51
|
Li R, Lin QX, Liang XZ, Liu GB, Tang H, Wang Y, Lu SB, Peng J. Stem cell therapy for treating osteonecrosis of the femoral head: From clinical applications to related basic research. Stem Cell Res Ther 2018; 9:291. [PMID: 30359305 PMCID: PMC6202807 DOI: 10.1186/s13287-018-1018-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a refractory disease that is associated with collapse of the femoral head, with a risk of hip arthroplasty in younger populations. Thus, there has been an increased focus on early interventions for ONFH that aim to preserve the native articulation. Stem cell therapy is a promising treatment, and an increasing number of recent studies have focused on this topic. Many clinical studies have reported positive outcomes of stem cell therapy for the treatment of ONFH. To improve the therapeutic effects of this approach, many related basic research studies have also been performed. However, some issues must be further explored, such as the appropriate patient selection procedure, the optimal stem cell selection protocol, the ideal injection number, and the safety of stem cell therapy. The purpose of this review is to summarize the available clinical studies and basic research related to stem cell therapy for ONFH.
Collapse
Affiliation(s)
- Rui Li
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| | - Qiu-Xia Lin
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| | - Xue-Zhen Liang
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, 250355 Shandong China
| | - Guang-Bo Liu
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| | - He Tang
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| | - Yu Wang
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| | - Shi-Bi Lu
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| | - Jiang Peng
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| |
Collapse
|
52
|
Hanna H, Mir LM, Andre FM. In vitro osteoblastic differentiation of mesenchymal stem cells generates cell layers with distinct properties. Stem Cell Res Ther 2018; 9:203. [PMID: 30053888 PMCID: PMC6063016 DOI: 10.1186/s13287-018-0942-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 06/04/2018] [Accepted: 06/26/2018] [Indexed: 02/06/2023] Open
Abstract
Background Differentiation of mesenchymal stem cells to osteoblasts is widely performed in research laboratories. Classical tests to prove this differentiation employ procedures such as cell fixation, cell lysis or cell scraping. Very few studies report gentle dissociation of mesenchymal stem cells undergoing an osteodifferentiation process. Here we used this technique to reveal the presence of several cell layers during osteogenesis and to study their different properties. Methods Through the sequential enzymatic detachment of the cells, we confirm the presence of several layers of differentiated cells and we compare them in terms of enzymatic sensitivity for dissociation, expression of cluster of differentiation, cytosolic calcium oscillations and osteogenic potential. Adipogenic and neurogenic differentiations were also performed in order to compare the cell layers. Results The cells undergoing differentiation formed one layer in the neurogenic differentiation, two layers in the adipogenic differentiation and at least four layers in the osteogenic differentiation. In the latter, the upper layers, maintained by a collagen I extracellular matrix, can be dissociated using collagenase I, while the remaining lowest layer, attached to the bottom of the dish, is sensitive only to trypsin-versene. The action of collagenase I is more efficient before the mineralization of the extracellular matrix. The collagenase-sensitive and trypsin-sensitive layers differ in their cluster of differentiation expression. The dissociation of the cells on day 15 reveals that cells could resume their growth (increase in cell number) and rapidly differentiate again in osteoblasts, in 2 weeks (instead of 4 weeks). Cells from the upper layers displayed a higher mineralization. Conclusions MSCs undergoing osteogenic differentiation form several layers with distinct osteogenic properties. This could allow the investigators to use upper layers to rapidly produce differentiated osteoblasts and the lowest layer to continue growth and differentiation until an ulterior dissociation. Electronic supplementary material The online version of this article (10.1186/s13287-018-0942-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hanna Hanna
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, PR2, 114 rue Edouard Vaillant, 94 805, Villejuif, France
| | - Lluis M Mir
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, PR2, 114 rue Edouard Vaillant, 94 805, Villejuif, France
| | - Franck M Andre
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, PR2, 114 rue Edouard Vaillant, 94 805, Villejuif, France.
| |
Collapse
|
53
|
Knitted 3D Scaffolds of Polybutylene Succinate Support Human Mesenchymal Stem Cell Growth and Osteogenesis. Stem Cells Int 2018; 2018:5928935. [PMID: 29853915 PMCID: PMC5964421 DOI: 10.1155/2018/5928935] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/01/2018] [Indexed: 01/09/2023] Open
Abstract
Polybutylene succinate (PBS) is a biodegradable polyester with better processability and different mechanical properties compared to polylactides (PLAs), the most commonly used synthetic polymers in tissue engineering (TE). Since only few studies have evaluated PBS-containing materials for bone TE, we prepared PLA-PBS blends and analyzed material properties as well as cell attachment, proliferation, and osteogenic differentiation of human mesenchymal stem cells (hMSCs) on scaffolds. In addition to PLA, PBS, and PLA-PBS blends, PLA-polycaprolactone and PLA-poly(trimethylene carbonate) blends were evaluated. Polymer fibers were prepared using melt spinning. Pure PBS was observed to have the highest crystallinity and strain at break compared to the tougher PLA and PLA blends. No degradation occurred during the 4-week hydrolysis in either of the materials. Knitted and rolled scaffolds were manufactured, seeded with hMSCs, and cultured for 27 days. Human MSC viability was good on all the materials, but cell spreading along the fibers was only detected in PBS-containing scaffolds. They also induced the strongest proliferative response and osteogenic differentiation, which diminished with decreasing PBS content. Based on these results, PBS is superior to PLA with respect to hMSC attachment, proliferation, and osteogenesis. This encourages utilizing PBS-based biomaterials more widely in bone TE applications.
Collapse
|
54
|
Tsang EJ, Wu B, Zuk P. MAPK signaling has stage-dependent osteogenic effects on human adipose-derived stem cells in vitro. Connect Tissue Res 2018; 59:129-146. [PMID: 28398098 PMCID: PMC6200338 DOI: 10.1080/03008207.2017.1313248] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 03/14/2017] [Indexed: 02/03/2023]
Abstract
OVERVIEW The use of pro-osteogenic growth factors, such as BMP2, in human adipose-derived stem cell (ASC) osteogenesis is well described. Because these growth factors work via signal transduction pathways, such as the mitogen-activated protein kinase (MAPK) cascade, a study of the relationship between MAPK signaling and ASC osteogenesis was conducted. MATERIALS AND METHODS ERK, JNK, and p38MAPK activation were measured in ASCs osteo-induced using either dexamethasone or vitamin D3 and correlated with mineralization. Activation and mineralization were also measured without dexamethasone or using the glucocorticoid, cortisone. The expression of the MAPK phosphatase, MKP1, and its relationship to mineralization was also assessed. The effect of decreasing MAPK activation on mineralization through the use of exogenous inhibitors was examined along with siRNA-knockdown and adenoviral overexpression of ERK1/2. Finally, the effect of ERK1/2 overexpression on ASCs induced on PLGA scaffolds was assessed. RESULTS ASC mineralization in dexamethasone or vitamin D3-induced ASCs correlated with both increased ERK1/2 and JNK1/2 activation. ASCs induced without dexamethasone also mineralized, with JNK1/2 signaling possibly mediating this event. No link between cortisone induction and MAPK signaling could be ascertained. ASCs treated with ERK, JNK, or p38MAPK inhibitors showed decreased osteogenic gene expression and diminished mineralization. Mineralization levels were also affected by viruses designed to inhibit or augment ERK1/2 expression and activity. Finally, ASC mineralization appeared to be a balance between the MAPK kinase activity and MKP1. CONCLUSIONS It is likely that MAPK signaling plays a significant role in ASC osteogenesis, affecting differentiation in kinase- and stage-specific manners.
Collapse
Affiliation(s)
- Eric J. Tsang
- Regenerative Bioengineering and Repair Laboratory, Department of Surgery, David Geffen School of Medicine at UCLA. University of California, Los Angeles, CA, USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences. University of California, Los Angeles, CA, USA
| | - Benjamin Wu
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences. University of California, Los Angeles, CA, USA
| | - Patricia Zuk
- Regenerative Bioengineering and Repair Laboratory, Department of Surgery, David Geffen School of Medicine at UCLA. University of California, Los Angeles, CA, USA
| |
Collapse
|
55
|
Kuismanen K, Juntunen M, Narra Girish N, Tuominen H, Huhtala H, Nieminen K, Hyttinen J, Miettinen S. Functional Outcome of Human Adipose Stem Cell Injections in Rat Anal Sphincter Acute Injury Model. Stem Cells Transl Med 2018; 7:295-304. [PMID: 29383878 PMCID: PMC5827744 DOI: 10.1002/sctm.17-0208] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 12/24/2017] [Indexed: 02/06/2023] Open
Abstract
Anal incontinence is a devastating condition that significantly reduces the quality of life. Our aim was to evaluate the effect of human adipose stem cell (hASC) injections in a rat model for anal sphincter injury, which is the main cause of anal incontinence in humans. Furthermore, we tested if the efficacy of hASCs could be improved by combining them with polyacrylamide hydrogel carrier, Bulkamid. Human ASCs derived from a female donor were culture expanded in DMEM/F12 supplemented with human platelet lysate. Female virgin Sprague‐Dawley rats were randomized into four groups (n = 14–15/group): hASCs in saline or Bulkamid (3 × 105/60 μl) and saline or Bulkamid without cells. Anorectal manometry (ARM) was performed before anal sphincter injury, at two (n = 58) and at four weeks after (n = 33). Additionally, the anal sphincter tissue was examined by micro‐computed tomography (μCT) and the histological parameters were compared between the groups. The median resting and peak pressure during spontaneous contraction measured by ARM were significantly higher in hASC treatment groups compared with the control groups without hASCs. There was no statistical difference in functional results between the hASC‐carrier groups (saline vs. Bulkamid). No difference was detected in the sphincter muscle continuation between the groups in the histology and μCT analysis. More inflammation was discovered in the group receiving saline with hASC. The hASC injection therapy with both saline and Bulkamid is a promising nonsurgical treatment for acute anal sphincter injury. Traditional histology combined with the 3D μCT image data lends greater confidence in assessing muscle healing and continuity. Stem Cells Translational Medicine2018;7:295–304
Collapse
Affiliation(s)
- Kirsi Kuismanen
- Tampere University Hospital, department of Obstetrics and Gynecology, Tampere, Finland.,University of Tampere, Faculty of Medicine and Life Sciences, Tampere, Finland
| | - Miia Juntunen
- University of Tampere, Faculty of Medicine and Life Sciences, Tampere, Finland
| | | | - Heikki Tuominen
- Tampere University Hospital, department of Clinical Physiology and Nuclear Medicine, Tampere, Finland
| | - Heini Huhtala
- University of Tampere, Faculty of Social Sciences, Tampere, Finland
| | - Kari Nieminen
- Tampere University Hospital, department of Obstetrics and Gynecology, Tampere, Finland
| | | | - Susanna Miettinen
- University of Tampere, Faculty of Medicine and Life Sciences, Tampere, Finland
| |
Collapse
|
56
|
Egro F, Marra KG. Soft Tissue Reconstruction. Methods Mol Biol 2018; 1773:203-213. [PMID: 29687392 DOI: 10.1007/978-1-4939-7799-4_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Autologous fat transplantation has revolutionized soft tissue reconstruction, but conventional methods remain unpredictable as graft resorption rates are high due to lack of vascularization. The advent of adipose-derived stem cells (ASCs) has led to improvement of fat grafting outcomes, in part to their ability to undergo facile differentiation into adipose tissue, their angiogenic properties, and their ability to express and secrete multiple growth factors. This chapter discusses the isolation and characterization of human ASCs, its expansion in vitro, and relevant in vivo models for adipose tissue engineering.
Collapse
|
57
|
Gentile P, Cervelli V. Adipose-Derived Stromal Vascular Fraction Cells and Platelet-Rich Plasma: Basic and Clinical Implications for Tissue Engineering Therapies in Regenerative Surgery. Methods Mol Biol 2018; 1773:107-122. [PMID: 29687384 DOI: 10.1007/978-1-4939-7799-4_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cell-based therapy and regenerative medicine offer a paradigm shift in regard to various diseases causing loss of substance or volume and tissue or organ damage. Recently, many authors have focused their attention on mesenchymal stem cells for their capacity to differentiate into many cell lineages. The most widely studied types are bone marrow mesenchymal stem cells and adipose derived stem cells (ADSCs), which display similar results. Based on the literature, we believe that the ADSCs offer advantages because of lower morbidity during the harvesting procedure. Additionally, platelet-rich plasma can be used in this field for its ability to stimulate tissue regeneration. The aim of this chapter is to describe ADSC preparation and isolation procedures, preparation of platelet-rich plasma, and the application of ADSCs in regenerative plastic surgery. We also discuss the mechanisms and future role of ADSCs in cell-based therapy and tissue engineering.
Collapse
Affiliation(s)
- Pietro Gentile
- Department of Plastic and Reconstructive Surgery, University of Rome "Tor Vergata", Rome, Italy.
- Catholic University, Tirane, Albania.
| | - Valerio Cervelli
- Department of Plastic and Reconstructive Surgery, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
58
|
Tabatabaei FS, Ai J. Mesenchymal endometrial stem/stromal cells for hard tissue engineering: a review of in vitro and in vivo evidence. Regen Med 2017; 12:983-995. [PMID: 29215321 DOI: 10.2217/rme-2017-0029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hard tissues including teeth, bone and cartilage have inability or poor capacity to self-renew, especially in large defects. Therefore, repair of damages in these tissues represents a huge challenge in the medical field today. Hard tissue engineering commonly utilizes different stem cell sources as a promising strategy for treating bone, cartilages and tooth defects or disorders. Decades ago, researchers successfully isolated and identified endometrial mesenchymal stem/stromal cells (EnSCs) and discovered their multidifferentiation potential. Current studies suggest that EnSCs have significant advantages compared with stem cells derived from other tissues. In this review article, we summarize the current in vitro and in vivo studies that utilize EnSCs or menstrual blood-derived stem cells for differentiation to osteoblasts, odontoblasts or chondroblasts in an effort to realize the potential of these cells in hard tissues regeneration.
Collapse
Affiliation(s)
- Fahimeh S Tabatabaei
- Department of Dental Biomaterials, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering & Applied Cell Sciences, Faculty of Advance Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
59
|
Hodges WM, O'Brien F, Fulzele S, Hamrick MW. Function of microRNAs in the Osteogenic Differentiation and Therapeutic Application of Adipose-Derived Stem Cells (ASCs). Int J Mol Sci 2017; 18:ijms18122597. [PMID: 29207475 PMCID: PMC5751200 DOI: 10.3390/ijms18122597] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 02/08/2023] Open
Abstract
Traumatic wounds with segmental bone defects represent substantial reconstructive challenges. Autologous bone grafting is considered the gold standard for surgical treatment in many cases, but donor site morbidity and associated post-operative complications remain a concern. Advances in regenerative techniques utilizing mesenchymal stem cell populations from bone and adipose tissue have opened the door to improving bone repair in the limbs, spine, and craniofacial skeleton. The widespread availability, ease of extraction, and lack of immunogenicity have made adipose-derived stem cells (ASCs) particularly attractive as a stem cell source for regenerative strategies. Recently it has been shown that small, non-coding miRNAs are involved in the osteogenic differentiation of ASCs. Specifically, microRNAs such as miR-17, miR-23a, and miR-31 are expressed during the osteogenic differentiation of ASCs, and appear to play a role in inhibiting various steps in bone morphogenetic protein-2 (BMP2) mediated osteogenesis. Importantly, a number of microRNAs including miR-17 and miR-31 that act to attenuate the osteogenic differentiation of ASCs are themselves stimulated by transforming growth factor β-1 (TGFβ-1). In addition, transforming growth factor β-1 is also known to suppress the expression of microRNAs involved in myogenic differentiation. These data suggest that preconditioning strategies to reduce TGFβ-1 activity in ASCs may improve the therapeutic potential of ASCs for musculoskeletal application. Moreover, these findings support the isolation of ASCs from subcutaneous fat depots that tend to have low endogenous levels of TGFβ-1 expression.
Collapse
Affiliation(s)
- Walter M Hodges
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - Frederick O'Brien
- Dwight D. Eisenhower Army Medical Center, Fort Gordon, Augusta, GA 30912, USA.
| | - Sadanand Fulzele
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - Mark W Hamrick
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
60
|
Mei H, González S, Nakatsu MN, Baclagon ER, Chen FV, Deng SX. Human adipose-derived stem cells support the growth of limbal stem/progenitor cells. PLoS One 2017; 12:e0186238. [PMID: 29020119 PMCID: PMC5636133 DOI: 10.1371/journal.pone.0186238] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 09/27/2017] [Indexed: 01/08/2023] Open
Abstract
The most efficient method to expand limbal stem cells (LSCs) in vitro for clinical transplantation is to culture single LSCs directly on growth-arrested mouse fibroblast 3T3 cells. To reduce possible xenobiotic contamination from 3T3s, primary human adipose-derived stem cells (ASCs) were examined as feeder cells to support the expansion of LSCs in vitro. To optimize the ASC-supported culture, freshly isolated limbal epithelial cells in the form of single cells (SC-ASC) or cell clusters (CC-ASC) were cultured using three different methods: LSCs seeded directly on feeder cells, a 3-dimensional (3D) culture system and a 3D culture system with fibrin (fibrin 3D). The expanded LSCs were examined at the end of a 2-week culture. The standard 3T3 culture served as control. Expansion of SC-ASC showed limited proliferation and exhibited differentiated morphology. CC-ASC generated epithelial cells with undifferentiated morphology in all culture methods, among which CC-ASC in 3D culture supported the highest cell doubling (cells doubled 9.0 times compared to cells doubled 4.9 times in control) while maintained the percentage of putative limbal stem/progenitor cells compared to the control. There were few cell-cell contacts between cultured LSCs and ASCs in 3D CC-ASC. In conclusion, ASCs support the growth of LSCs in the form of cell clusters but not in single cells. 3D CC-ASC could serve as a substitute for the standard 3T3 culture to expand LSCs.
Collapse
Affiliation(s)
- Hua Mei
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, California, United States of America
| | - Sheyla González
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, California, United States of America
| | - Martin N. Nakatsu
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, California, United States of America
| | - Elfren R. Baclagon
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, California, United States of America
| | - Felix V. Chen
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, California, United States of America
- UCLA College of Letters and Science, University of California, Los Angeles, California, United States of America
| | - Sophie X. Deng
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
61
|
Lee WJ, Lee SC, Lee JH, Rho GJ, Lee SL. Differential regulation of senescence and in vitro differentiation by 17β-estradiol between mesenchymal stem cells derived from male and female mini-pigs. J Vet Sci 2017; 17:159-70. [PMID: 26645340 PMCID: PMC4921664 DOI: 10.4142/jvs.2016.17.2.159] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 09/17/2015] [Accepted: 10/07/2015] [Indexed: 11/26/2022] Open
Abstract
The characterization and potential of mesenchymal stem cells (MSCs) are gender dependent and estrogen influences these properties. This study demonstrated that supplementation with 17β-estradiol (E2) increases the proliferation of bone marrow-MSCs derived from male and female mini-pigs (Mp- and Fp-BMSCs) in a concentration-dependent manner, with 10-12 M E2 suggested as the optimal dose of E2 that led to the greatest improvement in BMSCs proliferation. Supplementation of 10-12 M E2 resulted in down-regulation of β-galactosidase activity and pro-apoptotic activity in both BMSCs, while anti-apoptotic activity was up-regulated in only Fp-BMSCs. Further, E2 increased the osteogenic ability of Fp-BMSCs. Based on these findings, optimal utilization of E2 can improve cellular senescence and apoptosis, as well as in vitro osteogenesis of BMSCs, and could therefore be useful in stem cell therapy, particularly in bone regeneration for adult females.
Collapse
Affiliation(s)
- Won-Jae Lee
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea.,PWG Genetics Pte. Ltd., 15 Tech Park Crescent, Singapore 638117, Singapore
| | - Seung-Chan Lee
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea
| | - Jeong-Hyun Lee
- Advanced Therapy Products Research Division, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju 28159, Korea
| | - Gyu-Jin Rho
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea.,Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Korea
| | - Sung-Lim Lee
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea.,Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Korea
| |
Collapse
|
62
|
Barba M, Di Taranto G, Lattanzi W. Adipose-derived stem cell therapies for bone regeneration. Expert Opin Biol Ther 2017; 17:677-689. [PMID: 28374644 DOI: 10.1080/14712598.2017.1315403] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Cell-based therapies exploit the heterogeneous and self-sufficient biological environment of stem cells to restore, maintain and improve tissue functions. Adipose-derived stem cells (ASCs) are, to this aim, promising cell types thanks to advantageous isolation procedures, growth kinetics, plasticity and trophic properties. Specifically, bone regeneration represents a suitable, though often challenging, target setting to test and apply ASC-based therapeutic strategies. Areas covered: ASCs are extremely plastic and secrete bioactive peptides that mediate paracrine functions, mediating their trophic actions in vivo. Numerous preclinical studies demonstrated that ASCs improve bone healing. Clinical trials are ongoing to validate the clinical feasibility of these approaches. This review is intended to define the state-of-the-art on ASCs, encompassing the biological features that make them suitable for bone regenerative strategies, and to provide an update on existing preclinical and clinical applications. Expert opinion: ASCs offer numerous advantages over other stem cells in terms of feasibility of clinical translation. Data obtained from in vivo experimentation are encouraging, and clinical trials are ongoing. More robust validations are thus expected to be achieved during the next few years, and will likely pave the way to optimized patient-tailored treatments for bone regeneration.
Collapse
Affiliation(s)
- Marta Barba
- a Institute of Anatomy and Cell Biology , Università Cattolica del Sacro Cuore , Rome , Italy
| | - Giuseppe Di Taranto
- b Department of Plastic, Reconstructive and Aesthetic Surgery , University of Rome "Sapienza" , Policlinico Umberto I, Rome , Italy
| | - Wanda Lattanzi
- a Institute of Anatomy and Cell Biology , Università Cattolica del Sacro Cuore , Rome , Italy
| |
Collapse
|
63
|
Kokai LE, Traktuev DO, Zhang L, Merfeld-Clauss S, DiBernardo G, Lu H, Marra KG, Donnenberg A, Donnenberg V, Meyer EM, Fodor PB, March KL, Rubin JP. Adipose Stem Cell Function Maintained with Age: An Intra-Subject Study of Long-Term Cryopreserved Cells. Aesthet Surg J 2017; 37:454-463. [PMID: 28364523 DOI: 10.1093/asj/sjw197] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Background The progressive decline in tissue mechanical strength that occurs with aging is hypothesized to be due to a loss of resident stem cell number and function. As such, there is concern regarding use of autologous adult stem cell therapy in older patients. To abrogate this, many patients elect to cryopreserve the adipose stromal-vascular fraction (SVF) of lipoaspirate, which contains resident adipose stem cells (ASC). However, it is not clear yet if there is any clinical benefit from banking cells at a younger age. Objectives We performed a comparative analysis of SVF composition and ASC function from cells obtained under GMP conditions from the same three patients with time gap of 7 to 12 years. Methods SVF, cryobanked under good manufacturing practice (GMP) conditions, was thawed and cell yield, viability, and cellular composition were assessed. In parallel, ASC proliferation and efficiency of tri-lineage differentiation were evaluated. Results The results showed no significant differences existed in cell yield and SVF subpopulation composition within the same patient between harvest procedures 7 to 12 years apart. Further, no change in proliferation rates of cultured ASCs was found, and expanded cells from all patients were capable of tri-lineage differentiation. Conclusions By harvesting fat from the same patient at two time points, we have shown that despite the natural human aging process, the prevalence and functional activity of ASCs in an adult mesenchymal stem cell, is highly preserved. Level of Evidence 5.
Collapse
Affiliation(s)
- Lauren E Kokai
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dmitry O Traktuev
- Division of Cardiology, Indiana School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Liyong Zhang
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Hongyan Lu
- Division of Cardiology, Indiana School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Kacey G Marra
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Albert Donnenberg
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Vera Donnenberg
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - E Michael Meyer
- Flow Cytometry Facility, University of Pittsburgh Cancer Center, Pittsburgh, PA, USA
| | - Peter B Fodor
- Plastic surgeon in private practice in Santa Monica, CA, USA
| | - Keith L March
- Division of Cardiology, Indiana School of Medicine, Indiana University, Indianapolis, IN, USA
| | - J Peter Rubin
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
64
|
Varghese J, Griffin M, Mosahebi A, Butler P. Systematic review of patient factors affecting adipose stem cell viability and function: implications for regenerative therapy. Stem Cell Res Ther 2017; 8:45. [PMID: 28241882 PMCID: PMC5329955 DOI: 10.1186/s13287-017-0483-8] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The applications for fat grafting have increased recently, within both regenerative and reconstructive surgery. Although fat harvesting, processing and injection techniques have been extensively studied and standardised, this has not had a big impact on the variability of outcome following fat grafting. This suggests a possible larger role of patient characteristics on adipocyte and adipose-derived stem cell (ADSC) viability and function. This systematic review aims to collate current evidence on the effect of patient factors on adipocyte and ADSC behaviour. METHODS A systematic literature review was performed using MEDLINE, Cochrane Library and EMBASE. It includes outcomes observed in in vitro analyses, in vivo animal studies and clinical studies. Data from basic science work have been included in the discussion to enhance our understanding of the mechanism behind ADSC behaviour. RESULTS A total of 41 papers were included in this review. Accumulating evidence indicates decreased proliferation and differentiation potential of ADSCs with increasing age, body mass index, diabetes mellitus and exposure to radiotherapy and Tamoxifen, although this was not uniformly seen across all studies. Gender, donor site preference, HIV status and chemotherapy did not show a significant influence on fat retention. Circulating oestrogen levels have been shown to support both adipocyte function and graft viability. Evidence so far suggests no significant impact of total cholesterol, hypertension, renal disease, physical exercise and peripheral vascular disease on ADSC yield. CONCLUSIONS A more uniform comparison of all factors highlighted in this review, with the application of a combination of tests for each outcome measure, is essential to fully understand factors that affect adipocyte and ADSC viability, as well as functionality. As these patient factors interact, future studies looking at adipocyte viability need to take them into consideration for conclusions to be meaningful. This would provide crucial information for surgeons when deciding appropriate volumes of lipoaspirate to inject, improve patient selection, and counsel patient expectations with regards to outcomes and likelihood for repeat procedures. An improved understanding will also assist in identification of patient groups that would benefit from graft enrichment and cryopreservation techniques.
Collapse
Affiliation(s)
- Jajini Varghese
- Charles Wolfson Center for Reconstructive Surgery, Royal Free Hospital, London, UK.
| | - Michelle Griffin
- Charles Wolfson Center for Reconstructive Surgery, Royal Free Hospital, London, UK.,UCL Centre for Nanotechnology and Regenerative Medicine, Division of Surgery & Interventional Science, University College London, London, UK
| | - Afshin Mosahebi
- Charles Wolfson Center for Reconstructive Surgery, Royal Free Hospital, London, UK.,UCL Centre for Nanotechnology and Regenerative Medicine, Division of Surgery & Interventional Science, University College London, London, UK
| | - Peter Butler
- Charles Wolfson Center for Reconstructive Surgery, Royal Free Hospital, London, UK.,UCL Centre for Nanotechnology and Regenerative Medicine, Division of Surgery & Interventional Science, University College London, London, UK
| |
Collapse
|
65
|
Discussion: Homeotic and Embryonic Gene Expression in Breast Adipose Tissue and in Adipose Tissues Used as Donor Sites in Plastic Surgery. Plast Reconstr Surg 2017; 139:693e-694e. [PMID: 28234839 DOI: 10.1097/prs.0000000000003071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
66
|
Przekora A, Vandrovcova M, Travnickova M, Pajorova J, Molitor M, Ginalska G, Bacakova L. Evaluation of the potential of chitosan/
β
-1,3-glucan/hydroxyapatite material as a scaffold for living bone graft production
in vitro
by comparison of ADSC and BMDSC behaviour on its surface. Biomed Mater 2017; 12:015030. [DOI: 10.1088/1748-605x/aa56f9] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
67
|
Kim BS, Tilstam PV, Springenberg-Jung K, Boecker AH, Schmitz C, Heinrichs D, Hwang SS, Stromps JP, Ganse B, Kopp R, Knobe M, Bernhagen J, Pallua N, Bucala R. Characterization of adipose tissue macrophages and adipose-derived stem cells in critical wounds. PeerJ 2017; 5:e2824. [PMID: 28070458 PMCID: PMC5217526 DOI: 10.7717/peerj.2824] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 11/22/2016] [Indexed: 01/14/2023] Open
Abstract
Background Subcutaneous adipose tissue is a rich source of adipose tissue macrophages and adipose-derived stem cells which both play a key role in wound repair. While macrophages can be divided into the classically-activated M1 and the alternatively-activated M2 phenotype, ASCs are characterized by the expression of specific stem cell markers. Methods In the present study, we have investigated the expression of common macrophage polarization and stem cell markers in acutely inflamed adipose tissue. Subcutaneous adipose tissue adjacent to acutely inflamed wounds of 20 patients and 20 healthy subjects were harvested and underwent qPCR and flow cytometry analysis. Results Expression levels of the M1-specific markers CD80, iNOS, and IL-1b were significantly elevated in inflammatory adipose tissue when compared to healthy adipose tissue, whereas the M2-specific markers CD163 and TGF-β were decreased. By flow cytometry, a significant shift of adipose tissue macrophage populations towards the M1 phenotype was confirmed. Furthermore, a decrease in the mesenchymal stem cell markers CD29, CD34, and CD105 was observed whereas CD73 and CD90 remained unchanged. Discussion This is the first report describing the predominance of M1 adipose tissue macrophages and the reduction of stem cell marker expression in acutely inflamed, non-healing wounds.
Collapse
Affiliation(s)
- Bong-Sung Kim
- Plastic and Reconstructive Surgery, Hand Surgery-Burn Center, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany.,Department of Medicine, Yale University, New Haven, United States.,Institute of Biochemistry and Molecular Cell Biology, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | | | - Katrin Springenberg-Jung
- Plastic and Reconstructive Surgery, Hand Surgery-Burn Center, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Arne Hendrick Boecker
- Plastic and Reconstructive Surgery, Hand Surgery-Burn Center, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Corinna Schmitz
- Institute of Biochemistry and Molecular Cell Biology, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Daniel Heinrichs
- Institute of Biochemistry and Molecular Cell Biology, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Soo Seok Hwang
- Department of Immunology, Yale University, New Haven, United States
| | - Jan Philipp Stromps
- Plastic and Reconstructive Surgery, Hand Surgery-Burn Center, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Bergita Ganse
- Department of Orthopedic Trauma Surgery, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Ruedger Kopp
- Department of Intensive Care Medicine, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Matthias Knobe
- Department of Orthopedic Trauma Surgery, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Juergen Bernhagen
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig-Maximilians-Universität München (LMU), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Norbert Pallua
- Plastic and Reconstructive Surgery, Hand Surgery-Burn Center, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Richard Bucala
- Department of Medicine, Yale University, New Haven, United States
| |
Collapse
|
68
|
Gentile P, Scioli MG, Bielli A, Orlandi A, Cervelli V. Concise Review: The Use of Adipose-Derived Stromal Vascular Fraction Cells and Platelet Rich Plasma in Regenerative Plastic Surgery. Stem Cells 2017; 35:117-134. [PMID: 27641055 DOI: 10.1002/stem.2498] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 08/30/2016] [Indexed: 12/14/2022]
Abstract
Tissue engineering has emerged at the intersection of numerous disciplines to meet a global clinical need for technologies to promote the regeneration of tissues. Recently, many authors have focused their attention on mesenchymal stem/stromal cells (MSCs) for their capacity to differentiate into many cell lineages. The most widely studied cell types are bone marrow mesenchymal stem cells and adipose-derived stem cells (ASCs), which display similar results. Biomaterials, cells, and growth factors are needed to design a regenerative plastic surgery approach in the treatment of organ and tissue defects, but not all tissues are created equal. The aim of this article is to describe the advances in tissue engineering through the use of ASCs, platelet rich plasma, and biomaterials to enable regeneration of damaged complex tissue. Stem Cells 2017;35:117-134.
Collapse
Affiliation(s)
- Pietro Gentile
- Plastic and Reconstructive Surgery, University of Rome "Tor Vergata", Rome, Italy
- Plastic and Reconstructive Surgery, Catholic University "Our Lady of Good Counsel", Tirane, Albania
| | | | - Alessandra Bielli
- Anatomic Pathology Institute, University of Rome "Tor Vergata", Rome, Italy
| | - Augusto Orlandi
- Anatomic Pathology Institute, University of Rome "Tor Vergata", Rome, Italy
| | - Valerio Cervelli
- Plastic and Reconstructive Surgery, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
69
|
Saei Arezoumand K, Alizadeh E, Pilehvar-Soltanahmadi Y, Esmaeillou M, Zarghami N. An overview on different strategies for the stemness maintenance of MSCs. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:1255-1271. [DOI: 10.1080/21691401.2016.1246452] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Khatereh Saei Arezoumand
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Effat Alizadeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Younes Pilehvar-Soltanahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Esmaeillou
- Department of Medical Biotechnologies, Universita degli Studi di siena, Siena, Italy
| | - Nosratollah Zarghami
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
70
|
Guo SC, Tao SC, Yin WJ, Qi X, Sheng JG, Zhang CQ. Exosomes from Human Synovial-Derived Mesenchymal Stem Cells Prevent Glucocorticoid-Induced Osteonecrosis of the Femoral Head in the Rat. Int J Biol Sci 2016; 12:1262-1272. [PMID: 27766040 PMCID: PMC5069447 DOI: 10.7150/ijbs.16150] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/19/2016] [Indexed: 12/19/2022] Open
Abstract
Osteonecrosis of the femoral head (ONFH) represents a debilitating complication following glucocorticoid (GC)-based therapy. Synovial-derived mesenchymal stem cells (SMSCs) can exert protective effect in the animal model of GC-induced ONFH by inducing cell proliferation and preventing cell apoptosis. Recent studies indicate the transplanted cells exert therapeutic effects primarily via a paracrine mechanism and exosomes are an important paracrine factor that can be directly used as therapeutic agents for tissue engineering. Herein, we provided the first demonstration that the early treatment of exosomes secreted by human synovial-derived mesenchymal stem cells (SMSC-Exos) could prevent GC-induced ONFH in the rat model. Using a series of in vitro functional assays, we found that SMSC-Exos could be internalized into bone marrow derived stromal cells (BMSCs) and enhance their proliferation and have anti-apoptotic abilities. Finally, SMSC-Exos may be promising for preventing GC-induced ONFH.
Collapse
Affiliation(s)
- Shang-Chun Guo
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Shi-Cong Tao
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Wen-Jing Yin
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Xin Qi
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Jia-Gen Sheng
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Chang-Qing Zhang
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China;; Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| |
Collapse
|
71
|
Chondrogenic Potency Analyses of Donor-Matched Chondrocytes and Mesenchymal Stem Cells Derived from Bone Marrow, Infrapatellar Fat Pad, and Subcutaneous Fat. Stem Cells Int 2016; 2016:6969726. [PMID: 27781068 PMCID: PMC5066011 DOI: 10.1155/2016/6969726] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/04/2016] [Indexed: 12/25/2022] Open
Abstract
Autologous chondrocyte implantation (ACI) is a cell-based therapy that has been used clinically for over 20 years to treat cartilage injuries more efficiently in order to negate or delay the need for joint replacement surgery. In this time, very little has changed in the ACI procedure, but now many centres are considering or using alternative cell sources for cartilage repair, in particular mesenchymal stem cells (MSCs). In this study, we have tested the chondrogenic potential of donor-matched MSCs derived from bone marrow (BM), infrapatellar fat pad (FP), and subcutaneous fat (SCF), compared to chondrocytes. We have confirmed that there is a chondrogenic potency hierarchy ranging across these cell types, with the most potent being chondrocytes, followed by FP-MSCs, BM-MSCs, and lastly SCF-MSCs. We have also examined gene expression and surface marker profiles in a predictive model to identify cells with enhanced chondrogenic potential. In doing so, we have shown that Sox-9, Alk-1, and Coll X expressions, as well as immunopositivity for CD49c and CD39, have predictive value for all of the cell types tested in indicating chondrogenic potency. The findings from this study have significant clinical implications for the refinement and development of novel cell-based cartilage repair strategies.
Collapse
|
72
|
Nordberg RC, Zhang J, Griffith EH, Frank MW, Starly B, Loboa EG. Electrical Cell-Substrate Impedance Spectroscopy Can Monitor Age-Grouped Human Adipose Stem Cell Variability During Osteogenic Differentiation. Stem Cells Transl Med 2016; 6:502-511. [PMID: 28191763 PMCID: PMC5442814 DOI: 10.5966/sctm.2015-0404] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 07/28/2016] [Indexed: 01/01/2023] Open
Abstract
Human adipose stem cells (hASCs) are an attractive cell source for bone tissue engineering applications. However, a critical issue to be addressed before widespread hASC clinical translation is the dramatic variability in proliferative capacity and osteogenic potential among hASCs isolated from different donors. The goal of this study was to test our hypothesis that electrical cell-substrate impedance spectroscopy (ECIS) could track complex bioimpedance patterns of hASCs throughout proliferation and osteogenic differentiation to better understand and predict variability among hASC populations. Superlots composed of hASCs from young (aged 24-36 years), middle-aged (aged 48-55 years), and elderly (aged 60-81 years) donors were seeded on gold electrode arrays. Complex impedance measurements were taken throughout proliferation and osteogenic differentiation. During osteogenic differentiation, four impedance phases were identified: increase, primary stabilization, drop phase, and secondary stabilization. Matrix deposition was first observed 48-96 hours after the impedance maximum, indicating, for the first time, that ECIS can identify morphological changes that correspond to late-stage osteogenic differentiation. The impedance maximum was observed at day 10.0 in young, day 6.1 in middle-aged, and day 1.3 in elderly hASCs, suggesting that hASCs from younger donors require a longer time to differentiate than do hASCs from older donors, but young hASCs proliferated more and accreted more calcium long-term. This is the first study to use ECIS to predict osteogenic potential of multiple hASC populations and to show that donor age may temporally control onset of osteogenesis. These findings could be critical for development of patient-specific bone tissue engineering and regenerative medicine therapies. Stem Cells Translational Medicine 2017;6:502-511.
Collapse
Affiliation(s)
- Rachel C. Nordberg
- Joint Department of Biomedical Engineering, University of North Carolina Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
| | - Jianlei Zhang
- Edward P. Fitts Department of Industrial and Systems Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Emily H. Griffith
- Department of Statistics, North Carolina State University, Raleigh, North Carolina, USA
| | - Matthew W. Frank
- Joint Department of Biomedical Engineering, University of North Carolina Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
| | - Binil Starly
- Edward P. Fitts Department of Industrial and Systems Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Elizabeth G. Loboa
- Joint Department of Biomedical Engineering, University of North Carolina Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
- University of Missouri College of Engineering, Columbia, Missouri, USA
| |
Collapse
|
73
|
Grasys J, Kim BS, Pallua N. Content of Soluble Factors and Characteristics of Stromal Vascular Fraction Cells in Lipoaspirates from Different Subcutaneous Adipose Tissue Depots. Aesthet Surg J 2016; 36:831-41. [PMID: 26906346 DOI: 10.1093/asj/sjw022] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2016] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Although fat grafting has emerged as a major force in plastic, reconstructive, and aesthetic surgery, some questions regarding its reliability and regenerative potential remain unanswered. OBJECTIVES The authors examined the influence of three anatomic areas on various lipoaspirate properties to identify the most appropriate harvest site for fat-grafting procedures. METHODS Lipoaspirates from 25 healthy patients were harvested from the abdomen, inner thigh, and knee. The authors measured the content of soluble factors in the lipoaspirate followed by the assessment of the yield, adipogenic differentiation, proliferation of stromal vascular fraction (SVF) cells, and the percentage of adipose-derived stem cells (ASC) in the SVF. The results also were correlated with the age and body mass index of the donors. RESULTS Lipoaspirates from the abdomen showed significantly higher concentrations of matrix metalloproteinase (MMP)-9 compared with the knee. The content of basic fibroblast growth factor (b-FGF), platelet-derived growth factor (PDGF)-BB, and insulin-like growth factor (IGF)-1 tended to be highest in the abdomen but did not reach statistical significance. Vascular endothelial growth factor (VEGF)-A and bFGF-2 contents both correlated negatively with age in lipoaspirates from at least two different anatomic areas. CONCLUSIONS The authors' results indicate that the abdomen may be a slight favorite over the inner thigh and knee because of its richer content of soluble factors. However, because only the difference of MMP-9 content actually reached statistical significance and because no differences in SVF characteristics were observed, a decision primarily based on other criteria appears to be justifiable.
Collapse
Affiliation(s)
- Justinas Grasys
- From the Department of Plastic and Reconstructive Surgery, Hand Surgery - Burn Center, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Bong-Sung Kim
- From the Department of Plastic and Reconstructive Surgery, Hand Surgery - Burn Center, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Norbert Pallua
- From the Department of Plastic and Reconstructive Surgery, Hand Surgery - Burn Center, Medical Faculty, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
74
|
Quaade ML, Jensen CH, Andersen DC, Sheikh SP. A 3-month age difference profoundly alters the primary rat stromal vascular fraction phenotype. Acta Histochem 2016; 118:513-8. [PMID: 27265810 DOI: 10.1016/j.acthis.2016.05.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 05/20/2016] [Accepted: 05/20/2016] [Indexed: 01/01/2023]
Abstract
The stromal vascular fraction (SVF) is a heterogeneous population obtained from collagenase digestion of adipose tissue. When cultured the population becomes more homogeneous and the cells are then termed adipose stromal/stem cells (ASCs). Both the freshly isolated primary SVF population and the cultured ASC population possess regenerative abilities suggested to be mediated by paracrine mechanisms mainly. The use of ASCs and SVF cells, both in animal studies and human clinical studies, has dramatically increased during recent years. However, more knowledge regarding optimal donor characteristics such as age is demanded. Here we report that even a short age difference has an impact on the phenotype of primary SVF cells. We observed that a 3-month difference in relatively young adult rats affects the expression pattern of several mesenchymal stem cell markers in their primary SVF. The younger animals had significantly more CD90+/CD44+/CD29+/PDGFRα+primary cells, than the aged rats, suggesting an age dependent shift in the relative cell type distribution within the population. Taken together with recent studies of much more pronounced age differences, our data strongly suggest that donor age is a very critical parameter that should be taken into account in future stem cell studies, especially when using primary cells.
Collapse
Affiliation(s)
- Marlene Louise Quaade
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark; Department of Cardiovascular and Renal Research, Denmark.
| | - Charlotte Harken Jensen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark.
| | - Ditte Caroline Andersen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark.
| | - Søren Paludan Sheikh
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark; Department of Cardiovascular and Renal Research, Denmark.
| |
Collapse
|
75
|
Alves EG, Serakides R, Boeloni JN, Rosado IR, Ocarino NM, Oliveira HP, Góes AM, Rezende CM. Estudo comparativo da diferenciação osteogênica das células tronco mesenquimais da medula óssea e do tecido adiposo de cães adultos. PESQUISA VETERINARIA BRASILEIRA 2016. [DOI: 10.1590/s0100-736x201600130004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Resumo: O objetivo deste estudo foi comparar o potencial osteogênico das células tronco mesenquimais extraídas da medula óssea (CTM-MO) com as do tecido adiposo (CTM-AD) de cães adultos. As células foram caracterizadas fenotipicamente quanto à expressão de CD29, CD90, CD34 e CD45 e submetidas à diferenciação adipogênica e condrogênica por 21 dias e osteogênica por 7, 14 e 21 dias. Foram constituídos quatro grupos: 1) CTM-MO em meio osteogênico, 2) CTM-MO em meio basal, 3) CTM-AD em meio osteogênico e 4) CTM-AD em meio basal. Aos 7, 14 e 21 dias de diferenciação osteogênica as culturas foram submetidas às avaliações da conversão de MTT em formazan, da atividade da fosfatase alcalina (FA), da síntese de colágeno e de matriz mineralizada, avaliação do número de células por campo e foram quantificados os transcritos gênicos para osterix, sialoproteina óssea (BSP), osteonectina (ON) e osteocalcina (OC). Tanto as células extraídas da medula óssea quanto do tecido adiposo mostraram elevada expressão de marcadores para células tronco e baixa expressão de marcadores de células hematopoiéticas (menor que 2%). Além disso, foram capazes de se diferenciar em osteoblastos, condrócitos e adipócitos. As CTM-AD submetidas à diferenciação osteogênica mostraram maior conversão do MTT em formazan que as CTM-MO, sob mesmas condições aos 7 e 21 dias. O número de células por campo, a atividade da FA, a síntese de colágeno e de matriz mineralizada foram superior nas CTM-AD em diferenciação, em relação às CTM-MO sob as mesmas condições, em todos os tempos estudados. As expressões de osterix, BSP e OC foram predominantemente superiores nas CTM-MO diferenciadas, mas a expressão de ON foi superior nas CTM-AD diferenciadas aos 7, 14 e 21 dias. Conclui-se que as CTM-AD apresentam maior potencial osteogênico que as CTM-MO quando extraídas de cães adultos.
Collapse
|
76
|
New insight on obesity and adipose-derived stem cells using comprehensive metabolomics. Biochem J 2016; 473:2187-203. [PMID: 27208167 DOI: 10.1042/bcj20160241] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 05/19/2016] [Indexed: 12/11/2022]
Abstract
Obesity affects the functional capability of adipose-derived stem cells (ASCs) and their effective use in regenerative medicine through mechanisms that are still poorly understood. In the present study we used a multiplatform [LC/MS, GC/MS and capillary electrophoresis/MS (CE/MS)], metabolomics, untargeted approach to investigate the metabolic alteration underlying the inequalities observed in obesity-derived ASCs. The metabolic fingerprint (metabolites within the cells) and footprint (metabolites secreted in the culture medium), from obesity- and non-obesity-derived ASCs of humans or mice, were characterized to provide valuable information. Metabolites associated with glycolysis, the tricarboxylic acid cycle, the pentose phosphate pathway and the polyol pathway were increased in the footprint of obesity-derived human ASCs, indicating alterations in carbohydrate metabolism, whereas, from the murine model, deep differences in lipid and amino acid catabolism were highlighted. Therefore, new insights on the ASCs' metabolome were provided that enhance our understanding of the processes underlying ASCs' stemness capacity and its relationship with obesity, in different cell models.
Collapse
|
77
|
Duscher D, Luan A, Rennert RC, Atashroo D, Maan ZN, Brett EA, Whittam AJ, Ho N, Lin M, Hu MS, Walmsley GG, Wenny R, Schmidt M, Schilling AF, Machens HG, Huemer GM, Wan DC, Longaker MT, Gurtner GC. Suction assisted liposuction does not impair the regenerative potential of adipose derived stem cells. J Transl Med 2016; 14:126. [PMID: 27153799 PMCID: PMC4859988 DOI: 10.1186/s12967-016-0881-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 04/27/2016] [Indexed: 01/22/2023] Open
Abstract
Background Adipose-derived stem cells (ASCs) have been identified as a population of multipotent cells with promising applications in tissue engineering and regenerative medicine. ASCs are abundant in fat tissue, which can be safely harvested through the minimally invasive procedure of liposuction. However, there exist a variety of different harvesting methods, with unclear impact on ASC regenerative potential. The aim of this study was thus to compare the functionality of ASCs derived from the common technique of suction-assisted lipoaspiration (SAL) versus resection. Methods Human adipose tissue was obtained from paired abdominoplasty and SAL samples from three female donors, and was processed to isolate the stromal vascular fraction. Fluorescence-activated cell sorting was used to determine ASC yield, and cell viability was assayed. Adipogenic and osteogenic differentiation capacity were assessed in vitro using phenotypic staining and quantification of gene expression. Finally, ASCs were applied in an in vivo model of tissue repair to evaluate their regenerative potential. Results SAL specimens provided significantly fewer ASCs when compared to excised fat tissue, however, with equivalent viability. SAL-derived ASCs demonstrated greater expression of the adipogenic markers FABP-4 and LPL, although this did not result in a difference in adipogenic differentiation. There were no differences detected in osteogenic differentiation capacity as measured by alkaline phosphatase, mineralization or osteogenic gene expression. Both SAL- and resection-derived ASCs enhanced significantly cutaneous healing and vascularization in vivo, with no significant difference between the two groups. Conclusion SAL provides viable ASCs with full capacity for multi-lineage differentiation and tissue regeneration, and is an effective method of obtaining ASCs for cell-based therapies.
Collapse
Affiliation(s)
- Dominik Duscher
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA. .,Section of Plastic, Aesthetic and Reconstructive Surgery, Johannes Kepler University, Linz, Austria. .,Department of Plastic Surgery and Hand Surgery, Technical University Munich, Munich, Germany.
| | - Anna Luan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert C Rennert
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - David Atashroo
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Zeshaan N Maan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Elizabeth A Brett
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexander J Whittam
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Natalie Ho
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michelle Lin
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael S Hu
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Graham G Walmsley
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Raphael Wenny
- Section of Plastic, Aesthetic and Reconstructive Surgery, Johannes Kepler University, Linz, Austria
| | - Manfred Schmidt
- Section of Plastic, Aesthetic and Reconstructive Surgery, Johannes Kepler University, Linz, Austria
| | - Arndt F Schilling
- Department of Plastic Surgery and Hand Surgery, Technical University Munich, Munich, Germany
| | - Hans-Günther Machens
- Department of Plastic Surgery and Hand Surgery, Technical University Munich, Munich, Germany
| | - Georg M Huemer
- Section of Plastic, Aesthetic and Reconstructive Surgery, Johannes Kepler University, Linz, Austria
| | - Derrick C Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Geoffrey C Gurtner
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
78
|
Characterisation of synovial fluid and infrapatellar fat pad derived mesenchymal stromal cells: The influence of tissue source and inflammatory stimulus. Sci Rep 2016; 6:24295. [PMID: 27073003 PMCID: PMC4829842 DOI: 10.1038/srep24295] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/16/2016] [Indexed: 12/11/2022] Open
Abstract
The infrapatellar fat pad (FP) and synovial fluid (SF) in the knee serve as reservoirs of mesenchymal stromal cells (MSCs) with potential therapeutic benefit. We determined the influence of the donor on the phenotype of donor matched FP and SF derived MSCs and examined their immunogenic and immunomodulatory properties before and after stimulation with the pro-inflammatory cytokine interferon-gamma (IFN-γ). Both cell populations were positive for MSC markers CD73, CD90 and CD105, and displayed multipotency. FP-MSCs had a significantly faster proliferation rate than SF-MSCs. CD14 positivity was seen in both FP-MSCs and SF-MSCs, and was positively correlated to donor age but only for SF-MSCs. Neither cell population was positive for the co-stimulatory markers CD40, CD80 and CD86, but both demonstrated increased levels of human leukocyte antigen-DR (HLA-DR) following IFN-γ stimulation. HLA-DR production was positively correlated with donor age for FP-MSCs but not SF-MSCs. The immunomodulatory molecule, HLA-G, was constitutively produced by both cell populations, unlike indoleamine 2, 3-dioxygenase which was only produced following IFN-γ stimulation. FP and SF are accessible cell sources which could be utilised in the treatment of cartilage injuries, either by transplantation following ex-vivo expansion or endogenous targeting and mobilisation of cells close to the site of injury.
Collapse
|
79
|
Ock SA, Lee YM, Park JS, Shivakumar SB, Moon SW, Sung NJ, Lee WJ, Jang SJ, Park JM, Lee SC, Lee SL, Rho GJ. Evaluation of phenotypic, functional and molecular characteristics of porcine mesenchymal stromal/stem cells depending on donor age, gender and tissue source. J Vet Med Sci 2016; 78:987-95. [PMID: 26922917 PMCID: PMC4937159 DOI: 10.1292/jvms.15-0596] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The biological properties of mesenchymal stem cells (MSCs) are influenced by donor age, gender and/or tissue
sources. The present study investigated the cellular and molecular properties of porcine mesenchymal
stromal/stem cells (MSCs) isolated from different tissues (adipose & dermal skin) and sex at
different ages (1 week & 8 months after birth) with similar genetic and environmental backgrounds. MSCs
were analyzed for alkaline phosphatase (AP) activity, CD90 and Oct3/4 expression, in vitro
differentiation ability, senescence-associated β-galactosidase (SA-β-Gal)
activity, telomeric properties, cell cycle status and expression of senescence (IL6, c-myc, TGFβ, p53 and
p21)- and apoptosis (Bak and Bcl2)-related proteins. An age-dependent decline in AP activity and adipogenesis
was observed in all MSCs, except for male A-MSCs. CD90 expression did not change, but
SA-β-Gal activity increased with advancement in age, except in A-MSCs. Telomeric properties
were similar in all MSCs, whereas expression levels of Oct3/4 protein declined with the advancement in age.
p21 expression was increased with increase in donor age. Male derived cells have shown higher IL6 expression.
The expression of p53 was slightly lower in MSCs of dermal tissue than in adipose tissue. Bak was expressed in
all MSCs regardless of age, but up regulation of Bcl2 was observed in DS-MSCs derived at 1 week after birth.
In conclusion, adipose tissue-derived MSCs from young female individuals were found to be more resistant to
senescence under in vitro culture conditions.
Collapse
Affiliation(s)
- Sun-A Ock
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University, 501, Jinju-daero, Jinju 660-701, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Riis S, Nielsen FM, Pennisi CP, Zachar V, Fink T. Comparative Analysis of Media and Supplements on Initiation and Expansion of Adipose-Derived Stem Cells. Stem Cells Transl Med 2016; 5:314-24. [PMID: 26838270 DOI: 10.5966/sctm.2015-0148] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 11/02/2015] [Indexed: 12/18/2022] Open
Abstract
Adipose-derived stem cells (ASCs) are being tested in clinical trials related to cell-based regenerative therapies. Although most of the current expansion protocols for ASCs use fetal calf serum (FCS), xenogeneic-free medium supplements are greatly desired. This study aims to compare the effect of FCS, human platelet lysate (hPL), and a fully defined medium on the initiation and maintenance of ASC cultures. ASCs obtained from five donors were cultured in five different media: StemPro, Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% hPL, or α-minimum essential medium (A-MEM) supplemented with 5% hPL, 10% hPL, or 10% FCS. The effect of media on proliferation, colony-forming units (CFUs), attachment, and morphology was assessed along with cell size, granularity, and immunophenotype. StemPro greatly compromised the initiation of ASC cultures, which could not survive more than a few passages. Cells cultured in A-MEM proliferated at a faster rate than in DMEM, and hPL significantly enhanced cell size, granularity, and proliferation compared with FCS. All media except StemPro supported CFUs equally well. Analysis of surface markers revealed higher levels of CD73 and CD105 in FCS-cultured ASCs, whereas increased levels of CD146 were found in hPL-cultured cells. Multiparametric flow cytometric analysis performed after seven passages revealed the existence of four distinct ASC subpopulations, all positive for CD73, CD90, and CD105, which mainly differed by their expression of CD146 and CD271. Analysis of the different subpopulations might represent an important biological measure when assessing different medium formulations for a particular clinical application.
Collapse
Affiliation(s)
- Simone Riis
- Laboratory for Stem Cell Research, Department for Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Frederik Mølgaard Nielsen
- Laboratory for Stem Cell Research, Department for Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Cristian Pablo Pennisi
- Laboratory for Stem Cell Research, Department for Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Vladimir Zachar
- Laboratory for Stem Cell Research, Department for Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Trine Fink
- Laboratory for Stem Cell Research, Department for Health Science and Technology, Aalborg University, Aalborg, Denmark
| |
Collapse
|
81
|
Differential Effects of Processing Time and Duration of Collagenase Digestion on Human and Murine Fat Grafts. Plast Reconstr Surg 2015. [PMID: 26218393 DOI: 10.1097/prs.0000000000001446] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Autologous fat graft retention is unpredictable, and mechanisms of optimization are poorly understood. Attempts at improving retention use collagenase experimentally and clinically to isolate the stromal vascular fraction to "enhance" fat grafts. However, no standardized duration for collagenase digestion or time following fat graft harvest has been established. This study investigates the effect of (1) time after fat graft harvest and (2) collagenase digestion time on interstitial cell and adipocyte viability in murine fat and human lipoaspirate. METHODS Murine fat and human lipoaspirate were incubated ex vivo after harvest at room temperature for 120 minutes. Additional groups were incubated with collagenase for increasing 5-minute intervals from 30 to 60 minutes. Samples from each group were stained with BODIPY to quantify intact adipocytes and the LIVE/DEAD kit to quantify interstitial cell viability. RESULTS With increased time after harvest, the number of intact adipocytes in murine fat and human lipoaspirate remained unchanged. Human interstitial cells were resistant to the effect of increased time ex vivo, whereas murine interstitial cells decreased in viability. In both populations, increased collagenase digestion time significantly decreased the number of viable adipocytes (murine, p ≤ 0.001; human, p ≤ 0.001) and interstitial cells (murine, p ≤ 0.001; human, p ≤ 0.001). CONCLUSIONS Human and murine adipocytes and human interstitial cells appear resistant to deleterious effects of increasing time following harvest. However, murine interstitial cells are sensitive to increased time and prolonged collagenase digestion. These studies highlight the complex cellular components of fat grafts and how they respond differentially to time and collagenase digestion.
Collapse
|
82
|
Abstract
We have witnessed a rapid expansion of in vitro characterization and differentiation of adipose-derived stem cells, with increasing translation to both in vivo models and a breadth of clinical specialties. However, an appreciation of the truly heterogeneous nature of this unique stem cell group has identified a need to more accurately delineate subpopulations by any of a host of methods, to include functional properties or surface marker expression. Cells selected for improved proliferative, differentiative, angiogenic or ischemia-resistant properties are but a few attributes that could prove beneficial for targeted treatments or therapies. Optimizing cell culture conditions to permit re-introduction to patients is critical for clinical translation.
Collapse
Affiliation(s)
- Kavan S Johal
- Blond McIndoe Laboratories, Institute of Inflammation & Repair, School of Medicine, University of Manchester, M13 9PT, UK
| | | | | |
Collapse
|
83
|
Differential Effects of Processing Time and Duration of Collagenase Digestion on Human and Murine Fat Grafts. Plast Reconstr Surg 2015. [PMID: 26218393 DOI: 10.1097/prs.0000000000001446.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Autologous fat graft retention is unpredictable, and mechanisms of optimization are poorly understood. Attempts at improving retention use collagenase experimentally and clinically to isolate the stromal vascular fraction to "enhance" fat grafts. However, no standardized duration for collagenase digestion or time following fat graft harvest has been established. This study investigates the effect of (1) time after fat graft harvest and (2) collagenase digestion time on interstitial cell and adipocyte viability in murine fat and human lipoaspirate. METHODS Murine fat and human lipoaspirate were incubated ex vivo after harvest at room temperature for 120 minutes. Additional groups were incubated with collagenase for increasing 5-minute intervals from 30 to 60 minutes. Samples from each group were stained with BODIPY to quantify intact adipocytes and the LIVE/DEAD kit to quantify interstitial cell viability. RESULTS With increased time after harvest, the number of intact adipocytes in murine fat and human lipoaspirate remained unchanged. Human interstitial cells were resistant to the effect of increased time ex vivo, whereas murine interstitial cells decreased in viability. In both populations, increased collagenase digestion time significantly decreased the number of viable adipocytes (murine, p ≤ 0.001; human, p ≤ 0.001) and interstitial cells (murine, p ≤ 0.001; human, p ≤ 0.001). CONCLUSIONS Human and murine adipocytes and human interstitial cells appear resistant to deleterious effects of increasing time following harvest. However, murine interstitial cells are sensitive to increased time and prolonged collagenase digestion. These studies highlight the complex cellular components of fat grafts and how they respond differentially to time and collagenase digestion.
Collapse
|
84
|
Chen JY, Mou XZ, Du XC, Xiang C. Comparative analysis of biological characteristics of adult mesenchymal stem cells with different tissue origins. ASIAN PAC J TROP MED 2015; 8:739-46. [PMID: 26433660 DOI: 10.1016/j.apjtm.2015.07.022] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 07/20/2015] [Accepted: 07/20/2015] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE To invest the differences among mesenchymal stem cells (MSCs) derived from different tissues and their impacts on clinical applications. METHODS In this study, MSCs were isolated from adipose tissue (AD), umbilical cord tissue (UC), and menstrual blood (Men) and compared their biological characteristics in terms of proliferation capacity, passage capacity, colony formation, and surface markers were compared. RESULTS The stem cells (SCs) obtained from different sources were all characterized as MSCs, but demonstrated some differences. Umbilical cord-derived MSCs (UCMSCs) were able to overcome density inhibition. The proliferation rate decreased in the order UCMSCs > MenSCs > ADSCs, while the colony-forming ability decreased in the order MenSCs > ADSCs > UCMSCs. Based on gene-expression data for MSCs from different sources within the same donor, 768 MenSC genes were found that were specifically upregulated or downregulated compared with bone marrow-derived MSCs and UCMSCs, most of which were involved in cell function-related pathways. In addition, MenSCs appeared to be superior in terms of immune inflammation, stress response, and neural differentiation potentials, but weaker in terms of osteogenic and chondrogenic differentiation capacities, compared with UCMSCs and bone marrow-derived MSCs. CONCLUSIONS MenSCs have higher extraction efficiency, colony-forming ability, and long time passage capacity. Although the proliferation capacity is inferior to UCMSCs.
Collapse
Affiliation(s)
- Jin-Yang Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Xiao-Zhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | | | - Charlie Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China; S-Evans Biosciences, Hangzhou 311121, China.
| |
Collapse
|
85
|
Human adipose-derived mesenchymal stem cells attenuate collagen antibody-induced autoimmune arthritis by inducing expression of FCGIIB receptors. BMC Musculoskelet Disord 2015. [PMID: 26210906 PMCID: PMC4515315 DOI: 10.1186/s12891-015-0634-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background Adipose-derived stem cells (ASCs) are mesenchymal stem cells (MSCs) derived from adipose tissue. MSCs have multiple properties including anti-inflammatory and immunomodulatory effects in various disease models and human diseases. However, the mechanisms underlying this wide range of effects need to be explored. Methods Collagen antibody-induced arthritis (CAIA) is a unique model in which arthritis is rapidly and strongly induced. ASCs were intraperitoneally infused into CAIA mice before or after arthritis induction. The serum levels of various cytokines, adipokines, and chemokines were measured. The expression of FC gamma receptors (FCGRs) was investigated in peritoneal macrophages ex vivo. RAW264.7 cells and ASCs were co-cultured to elucidate the direct and indirect role of ASCs on FCGR expression. Results ASCs attenuated arthritis in CAIA mice. Serum levels of tumor necrosis factor α, interleukin (IL)-15, resistin, and leptin were reduced in ASC-treated CAIA mice, whereas serum levels of IL-6 and adiponectin were not affected. In peritoneal macrophages isolated from ASC-treated mice, expression of FCGRIIB, which is immunoinhibitory, was higher than that of FCGRI. Co-culture of ASCs with RAW264.7 cells modulated the expression of FCGRs. The expression patterns and timings of peak expression differed among FCGRs. Expression of FCGRIIB was higher and peaked earlier than that of FCGRI. FCGRIII expression was not affected by this co-culture. Conclusions This is a study to show that ASCs have anti-arthritic effects in CAIA mice. Modulation of FCGRs by ASCs might be a therapeutic mechanism in this antibody-associated arthritis model.
Collapse
|
86
|
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
|
87
|
Di Taranto G, Cicione C, Visconti G, Isgrò MA, Barba M, Di Stasio E, Stigliano E, Bernardini C, Michetti F, Salgarello M, Lattanzi W. Qualitative and quantitative differences of adipose-derived stromal cells from superficial and deep subcutaneous lipoaspirates: a matter of fat. Cytotherapy 2015; 17:1076-89. [PMID: 26002819 DOI: 10.1016/j.jcyt.2015.04.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 04/08/2015] [Accepted: 04/09/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND AIMS Subcutaneous fat represents a valuable reservoir of adipose-derived stem cells (ASCs) in the stromal vascular fraction (SVF), widely exploited in regenerative medicine applications, being easily harvested through lipoaspiration. The lack of standardized procedures for autologous fat grafting guided research efforts aimed at identifying possible differences related to the harvesting site, which may affect cell isolation yield, cell growth properties and clinical outcomes. Subcutaneous fat features a complex architecture: the superficial fascia separates superficial adipose tissue (SAT) from deep layer tissue (DAT). We aimed to unravel the differences between SAT and DAT, considering morphological structure, SVF composition, and ASC properties. METHODS SAT and DAT were collected from female donors and comparatively analyzed to evaluate cellular yield and viability, morphology, immunophenotype and molecular profile. ASCs were isolated in primary culture and used for in vitro differentiation assays. SAT and DAT from cadaver donors were also analyzed through histology and immunohistochemistry to assess morphology and cell localization within the hypoderm. RESULTS Liposuctioned SAT contained a higher stromal tissue compound, along with a higher proportion of CD105-positive cells, compared with DAT from the same harvesting site. Also, cells isolated from SAT displayed increased multipotency and stemness features. All differences were mainly evidenced in specimens harvested from the abdominal region. According to our results, SAT features overall increased stem properties. CONCLUSIONS Given that subcutaneous adipose tissue is currently exploited as the gold standard source for high-yield isolation of adult stem cells, these results may provide precious hints toward the definition of standardized protocols for microharvesting.
Collapse
Affiliation(s)
- Giuseppe Di Taranto
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Plastic and Reconstructive Surgery, Università Cattolica del Sacro Cuore University Hospital A. Gemelli Rome, Italy
| | - Claudia Cicione
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giuseppe Visconti
- Department of Plastic and Reconstructive Surgery, Università Cattolica del Sacro Cuore University Hospital A. Gemelli Rome, Italy
| | - Maria A Isgrò
- Institute of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore University Hospital A. Gemelli Rome, Italy
| | - Marta Barba
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Enrico Di Stasio
- Institute of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore University Hospital A. Gemelli Rome, Italy
| | - Egidio Stigliano
- Institute of Pathology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Camilla Bernardini
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Fabrizio Michetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy; Latium Musculoskeletal Tissue Bank, Rome, Italy
| | - Marzia Salgarello
- Department of Plastic and Reconstructive Surgery, Università Cattolica del Sacro Cuore University Hospital A. Gemelli Rome, Italy
| | - Wanda Lattanzi
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy; Latium Musculoskeletal Tissue Bank, Rome, Italy.
| |
Collapse
|
88
|
The role of adipose-derived stem cells in breast cancer progression and metastasis. Stem Cells Int 2015; 2015:120949. [PMID: 26000019 PMCID: PMC4427098 DOI: 10.1155/2015/120949] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 12/26/2014] [Indexed: 12/15/2022] Open
Abstract
Conventional breast cancer extirpation involves resection of parts of or the whole gland, resulting in asymmetry and disfiguration. Given the unsatisfactory aesthetic outcomes, patients often desire postmastectomy reconstructive procedures. Autologous fat grafting has been proposed for reconstructive purposes for decades to restore form and anatomy after mastectomy. Fat has the inherent advantage of being autologous tissue and the most natural-appearing filler, but given its inconsistent engraftment and retention rates, it lacks reliability. Implementation of autologous fat grafts with cellular adjuncts, such as multipotent adipose-derived stem cells (ADSCs), has shown promising results. However, it is pertinent and critical to question whether these cells could promote any residual tumor cells to proliferate, differentiate, or metastasize or even induce de novo carcinogenesis. Thus far, preclinical and clinical study findings are discordant. A trend towards potential promotion of both breast cancer growth and invasion by ADSCs found in basic science studies was indeed not confirmed in clinical trials. Whether experimental findings eventually correlate with or will be predictive of clinical outcomes remains unclear. Herein, we aimed to concisely review current experimental findings on the interaction of mesenchymal stem cells and breast cancer, mainly focusing on ADSCs as a promising tool for regenerative medicine, and discuss the implications in clinical translation.
Collapse
|
89
|
Minteer DM, Young MT, Lin YC, Over PJ, Rubin JP, Gerlach JC, Marra KG. Analysis of type II diabetes mellitus adipose-derived stem cells for tissue engineering applications. J Tissue Eng 2015; 6:2041731415579215. [PMID: 26090087 PMCID: PMC4456321 DOI: 10.1177/2041731415579215] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 02/26/2015] [Indexed: 11/28/2022] Open
Abstract
To address the functionality of diabetic adipose-derived stem cells in tissue engineering applications, adipose-derived stem cells isolated from patients with and without type II diabetes mellitus were cultured in bioreactor culture systems. The adipose-derived stem cells were differentiated into adipocytes and maintained as functional adipocytes. The bioreactor system utilizes a hollow fiber–based technology for three-dimensional perfusion of tissues in vitro, creating a model in which long-term culture of adipocytes is feasible, and providing a potential tool useful for drug discovery. Daily metabolic activity of the adipose-derived stem cells was analyzed within the medium recirculating throughout the bioreactor system. At experiment termination, tissues were extracted from bioreactors for immunohistological analyses in addition to gene and protein expression. Type II diabetic adipose-derived stem cells did not exhibit significantly different glucose consumption compared to adipose-derived stem cells from patients without type II diabetes (p > 0.05, N = 3). Expression of mature adipocyte genes was not significantly different between diabetic/non-diabetic groups (p > 0.05, N = 3). Protein expression of adipose tissue grown within all bioreactors was verified by Western blotting.The results from this small-scale study reveal adipose-derived stem cells from patients with type II diabetes when removed from diabetic environments behave metabolically similar to the same cells of non-diabetic patients when cultured in a three-dimensional perfusion bioreactor, suggesting that glucose transport across the adipocyte cell membrane, the hindrance of which being characteristic of type II diabetes, is dependent on environment. The presented observation describes a tissue-engineered tool for long-term cell culture and, following future adjustments to the culture environment and increased sample sizes, potentially for anti-diabetic drug testing.
Collapse
Affiliation(s)
| | - Matthew T Young
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yen-Chih Lin
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA ; Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patrick J Over
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - J Peter Rubin
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA ; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA ; Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jorg C Gerlach
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA ; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA ; Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kacey G Marra
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA ; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA ; Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
90
|
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
|
91
|
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
|
92
|
Choudhery MS, Badowski M, Muise A, Pierce J, Harris DT. Subcutaneous Adipose Tissue-Derived Stem Cell Utility Is Independent of Anatomical Harvest Site. Biores Open Access 2015; 4:131-145. [PMID: 26309790 PMCID: PMC4497709 DOI: 10.1089/biores.2014.0059] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
One of the challenges for tissue engineering and regenerative medicine is to obtain suitably large cell numbers for therapy. Mesenchymal stem cells (MSCs) can easily be expanded in vitro to obtain large numbers of cells, but this approach may induce cellular senescence. The characteristics of cells are dependent on variables like age, body mass index (BMI), and disease conditions, however, and in the case of adipose tissue-derived stem cells (ASCs), anatomical harvest site is also an important variable that can affect the regenerative potential of isolated cells. We therefore had kept the parameters (age, BMI, disease conditions) constant in this study to specifically assess influence of anatomical sites of individual donors on utility of ASCs. Adipose tissue was obtained from multiple anatomical sites in individual donors, and viability and nucleated cell yield were determined. MSC frequency was enumerated using colony forming unit assay and cells were characterized by flow cytometry. Growth characteristics were determined by long-term population doubling analysis of each sample. Finally, MSCs were induced to undergo adipogenic, osteogenic, and chondrogenic differentiation. To validate the findings, these results were compared with similar single harvest sites from multiple individual patients. The results of the current study indicated that MSCs obtained from multiple harvest sites in a single donor have similar morphology and phenotype. All adipose depots in a single donor exhibited similar MSC yield, viability, frequency, and growth characteristics. Equivalent differentiation capacity into osteocytes, adipocytes, and chondrocytes was also observed. On the basis of results, we conclude that it is acceptable to combine MSCs obtained from various anatomical locations in a single donor to obtain suitably large cell numbers required for therapy, avoiding in vitro senescence and lengthy and expensive in vitro culturing and expansion steps.
Collapse
Affiliation(s)
- Mahmood S. Choudhery
- Tissue Engineering and Regenerative Medicine Laboratory, Advance Research Center of Biomedical Sciences, King Edward Medical University, Lahore, Pakistan
- Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona
| | - Michael Badowski
- Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona
| | - Angela Muise
- Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona
| | - John Pierce
- Aesthetic Surgery of Tucson, Tucson, Arizona
| | - David T. Harris
- Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona
| |
Collapse
|
93
|
Chen C, Watkins-Curry P, Smoak M, Hogan K, Deese S, McCandless GT, Chan JY, Hayes DJ. Targeting Calcium Magnesium Silicates for Polycaprolactone/Ceramic Composite Scaffolds. ACS Biomater Sci Eng 2015. [DOI: 10.1021/ab500011x] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Cong Chen
- Department
of Biological Engineering, Louisiana State University and Louisiana State University Agricultural Center, Baton Rouge, Louisiana 70803, United States
| | - Pilanda Watkins-Curry
- Department
of Chemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, Texas 75080, United States
- Department
of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803, United States
| | - Mollie Smoak
- Department
of Biological Engineering, Louisiana State University and Louisiana State University Agricultural Center, Baton Rouge, Louisiana 70803, United States
| | - Katie Hogan
- Department
of Biological Engineering, Louisiana State University and Louisiana State University Agricultural Center, Baton Rouge, Louisiana 70803, United States
| | - Steve Deese
- Department
of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803, United States
| | - Gregory T. McCandless
- Department
of Chemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, Texas 75080, United States
| | - Julia Y. Chan
- Department
of Chemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, Texas 75080, United States
| | - Daniel J. Hayes
- Department
of Biological Engineering, Louisiana State University and Louisiana State University Agricultural Center, Baton Rouge, Louisiana 70803, United States
| |
Collapse
|
94
|
Kang SJ, Park YI, Kwon MJ, Yang YH, Bang SI, Sohn SH, Park YH, So B, Kang HG. Adipose Stromal Cells are a More Efficient Source than Adipose Stem Cells in Retrovirus-Mediated iPS Induction. Cell Mol Bioeng 2015. [DOI: 10.1007/s12195-014-0374-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
95
|
Lewallen EA, Riester SM, Bonin CA, Kremers HM, Dudakovic A, Kakar S, Cohen RC, Westendorf JJ, Lewallen DG, van Wijnen AJ. Biological strategies for improved osseointegration and osteoinduction of porous metal orthopedic implants. TISSUE ENGINEERING PART B-REVIEWS 2014; 21:218-30. [PMID: 25348836 DOI: 10.1089/ten.teb.2014.0333] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The biological interface between an orthopedic implant and the surrounding host tissue may have a dramatic effect upon clinical outcome. Desired effects include bony ingrowth (osseointegration), stimulation of osteogenesis (osteoinduction), increased vascularization, and improved mechanical stability. Implant loosening, fibrous encapsulation, corrosion, infection, and inflammation, as well as physical mismatch may have deleterious clinical effects. This is particularly true of implants used in the reconstruction of load-bearing synovial joints such as the knee, hip, and the shoulder. The surfaces of orthopedic implants have evolved from solid-smooth to roughened-coarse and most recently, to porous in an effort to create a three-dimensional architecture for bone apposition and osseointegration. Total joint surgeries are increasingly performed in younger individuals with a longer life expectancy, and therefore, the postimplantation lifespan of devices must increase commensurately. This review discusses advancements in biomaterials science and cell-based therapies that may further improve orthopedic success rates. We focus on material and biological properties of orthopedic implants fabricated from porous metal and highlight some relevant developments in stem-cell research. We posit that the ideal primary and revision orthopedic load-bearing metal implants are highly porous and may be chemically modified to induce stem cell growth and osteogenic differentiation, while minimizing inflammation and infection. We conclude that integration of new biological, chemical, and mechanical methods is likely to yield more effective strategies to control and modify the implant-bone interface and thereby improve long-term clinical outcomes.
Collapse
|
96
|
Tsuji W, Rubin JP, Marra KG. Adipose-derived stem cells: Implications in tissue regeneration. World J Stem Cells 2014; 6:312-321. [PMID: 25126381 PMCID: PMC4131273 DOI: 10.4252/wjsc.v6.i3.312] [Citation(s) in RCA: 265] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 05/16/2014] [Accepted: 06/11/2014] [Indexed: 02/06/2023] Open
Abstract
Adipose-derived stem cells (ASCs) are mesenchymal stem cells (MSCs) that are obtained from abundant adipose tissue, adherent on plastic culture flasks, can be expanded in vitro, and have the capacity to differentiate into multiple cell lineages. Unlike bone marrow-derived MSCs, ASCs can be obtained from abundant adipose tissue by a minimally invasive procedure, which results in a high number of cells. Therefore, ASCs are promising for regenerating tissues and organs damaged by injury and diseases. This article reviews the implications of ASCs in tissue regeneration.
Collapse
|
97
|
Estrogen as a Novel Agent for Induction of Adipose-Derived Mesenchymal Stem Cells for Osteogenic Differentiation. Plast Reconstr Surg 2014; 133:499e-510e. [DOI: 10.1097/prs.0000000000000056] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
98
|
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: 207] [Impact Index Per Article: 18.8] [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
|
99
|
Zanotti S, Kalajzic I, Aguila HL, Canalis E. Sex and genetic factors determine osteoblastic differentiation potential of murine bone marrow stromal cells. PLoS One 2014; 9:e86757. [PMID: 24489784 PMCID: PMC3904935 DOI: 10.1371/journal.pone.0086757] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 12/16/2013] [Indexed: 01/08/2023] Open
Abstract
Sex and genetic factors determine skeletal mass, and we tested whether bone histomorphometric parameters were sexually dimorphic in femurs from 1 to 6 month old C57BL/6 mice. Trabecular bone volume declined more rapidly in female mice than in male littermates because of enhanced bone resorption. Although bone formation was not different between sexes, female mice exhibited a higher number of osteoblasts than male littermates, suggesting that osteoblasts from female mice may have a reduced ability to form bone. To determine the impact of sex on osteoblastogenesis, we investigated the potential for osteoblastic differentiation of bone marrow stromal cells from C57BL/6, Friend leukemia virus-B (FVB), C3H/HeJ and BALB/c mice of both sexes. Bone marrow stromal cells from female FVB, C57BL/6 and C3H/HeJ mice exhibited lower Alpl and Osteocalcin expression and alkaline phosphatase activity, and formed fewer mineralized nodules than cells from male littermates. Proliferative capacity was greater in cells from male than female C57BL/6, but not FVB, mice. Sorting of bone marrow stromal cells from mice expressing an α-Smooth muscle actin-green fluorescent protein transgene, revealed a higher yield of mesenchymal stem cells in cultures from male mice than in those from female littermates. Sex had a modest impact on osteoblastic differentiation of mesenchymal stem cells. To determine the influence of sex and genetic factors on osteoblast function, calvarial osteoblasts were harvested from C57BL/6, FVB, C3H/HeJ and BALB/c mice. Alpl expression and activity were lower in osteoblasts from C57BL/6 and C3H/HeJ, but not FVB or BALB/c, female mice than in cells from littermates. Sex had no effect on osteoclastogenesis of bone marrow cultures of C57BL/6 mice, but osteoblasts from female mice exhibited higher Rankl and lower Opg expression than cells from male littermates. In conclusion, osteoblastogenesis is sexually dimorphic and influenced by genetic factors.
Collapse
Affiliation(s)
- Stefano Zanotti
- Department of Research, Saint Francis Hospital and Medical Center, Hartford, Connecticut, United States of America
- University of Connecticut School of Medicine, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- * E-mail:
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Hector Leonardo Aguila
- Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Ernesto Canalis
- Department of Research, Saint Francis Hospital and Medical Center, Hartford, Connecticut, United States of America
- University of Connecticut School of Medicine, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| |
Collapse
|
100
|
Russo V, Yu C, Belliveau P, Hamilton A, Flynn LE. Comparison of human adipose-derived stem cells isolated from subcutaneous, omental, and intrathoracic adipose tissue depots for regenerative applications. Stem Cells Transl Med 2013; 3:206-17. [PMID: 24361924 DOI: 10.5966/sctm.2013-0125] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Adipose tissue is an abundant source of multipotent progenitor cells that have shown promise in regenerative medicine. In humans, fat is primarily distributed in the subcutaneous and visceral depots, which have varying biochemical and functional properties. In most studies to date, subcutaneous adipose tissue has been investigated as the adipose-derived stem cell (ASC) source. In this study, we sought to develop a broader understanding of the influence of specific adipose tissue depots on the isolated ASC populations through a systematic comparison of donor-matched abdominal subcutaneous fat and omentum, and donor-matched pericardial adipose tissue and thymic remnant samples. We found depot-dependent and donor-dependent variability in the yield, viability, immunophenotype, clonogenic potential, doubling time, and adipogenic and osteogenic differentiation capacities of the ASC populations. More specifically, ASCs isolated from both intrathoracic depots had a longer average doubling time and a significantly higher proportion of CD34(+) cells at passage 2, as compared with cells isolated from subcutaneous fat or the omentum. Furthermore, ASCs from subcutaneous and pericardial adipose tissue demonstrated enhanced adipogenic differentiation capacity, whereas ASCs isolated from the omentum displayed the highest levels of osteogenic markers in culture. Through cell culture analysis under hypoxic (5% O(2)) conditions, oxygen tension was shown to be a key mediator of colony-forming unit-fibroblast number and osteogenesis for all depots. Overall, our results suggest that depot selection is an important factor to consider when applying ASCs in tissue-specific cell-based regenerative therapies, and also highlight pericardial adipose tissue as a potential new ASC source.
Collapse
Affiliation(s)
- Valerio Russo
- Departments of Chemical Engineering and Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada; Human Mobility Research Centre and Department of Surgery, Kingston General Hospital, Kingston, Ontario, Canada
| | | | | | | | | |
Collapse
|