with assessments made on their safety and efficacy. With the progress of ASCs into clinical applications, the aim towards producing clinical-grade ASCs becomes increasingly important. Several countries have recognised the growing industry of cell therapies and have developed regulatory frameworks to assure their safety. With more research efforts made to understand their effects in both scientific and clinical settings, ASCs hold great promise as a future therapeutic strategy in treating a wide variety of diseases. Therefore, this review seeks to highlight the clinical applicability of ASCs as well as their progress in clinical trials across various medical disciplines.
Collapse
|
184
|
Mueller S, Hohlweg-Majert B, Buergers R, Steiner T, Reichert TE, Wolff KD, Gosau M. The functional and aesthetic reconstruction of midfacial and orbital defects by combining free flap transfer and craniofacial prosthesis. Clin Oral Investig 2014; 19:413-9. [DOI: 10.1007/s00784-014-1243-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 04/03/2014] [Indexed: 12/15/2022]
|
185
|
Yamaguchi DT. “Ins” and “Outs” of mesenchymal stem cell osteogenesis in regenerative medicine. World J Stem Cells 2014; 6:94-110. [PMID: 24772237 PMCID: PMC3999785 DOI: 10.4252/wjsc.v6.i2.94] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Accepted: 01/20/2014] [Indexed: 02/06/2023] Open
Abstract
Repair and regeneration of bone requires mesenchymal stem cells that by self-renewal, are able to generate a critical mass of cells with the ability to differentiate into osteoblasts that can produce bone protein matrix (osteoid) and enable its mineralization. The number of human mesenchymal stem cells (hMSCs) diminishes with age and ex vivo replication of hMSCs has limited potential. While propagating hMSCs under hypoxic conditions may maintain their ability to self-renew, the strategy of using human telomerase reverse transcriptase (hTERT) to allow for hMSCs to prolong their replicative lifespan is an attractive means of ensuring a critical mass of cells with the potential to differentiate into various mesodermal structural tissues including bone. However, this strategy must be tempered by the oncogenic potential of TERT-transformed cells, or their ability to enhance already established cancers, the unknown differentiating potential of high population doubling hMSCs and the source of hMSCs (e.g., bone marrow, adipose-derived, muscle-derived, umbilical cord blood, etc.) that may provide peculiarities to self-renewal, differentiation, and physiologic function that may differ from non-transformed native cells. Tissue engineering approaches to use hMSCs to repair bone defects utilize the growth of hMSCs on three-dimensional scaffolds that can either be a base on which hMSCs can attach and grow or as a means of sequestering growth factors to assist in the chemoattraction and differentiation of native hMSCs. The use of whole native extracellular matrix (ECM) produced by hMSCs, rather than individual ECM components, appear to be advantageous in not only being utilized as a three-dimensional attachment base but also in appropriate orientation of cells and their differentiation through the growth factors that native ECM harbor or in simulating growth factor motifs. The origin of native ECM, whether from hMSCs from young or old individuals is a critical factor in “rejuvenating” hMSCs from older individuals grown on ECM from younger individuals.
Collapse
|
186
|
Romagnoli C, Brandi ML. Adipose mesenchymal stem cells in the field of bone tissue engineering. World J Stem Cells 2014; 6:144-152. [PMID: 24772241 PMCID: PMC3999772 DOI: 10.4252/wjsc.v6.i2.144] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 12/19/2013] [Accepted: 03/04/2014] [Indexed: 02/06/2023] Open
Abstract
Bone tissue engineering represents one of the most challenging emergent fields for scientists and clinicians. Current failures of autografts and allografts in many pathological conditions have prompted researchers to find new biomaterials able to promote bone repair or regeneration with specific characteristics of biocompatibility, biodegradability and osteoinductivity. Recent advancements for tissue regeneration in bone defects have occurred by following the diamond concept and combining the use of growth factors and mesenchymal stem cells (MSCs). In particular, a more abundant and easily accessible source of MSCs was recently discovered in adipose tissue. These adipose stem cells (ASCs) can be obtained in large quantities with little donor site morbidity or patient discomfort, in contrast to the invasive and painful isolation of bone marrow MSCs. The osteogenic potential of ASCs on scaffolds has been examined in cell cultures and animal models, with only a few cases reporting the use of ASCs for successful reconstruction or accelerated healing of defects of the skull and jaw in patients. Although these reports extend our limited knowledge concerning the use of ASCs for osseous tissue repair and regeneration, the lack of standardization in applied techniques makes the comparison between studies difficult. Additional clinical trials are needed to assess ASC therapy and address potential ethical and safety concerns, which must be resolved to permit application in regenerative medicine.
Collapse
|
187
|
Al-Saqi SH, Saliem M, Asikainen S, Quezada HC, Ekblad A, Hovatta O, Le Blanc K, Jonasson AF, Götherström C. Defined serum-free media for in vitro expansion of adipose-derived mesenchymal stem cells. Cytotherapy 2014; 16:915-26. [PMID: 24726655 DOI: 10.1016/j.jcyt.2014.02.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 02/02/2014] [Accepted: 02/16/2014] [Indexed: 01/01/2023]
Abstract
BACKGROUND There is a growing interest in mesenchymal stem cells (MSCs) because they are regarded as good candidates for cell therapy. Adipose tissue represents an easily accessible source to derive mesenchymal stem cells (Ad-MSCs) non-invasively in large numbers. The aim of this study was to evaluate a defined serum-free medium for in vitro expansion of MSCs as a prerequisite for their clinical use. METHODS Adipose tissue was isolated from healthy donors. Cells were isolated and expanded for five passages in serum-free medium (Mesencult-XF) and Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum (DMEM-FBS). MSC morphology, marker expression, viability, population doubling time and differentiation potential toward osteogenic and adipogenic lineages were evaluated. Bone marrow MSCs were included as controls. RESULTS Ad-MSCs cultured in Mesencult-XF had shorter population doubling time (33.3 ± 13.7 h) compared with those cultured in DMEM-FBS (54.3 ± 41.0 h, P < 0.05). Ad-MSCs cultured in Mesencult-XF displayed a stable morphology and surface marker expression and a higher differentiation potential in comparison to Ad-MSCs cultured in DMEM-FBS. CONCLUSIONS The defined serum-free and xeno-free Mesencult-XF media appear to be a good choice for Ad-MSCs, but it is not as good in supporting culture of bone marrow MSCs when the cells are to be used for clinical purposes.
Collapse
Affiliation(s)
- Shahla Hamza Al-Saqi
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| | - Mohammed Saliem
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Suvi Asikainen
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Hernan Concha Quezada
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Asa Ekblad
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden; Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Outi Hovatta
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Katarina Le Blanc
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden; Division of Clinical Immunology and Transfusion Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Aino Fianu Jonasson
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Cecilia Götherström
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden; Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
188
|
Da Costa G, Pare A, Goga D, Sury F, Laure B. [CT-scan evaluation of calvarial bone donor site reconstruction using calcium phosphate cement]. ACTA ACUST UNITED AC 2014; 115:140-4. [PMID: 24703439 DOI: 10.1016/j.revsto.2014.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 10/02/2013] [Accepted: 02/12/2014] [Indexed: 10/25/2022]
Abstract
UNLABELLED The aim of reconstructing a calvarial donor site with biomaterial is to reconstruct the skull vault. The aim of this retrospective study was to assess the quality of reconstruction of calvarial bone with Hydroset™ (Stryker™) in patients having undergone monocortical parietal bone graft harvesting. PATIENTS AND METHOD The donor sites of patients having undergone calvarial bone harvesting had been reconstructed with Hydroset™ cement over a period of four years. Calvarial bone reconstruction and the thickness of the parietal bone were evaluated by CT scan. RESULTS Twenty-six patients had undergone reconstruction. The CT scan revealed a good integration of Hydroset™ with maintained thickness of the biomaterial. The parietal bone thickness was increased by 0.67 mm on average (P=0.002). DISCUSSION The reconstruction of calvarial donor site bone defect with Hydroset™, after a monocortical harvesting, demonstrates parietal osseous thickness maintained in time.
Collapse
Affiliation(s)
- G Da Costa
- Service de chirurgie maxillo-faciale et plastique de la face, université François-Rabelais de Tours, CHU Trousseau, 37044 Tours cedex, France.
| | - A Pare
- Service de chirurgie maxillo-faciale et plastique de la face, université François-Rabelais de Tours, CHU Trousseau, 37044 Tours cedex, France
| | - D Goga
- Service de chirurgie maxillo-faciale et plastique de la face, université François-Rabelais de Tours, CHU Trousseau, 37044 Tours cedex, France
| | - F Sury
- Service de chirurgie maxillo-faciale et plastique de la face, université François-Rabelais de Tours, CHU Trousseau, 37044 Tours cedex, France
| | - B Laure
- Service de chirurgie maxillo-faciale et plastique de la face, université François-Rabelais de Tours, CHU Trousseau, 37044 Tours cedex, France
| |
Collapse
|
189
|
Jalali M, Kirkpatrick WNA, Cameron MG, Pauklin S, Vallier L. Human stem cells for craniomaxillofacial reconstruction. Stem Cells Dev 2014; 23:1437-51. [PMID: 24564584 DOI: 10.1089/scd.2013.0576] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Human stem cell research represents an exceptional opportunity for regenerative medicine and the surgical reconstruction of the craniomaxillofacial complex. The correct architecture and function of the vastly diverse tissues of this important anatomical region are critical for life supportive processes, the delivery of senses, social interaction, and aesthetics. Craniomaxillofacial tissue loss is commonly associated with inflammatory responses of the surrounding tissue, significant scarring, disfigurement, and psychological sequelae as an inevitable consequence. The in vitro production of fully functional cells for skin, muscle, cartilage, bone, and neurovascular tissue formation from human stem cells, may one day provide novel materials for the reconstructive surgeon operating on patients with both hard and soft tissue deficit due to cancer, congenital disease, or trauma. However, the clinical translation of human stem cell technology, including the application of human pluripotent stem cells (hPSCs) in novel regenerative therapies, faces several hurdles that must be solved to permit safe and effective use in patients. The basic biology of hPSCs remains to be fully elucidated and concerns of tumorigenicity need to be addressed, prior to the development of cell transplantation treatments. Furthermore, functional comparison of in vitro generated tissue to their in vivo counterparts will be necessary for confirmation of maturity and suitability for application in reconstructive surgery. Here, we provide an overview of human stem cells in disease modeling, drug screening, and therapeutics, while also discussing the application of regenerative medicine for craniomaxillofacial tissue deficit and surgical reconstruction.
Collapse
Affiliation(s)
- Morteza Jalali
- 1 Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge , Cambridge, United Kingdom
| | | | | | | | | |
Collapse
|
190
|
Kokai LE, Marra K, Rubin JP. Adipose stem cells: biology and clinical applications for tissue repair and regeneration. Transl Res 2014; 163:399-408. [PMID: 24361334 DOI: 10.1016/j.trsl.2013.11.009] [Citation(s) in RCA: 206] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 11/06/2013] [Accepted: 11/18/2013] [Indexed: 12/17/2022]
Abstract
There is a clear clinical need for cell therapies to repair or regenerate tissue lost to disease or trauma. Adipose tissue is a renewable source of stem cells, called adipose-derived stem cells (ASCs), that release important growth factors for wound healing, modulate the immune system, decrease inflammation, and home in on injured tissues. Therefore, ASCs may offer great clinical utility in regenerative therapies for afflictions such as Parkinson's disease and Alzheimer's disease, spinal cord injury, heart disease, and rheumatoid arthritis, or for replacing lost tissue from trauma or tumor removal. This article discusses the regenerative properties of ASCs that can be harnessed for clinical applications, and explores current and future challenges for ASC clinical use. Such challenges include knowledge-based deficiencies, hurdles for translating research to the clinic, and barriers to establishing a new paradigm of medical care. Clinical experience with ASCs, ASCs as a portion of the heterogeneous stromal cell population extracted enzymatically from adipose tissue, and stromal vascular fraction are also described.
Collapse
Affiliation(s)
- Lauren E Kokai
- Department of Plastic Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Kacey Marra
- Department of Plastic Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - J Peter Rubin
- Department of Plastic Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA.
| |
Collapse
|
191
|
Josh F, Tobita M, Tanaka R, Orbay H, Ogata K, Suzuki K, Hyakusoku H, Mizuno H. Concentration of PDGF-AB, BB and TGF-β1 as valuable human serum parameters in adipose-derived stem cell proliferation. J NIPPON MED SCH 2014; 80:140-7. [PMID: 23657067 DOI: 10.1272/jnms.80.140] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Human serum (HS) has attributes similar to fetal bovine serum (FBS) in the proliferation and differentiation of human adipose-derived stem cells (hASCs) when compared in vitro. The purpose of this study was to determine what types of HS, with respect to the concentrations of endogenous growth factors, could be made available for hASC proliferation. METHODS HS was collected from 2 groups of healthy donor (freshly isolated HS [n=9], and HS preserved for 4 years [n=7]). All sera were isolated with a Cellaid HS isolation device (JMS Co., Ltd, Hiroshima, Japan) and then classified into 3 groups based on the concentrations (high, middle, and low) of platelet-derived growth factor (PDGF)-AB, PDGF-BB, and transforming growth factor-beta 1 (TGF-β1) by means of enzyme-linked immunoassay screening. The hASCs were isolated from subcutaneous fat using a collagenase enzymatic digestion process and were cultured in control media, each supplemented with HS from a different group. Cell numbers were counted on days 2, 4, 7, and 14, and the relationship between cell proliferation and the level of each growth factor was investigated. RESULTS The proliferation of hASCs correlated with the concentration of each growth factor. The cut-off points for PDGF-AB, PDGF-BB, and TGF-β1 in HS [necessary for hASC proliferation when compared with FBS] were 10 ng/mL, 1.5 ng/mL, and 15 ng/mL, respectively. There was no correlation between the storage period of HS and the proliferation potential of hASCs. CONCLUSIONS These results suggest that the effectiveness of HS on hASC proliferation depends on the concentrations of endogenous PDGFs. In addition, the Cellaid device used in this study allows the simultaneous release of several growth factors from platelets, and our results have shown that it can be used to collect HS for future hASC-based therapies.
Collapse
Affiliation(s)
- Fonny Josh
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
192
|
Koellensperger E, Bollinger N, Dexheimer V, Gramley F, Germann G, Leimer U. Choosing the right type of serum for different applications of human adipose tissue-derived stem cells: influence on proliferation and differentiation abilities. Cytotherapy 2014; 16:789-99. [PMID: 24642018 DOI: 10.1016/j.jcyt.2014.01.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 01/13/2014] [Accepted: 01/14/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND AIMS Adipose tissue-derived stem cells (ADSCs) are thought to have great potential in regenerative medicine. A xenoprotein-free culture and handling system is desirable. To date, there is only little and contradictory information about the influence of the different types of human serum on ADSC proliferation and differentiation. METHODS First, ADSCs were cultured in media containing regular human serum (HS plus) or fetal calf serum (FCS plus) with supplementation of growth factors for three passages. During passage 4, ADSC proliferative activity and adipogenic, osteogenic and chondrogenic differentiation ability was quantified. Second, ADSCs were cultured with three different human sera (regular human serum [HS], human serum from platelet-poor plasma [SPPP] or human serum from platelet-rich plasma [SPRP]) without supplementation of platelet-derived growth factor and assessed accordingly. The growth factor content of the different types of human sera was determined by means of multiplex protein assay and enzyme-linked immunosorbent assay. RESULTS The different sera did not affect ADSC doubling time significantly (P < 0.05). Specific glycerol-3-phosphat-dehydrogenase activity was significantly lower in cultures with SPRP (P < 0.01) compared with the other media compositions. Extracellular calcium deposition was significantly higher in cells differentiated in cultures with HS or SPPP compared with those with SPRP, HS plus or FCS (P < 0.01). Glycosaminoglycan content and collagen 2 were highest in cells cultured with SPRP (P < 0.001). CONCLUSIONS Culturing ADSCs in human serum appears to be a reasonable and efficient alternative compared with FCS. With respect to the outcome of a sighted clinical application, it appears to be feasible to handle the cells in a serum suitable for the intended later use.
Collapse
Affiliation(s)
- Eva Koellensperger
- Clinic for Plastic and Reconstructive Surgery, Aesthetic and Preventive Medicine at Heidelberg University Hospital-ETHIANUM, Heidelberg, Germany.
| | - Nils Bollinger
- Clinic for Plastic and Reconstructive Surgery, Aesthetic and Preventive Medicine at Heidelberg University Hospital-ETHIANUM, Heidelberg, Germany
| | - Verena Dexheimer
- Clinic for Plastic and Reconstructive Surgery, Aesthetic and Preventive Medicine at Heidelberg University Hospital-ETHIANUM, Heidelberg, Germany
| | - Felix Gramley
- Department of Cardiology, University of Frankfurt, Frankfurt, Germany
| | - Guenter Germann
- Clinic for Plastic and Reconstructive Surgery, Aesthetic and Preventive Medicine at Heidelberg University Hospital-ETHIANUM, Heidelberg, Germany
| | - Uwe Leimer
- Clinic for Plastic and Reconstructive Surgery, Aesthetic and Preventive Medicine at Heidelberg University Hospital-ETHIANUM, Heidelberg, Germany
| |
Collapse
|
193
|
Sándor GK, Numminen J, Wolff J, Thesleff T, Miettinen A, Tuovinen VJ, Mannerström B, Patrikoski M, Seppänen R, Miettinen S, Rautiainen M, Öhman J. Adipose stem cells used to reconstruct 13 cases with cranio-maxillofacial hard-tissue defects. Stem Cells Transl Med 2014; 3:530-40. [PMID: 24558162 DOI: 10.5966/sctm.2013-0173] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Although isolated reports of hard-tissue reconstruction in the cranio-maxillofacial skeleton exist, multipatient case series are lacking. This study aimed to review the experience with 13 consecutive cases of cranio-maxillofacial hard-tissue defects at four anatomically different sites, namely frontal sinus (3 cases), cranial bone (5 cases), mandible (3 cases), and nasal septum (2 cases). Autologous adipose tissue was harvested from the anterior abdominal wall, and adipose-derived stem cells were cultured, expanded, and then seeded onto resorbable scaffold materials for subsequent reimplantation into hard-tissue defects. The defects were reconstructed with either bioactive glass or β-tricalcium phosphate scaffolds seeded with adipose-derived stem cells (ASCs), and in some cases with the addition of recombinant human bone morphogenetic protein-2. Production and use of ASCs were done according to good manufacturing practice guidelines. Follow-up time ranged from 12 to 52 months. Successful integration of the construct to the surrounding skeleton was noted in 10 of the 13 cases. Two cranial defect cases in which nonrigid resorbable containment meshes were used sustained bone resorption to the point that they required the procedure to be redone. One septal perforation case failed outright at 1 year because of the postsurgical resumption of the patient's uncontrolled nasal picking habit.
Collapse
Affiliation(s)
- George K Sándor
- Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere, Tampere, Finland; Department of Oral and Maxillofacial Surgery, University of Oulu, Oulu, Finland; Oulu University Hospital, Oulu, Finland; Department of Otolaryngology, Head and Neck Surgery and Oral Diseases and Department of Neurosciences and Rehabilitation, Tampere University Hospital, Tampere, Finland; Department of Oral and Maxillofacial Surgery, Central Hospital of Central Finland Health Care District, Jyväskylä, Finland; Department of Biomedical Engineering, Tampere University of Technology, Tampere, Finland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Salamon A, van Vlierberghe S, van Nieuwenhove I, Baudisch F, Graulus GJ, Benecke V, Alberti K, Neumann HG, Rychly J, Martins JC, Dubruel P, Peters K. Gelatin-Based Hydrogels Promote Chondrogenic Differentiation of Human Adipose Tissue-Derived Mesenchymal Stem Cells In Vitro. MATERIALS 2014; 7:1342-1359. [PMID: 28788517 PMCID: PMC5453082 DOI: 10.3390/ma7021342] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 01/24/2014] [Accepted: 01/27/2014] [Indexed: 12/22/2022]
Abstract
Due to the weak regeneration potential of cartilage, there is a high clinical incidence of articular joint disease, leading to a strong demand for cartilaginous tissue surrogates. The aim of this study was to evaluate a gelatin-based hydrogel for its suitability to support chondrogenic differentiation of human mesenchymal stem cells. Gelatin-based hydrogels are biodegradable, show high biocompatibility, and offer possibilities to introduce functional groups and/or ligands. In order to prove their chondrogenesis-supporting potential, a hydrogel film was developed and compared with standard cell culture polystyrene regarding the differentiation behavior of human mesenchymal stem cells. Cellular basis for this study were human adipose tissue-derived mesenchymal stem cells, which exhibit differentiation potential along the adipogenic, osteogenic and chondrogenic lineage. The results obtained show a promotive effect of gelatin-based hydrogels on chondrogenic differentiation of mesenchymal stem cells in vitro and therefore encourage subsequent in vivo studies.
Collapse
Affiliation(s)
- Achim Salamon
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, Rostock D-18057,
Germany; E-Mails: (F.B.); (V.B.); (J.R.); (K.P.)
- Authors to whom correspondence should be addressed; E-Mails: (A.S.); (S.V.)
| | - Sandra van Vlierberghe
- Polymer Chemistry and Biomaterials Group, Gent University, Krijgslaan 281, Building S4, Gent BE-9000,
Belgium; E-Mails: (I.N.); (G.-J.G.); (P.D.)
- Authors to whom correspondence should be addressed; E-Mails: (A.S.); (S.V.)
| | - Ine van Nieuwenhove
- Polymer Chemistry and Biomaterials Group, Gent University, Krijgslaan 281, Building S4, Gent BE-9000,
Belgium; E-Mails: (I.N.); (G.-J.G.); (P.D.)
| | - Frank Baudisch
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, Rostock D-18057,
Germany; E-Mails: (F.B.); (V.B.); (J.R.); (K.P.)
| | - Geert-Jan Graulus
- Polymer Chemistry and Biomaterials Group, Gent University, Krijgslaan 281, Building S4, Gent BE-9000,
Belgium; E-Mails: (I.N.); (G.-J.G.); (P.D.)
| | - Verena Benecke
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, Rostock D-18057,
Germany; E-Mails: (F.B.); (V.B.); (J.R.); (K.P.)
| | - Kristin Alberti
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, Rostock D-18057,
Germany; E-Mails: (F.B.); (V.B.); (J.R.); (K.P.)
| | | | - Joachim Rychly
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, Rostock D-18057,
Germany; E-Mails: (F.B.); (V.B.); (J.R.); (K.P.)
| | - José C. Martins
- NMR and Structure Analysis Research Group, Gent University, Krijgslaan 281, Building S4, Gent BE-9000,
Belgium; E-Mail:
| | - Peter Dubruel
- Polymer Chemistry and Biomaterials Group, Gent University, Krijgslaan 281, Building S4, Gent BE-9000,
Belgium; E-Mails: (I.N.); (G.-J.G.); (P.D.)
| | - Kirsten Peters
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, Rostock D-18057,
Germany; E-Mails: (F.B.); (V.B.); (J.R.); (K.P.)
| |
Collapse
|
195
|
|
196
|
Araña M, Mazo M, Aranda P, Pelacho B, Prosper F. Adipose tissue-derived mesenchymal stem cells: isolation, expansion, and characterization. Methods Mol Biol 2014; 1036:47-61. [PMID: 23807785 DOI: 10.1007/978-1-62703-511-8_4] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Over the last decade, cell therapy has emerged as a potentially new approach for the treatment of cardiovascular diseases. Among the wide range of cell types and sources, adipose-derived mesenchymal stem cells have shown promise, mainly due to its plasticity and remarkable paracrine-secretion capacity, largely demonstrated at the in vitro and in vivo levels. Furthermore, its accessibility and abundance, the low morbidity of the surgical procedure, its easy isolation, culture, and long-term passaging capacity added to its immunomodulatory properties that could allow its allogeneic transplantation, making it one of the most attractive candidates for clinical application. In this chapter, we will focus on the methodology for the isolation, expansion, phenotypical characterization, differentiation, and storage of the adipose-derived stem cells.
Collapse
Affiliation(s)
- Miriam Araña
- Laboratory of Cell Therapy, Division of Cancer, Foundation for Applied Medical Research, Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | | | | | | | | |
Collapse
|
197
|
Adipose-Derived Stem Cells as a Novel Tool for Future Regenerative Medicine. STEM CELLS AND CANCER STEM CELLS, VOLUME 12 2014. [DOI: 10.1007/978-94-017-8032-2_15] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
198
|
Asahina I. Bone Morphogenetic Proteins: Their History and Characteristics. J HARD TISSUE BIOL 2014. [DOI: 10.2485/jhtb.23.283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
199
|
Ohgushi H. Osteogenically differentiated mesenchymal stem cells and ceramics for bone tissue engineering. Expert Opin Biol Ther 2013; 14:197-208. [PMID: 24308323 DOI: 10.1517/14712598.2014.866086] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION In the human body, cells having self-renewal and multi-differentiation capabilities reside in many tissues and are called adult stem cells. In bone marrow tissue, two types of stem cells are well known: hematopoietic stem cells and mesenchymal stem cells (MSCs). Though the number of MSCs in bone marrow tissue is very low, it can be increased by in vitro culture of the marrow, and culture-expanded MSCs are available for various tissue regeneration. AREAS COVERED The culture-expanded MSCs can further differentiate into osteogenic cells such as bone forming osteoblasts by culturing the MSCs in an osteogenic medium. This paper discusses osteogenically differentiated MSCs derived from the bone marrow of patients. Importantly, the differentiation can be achieved on ceramic surfaces which demonstrate mineralized bone matrix formation as well as appearance of osteogenic cells. The cell/matrix/ceramic constructs could show immediate in vivo bone formation and are available for bone reconstruction surgery. EXPERT OPINION Currently, MSCs are clinically available for the regeneration of various tissues due to their high proliferation/differentiation capabilities. However, the capabilities are still limited and thus technologies to improve or recover the inherent capabilities of MSCs are needed.
Collapse
Affiliation(s)
- Hajime Ohgushi
- Department Head, Ookuma Hospital, Department of Orthopedics , 2-17-13 Kuise-honmachi, Amagasaki City, Hyogo 660-0814 , Japan +81-6-6481-1667 ; +81-6-6481-4234
| |
Collapse
|
200
|
Ma J, Both SK, Yang F, Cui FZ, Pan J, Meijer GJ, Jansen JA, van den Beucken JJJP. Concise review: cell-based strategies in bone tissue engineering and regenerative medicine. Stem Cells Transl Med 2013; 3:98-107. [PMID: 24300556 DOI: 10.5966/sctm.2013-0126] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cellular strategies play an important role in bone tissue engineering and regenerative medicine (BTE/RM). Variability in cell culture procedures (e.g., cell types, cell isolation and expansion, cell seeding methods, and preculture conditions before in vivo implantation) may influence experimental outcome. Meanwhile, outcomes from initial clinical trials are far behind those of animal studies, which is suggested to be related to insufficient nutrient and oxygen supply inside the BTE/RM constructs as some complex clinical implementations require bone regeneration in too large a quantity. Coculture strategies, in which angiogenic cells are introduced into osteogenic cell cultures, might provide a solution for improving vascularization and hence increasing bone formation for cell-based constructs. So far, preclinical studies have demonstrated that cell-based tissue-engineered constructs generally induce more bone formation compared with acellular constructs. Further, cocultures have been shown to enhance vascularization and bone formation compared with monocultures. However, translational efficacy from animal studies to clinical use requires improvement, and the role implanted cells play in clinical bone regeneration needs to be further elucidated. In view of this, the present review provides an overview of the critical procedures during in vitro and in vivo phases for cell-based strategies (both monoculture and coculture) in BTE/RM to achieve more standardized culture conditions for future studies, and hence enhance bone formation.
Collapse
Affiliation(s)
- Jinling Ma
- Department of VIP Service and Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, Capital Medical University, Beijing, China; Department of Biomaterials and Department of Oral and Maxillofacial Surgery, Radboud University Medical Center, Nijmegen, The Netherlands; State Key Laboratory of New Ceramics and Fine Processing, Department of Materials Science & Engineering, Tsinghua University, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|