1
|
Jafrani A, Mehmood H, Shaikh MS, Lone MA, Fareed MA, Zafar MS. A narrative review on the role of adipose-derived stem cells in periodontal regeneration. J Dent 2025; 158:105801. [PMID: 40324579 DOI: 10.1016/j.jdent.2025.105801] [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] [Received: 03/07/2025] [Revised: 04/05/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025] Open
Abstract
OBJECTIVES Recent research suggests that periodontal regeneration (PR) through the application of adipose-derived stem cells (ADSCs) has depicted some encouraging outcomes. They have the benefit of convenient access and are harvested through minimally invasive and inexpensive procedures. This review aimed to explore the role of ADSCs in PR. DATA AND SOURCES The recommendation from the Scale for the Assessment of Narrative Review Articles (SANRA) was utilised to conduct this review. It was carried out by extensively searching the literature for evidence on PR using ADSCs by electronic search through PubMed and Google Scholar databases. STUDY SELECTION The search terms included "adipose-derived stem cells" OR "ADSCs" AND "periodontal regeneration" OR "alveolar bone regeneration". A literature review was established to include articles related to the use of ADSCs in PR, after refining the inclusion criteria. CONCLUSION The relevant studies demonstrated that ADSCs have a significant regenerative and therapeutic potential in periodontal tissues. Specifically, ADSCs were shown to differentiate into periodontal structures. However, the data is scarce for assessing their effectiveness in human models. Consequently, further research is required, including randomised controlled trials on human and animal models to further verify their regenerative capability in periodontal restoration. CLINICAL SIGNIFICANCE Adipose-derived stem cells exhibited their capability to differentiate into key periodontal structures and enhance tissue repair indicating their potential as a novel therapeutic method for improving clinical results in periodontal treatment.
Collapse
Affiliation(s)
- Anosha Jafrani
- Sindh Institute of Oral Health Sciences, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Huda Mehmood
- Sindh Institute of Oral Health Sciences, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Muhammad Saad Shaikh
- Department of Oral Biology, Sindh Institute of Oral Health Sciences, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Mohid Abrar Lone
- Department of Oral Pathology, Sindh Institute of Oral Health Sciences, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Muhammad Amber Fareed
- Department of Clinical Sciences, College of Dentistry, Ajman University, Ajman, UAE; Center of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, UAE
| | - Muhammad Sohail Zafar
- Department of Clinical Sciences, College of Dentistry, Ajman University, Ajman, UAE; Center of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, UAE; School of Dentistry, University of Jordan, Amman 11942, Jordan.
| |
Collapse
|
2
|
Soltanmohammadi F, Mahmoudi Gharehbaba A, Alizadeh E, Javadzadeh Y. Innovative approaches to tissue engineering: Utilizing decellularized extracellular matrix hydrogels for mesenchymal stem cell transport. Int J Biol Macromol 2025; 290:138893. [PMID: 39706433 DOI: 10.1016/j.ijbiomac.2024.138893] [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] [Received: 09/24/2024] [Revised: 12/07/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
In recent years, the realm of tissue regeneration experienced significant advancements, leading to the development of innovative therapeutic agents. The systemic delivery of mesenchymal stem cells (MSCs) emerged as a promising strategy for promoting tissue regeneration. However, this approach is hindered by hurdles such as poor cell survival, limited cell propagation, and inadequate cell integration. Decellularized extracellular matrix (dECM) hydrogel serves as an innovative carrier that protects MSCs from the detrimental effects of the hostile microenvironment, facilitates their localization and retention at the injection site, and preserves their viability. Regarding its low immunogenicity, low cytotoxicity, high biocompatibility, and its ability to mimic natural extracellular matrix (ECM), this natural hydrogel offers a new avenue for systemic delivery of MSCs. This review digs into the properties of dECM hydrogels (dECMHs), the methods employed for decellularization and the utilization of dECMH as carriers for various types of MSCs for tissue regeneration purposes. This review also sheds light on the benefits of hybrid hydrogels composed of dECMH and other components such as proteins and polysaccharides. By addressing the limitations of conventional hydrogels and enhancing efficacy of cell therapy, dECMH opens new pathways for the future of tissue regeneration.
Collapse
Affiliation(s)
- Fatemeh Soltanmohammadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Adel Mahmoudi Gharehbaba
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Effat Alizadeh
- Endocrin Research Center and Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Yousef Javadzadeh
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
Tseng SL, Kang L, Li ZJ, Wang LQ, Li ZM, Li TH, Xiang JY, Huang JZ, Yu NZ, Long X. Adipose-derived stem cells in diabetic foot care: Bridging clinical trials and practical application. World J Diabetes 2024; 15:1162-1177. [PMID: 38983804 PMCID: PMC11229965 DOI: 10.4239/wjd.v15.i6.1162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/03/2024] [Accepted: 04/19/2024] [Indexed: 06/11/2024] Open
Abstract
Diabetic foot ulcers (DFUs) pose a critical medical challenge, significantly im-pairing the quality of life of patients. Adipose-derived stem cells (ADSCs) have been identified as a promising therapeutic approach for improving wound healing in DFUs. Despite extensive exploration of the mechanical aspects of ADSC therapy against DFU, its clinical applications remain elusive. In this review, we aimed to bridge this gap by evaluating the use and advancements of ADSCs in the clinical management of DFUs. The review begins with a discussion of the classification and clinical management of diabetic foot conditions. It then discusses the current landscape of clinical trials, focusing on their geographic distribution, reported efficacy, safety profiles, treatment timing, administration techniques, and dosing considerations. Finally, the review discusses the preclinical strategies to enhance ADSC efficacy. This review shows that many trials exhibit biases in study design, unclear inclusion criteria, and intervention protocols. In conclusion, this review underscores the potential of ADSCs in DFU treatment and emphasizes the critical need for further research and refinement of therapeutic approaches, with a focus on improving the quality of future clinical trials to enhance treatment outcomes and advance the field of diabetic wound care.
Collapse
Affiliation(s)
- Song-Lu Tseng
- Department of Plastic and Reconstructive Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Lin Kang
- Biomedical Engineering Facility, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences Beijing, Beijing 100021, China
| | - Zhu-Jun Li
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Li-Quan Wang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Zi-Ming Li
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Tian-Hao Li
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jie-Yu Xiang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jiu-Zuo Huang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Nan-Ze Yu
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xiao Long
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
4
|
Park JS, Kim DY, Hong HS. FGF2/HGF priming facilitates adipose-derived stem cell-mediated bone formation in osteoporotic defects. Heliyon 2024; 10:e24554. [PMID: 38304814 PMCID: PMC10831751 DOI: 10.1016/j.heliyon.2024.e24554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/14/2023] [Accepted: 01/10/2024] [Indexed: 02/03/2024] Open
Abstract
Aims The activity of adipose-derived stem cells (ADSCs) is susceptible to the physiological conditions of the donor. Therefore, employing ADSCs from donors of advanced age or with diseases for cell therapy necessitates a strategy to enhance therapeutic efficacy before transplantation. This study aims to investigate the impact of supplementing Fibroblast Growth Factor 2 (FGF2) and Hepatocyte Growth Factor (HGF) on ADSC-mediated osteogenesis under osteoporotic conditions and to explore the underlying mechanisms of action. Main methods Adipose-derived stem cells (ADSCs) obtained from ovariectomized (OVX) rats were cultured ex vivo. These cells were cultured in an osteogenic medium supplemented with FGF2 and HGF and subsequently autologously transplanted into osteoporotic femur defects using Hydroxyapatite-Tricalcium Phosphate. The assessment of bone formation was conducted four weeks post-transplantation. Key findings Osteoporosis detrimentally affects the viability and osteogenic differentiation potential of ADSCs, often accompanied by a deficiency in FGF2 and HGF signaling. However, priming with FGF2 and HGF facilitated the formation of immature osteoblasts from OVX ADSCs in vitro, promoting the expression of osteoblastogenic proteins, including Runx-2, osterix, and ALP, during the early phase of osteogenesis. Furthermore, FGF2/HGF priming augmented the levels of VEGF and SDF-1α in the microenvironment of OVX ADSCs under osteogenic induction. Importantly, transplantation of OVX ADSCs primed with FGF2/HGF for 6 days significantly enhanced bone formation compared to non-primed cells. The success of bone regeneration was confirmed by the expression of type-1 collagen and osteocalcin in the bone tissue of the deficient area. Significance Our findings corroborate that priming with FGF2/HGF can improve the differentiation potential of ADSCs. This could be applied in autologous stem cell therapy for skeletal disease in the geriatric population.
Collapse
Affiliation(s)
- Jeong Seop Park
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, 02447, South Korea
| | - Do Young Kim
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, 02447, South Korea
| | - Hyun Sook Hong
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, 02447, South Korea
- East-West Medical Research Institute, Kyung Hee University, Seoul, 02447, South Korea
- Kyung Hee Institute of Regenerative Medicine (KIRM), Medical Science Research Institute, Kyung Hee University Medical Center, Seoul, 02447, South Korea
| |
Collapse
|
5
|
Chen CF, Chen YC, Fu YS, Tsai SW, Wu PK, Chen CM, Chen WM, Wu HTH, Lee CH, Chang CL, Lin PC, Kao YC, Chen CH, Chuang MH. Safety and Tolerability of Intra-Articular Injection of Adipose-Derived Mesenchymal Stem Cells GXCPC1 in 11 Subjects With Knee Osteoarthritis: A Nonrandomized Pilot Study Without a Control Arm. Cell Transplant 2024; 33:9636897231221882. [PMID: 38205679 PMCID: PMC10785714 DOI: 10.1177/09636897231221882] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 11/30/2023] [Accepted: 12/06/2023] [Indexed: 01/12/2024] Open
Abstract
The current study aimed to determine the safety profile of intra-articular-injected allogeneic adipose-derived mesenchymal stem cells (ADSCs) GXCPC1 in subjects with knee osteoarthritis (OA) and its preliminary efficacy outcome. The 3 + 3 phase I study was designed with two dose-escalation cohorts: low dose (6.7 × 106 GXCPC1, N = 5) and high dose (4 × 107 GXCPC1, N = 6). The primary endpoint was safety, which was evaluated by recording adverse events throughout the trial; the secondary endpoints included total, pain, stiffness, and function subscales of the Western Ontario and McMaster Universities Arthritis Index (WOMAC), Visual Analogue Scale (VAS) for pain, and 12-Item Short Form (SF-12) health survey questionnaire. The GXCPC1 treatment was found to be safe after 1 year of follow-up with no treatment-related severe adverse events observed. When compared to baseline, subjects in both the low- and high-dose cohorts demonstrated improving trends in pain and knee function after receiving GXCPC1 treatment. Generally, the net change in pain (95% confidence interval (CI) = -7.773 to -2.561t at 12 weeks compared to baseline) and knee function (95% CI = -24.297 to -10.036t at 12 weeks compared to baseline) was better in subjects receiving high-dose GXCPC1. Although this study included a limited number of subjects without a placebo arm, it showed that the intra-articular injection of ADSCs was safe and well-tolerated in subjects with therapeutic alternatives to treat knee OA. However, a larger scale study with an appropriate control would be necessary for clinical efficacy in the following study.
Collapse
Affiliation(s)
- Cheng-Fong Chen
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Therapeutical and Research Center of Musculoskeletal Tumor, Department of Orthopaedics, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yi-Chung Chen
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Therapeutical and Research Center of Musculoskeletal Tumor, Department of Orthopaedics, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yu-Show Fu
- Department of Anatomy and Cell Biology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Shang-Wen Tsai
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Po-Kuei Wu
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Therapeutical and Research Center of Musculoskeletal Tumor, Department of Orthopaedics, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Chao-Ming Chen
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Therapeutical and Research Center of Musculoskeletal Tumor, Department of Orthopaedics, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Wei-Ming Chen
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Therapeutical and Research Center of Musculoskeletal Tumor, Department of Orthopaedics, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Hung-Ta Hondar Wu
- Department of Radiology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Radiology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Chia-Hsin Lee
- Gwo Xi Stem Cell Applied Technology Co., Ltd., Hsinchu, Taiwan, ROC
| | - Chao-Liang Chang
- Gwo Xi Stem Cell Applied Technology Co., Ltd., Hsinchu, Taiwan, ROC
| | - Po-Cheng Lin
- Gwo Xi Stem Cell Applied Technology Co., Ltd., Hsinchu, Taiwan, ROC
| | - Yong-Cheng Kao
- Gwo Xi Stem Cell Applied Technology Co., Ltd., Hsinchu, Taiwan, ROC
| | - Chun-Hung Chen
- Gwo Xi Stem Cell Applied Technology Co., Ltd., Hsinchu, Taiwan, ROC
| | - Ming-Hsi Chuang
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- College of Management, Chung Hua University, Hsinchu, Taiwan, ROC
| |
Collapse
|
6
|
Janarthanan R, Jayakumar R, Iyer S. Injectable Pectin-Alginate Hydrogels for Improving Vascularization and Adipogenesis of Human Fat Graft. J Funct Biomater 2023; 14:409. [PMID: 37623654 PMCID: PMC10455938 DOI: 10.3390/jfb14080409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/26/2023] Open
Abstract
Autologous fat grafting (AFG) is the most prevailing tool for soft tissue regeneration in clinics, although efficiency is limited to unpredictable volume resorption due to poor vascularization and eventual necrosis. This study sought to improve the AFG efficiency using a hydrogel as a carrier for human fat graft (F) with and without platelet-rich plasma (PRP). PRP is clinically well known for the local release of several endogenous growth factors and has been in clinical use already. A human-fat-graft-encapsulated pectin-alginate hydrogel (FG) was developed and characterized. PRP was added to F to develop a human fat graft with PRP (FP). FP was admixed with a pectin-alginate hydrogel to develop FGP. FG and FGP showed the smooth injectable, elastic, and shear-thinning properties. FG and FGP groups showed enhanced cell viability and proliferation compared to the control F in vitro. We also investigated the in vivo angiogenesis and neo-adipogenesis ability of F, FG, FGP, and FP in nude mice after subcutaneous injection. After 2 and 4 weeks, an MRI of the mice was conducted, followed by graft explantation. The explanted grafts were also assessed histologically and with immunohistochemistry (IHC) studies. MRI and histology results revealed better vascularity of the FG and FGP system compared to fat graft alone. Further, the IHC studies, CD 31, and perilipin staining also revealed better vasculature and adipogenesis of FG and FGP systems. These results indicate the enhanced angiogenesis and adipogenesis of FG and FGP. Thus, developed pectin-alginate hydrogel-based fat graft systems FG and FGP replenish the native microenvironment by mediating angiogenesis and adipogenesis, thereby maximizing the clinical outcomes of autologous fat grafting.
Collapse
Affiliation(s)
- Ramu Janarthanan
- Department of Plastic and Reconstructive Surgery, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi 682041, India;
| | - Rangasamy Jayakumar
- Polymeric Biomaterials Lab, School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682041, India;
| | - Subramania Iyer
- Department of Plastic and Reconstructive Surgery, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi 682041, India;
| |
Collapse
|
7
|
Zhang Y, Lv P, Li Y, Zhang Y, Cheng C, Hao H, Yue H. Inflammatory Cytokine Interleukin-6 (IL-6) Promotes the Proangiogenic Ability of Adipose Stem Cells from Obese Subjects via the IL-6 Signaling Pathway. Curr Stem Cell Res Ther 2023; 18:93-104. [PMID: 36883256 DOI: 10.2174/1574888x17666220429103935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 01/05/2022] [Accepted: 03/01/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND The prevalence of obesity, as well as obesity-induced chronic inflammatory diseases, is increasing worldwide. Chronic inflammation is related to the complex process of angiogenesis, and we found that adipose-derived stem cells from obese subjects (obADSCs) had proangiogenic features, including higher expression levels of interleukin-6 (IL-6), Notch ligands and receptors, and proangiogenic cytokines, than those from control subjects. We hypothesized that IL-6 and Notch signaling pathways are essential for regulating the proangiogenic characteristics of obADSCs. OBJECTIVE This study aimed to investigate whether the inflammatory cytokine interleukin 6 (IL-6) promotes the proangiogenic capacity of adipose stem cells in obese subjects via the IL-6 signaling pathway. METHODS We compared the phenotype analysis as well as cell doubling time, proliferation, migration, differentiation, and proangiogenic properties of ADSCs in vitro. Moreover, we used small interfering RNAs to inhibit the gene and protein expression of IL-6. RESULTS We found that ADSCs isolated from control individuals (chADSCs) and obADSCs had similar phenotypes and growth characteristics, and chADSCs had a stronger differentiation ability than obADSCs. However, obADSCs were more potent in promoting EA.hy926 cell migration and tube formation than chADSCs in vitro. We confirmed that IL-6 siRNA significantly reduced the transcriptional level of IL-6 in obADSCs, thereby reducing the expression of vascular endothelial growth factor (VEGF)- A, VEGF receptor 2, transforming growth factor β, and Notch ligands and receptors in obADSCs. CONCLUSION The finding suggests that inflammatory cytokine interleukin-6 (IL-6) promotes the proangiogenic ability of obADSCs via the IL-6 signaling pathway.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Translational Medicine Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, 450007, China
| | - Pengju Lv
- Translational Medicine Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, 450007, China
| | - Yalong Li
- Stem Cell Research Center, Henan Key Laboratory of Stem Cell Differentiation and Modification Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China.,People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| | - Yonghui Zhang
- Stem Cell Research Center, Henan Key Laboratory of Stem Cell Differentiation and Modification Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China.,People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| | - Chaofei Cheng
- Stem Cell Research Center, Henan Key Laboratory of Stem Cell Differentiation and Modification Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China.,People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| | - Hongbo Hao
- Neuroscience Initiative, Advanced Science Research Center at the Graduate Center, City University of New York, New York, 10031, USA
| | - Han Yue
- Stem Cell Research Center, Henan Key Laboratory of Stem Cell Differentiation and Modification Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China.,People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| |
Collapse
|
8
|
Park JS, Kim D, Hong HS. Priming with a Combination of FGF2 and HGF Restores the Impaired Osteogenic Differentiation of Adipose-Derived Stem Cells. Cells 2022; 11:cells11132042. [PMID: 35805126 PMCID: PMC9265418 DOI: 10.3390/cells11132042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/20/2022] [Accepted: 06/26/2022] [Indexed: 02/05/2023] Open
Abstract
Classical aging-associated diseases include osteoporosis, diabetes, hypertension, and arthritis. Osteoporosis causes the bone to become brittle, increasing fracture risk. Among the various treatments for fractures, stem cell transplantation is currently in the spotlight. Poor paracrine/differentiation capacity, owing to donor age or clinical history, limits efficacy. Lower levels of fibroblast growth factor 2 (FGF2) and hepatocyte growth factor (HGF) are involved in cell repopulation, angiogenesis, and bone formation in the elderly ADSCs (ADSC-E) than in the young ADSCs (ADSC-Y). Here, we study the effect of FGF2/HGF priming on the osteogenic potential of ADSC-E, determined by calcium deposition in vitro and ectopic bone formation in vivo. Age-induced FGF2/HGF deficiency was confirmed in ADSCs, and their supplementation enhanced the osteogenic differentiation ability of ADSC-E. Priming with FGF2/HGF caused an early shift of expression of osteogenic markers, including Runt-related transcription factor 2 (Runx-2), osterix, and alkaline phosphatase (ALP) during osteogenic differentiation. FGF2/HGF priming also created an environment favorable to osteogenesis by facilitating the secretion of bone morphogenetic protein 2 (BMP-2) and vascular endothelial growth factor (VEGF). Bone tissue of ADSC-E origin was observed in mice transplanted with FGF/HGF-primed ADSC-E. Collectively, FGF2/HGF priming could enhance the bone-forming capacity in ADSC-E. Therefore, growth factor-mediated cellular priming can enhance ADSC differentiation in bone diseases and thus contributes to the increased efficacy in vivo.
Collapse
Affiliation(s)
- Jeong Seop Park
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.S.P.); (D.K.)
| | - Doyoung Kim
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.S.P.); (D.K.)
| | - Hyun Sook Hong
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.S.P.); (D.K.)
- East-West Medical Research Institute, Kyung Hee University, Seoul 02447, Korea
- Kyung Hee Institute of Regenerative Medicine (KIRM), Medical Science Research Institute, Kyung Hee University Medical Center, Seoul 02447, Korea
- Correspondence: ; Tel.: +82-2-958-1828
| |
Collapse
|
9
|
Sok D, Raval S, McKinney J, Drissi H, Mason A, Mautner K, Kaiser JM, Willett NJ. NSAIDs Reduce Therapeutic Efficacy of Mesenchymal Stromal Cell Therapy in a Rodent Model of Posttraumatic Osteoarthritis. Am J Sports Med 2022; 50:1389-1398. [PMID: 35420503 DOI: 10.1177/03635465221083610] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Intra-articular injections of human mesenchymal stromal cells (hMSCs) have shown promise in slowing cartilage degradation in posttraumatic osteoarthritis (PTOA). Clinical use of cell therapies for osteoarthritis has accelerated in recent years without sufficient scientific evidence defining best-use practices. Common recommendations advise patients to avoid nonsteroidal anti-inflammatory drug (NSAID) use before and after cell injection over concerns that NSAIDs may affect therapeutic efficacy. Recommendations to restrict NSAID use are challenging for patients, and it is unclear if patients are compliant. HYPOTHESIS NSAIDs will reduce the efficacy of hMSC therapy in treating a preclinical model of PTOA. STUDY DESIGN Controlled laboratory study. METHODS Lewis rats underwent medial meniscal transection (MMT) surgery to induce PTOA or a sham (sham group) surgery that did not progress to PTOA. Rats received naproxen solution orally daily before (Pre-NSAID group) or after (Post-NSAID group) hMSC treatment, throughout the course of the experiment (Full-NSAID group), or received hMSCs without NSAIDs (No NSAID). Cartilage morphology and composition were quantified using contrast-enhanced micro-computed tomography and histology. Pain (secondary allodynia) was measured using a von Frey filament. RESULTS Injection of hMSCs attenuated cartilage degeneration associated with MMT. hMSCs prevented proteoglycan loss, maintained smooth cartilage surfaces, reduced cartilage lesions, reduced mineralized osteophyte formation, and reduced pain by week 7. The Pre-NSAID group had decreased proteoglycan levels compared with the hMSC group, although there were no other significant differences. Thus, pretreatment with NSAIDs had minimal effects on the therapeutic benefits of hMSC injections. The Post-NSAID and Full-NSAID groups, however, exhibited significantly worse osteoarthritis than the hMSC-only group, with greater proteoglycan loss, surface roughness, osteophyte volume, and pain. CONCLUSION Use of NSAIDs before hMSC injection minimally reduced the therapeutic benefits for PTOA, which included preservation of cartilage surface integrity as well as a reduction in osteophytes. Use of NSAIDs after injections, however, substantially reduced the therapeutic efficacy of cellular treatment. CLINICAL RELEVANCE Our data support the clinical recommendation of avoiding NSAID use after hMSC injection but suggest that using NSAIDs before treatment may not substantially diminish the therapeutic efficacy of cell treatment.
Collapse
Affiliation(s)
- Daniel Sok
- Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sarvgna Raval
- Emory University School of Medicine, Atlanta, Georgia, USA.,Atlanta Veterans Affairs Hospital, Atlanta, Georgia, USA
| | - Jay McKinney
- Emory University School of Medicine, Atlanta, Georgia, USA.,Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Hicham Drissi
- Emory University School of Medicine, Atlanta, Georgia, USA.,Atlanta Veterans Affairs Hospital, Atlanta, Georgia, USA
| | - Amadeus Mason
- Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ken Mautner
- Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jarred M Kaiser
- Emory University School of Medicine, Atlanta, Georgia, USA.,Atlanta Veterans Affairs Hospital, Atlanta, Georgia, USA
| | - Nick J Willett
- Emory University School of Medicine, Atlanta, Georgia, USA.,Georgia Institute of Technology, Atlanta, Georgia, USA.,Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, USA
| |
Collapse
|
10
|
Chiu TL, Baskaran R, Tsai ST, Huang CY, Chuang MH, Syu WS, Harn HJ, Lin YC, Chen CH, Huang PC, Wang YF, Chuang CH, Lin PC, Lin SZ. Intracerebral transplantation of autologous adipose-derived stem cells for chronic ischemic stroke: A phase I study. J Tissue Eng Regen Med 2021; 16:3-13. [PMID: 34644444 DOI: 10.1002/term.3256] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 09/14/2021] [Accepted: 10/09/2021] [Indexed: 11/08/2022]
Abstract
Current therapy does not provide significant benefits for patients with chronic stroke. Pre-clinical studies suggested that autologous adipose-derived stem cells have benefits for the treatment of chronic stroke. This Phase I open-label study was conducted to demonstrate the safety and efficacy of autologous adipose-derived stem cells (GXNPC1) in chronic stroke. Three patients with chronic stroke were treated with stereotactic implantation of autologous adipose-derived stem cells (1 × 108 cells). The primary endpoints of safety evaluation included adverse events, over a 6 months post-implantation period. The secondary endpoints included improvements in neurological functions. Evolutional change of brain parenchyma was also followed with magnetic resonance imaging (MRI). All three participants improved significantly at 6 months follow-up. The extent of improvement from pre-treatment was: National Institutes of Health Stroke Scale improved 5-15 points, Barthel Index: 25-50 points, Berg balance scale 0-21 points and Fugl-Meyer modified sensation 3-28 points. All three patients had signal change along the implantation tract on MRI one month after surgery. There is no related safety issue through 6 months observation. Clinical measures of neurological symptoms of these patients with chronic stroke improved at 6 months without adverse effects after implantation of autologous adipose-derived stem cells (GXNPC1), which might be correlated with post-implantation changes on brain MRI. Clinical Trial Registration-URL: https://clinicaltrials.gov/ct2/show/NCT02813512?term=ADSC&cond=Stroke&cntry=TW&draw=2&rank=1 Unique identifier: NCT02813512.
Collapse
Affiliation(s)
- Tsung-Lang Chiu
- Department of Neurosurgery, Bioinnovation Center, Tzu Chi Foundation, Buddhist Tzu Chi General Hospital, Tzu Chi University, Hualien, Taiwan, ROC
| | - Rathinasamy Baskaran
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan, ROC
| | - Sheng-Tzung Tsai
- Department of Neurosurgery, Bioinnovation Center, Tzu Chi Foundation, Buddhist Tzu Chi General Hospital, Tzu Chi University, Hualien, Taiwan, ROC
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, ROC.,Department of Biological Science and Technology, Asia University, Taichung, Taiwan, ROC.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan, ROC
| | - Ming-Hsi Chuang
- Department of Technology Management, Chung Hwa University, Hsinchu, Taiwan, ROC
| | - Wan-Sin Syu
- Department of Stem Cell Applied Technology, Gwo Xi Stem Cell Applied Technology, Hsinchu, Taiwan, ROC
| | - Horng-Jyh Harn
- Bioinnovation Center, Tzu Chi foundation; Department of Pathology, Buddhist Tzu Chi General Hospital, Tzu Chi University, Hualien, Taiwan, ROC
| | - Yi-Chun Lin
- Department of Stem Cell Applied Technology, Gwo Xi Stem Cell Applied Technology, Hsinchu, Taiwan, ROC
| | - Chun-Hung Chen
- Department of Stem Cell Applied Technology, Gwo Xi Stem Cell Applied Technology, Hsinchu, Taiwan, ROC
| | - Pi-Chun Huang
- Department of Stem Cell Applied Technology, Gwo Xi Stem Cell Applied Technology, Hsinchu, Taiwan, ROC
| | - Yi-Fen Wang
- Department of Neurosurgery, Bioinnovation Center, Tzu Chi Foundation, Buddhist Tzu Chi General Hospital, Tzu Chi University, Hualien, Taiwan, ROC
| | | | - Po-Cheng Lin
- Department of Stem Cell Applied Technology, Gwo Xi Stem Cell Applied Technology, Hsinchu, Taiwan, ROC
| | - Shinn-Zong Lin
- Department of Neurosurgery, Bioinnovation Center, Tzu Chi Foundation, Buddhist Tzu Chi General Hospital, Tzu Chi University, Hualien, Taiwan, ROC
| |
Collapse
|
11
|
Platoff R, Villalobos MA, Hagaman AR, Liu Y, Matthews M, DiSanto ME, Carpenter JP, Zhang P. Effects of radiation and chemotherapy on adipose stem cells: Implications for use in fat grafting in cancer patients. World J Stem Cells 2021; 13:1084-1093. [PMID: 34567427 PMCID: PMC8422936 DOI: 10.4252/wjsc.v13.i8.1084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/30/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023] Open
Abstract
Autologous fat transplantation is a versatile tool in reconstructive surgery. Adipose-derived stem cells (ASCs) increase survival of fat grafts and thus are increasingly used for breast reconstruction in breast cancer patients. However, radiation and/or chemotherapy have been proposed to inhibit soft tissue regeneration in wound healing thus suggesting alteration in stem cell pathways. Therefore, elucidating effects of radiation and chemotherapy on ASCs is critical if one desires to enhance the survival of fat grafts in patients. This review outlines our work evaluating the function and recoverability of ASCs from radiation or chemotherapy patients, focusing specifically on their availability as a source of autologous stem cells for fat grafting and breast reconstruction in cancer patients. Even though evidence suggests radiation and chemotherapy negatively influence ASCs at the cellular level, the efficiency of the isolation and differentiation capacity did not appear influenced in patients after receiving chemotherapy treatment, although fat from radiated patients exhibited significantly altered ASC differentiation into endothelial-like cells. Further, the in vitro growth rates of patient’s ASCs do not differ significantly before or after treatment. Taken together, these studies suggest ASCs as an important new tool for grafting and reconstruction even when radiation and chemotherapy treatment are involved.
Collapse
Affiliation(s)
- Rebecca Platoff
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, United States
| | - Miguel A Villalobos
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, United States
| | - Ashleigh Rapp Hagaman
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, United States
| | - Yuan Liu
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, United States
- Department of Surgery, Cooper Medical School of Rowan University, Camden, NJ 08103, United States
| | - Martha Matthews
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, United States
- Department of Surgery, Cooper Medical School of Rowan University, Camden, NJ 08103, United States
| | - Michael E DiSanto
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, United States
| | - Jeffrey P Carpenter
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, United States
- Department of Surgery, Cooper Medical School of Rowan University, Camden, NJ 08103, United States
| | - Ping Zhang
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, United States
- Department of Surgery, Cooper Medical School of Rowan University, Camden, NJ 08103, United States
| |
Collapse
|
12
|
Wang MY, Wang YX, Li-Ling J, Xie HQ. Adult Stem Cell Therapy for Premature Ovarian Failure: From Bench to Bedside. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:63-78. [PMID: 33427039 DOI: 10.1089/ten.teb.2020.0205] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Premature ovarian failure (POF) is a devastating condition for women of childbearing age with serious health consequences, including distress, infertility, osteoporosis, autoimmune disorders, ischemic heart disease, and increased mortality. In addition to the mainstay estrogen therapy, stem cell therapy has been tested as the result of rapid progress in cell biology and reprogramming research. We hereby provide a review for the latest research and issues related with stem cell-based therapy for POF, and provide a commentary on various methods for enhancing its effect. Large amount of animal studies have demonstrated an extensive benefit of stem cells for failed ovarian recovering. As shown by such studies, stem cell therapy can result in recovery of hormonal levels, follicular activation, ovarian angiogenesis, and functional restoration. Meanwhile, a study of molecular pathways revealed that the function of stem cells mainly depends on their paracrine actions, which can produce multiple factors for the promotion of ovarian angiogenesis and regulation of cellular functions. Nevertheless, studies using disease models also revealed certain drawbacks. Clinical trials have shown that menstrual cycle and even pregnancy may occur in POF patients following transplantation of stem cells, although the limitations, including inadequate number of cases and space for the improvement of transplantation methodology. Only with its safety and effect get substantial improvement through laboratory experiments and clinical trials, can stem cell therapy really bring benefits to more patients. Additionally, effective pretreatment and appropriate transplantation methods for stem cells are also required. Taken together, stem cell therapy has shown a great potential for the reversal of POF and is stepping from bench to bedside. Impact statement Premature ovarian failure (POF) is a devastating condition with serious clinical consequences. The purpose of this review was to summarize the current status of stem cell therapy for POF. Considering the diversity of cell types and functions, a rigorous review is required for the guidance for further research into this field. Meanwhile, the challenges and prospect for clinical application of stem cell treatment, methodological improvements, and innovations are addressed.
Collapse
Affiliation(s)
- Ming-Yao Wang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Yi-Xuan Wang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Jesse Li-Ling
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Hui-Qi Xie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| |
Collapse
|
13
|
Quiroz YY, Llorens E, Motta G, Bujons A. Autologous fat grafting with stem cell transplantation in an exstrophic patient. A case. J Pediatr Urol 2021; 17:119-121. [PMID: 33390329 DOI: 10.1016/j.jpurol.2020.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/14/2020] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Lipofilling has been used in plastic surgery for breast reconstruction and treatment of burns. Carrying out minimally invasive techniques would allow treatment of hypertrophic scars and depressed suprapubic area in exstrophic patients to improve their aesthetic appearance. MATERIAL AND METHODS This video presents the case of a female 16 year old patient with bladder exstrophy, who initially had an urinary reconstruction with ureterosigmoidostomy and posteriorly bladder augmentation and Mitrofanoff, with hypertrophic scars and depressed abdominal wall in suprapubic area. We present this procedure for autologous fat grafting with stem cells in the abdominal area. RESULTS The lipofilling procedure was carried out successfully, without intraoperative complications in 120 min. The liposuction was 250 cc and 80 cc was used for injection, achieving the improvement of the aesthetic appearance of the scars in a short time. The imaging control was follow up with abdominal wall ultrasound, three months later, increasing its thickness by 42%. CONCLUSION The autologous fat grafting with stem cells is a safe and feasible procedure in the exstrophic population, with excellent aesthetic results, but we need long term follow ups to determine how long the effect of fat grafting remains.
Collapse
Affiliation(s)
- Yesica Y Quiroz
- Urology Department, Fundación Puigvert, Universidad Autónoma de Barcelona, Barcelona, Spain.
| | - Erika Llorens
- Urology Department, Fundación Puigvert, Universidad Autónoma de Barcelona, Barcelona, Spain.
| | - Guilherme Motta
- Urology Department, Fundación Puigvert, Universidad Autónoma de Barcelona, Barcelona, Spain.
| | - Anna Bujons
- Urology Department, Fundación Puigvert, Universidad Autónoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
14
|
Ono‐Uruga Y, Ikeda Y, Matsubara Y. Platelet production using adipose-derived mesenchymal stem cells: Mechanistic studies and clinical application. J Thromb Haemost 2021; 19:342-350. [PMID: 33217130 PMCID: PMC7898515 DOI: 10.1111/jth.15181] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/29/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022]
Abstract
Megakaryocytes (MKs) are platelet progenitor stem cells found in the bone marrow. Platelets obtained from blood draws can be used for therapeutic applications, especially platelet transfusion. The needs for platelet transfusions for clinical situation is increasing, due in part to the growing number of patients undergoing chemotherapy. Platelets obtained from donors, however, have the disadvantages of a limited storage lifespan and the risk of donor-related infection. Extensive effort has therefore been directed at manufacturing platelets ex vivo. Here, we review ex vivo technologies for MK development, focusing on human adipose tissue-derived mesenchymal stem/stromal cell line (ASCL)-based strategies and their potential clinical application. Bone marrow and adipose tissues contain mesenchymal stem/stromal cells that have an ability to differentiate into MKs, which release platelets. Taking advantage of this mechanism, we developed a donor-independent system for manufacturing platelets for clinical application using ASCL established from adipose-derived mesenchymal stem/stromal cells (ASCs). Culture of ASCs with endogenous thrombopoietin and its receptor c-MPL, and endogenous genes such as p45NF-E2 leads to MK differentiation and subsequent platelet production. ASCs compose heterogeneous cells, however, and are not suitable for clinical application. Thus, we established ASCLs, which expand into a more homogeneous population, and fulfill the criteria for mesenchymal stem cells set by the International Society for Cellular Therapy. Using our ASCL culture system with MK lineage induction medium without recombinant thrombopoietin led to peak production of platelets within 12 days, which may be sufficient for clinical application.
Collapse
Affiliation(s)
- Yukako Ono‐Uruga
- Clinical and Translational Research CenterKeio University School of MedicineTokyoJapan
| | - Yasuo Ikeda
- Department of HematologyKeio University School of MedicineTokyoJapan
- Life Science and Medical BioscienceWaseda UniversityTokyoJapan
| | - Yumiko Matsubara
- Clinical and Translational Research CenterKeio University School of MedicineTokyoJapan
- Department of Laboratory MedicineKeio University School of MedicineTokyoJapan
| |
Collapse
|
15
|
Banani MA, Rahmatullah M, Farhan N, Hancox Z, Yousaf S, Arabpour Z, Moghaddam ZS, Mozafari M, Sefat F. Adipose tissue-derived mesenchymal stem cells for breast tissue regeneration. Regen Med 2021; 16:47-70. [PMID: 33533667 DOI: 10.2217/rme-2020-0045] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 01/08/2021] [Indexed: 02/06/2023] Open
Abstract
With an escalating incidence of breast cancer cases all over the world and the deleterious psychological impact that mastectomy has on patients along with several limitations of the currently applied modalities, it's plausible to seek unconventional approaches to encounter such a burgeoning issue. Breast tissue engineering may allow that chance via providing more personalized solutions which are able to regenerate, mimicking natural tissues also facing the witnessed limitations. This review is dedicated to explore the utilization of adipose tissue-derived mesenchymal stem cells for breast tissue regeneration among postmastectomy cases focusing on biomaterials and cellular aspects in terms of harvesting, isolation, differentiation and new tissue formation as well as scaffolds types, properties, material-host interaction and an in vitro breast tissue modeling.
Collapse
Affiliation(s)
- Mohammed A Banani
- Division of Surgery & Interventional Science, University College London, London, NW3 2PS, UK
| | - Mohammed Rahmatullah
- Division of Surgery & Interventional Science, University College London, London, NW3 2PS, UK
| | - Nawras Farhan
- Division of Surgery & Interventional Science, University College London, London, NW3 2PS, UK
| | - Zoe Hancox
- Department of Biomedical & Electronics Engineering, School of Engineering, University of Bradford, Bradford, BD7 1DP, UK
| | - Safiyya Yousaf
- Department of Biomedical & Electronics Engineering, School of Engineering, University of Bradford, Bradford, BD7 1DP, UK
| | - Zohreh Arabpour
- Department of Biomedical & Electronics Engineering, School of Engineering, University of Bradford, Bradford, BD7 1DP, UK
| | - Zoha Salehi Moghaddam
- Department of Biomedical & Electronics Engineering, School of Engineering, University of Bradford, Bradford, BD7 1DP, UK
- Interdisciplinary Research Centre in Polymer Science & Technology (IRC Polymer), University of Bradford, Bradford, BD7 1DP, UK
| | - Masoud Mozafari
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, M5G 1X5, Canada
| | - Farshid Sefat
- Department of Biomedical & Electronics Engineering, School of Engineering, University of Bradford, Bradford, BD7 1DP, UK
- Interdisciplinary Research Centre in Polymer Science & Technology (IRC Polymer), University of Bradford, Bradford, BD7 1DP, UK
| |
Collapse
|
16
|
Meyfour A, Pahlavan S, Mirzaei M, Krijgsveld J, Baharvand H, Salekdeh GH. The quest of cell surface markers for stem cell therapy. Cell Mol Life Sci 2021; 78:469-495. [PMID: 32710154 PMCID: PMC11073434 DOI: 10.1007/s00018-020-03602-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/10/2020] [Accepted: 07/17/2020] [Indexed: 12/15/2022]
Abstract
Stem cells and their derivatives are novel pharmaceutics that have the potential for use as tissue replacement therapies. However, the heterogeneous characteristics of stem cell cultures have hindered their biomedical applications. In theory and practice, when cell type-specific or stage-specific cell surface proteins are targeted by unique antibodies, they become highly efficient in detecting and isolating specific cell populations. There is a growing demand to identify reliable and actionable cell surface markers that facilitate purification of particular cell types at specific developmental stages for use in research and clinical applications. The identification of these markers as very important members of plasma membrane proteins, ion channels, transporters, and signaling molecules has directly benefited from proteomics and tools for proteomics-derived data analyses. Here, we review the methodologies that have played a role in the discovery of cell surface markers and introduce cutting edge single cell proteomics as an advanced tool. We also discuss currently available specific cell surface markers for stem cells and their lineages, with emphasis on the nervous system, heart, pancreas, and liver. The remaining gaps that pertain to the discovery of these markers and how single cell proteomics and identification of surface markers associated with the progenitor stages of certain terminally differentiated cells may pave the way for their use in regenerative medicine are also discussed.
Collapse
Affiliation(s)
- Anna Meyfour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mehdi Mirzaei
- Department of Molecular Sciences, Macquarie University, Sydney, NSW, Australia
- Australian Proteome Analysis Facility, Macquarie University, Sydney, NSW, Australia
| | - Jeroen Krijgsveld
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, Heidelberg, Germany
- Medical Faculty, Heidelberg University, Im Neuenheimer Feld 672, Heidelberg, Germany
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Ghasem Hosseini Salekdeh
- Department of Molecular Sciences, Macquarie University, Sydney, NSW, Australia.
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Banihashem St, P.O. Box: 16635-148, 1665659911, Tehran, Iran.
| |
Collapse
|
17
|
Sharath SS, Ramu J, Nair SV, Iyer S, Mony U, Rangasamy J. Human Adipose Tissue Derivatives as a Potent Native Biomaterial for Tissue Regenerative Therapies. Tissue Eng Regen Med 2020; 17:123-140. [PMID: 31953618 PMCID: PMC7105544 DOI: 10.1007/s13770-019-00230-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/07/2019] [Accepted: 11/15/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Human adipose tissue is a great source of translatable biomaterials owing to its ease of availability and simple processing. Reusing discardable adipose tissue for tissue regeneration helps in mimicking the exact native microenvironment of tissue. Over the past 10 years, extraction, processing, tuning and fabrication of adipose tissue have grabbed the attention owing to their native therapeutic and regenerative potential. The present work gives the overview of next generation biomaterials derived from human adipose tissue and their development with clinical relevance. METHODS Around 300 articles have been reviewed to widen the knowledge on the isolation, characterization techniques and medical applications of human adipose tissue and its derivatives from bench to bedside. The prospective applications of adipose tissue derivatives like autologous fat graft, stromal vascular fraction, stem cells, preadipocyte, adipokines and extracellular matrix, their behavioural mechanism, rational property of providing native bioenvironment, circumventing their translational abilities, recent advances in featuring them clinically have been reviewed extensively to reveal the dormant side of human adipose tissue. RESULTS Basic understanding about the molecular and structural aspect of human adipose tissue is necessary to employ it constructively. This review has nailed the productive usage of human adipose tissue, in a stepwise manner from exploring the methods of extracting derivatives, concerns during processing and its formulations to turning them into functional biomaterials. Their performance as functional biomaterials for skin regeneration, wound healing, soft tissue defects, stem cell and other regenerative therapies under in vitro and in vivo conditions emphasizes the translational efficiency of adipose tissue derivatives. CONCLUSION In the recent years, research interest has inclination towards constructive tissue engineering and regenerative therapies. Unravelling the maximum utilization of human adipose tissue derivatives paves a way for improving existing tissue regeneration and cellular based therapies and other biomedical applications.
Collapse
Affiliation(s)
- Siva Sankari Sharath
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - Janarthanan Ramu
- Department of Plastic and Reconstructive Surgery, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - Shantikumar Vasudevan Nair
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - Subramaniya Iyer
- Department of Plastic and Reconstructive Surgery, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - Ullas Mony
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041, India.
| | - Jayakumar Rangasamy
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041, India.
| |
Collapse
|
18
|
The Role of Marine Organic Extract in Bone Regeneration: A Pilot Study. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2925879. [PMID: 32149098 PMCID: PMC7049417 DOI: 10.1155/2020/2925879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 12/22/2019] [Accepted: 01/07/2020] [Indexed: 11/25/2022]
Abstract
Novel biomaterials capable of accelerating the healing process of skeletal tissues are urgently needed in dentistry. The present in vivo study assessed the osteoconductive and osteoinductive properties of experimental biphasic bioceramics (HA-TCP) modified or not by a nacre extract (marine organic extract, MOE) in a sheep model. Fabrication of MOE involved mixing ground nacre (0.05 g, particle sizes < 0.1 mm) with glacial ethanoic acid (5 mL, pH 7) for 72 hours using external magnetic stirring (25°C). Nonreactive carriers (sterile polythene tubes; 3/animal, radius: 2.5 mm, length: 10.0 mm) pertaining to the control (empty) or experimental groups (HA-TCP or MOE-modified HA-TCP) were implanted intramuscularly into the abdominal segment of the torso in sheep (n = 8, age: 2 years, weight: 45 kg). Euthanization of animals was performed at 3 and 6 months after surgery. Tissues harvested were subjected to macroscopic and radiographic assessments. Specimens were then stained for histological analysis. Both control and experimental animals were capable of inducing the neoformation of fibrous connective tissue at both time points where superior amounts of tissue formation and mineralization were detected for experimental groups (unaltered (at 3 and 6 mos) and MOE-modified HA-TCP (at 3 mos)). Histological results, however, revealed that mature bone formation was only observed for specimens fabricated with MOE-modified HA-TCP in a time-dependent manner. The present study has successfully demonstrated the in vivo utility of experimental biphasic bioceramics modified by MOE in an ectopic grafting sheep model. Promising osteoconductive and osteoinductive properties must be further developed and confirmed by subsequent research.
Collapse
|
19
|
Liu GB, Pan YM, Liu YS, Hu JH, Zhang XD, Zhang DW, Wang Y, Feng YK, Yu JB, Cheng YX. Ghrelin promotes neural differentiation of adipose tissue-derived mesenchymal stem cell via AKT/mTOR and β-catenin signaling pathways. Kaohsiung J Med Sci 2020; 36:405-416. [PMID: 32003536 DOI: 10.1002/kjm2.12188] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 01/07/2020] [Indexed: 02/04/2023] Open
Abstract
Adipose tissue-derived mesenchymal stem cells (ADSCs) are multipotent cells that can differentiate into various cell types. This study aimed to investigate the effect of ghrelin on the neural differentiation of rat ADSCs and underlying molecular mechanisms. Rat ADSCs were isolated and third-passage ADSCs were used in this study. The isolated ADSCs were characterized by flow cytometry analysis for MSCs' surface expression markers as evidenced by positive for CD90, CD44, and CD29 and negative for CD34, CD45, and CD11b/2f/c. The multilineage differentiation of ADSCs was confirmed by adipogenic, osteogenic, and neural differentiation. After induction of neurogenesis, the differentiated cells were identified by development of neuron-like morphology and expression of neural markers including glial fibrillary acidic protein, Nestin, MAP2, and β-Tubulin III using immunofluorescence and western blot. Ghrelin concentration dependently elevated the proportion of neural-like cells and branching dendrites, as well as upregulated the expression of neural markers. Further, the expression of nuclear β-catenin, p-GSK-3β, p-AKT, and p-mTOR was increased by ghrelin, indicating an activation of β-catenin and AKT/mTOR signaling after the ghrelin treatment. Importantly, inhibition of β-catenin or AKT/mTOR signaling suppressed ghrelin-induced neurogenesis. Therefore, we demonstrate that ghrelin promotes neural differentiation of ADSCs through the activation of β-catenin and AKT/mTOR signaling pathways.
Collapse
Affiliation(s)
- Gui-Bo Liu
- Department of Anatomy, School of Basic Medical Sciences, Mudanjiang Medical College, Mudanjiang, People's Republic of China.,Institute of Neural Tissue Engineering, Mudanjiang Medical College, Mudanjiang, People's Republic of China
| | - Yan-Ming Pan
- Department of Anatomy, School of Basic Medical Sciences, Mudanjiang Medical College, Mudanjiang, People's Republic of China.,Key Laboratory of Cancer Prevention and Treatment of Heilongjiang Province, Mudanjiang Medical College, Mudanjiang, People's Republic of China
| | - Yun-Shuang Liu
- Department of Medical Imaging, Hongqi Hospital of Mudanjiang Medical College, Mudanjiang, People's Republic of China
| | - Jia-Hang Hu
- Department of Medical Imaging, Hongqi Hospital of Mudanjiang Medical College, Mudanjiang, People's Republic of China
| | - Xiao-Dong Zhang
- Department of Infectious Diseases, Hongqi Hospital of Mudanjiang Medical College, Mudanjiang, People's Republic of China
| | - Da-Wei Zhang
- Department of Anatomy, School of Basic Medical Sciences, Mudanjiang Medical College, Mudanjiang, People's Republic of China
| | - Ying Wang
- Department of Anatomy, School of Basic Medical Sciences, Mudanjiang Medical College, Mudanjiang, People's Republic of China.,Institute of Neural Tissue Engineering, Mudanjiang Medical College, Mudanjiang, People's Republic of China
| | - Yu-Kuan Feng
- Department of Anatomy, School of Basic Medical Sciences, Mudanjiang Medical College, Mudanjiang, People's Republic of China
| | - Jian-Bo Yu
- Key Laboratory of Cancer Prevention and Treatment of Heilongjiang Province, Mudanjiang Medical College, Mudanjiang, People's Republic of China.,Pathology Diagnosis Center, The First Clinical Medical School of Mudanjiang Medical College, Mudanjiang, People's Republic of China
| | - Yong-Xia Cheng
- Key Laboratory of Cancer Prevention and Treatment of Heilongjiang Province, Mudanjiang Medical College, Mudanjiang, People's Republic of China.,Pathology Diagnosis Center, The First Clinical Medical School of Mudanjiang Medical College, Mudanjiang, People's Republic of China.,Institute of Stem Cells, Mudanjiang Medical College, Mudanjiang, People's Republic of China
| |
Collapse
|
20
|
Huang KC, Chuang MH, Lin ZS, Lin YC, Chen CH, Chang CL, Huang PC, Syu WS, Chiou TW, Hong ZH, Tsai YC, Harn HJ, Lin PC, Lin SZ. Transplantation with GXHPC1 for Liver Cirrhosis: Phase 1 Trial. Cell Transplant 2019; 28:100S-111S. [PMID: 31722556 PMCID: PMC7016466 DOI: 10.1177/0963689719884885] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Currently, the only effective therapy for cirrhosis of the liver is liver transplantation. However, finding a compatible liver is difficult due to the low supply of healthy livers and the ever-increasing demand. However, stem-cell therapy may offer a solution for liver cirrhosis; for example, GXHPC1 therapy preparation contains adipose-derived mesenchymal stem cells (AD-MSCs) and was developed for the treatment of liver cirrhosis. In our previous report, animal studies suggested that treatment of a diseased liver via GXHPC1 transplantation can abrogate liver fibrosis and facilitate recovery of liver function. In our current human trial, patients with liver cirrhosis were included. Their adipose tissue was harvested from the subcutaneous fat of the abdominal wall during surgery. AD-MSCs were cultured and suspended at a concentration of 100 million cells in 1 ml of physiological saline (i.e., GXHPC1). This human study passed the Taiwan Food and Drug Administration IND inspection and received Phase I clinical trial permission. The trial was conducted with six patients with liver cirrhosis to demonstrate the safety and efficacy of administering GXHPC1. Intrahepatic injection of GXHPC1 did not cause any safety issues in the analysis of adverse drug reactions and suspected unexpected serious adverse reactions, and showed a tendency for improvement of liver function, METAVIR score, Child–Pugh score, MELD score, and quality of life for patients with liver cirrhosis.
Collapse
Affiliation(s)
- Ko-Chang Huang
- Department of Gastroenterological, China Medical University Beigan Hospital, Yunlin
| | - Ming-Hsi Chuang
- PhD Program of Technology Management, Chung Hua University, Hsinchu.,Department of Bioinformatics, Chung Hua University, Hsinchu
| | - Zung-Sheng Lin
- Department of General Surgery, China Medical University Beigan Hospital, Yunlin
| | - Yi-Chun Lin
- Department of Stem Cell Applied Technology, Gwo Xi Stem Cell Applied Technology, Hsinchu
| | | | | | - Pi-Chun Huang
- Department of Stem Cell Applied Technology, Gwo Xi Stem Cell Applied Technology, Hsinchu
| | - Wan-Sin Syu
- Department of Stem Cell Applied Technology, Gwo Xi Stem Cell Applied Technology, Hsinchu
| | - Tzyy-Wen Chiou
- Department of Life Science and Graduate Institute of Biotechnology, National Dong Hwa University, Hualien
| | - Zih-Han Hong
- Department of Stem Cell Applied Technology, Gwo Xi Stem Cell Applied Technology, Hsinchu
| | - Yu-Chen Tsai
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei
| | - Horng-Jyh Harn
- Bioinnovation Center, Tzu Chi foundation; Department of Pathology, Buddhist Tzu Chi General hospital, Tzu Chi University, Hualien
| | - Po-Cheng Lin
- Department of Stem Cell Applied Technology, Gwo Xi Stem Cell Applied Technology, Hsinchu
| | - Shinn-Zong Lin
- Bioinnovation Center, Tzu Chi foundation; Department of Neurosurgery, Buddhist Tzu Chi General hospital, Tzu Chi University, Hualien
| |
Collapse
|
21
|
Interleukin-6 from Adipose-Derived Stem Cells Promotes Tissue Repair by the Increase of Cell Proliferation and Hair Follicles in Ischemia/Reperfusion-Treated Skin Flaps. Mediators Inflamm 2019; 2019:2343867. [PMID: 31814799 PMCID: PMC6877947 DOI: 10.1155/2019/2343867] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/25/2019] [Accepted: 09/27/2019] [Indexed: 12/21/2022] Open
Abstract
The most common postoperative complication after reconstructive surgery is flap necrosis. Adipose-derived stem cells (ADSCs) and their secretomes are reported to mediate skin repair. This study was designed to investigate whether conditioned media from ADSCs (ADSC-CM) protects ischemia/reperfusion- (I/R-) induced injury in skin flaps by promoting cell proliferation and increasing the number of hair follicles. The mouse flap model of ischemia was ligating the long thoracic vessels for 3 h, followed by blood reperfusion. ADSC-CM was administered to the flaps, and their survival was observed on postoperative day 5. ADSC-CM treatment led to a significant increase in cell proliferation and the number of hair follicles. IL-6 levels in the lysate and CM from ADSCs were significantly higher than those from Hs68 fibroblasts. Furthermore, a strong decrease in cell proliferation and the number of hair follicles was observed after treatment with IL-6-neutralizing antibodies or si-IL-6-ADSC. In addition, ADSC transplantation increased flap repair, cell proliferation, and hair follicle number in I/R injury of IL-6-knockout mice. In conclusion, IL-6 secreted from ADSCs promotes the survival of I/R-induced flaps by increasing cell proliferation and the number of hair follicles. ADSCs represent a promising therapy for preventing skin flap necrosis following reconstructive and plastic surgery.
Collapse
|
22
|
Yamasaki A, Kunitomi Y, Murata D, Sunaga T, Kuramoto T, Sogawa T, Misumi K. Osteochondral regeneration using constructs of mesenchymal stem cells made by bio three-dimensional printing in mini-pigs. J Orthop Res 2019; 37:1398-1408. [PMID: 30561041 DOI: 10.1002/jor.24206] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 12/06/2018] [Indexed: 02/04/2023]
Abstract
Osteoarthritis is a major joint disease that has been extensively investigated in humans and in model animals. In this study, we examined the regeneration of articular cartilage and subchondral bone using artificial scaffold-free constructs composed of adipose tissue-derived mesenchymal stem cells (AT-MSCs) created using bio three-dimensional (3D) printing with a needle-array. Printed constructs were implanted into osteochondral defects created in the right femoral trochlear groove of six mini-pigs, using femoral defects created in the left femurs as controls. Repair within the defects was evaluated at 3 and 6 months post-implantation using computed tomography (CT) and magnetic resonance (MR) imaging. The radiolucent volume (RV, mm3 ) in the defects was calculated using multi-planar reconstruction of CT images. MR images were evaluated based on a modified 2D- MOCART (magnetic resonance observation of cartilage repair tissue) grading system. Gross and microscopic pathology were scored according to the ICRS (International Cartilage Repair Society) scale at 6 months after implantation. The percentage RV at 3 months postoperation was significantly lower in the implanted defects than in the controls, whereas total scores based on the MOCART system were significantly higher in the implanted defects as compared with the controls. Although there were no statistical differences in the gross scores, the average histological scores were significantly higher in the implanted defects than in the controls. To our knowledge, this is the first report to suggest that artificial scaffold-free 3D-printed constructs of autologous AT-MSCs can be aid in the osteochondral regeneration in pigs. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1398-1408, 2019.
Collapse
Affiliation(s)
- Atsushi Yamasaki
- Veterinary Surgery, Department of Veterinary Clinical Science, Joint Faculty of Veterinary Medicine, Kagoshima University, 21-24 Korimoto 1-chome, Kagoshima, 890-0065, Japan
| | - Yoshihiro Kunitomi
- Cyfuse Biomedical K.K., 3-1 Hongo 7-chome, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Daiki Murata
- Department of Regenerative Medicine and Biomedical Engineering, Faculty of Medicine, Saga University, Honjyo 1-chome, Honjyo-cho, Saga, 840-8502, Japan
| | - Takafumi Sunaga
- Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, 21-24 Korimoto 1-chome, Kagoshima, 890-0065, Japan
| | - Tomohide Kuramoto
- Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, 21-24 Korimoto 1-chome, Kagoshima, 890-0065, Japan
| | - Takeshi Sogawa
- Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, 21-24 Korimoto 1-chome, Kagoshima, 890-0065, Japan
| | - Kazuhiro Misumi
- Veterinary Surgery, Department of Veterinary Clinical Science, Joint Faculty of Veterinary Medicine, Kagoshima University, 21-24 Korimoto 1-chome, Kagoshima, 890-0065, Japan
| |
Collapse
|
23
|
Use of artificial neural networks to identify the predictive factors of extracorporeal shock wave therapy treating patients with chronic plantar fasciitis. Sci Rep 2019; 9:4207. [PMID: 30862876 PMCID: PMC6414656 DOI: 10.1038/s41598-019-39026-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 01/11/2019] [Indexed: 02/06/2023] Open
Abstract
The purpose of our study is to identify the predictive factors for a minimum clinically successful therapy after extracorporeal shock wave therapy for chronic plantar fasciitis. The demographic and clinical characteristics were evaluated. The artificial neural networks model was used to choose the significant variables and model the effect of achieving the minimum clinically successful therapy at 6-months’ follow-up. The multilayer perceptron model was selected. Higher VAS (Visual Analogue Score) when taking first steps in the morning, presence of plantar fascia spur, shorter duration of symptom had statistical significance in increasing the odd. The artificial neural networks model shows that the sensitivity of predictive factors was 84.3%, 87.9% and 61.4% for VAS, spurs and duration of symptom, respectively. The specificity 35.7%, 37.4% and 22.3% for VAS, spurs and duration of symptom, respectively. The positive predictive value was 69%, 72% and 57% for VAS, spurs and duration of symptom, respectively. The negative predictive value was 82%, 84% and 59%, for VAS, spurs and duration of symptom respectively. The area under the curve was 0.738, 0.882 and 0.520 for VAS, spurs and duration of symptom, respectively. The predictive model showed a good fitting of with an overall accuracy of 92.5%. Higher VAS symptomatized by short-duration, severer pain or plantar fascia spur are important prognostic factors for the efficacy of extracorporeal shock wave therapy. The artificial neural networks predictive model is reasonable and accurate model can help the decision-making for the application of extracorporeal shock wave therapy.
Collapse
|
24
|
Megakaryocytes and platelets from a novel human adipose tissue-derived mesenchymal stem cell line. Blood 2018; 133:633-643. [PMID: 30487128 DOI: 10.1182/blood-2018-04-842641] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 10/02/2018] [Indexed: 12/24/2022] Open
Abstract
The clinical need for platelet transfusions is increasing; however, donor-dependent platelet transfusions are associated with practical problems, such as the limited supply and the risk of infection. Thus, we developed a manufacturing system for platelets from a donor-independent cell source: a human adipose-derived mesenchymal stromal/stem cell line (ASCL). The ASCL was obtained using an upside-down culture flask method and satisfied the minimal criteria for defining mesenchymal stem cells (MSCs) by The International Society for Cellular Therapy. The ASCL showed its proliferation capacity for ≥2 months without any abnormal karyotypes. The ASCL was cultured in megakaryocyte induction media. ASCL-derived megakaryocytes were obtained, with a peak at day 8 of culture, and ASCL-derived platelets (ASCL-PLTs) were obtained, with a peak at day 12 of culture. We observed that CD42b+ cells expressed an MSC marker (CD90) which is related to cell adhesion. Compared with peripheral platelets, ASCL-PLTs exhibit higher levels of PAC1 binding, P-selectin surface exposure, ristocetin-induced platelet aggregation, and ADP-induced platelet aggregation, as well as similar levels of fibrinogen binding and collagen-induced platelet aggregation. ASCL-PLTs have lower epinephrine-induced platelet aggregation. The pattern of in vivo kinetics after infusion into irradiated immunodeficient NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ mice was similar to that of platelet concentrates. ASCL-PLTs have similar characteristics to those of peripheral platelets and might have an additional function as MSCs. The establishment of the ASCL and its differentiation into ASCL-PLTs do not require gene transfer, and endogenous thrombopoietin is used for differentiation. The present protocol is a simple method that does not require feeder cells, further enhancing the clinical application of our approach.
Collapse
|
25
|
Parshyna I, Lehmann S, Grahl K, Pahlke C, Frenzel A, Weidlich H, Morawietz H. Impact of omega-3 fatty acids on expression of angiogenic cytokines and angiogenesis by adipose-derived stem cells. ATHEROSCLEROSIS SUPP 2018; 30:303-310. [PMID: 29096855 DOI: 10.1016/j.atherosclerosissup.2017.05.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS Human adipose-tissue derived stem cells (ADSC) are interesting novel targets in tissue engineering and regenerative medicine with pronounced angiogenic capacities. Furthermore, omega-3 fatty acids have been described to mediate cardioprotective effects, but their role in angiogenesis and vascular regeneration is not well-understood. Here, we analyzed the impact of different omega-3 fatty acids on angiogenesis by ADSCs. METHODS Stem cells were cultured as monolayers or in 3D models, in spheroids embedded in collagen matrix or in co-cultures with human umbilical vein endothelial cells (HUVECs) in the Matrigel™ assay. The angiogenic properties of ADSCs were assessed by their sprouting and paracrine activities, gene expression by RT-PCR, Western blot, and enzyme immunoassay. RESULTS Stimulation of undifferentiated ADSCs with docosahexaenoic acid (DHA) strongly upregulated angiopoietin-1 mRNA levels up to 4.6 ± 0.3 fold. Furthermore, Il-6 and Il-8 mRNAs were increased 4.2 ± 0.5 fold and 7.1 ± 1.1 fold, respectively. On the other hand, addition of DHA significantly decreased the cumulative sprout length by 2.7 ± 0.8 fold and reduced the total number of sprouts by 2.3 ± 0.9 fold in the in vitro angiogenesis assay. Moreover, excretion of IL-8 into the medium rapidly increased up to 1.7 ± 0.3 fold in response to treatment of ADSCs with DHA. Finally, protein kinase C inhibitor RO-31-8220 abrogated DHA-mediated up-regulation of angiopoietin-1 without significantly affecting ADSCs cell viability. CONCLUSION In conclusion, ADSCs might regulate the formation and function of microvascular networks.
Collapse
Affiliation(s)
- Iryna Parshyna
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital and Medical Faculty Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Susann Lehmann
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital and Medical Faculty Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Katrin Grahl
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital and Medical Faculty Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Claudia Pahlke
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital and Medical Faculty Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Annika Frenzel
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital and Medical Faculty Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | | | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital and Medical Faculty Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| |
Collapse
|
26
|
Dong M, Lin J, Lim W, Jin W, Lee HJ. Role of brown adipose tissue in metabolic syndrome, aging, and cancer cachexia. Front Med 2017; 12:130-138. [PMID: 29119382 DOI: 10.1007/s11684-017-0555-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 04/29/2017] [Indexed: 02/07/2023]
Abstract
Brown adipose tissue (BAT) plays a fundamental role in maintaining body temperature by producing heat. BAT that had been know to exist only in mammals and the human neonate has received great attention for the treatment of obesity and diabetes due to its important function in energy metabolism, ever since it is recently reported that human adults have functional BAT. In addition, beige adipocytes, brown adipocytes in white adipose tissue (WAT), have also been shown to take part in whole body metabolism. Multiple lines of evidence demonstrated that transplantation or activation of BAT or/and beige adipocytes reversed obesity and improved insulin sensitivity. Furthermore, many genes involved in BATactivation and/or the recruitment of beige cells have been found, thereby providing new promising strategies for future clinical application of BAT activation to treat obesity and metabolic diseases. This review focuses on recent advances of BAT function in the metabolic aspect and the relationship between BAT and cancer cachexia, a pathological process accompanied with decreased body weight and increased energy expenditure in cancer patients. The underlying possible mechanisms to reduce BAT mass and its activity in the elderly are also discussed.
Collapse
Affiliation(s)
- Meng Dong
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,The University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Jun Lin
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,The University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Wonchung Lim
- Department of Sports Medicine, College of Health Science, Cheongju University, Cheongju, 363-764, Republic of Korea
| | - Wanzhu Jin
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hyuek Jong Lee
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China. .,Center for Vascular Research, Institute for Basic Science (IBS), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
27
|
Gonçalves AI, Gershovich PM, Rodrigues MT, Reis RL, Gomes ME. Human adipose tissue-derived tenomodulin positive subpopulation of stem cells: A promising source of tendon progenitor cells. J Tissue Eng Regen Med 2017; 12:762-774. [PMID: 28593712 DOI: 10.1002/term.2495] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/14/2017] [Accepted: 06/03/2017] [Indexed: 01/05/2023]
Abstract
Cell-based therapies are of particular interest for tendon and ligament regeneration given the low regenerative potential of these tissues. Adipose tissue is an abundant source of stem cells, which may be employed for the healing of tendon lesions. However, human adult multipotent adipose-derived stem cells (hASCs) isolated from the stromal vascular fraction of adipose tissue originate highly heterogeneous cell populations that hinder their use in specific tissue-oriented applications. In this study, distinct subpopulations of hASCs were immunomagnetic separated and their tenogenic differentiation capacity evaluated in the presence of several growth factors (GFs), namely endothelial GF, basic-fibroblast GF, transforming GF-β1 and platelet-derived GF-BB, which are well-known regulators of tendon development, growth and healing. Among the screened hASCs subpopulations, tenomodulin-positive cells were shown to be more promising for tenogenic applications and therefore this subpopulation was further studied, assessing tendon-related markers (scleraxis, tenomodulin, tenascin C and decorin) both at gene and protein level. Additionally, the ability for depositing collagen type I and III forming extracellular matrix structures were weekly assessed up to 28 days. The results obtained indicated that tenomodulin-positive cells exhibit phenotypical features of tendon progenitor cells and can be biochemically induced towards tenogenic lineage, demonstrating that this subset of hASCs can provide a reliable source of progenitor cells for therapies targeting tendon regeneration.
Collapse
Affiliation(s)
- A I Gonçalves
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - P M Gershovich
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - M T Rodrigues
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - R L Reis
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.,The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| | - M E Gomes
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.,The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| |
Collapse
|
28
|
The Use of Adipose-Derived Stem Cells in Selected Skin Diseases (Vitiligo, Alopecia, and Nonhealing Wounds). Stem Cells Int 2017; 2017:4740709. [PMID: 28904532 PMCID: PMC5585652 DOI: 10.1155/2017/4740709] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 06/03/2017] [Accepted: 06/18/2017] [Indexed: 12/15/2022] Open
Abstract
The promising results derived from the use of adipose-derived stem cells (ADSCs) in many diseases are a subject of observation in preclinical studies. ADSCs seem to be the ideal cell population for the use in regenerative medicine due to their easy isolation, nonimmunogenic properties, multipotential nature, possibilities for differentiation into various cell lines, and potential for angiogenesis. This article reviews the current data on the use of ADSCs in the treatment of vitiligo, various types of hair loss, and the healing of chronic wounds.
Collapse
|
29
|
Zhang C, Li M, Zhu J, Luo F, Zhao J. Enhanced bone repair induced by human adipose-derived stem cells on osteogenic extracellular matrix ornamented small intestinal submucosa. Regen Med 2017; 12:541-552. [PMID: 28718708 DOI: 10.2217/rme-2017-0024] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
AIM Our aim was to design an osteogenic extracellular matrix (ECM) coated bioscaffold and to apply it to critical bone defect repair with adipose-derived stem cells (ADSCs). MATERIALS & METHODS Morphology of scaffolds was scanned by scanning electron microscope. Cell adhesion, proliferation and osteogenic differentiation of ADSCs on ECM-small intestinal submucosa (SIS) were evaluated by immunofluorescences staining, cell counting kit-8 and real-time qPCR, respectively. A mouse calvarial defect model was used to assess effects on bone regeneration in vivo. RESULTS Abundant ECM was coated on SIS, which facilitated cell adhesion and proliferation of ADSCs. ECM-SIS induced osteogenic differentiation of ADSCs even without osteogenic inductive factors. Bone regeneration in vivo was enhanced by ECM-SIS + ADSCs via BMP/SMAD pathway. CONCLUSION This work suggested a biofabricated SIS scaffold coated with osteogenic ECM-facilitated bone regeneration with ADSCs synergistically.
Collapse
Affiliation(s)
- Chi Zhang
- Zhejiang Key Laboratory of Pathophysiology, Medical School, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
| | - Mei Li
- Zhejiang Key Laboratory of Pathophysiology, Medical School, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China.,Ningbo Institute of Medical Sciences, Ningbo, Zhejiang 315020, People's Republic of China
| | - Jinjin Zhu
- Zhejiang Key Laboratory of Pathophysiology, Medical School, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
| | - Fangmiao Luo
- Zhejiang Key Laboratory of Pathophysiology, Medical School, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
| | - Jiyuan Zhao
- Zhejiang Key Laboratory of Pathophysiology, Medical School, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
| |
Collapse
|
30
|
Wang G, Yuan N, Huang S, Feng L, Han R, Zhang Y, Ren J, Meng M, Zhao X. The CNGRCLLII(KLAKLAK)2 peptide shows cytotoxicity against HUVECs by inducing apoptosis: An in vitro and in vivo study. Tumour Biol 2017; 39:1010428317701649. [PMID: 28475015 DOI: 10.1177/1010428317701649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Fibrinogen Asn-Gly-Arg motif can specifically recognize and bind to Aminopeptidase N (CD13) on vascular endothelial cells in newly formed tumor vessels. Adipose-derived stem cells can serve as ideal vectors for gene therapy because of their ability of migrating to tumor tissues. First, this study was aimed to design a new peptide (CNGRCLLII(KLAKLAK)2) named CNAK which contains cyclic Asn-Gly-Arg motif and test its biological activity against human umbilical vein endothelial cells. Second, we aimed to construct stably transfected adipose-derived stem cells which express the CNAK peptide and investigate their anti-angiogenic activity in vivo. Adipose-derived stem cells were employed to localize CNAK on vascular endothelial cells in tumors based on their homing property. First of all, the new peptide was synthesized, which effectively entered into CD13+ human umbilical vein endothelial cells and showed cytotoxicity against human umbilical vein endothelial cells. The peptide induced apoptosis of human umbilical vein endothelial cells in a time- and dose-dependent manner, inhibited the expression of Bcl-2, and promoted the expression of Caspase-3 in human umbilical vein endothelial cells. Furthermore, the migration and tube formation of human umbilical vein endothelial cells were inhibited by CNAK. Primary adipose-derived stem cells were then isolated and identified. Stably transfected adipose-derived stem cells which express CNAK peptide (CNAK-ASCs) were successfully established, and the migration of CNAK-ASCs was assessed. In vivo, CNAK-ASCs were found to inhibit the growth and angiogenesis of breast cancer xenografts. This effect may be through inhibiting the secretion of matrix metalloproteinase-2 and membrane type 1-matrix metalloproteinase in vivo. It was also found that CNAK-ASCs reduced the quantity of breast cancer stem cells in tumor tissues. Our data suggested that the new peptide CNAK containing Asn-Gly-Arg motif had anti-angiogenic activity in vitro and in vivo.
Collapse
Affiliation(s)
- Guanying Wang
- 1 Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Na Yuan
- 2 Department of Ultrasound, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Shangke Huang
- 1 Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Lu Feng
- 1 Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Rui Han
- 1 Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yujiao Zhang
- 1 Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Juan Ren
- 3 Department of Radiotherapy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Min Meng
- 4 Department of Oncology, Shandong Provincial Hospital Affiliated with Shandong University, Jinan, People's Republic of China
| | - Xinhan Zhao
- 1 Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| |
Collapse
|
31
|
Jin R, Shen M, Yu L, Wang X, Lin X. Adipose-Derived Stem Cells Suppress Inflammation Induced by IL-1β through Down-Regulation of P2X7R Mediated by miR-373 in Chondrocytes of Osteoarthritis. Mol Cells 2017; 40:222-229. [PMID: 28343378 PMCID: PMC5386960 DOI: 10.14348/molcells.2017.2314] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 01/17/2017] [Accepted: 02/07/2017] [Indexed: 12/12/2022] Open
Abstract
Adipose-derived stem cells (ADSCs) were previously considered to have an anti-inflammatory effect, and Interleukin-1β (IL-1β) was found to be a pro-inflammatory factor in chondrocytes, but the mechanism underlying ADSCs and IL-1β is unclear. In this study, we investigate whether P2X7 receptor (P2X7R) signalling, regulated by microRNA 373 (miR-373), was involved in the ADSCs and IL-1β mediated inflammation in osteoarthritis (OA). Chondrocytes were collected from 20 OA patients and 20 control participants, and ADSCs were collected from patients who had undergone abdominal surgery. The typical surface molecules of ASDCs were detected by flow cytometry. The level of nitric oxide (NO) was determined by Griess reagent. Concentrations of prostaglandin E2 (PGE2), interleukin 6 (IL-6), matrix metallopeptidase 3 (MMP-3) were detected by enzyme-linked immunosorbent assay (ELISA). The expressions of IL-6, MMP-3, miR-373 and P2X7R were determined by real-time polymerase chain reaction (PCR), and Western blot was used to detect the protein expression of P2X7R. The typical potential characters of ADSCs were verified. In chondrocytes or OA tissues, the miR-373 expression level was decreased, but the P2X7R expression was increased. IL-1β stimulation increased the level of inflammatory factors in OA chondrocytes, and ADSCs co-cultured with IL-1β-stimulated chondrocytes decreased the inflammation. OA chondrocytes transfected with the miR-373 inhibitor increased the inflammation level. The miR-373 mimic suppressed the inflammation by targeting P2X7R and regulated its expression, while its effect was reversed by overexpression of P2X7R. IL-1β induced inflammation in OA chondrocytes, while ADSCs seemed to inhibit the expression of P2X7R that was regulated by miR-373 and involved in the anti-inflammatory process in OA.
Collapse
Affiliation(s)
- Rilong Jin
- Department of Orthopedics Surgery, The First Affiliated Hospital, College of Medical Zhejiang University, Hangzhou, 310003
China
| | - Miaoda Shen
- Department of Orthopedics Surgery, The First Affiliated Hospital, College of Medical Zhejiang University, Hangzhou, 310003
China
| | - Liedao Yu
- Department of Orthopedics Surgery, The First Affiliated Hospital, College of Medical Zhejiang University, Hangzhou, 310003
China
| | - Xuanwei Wang
- Department of Orthopedics Surgery, The First Affiliated Hospital, College of Medical Zhejiang University, Hangzhou, 310003
China
| | - Xiangjin Lin
- Department of Orthopedics Surgery, The First Affiliated Hospital, College of Medical Zhejiang University, Hangzhou, 310003
China
| |
Collapse
|
32
|
|
33
|
Pu CM, Liu CW, Liang CJ, Yen YH, Chen SH, Jiang-Shieh YF, Chien CL, Chen YC, Chen YL. Adipose-Derived Stem Cells Protect Skin Flaps against Ischemia/Reperfusion Injury via IL-6 Expression. J Invest Dermatol 2017; 137:1353-1362. [PMID: 28163069 DOI: 10.1016/j.jid.2016.12.030] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 12/19/2016] [Accepted: 12/29/2016] [Indexed: 12/26/2022]
Abstract
Flap necrosis is the most frequent postoperative complication encountered in reconstructive surgery. We elucidated whether adipose-derived stem cells (ADSCs) and their derivatives might induce neovascularization and protect skin flaps during ischemia/reperfusion (I/R) injury. Flaps were subjected to 3 hours of ischemia by ligating long thoracic vessels and then to blood reperfusion. Qtracker-labeled ADSCs, ADSCs in conditioned medium (ADSC-CM), or ADSC exosomes (ADSC-Exo) were injected into the flaps. These treatments led to significantly increased flap survival and capillary density compared with I/R on postoperative day 5. IL-6 levels in the cell lysates or in conditioned medium were significantly higher in ADSCs than in Hs68 fibroblasts. ADSC-CM and ADSC-Exo increased tube formation. This result was corroborated by a strong decrease in skin repair after adding IL-6-neutralizing antibodies or small interfering RNA for IL-6 ADSCs. ADSC transplantation also increased flap recovery in I/R injury of IL-6-knockout mice. IL-6 was secreted from ADSCs through signal transducer and activator of transcription phosphorylation, and then IL-6 stimulated angiogenesis and enhanced recovery after I/R injury by the classic signaling pathway. The mechanism of skin recovery includes the direct differentiation of ADSCs into endothelial cells and the indirect effect of IL-6 released from ADSCs. ADSC-CM and ADSC-Exo could be used as off-the-shelf products for this therapy.
Collapse
Affiliation(s)
- Chi-Ming Pu
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan; Division of Plastic Surgery, Department of Surgery, Cathay General Hospital, Taipei, Taiwan
| | - Chen-Wei Liu
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chan-Jung Liang
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Hsiu Yen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan; Division of Plastic Surgery, Department of Surgery, Cathay General Hospital, Taipei, Taiwan
| | - Shun-Hua Chen
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Fen Jiang-Shieh
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chung-Liang Chien
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ya-Chun Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yuh-Lien Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
34
|
|
35
|
Haack-Sørensen M, Follin B, Juhl M, Brorsen SK, Søndergaard RH, Kastrup J, Ekblond A. Culture expansion of adipose derived stromal cells. A closed automated Quantum Cell Expansion System compared with manual flask-based culture. J Transl Med 2016; 14:319. [PMID: 27852267 PMCID: PMC5112664 DOI: 10.1186/s12967-016-1080-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/08/2016] [Indexed: 01/01/2023] Open
Abstract
Background Adipose derived stromal cells (ASCs) are a rich and convenient source of cells for clinical regenerative therapeutic approaches. However, applications of ASCs often require cell expansion to reach the needed dose. In this study, cultivation of ASCs from stromal vascular fraction (SVF) over two passages in the automated and functionally closed Quantum Cell Expansion System (Quantum system) is compared with traditional manual cultivation. Methods Stromal vascular fraction was isolated from abdominal fat, suspended in α-MEM supplemented with 10% Fetal Bovine Serum and seeded into either T75 flasks or a Quantum system that had been coated with cryoprecipitate. The cultivation of ASCs from SVF was performed in 3 ways: flask to flask; flask to Quantum system; and Quantum system to Quantum system. In all cases, quality controls were conducted for sterility, mycoplasmas, and endotoxins, in addition to the assessment of cell counts, viability, immunophenotype, and differentiation potential. Results The viability of ASCs passage 0 (P0) and P1 was above 96%, regardless of cultivation in flasks or Quantum system. Expression of surface markers and differentiation potential was consistent with ISCT/IFATS standards for the ASC phenotype. Sterility, mycoplasma, and endotoxin tests were consistently negative. An average of 8.0 × 107 SVF cells loaded into a Quantum system yielded 8.96 × 107 ASCs P0, while 4.5 × 106 SVF cells seeded per T75 flask yielded an average of 2.37 × 106 ASCs—less than the number of SVF cells seeded. ASCs P1 expanded in the Quantum system demonstrated a population doubling (PD) around 2.2 regardless of whether P0 was previously cultured in flasks or Quantum, while ASCs P1 in flasks only reached a PD of 1.0. Conclusion: Manufacturing of ASCs in a Quantum system enhances ASC expansion rate and yield significantly relative to manual processing in T-flasks, while maintaining the purity and quality essential to safe and robust cell production. Notably, the use of the Quantum system entails significantly reduced working hours and thereby costs. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-1080-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mandana Haack-Sørensen
- Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet University of Copenhagen, Juliane Maries Vej 20, Dept. 9302, 2100, Copenhagen, Denmark
| | - Bjarke Follin
- Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet University of Copenhagen, Juliane Maries Vej 20, Dept. 9302, 2100, Copenhagen, Denmark.
| | - Morten Juhl
- Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet University of Copenhagen, Juliane Maries Vej 20, Dept. 9302, 2100, Copenhagen, Denmark
| | - Sonja K Brorsen
- Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet University of Copenhagen, Juliane Maries Vej 20, Dept. 9302, 2100, Copenhagen, Denmark
| | - Rebekka H Søndergaard
- Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet University of Copenhagen, Juliane Maries Vej 20, Dept. 9302, 2100, Copenhagen, Denmark
| | - Jens Kastrup
- Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet University of Copenhagen, Juliane Maries Vej 20, Dept. 9302, 2100, Copenhagen, Denmark
| | - Annette Ekblond
- Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet University of Copenhagen, Juliane Maries Vej 20, Dept. 9302, 2100, Copenhagen, Denmark
| |
Collapse
|
36
|
Abstract
Bone marrow stromal stem cells (BMSCs) are adult multipotent cells, which have the potential to differentiate into cell types of mesodermal origin, namely osteocytes, adipocytes, and chondrocytes. Due to their accessibility and expansion potential, BMSCs have historically held therapeutic promise in tissue engineering and regenerative medicine applications. More recently, it has been demonstrated that not only can bone marrow stromal stem cells directly participate in tissue regeneration, but they also have the capacity to migrate to distant sites of tissue injury, where they can participate in tissue repair either directly through their differentiation or indirectly through paracrine mechanisms. Additionally, they can elicit various immunomodulatory signals, which can attenuate the inflammatory and immune responses. As such, bone marrow stromal stem cells have been explored clinically for treatment of a wide variety of different conditions including bone defects, graft-vs.-host disease, cardiovascular diseases, autoimmune diseases, diabetes, neurological diseases, and liver and kidney diseases. This review provides an overview of current clinical applications of bone marrow stromal stem cells and discusses their therapeutic properties, while also addressing limitations of their use. PubMed, Ovid, and Google Scholar online databases were searched using several keywords, including "stem cells", "tissue engineering", tissue regeneration" and "clinical trials". Additionally, Clinical trials.gov was used to locate completed clinical trials using bone marrow derived stem cells.
Collapse
Affiliation(s)
- A. Polymeri
- Department of Periodontics and Oral Medicine, University of Michigan, School of Dentistry, Ann Arbor, Michigan, USA
| | - W. V. Giannobile
- Department of Periodontics and Oral Medicine, University of Michigan, School of Dentistry, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, University of Michigan, College of Engineering, Ann Arbor, Michigan, USA
| | - D. Kaigler
- Department of Periodontics and Oral Medicine, University of Michigan, School of Dentistry, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, University of Michigan, College of Engineering, Ann Arbor, Michigan, USA
| |
Collapse
|
37
|
Rathinasabapathy A, Bruce E, Espejo A, Horowitz A, Sudhan DR, Nair A, Guzzo D, Francis J, Raizada MK, Shenoy V, Katovich MJ. Therapeutic potential of adipose stem cell-derived conditioned medium against pulmonary hypertension and lung fibrosis. Br J Pharmacol 2016; 173:2859-79. [PMID: 27448286 PMCID: PMC5275771 DOI: 10.1111/bph.13562] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 07/07/2016] [Accepted: 07/10/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE Pulmonary hypertension (PH) and pulmonary fibrosis (PF) are life threatening cardiopulmonary diseases. Existing pharmacological interventions have failed to improve clinical outcomes or reduce disease-associated mortality. Emerging evidence suggests that stem cells offer an effective treatment approach against various pathological conditions. It has been proposed that their beneficial actions may be mediated via secretion of paracrine factors. Herein, we evaluated the therapeutic potential of conditioned media (CM) from adipose stem cells (ASCs) against experimental models of PH and PF. EXPERIMENTAL APPROACH Monocrotaline (MCT) or bleomycin (Bleo) was injected into male Sprague-Dawley rats to induce PH or PF respectively. A subset of MCT and Bleo animals were treated with ASCs or CM. Echocardiographic and haemodynamic measurements were performed at the end of the study. Lung and heart tissues were harvested for RNA, protein and histological measurements. KEY RESULTS CM treatment attenuated MCT-induced PH by improving pulmonary blood flow and inhibiting cardiac remodelling. Further, histological studies revealed that right ventricular fibrosis, pulmonary vessel wall thickness and pericyte distribution were significantly decreased by CM administration. Likewise, CM therapy arrested the progression of PF in the Bleo model by reducing collagen deposition. Elevated expression of markers associated with tissue remodelling and inflammation were significantly reduced in both PF and PH lungs. Similar results were obtained with ASCs administration. CONCLUSIONS AND IMPLICATIONS Our study indicates that CM treatment is as effective as ASCs in treating PH and PF. These beneficial effects of CM may provide an innovative approach to treat cardiopulmonary disorders.
Collapse
Affiliation(s)
- Anandharajan Rathinasabapathy
- Pharmacodynamics, University of Florida, Gainesville, FL, USA
- Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Erin Bruce
- Pharmacodynamics, University of Florida, Gainesville, FL, USA
| | - Andrew Espejo
- Pharmacodynamics, University of Florida, Gainesville, FL, USA
| | - Alana Horowitz
- Pharmacodynamics, University of Florida, Gainesville, FL, USA
| | - Dhivya R Sudhan
- Radiation Oncology, University of Florida, Gainesville, FL, USA
| | - Anand Nair
- Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA, USA
- Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Dominic Guzzo
- Pharmacodynamics, University of Florida, Gainesville, FL, USA
| | - Joseph Francis
- Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - Mohan K Raizada
- Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA
| | - Vinayak Shenoy
- Pharmacodynamics, University of Florida, Gainesville, FL, USA.
- Pharmaceutical and Biomedical Sciences, California Health Sciences University, Clovis, CA, USA.
| | | |
Collapse
|
38
|
Nurkovic J, Dolicanin Z, Mustafic F, Mujanovic R, Memic M, Grbovic V, Skevin AJ, Nurkovic S. Mesenchymal stem cells in regenerative rehabilitation. J Phys Ther Sci 2016; 28:1943-8. [PMID: 27390452 PMCID: PMC4932093 DOI: 10.1589/jpts.28.1943] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/12/2016] [Indexed: 02/06/2023] Open
Abstract
[Purpose] Regenerative medicine and rehabilitation contribute in many ways to a specific
plan of care based on a patient’s medical status. The intrinsic self-renewing,
multipotent, regenerative, and immunosuppressive properties of mesenchymal stem cells
offer great promise in the treatment of numerous autoimmune, degenerative, and
graft-versus-host diseases, as well as tissue injuries. As such, mesenchymal stem cells
represent a therapeutic fortune in regenerative medicine. The aim of this review is to
discuss possibilities, limitations, and future clinical applications of mesenchymal stem
cells. [Subjects and Methods] The authors have identified and discussed clinically and
scientifically relevant articles from PubMed that have met the inclusion criteria.
[Results] Direct treatment of muscle injuries, stroke, damaged peripheral nerves, and
cartilage with mesenchymal stem cells has been demonstrated to be effective, with
synergies seen between cellular and physical therapies. Over the past few years, several
researchers, including us, have shown that there are certain limitations in the use of
mesenchymal stem cells. Aging and spontaneous malignant transformation of mesenchymal stem
cells significantly affect the functionality of these cells. [Conclusion] Definitive
conclusions cannot be made by these studies because limited numbers of patients were
included. Studies clarifying these results are expected in the near future.
Collapse
Affiliation(s)
- Jasmin Nurkovic
- Department of Biomedical Sciences, State University of Novi Pazar, Serbia; Center for Physical Medicine and Rehabilitation, Clinical Center Kragujevac, Serbia; Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Serbia
| | - Zana Dolicanin
- Department of Biomedical Sciences, State University of Novi Pazar, Serbia; General Hospital Novi Pazar, Serbia
| | | | - Rifat Mujanovic
- Department of Biomedical Sciences, State University of Novi Pazar, Serbia
| | - Mensur Memic
- Department of Biomedical Sciences, State University of Novi Pazar, Serbia
| | - Vesna Grbovic
- Center for Physical Medicine and Rehabilitation, Clinical Center Kragujevac, Serbia; Faculty of Medical Sciences, University of Kragujevac, Serbia
| | - Aleksandra Jurisic Skevin
- Center for Physical Medicine and Rehabilitation, Clinical Center Kragujevac, Serbia; Faculty of Medical Sciences, University of Kragujevac, Serbia
| | - Selmina Nurkovic
- Faculty of Medical Sciences, University of Kragujevac, Serbia; General Hospital Novi Pazar, Serbia
| |
Collapse
|
39
|
Ono-Uruga Y, Tozawa K, Horiuchi T, Murata M, Okamoto S, Ikeda Y, Suda T, Matsubara Y. Human adipose tissue-derived stromal cells can differentiate into megakaryocytes and platelets by secreting endogenous thrombopoietin. J Thromb Haemost 2016; 14:1285-97. [PMID: 26990635 DOI: 10.1111/jth.13313] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Indexed: 12/13/2022]
Abstract
UNLABELLED Essentials Manufacturing platelets from a donor-independent source is highlighted in transfusion medicine. We examined the differentiation of adipose tissue-derived stromal cells (ASCs) into platelets. Endogenous thrombopoietin (TPO) induced ASCs differentiation into megakaryocytes and platelets. TPO secretion from ASCs was due to an interaction of transferrin with its receptor CD71. SUMMARY Background Ex vivo production of megakaryocytes (MKs) and platelets from a donor-independent source is currently of intense interest in transfusion medicine. Adipose tissue-derived stromal cells (ASCs) constitute an attractive candidate cell source, because inducing these cells into MK lineages requires no gene transfer and only endogenous transcription factors containing p45NF-E2/Maf, an MK-inducing factor. Objectives To examine whether ASCs differentiate into MK lineages by using endogenous thrombopoietin (TPO), a primary cytokine that drives MK lineages. Methods TPO levels were measured by quantitative real-time PCR and ELISA. To investigate the effects of endogenous TPO on MK and platelet production, surface marker expression and functions for platelets were analyzed in ASC-derived cells cultured in the presence or absence of recombinant TPO. Based on a screening test, the role of transferrin receptor CD71 in TPO production and MK differentiation was examined with anti-CD71 antibody, small interfering RNA (siRNA) against CD71 (siRNA-CD71), and CD71-positive/negative cells. Results ASCs secreted TPO during MK differentiation, and the endogenous TPO facilitated MK and platelet production from ASCs. TPO secretion from ASCs occurred in a transferrin-dependent manner. ASCs treated with anti-CD71 antibody or transfected with siRNA-CD71 produced markedly less TPO. The TPO levels and MK yield were significantly higher when CD71-positive ASCs were used than when CD71-negative ASCs were used. Conclusions CD71 might be an appropriate marker for MK progenitor cells among human ASCs, because of the higher capacity of CD71-positive cells to produce TPO and their ability to differentiate into MKs. These findings could help to establish an efficient method for platelet production.
Collapse
Affiliation(s)
- Y Ono-Uruga
- Clinical and Translational Research Center, Keio University School of Medicine, Tokyo, Japan
- Kanagawa Academy of Science and Technology, Kanagawa, Japan
- Division of Hematology, Keio University School of Medicine, Tokyo, Japan
| | - K Tozawa
- Division of Hematology, Keio University School of Medicine, Tokyo, Japan
| | - T Horiuchi
- Clinical and Translational Research Center, Keio University School of Medicine, Tokyo, Japan
- Kanagawa Academy of Science and Technology, Kanagawa, Japan
| | - M Murata
- Department of Laboratory Medicine, Keio University School of Medicine, Tokyo, Japan
| | - S Okamoto
- Division of Hematology, Keio University School of Medicine, Tokyo, Japan
| | - Y Ikeda
- Division of Hematology, Keio University School of Medicine, Tokyo, Japan
- Faculty of Science and Engineering, Life Science and Medical Bioscience, Waseda University, Tokyo, Japan
| | - T Suda
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Y Matsubara
- Clinical and Translational Research Center, Keio University School of Medicine, Tokyo, Japan
- Kanagawa Academy of Science and Technology, Kanagawa, Japan
- Department of Laboratory Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
40
|
Hou X, Shi C, Chen W, Chen B, Jia W, Guo Y, Ma C, Ye G, Kang J, Dai J. Transplantation of human adipose-derived mesenchymal stem cells on a bladder acellular matrix for bladder regeneration in a canine model. Biomed Mater 2016; 11:031001. [DOI: 10.1088/1748-6041/11/3/031001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
41
|
Hashemibeni B, Dehghani L, Sadeghi F, Esfandiari E, Gorbani M, Akhavan A, Tahani ST, Bahramian H, Goharian V. Bone Repair with Differentiated Osteoblasts from Adipose-derived Stem Cells in Hydroxyapatite/Tricalcium Phosphate In vivo. Int J Prev Med 2016; 7:62. [PMID: 27141281 PMCID: PMC4837802 DOI: 10.4103/2008-7802.179510] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 11/07/2015] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Recently, tissue engineering has developed approaches for repair and restoration of damaged skeletal system based on different scaffolds and cells. This study evaluated the ability of differentiated osteoblasts from adipose-derived stem cells (ADSCs) seeded into hydroxyapatite/tricalcium phosphate (HA-TCP) to repair bone. METHODS In this study, ADSCs of 6 canines were seeded in HA-TCP and differentiated into osteoblasts in osteogenic medium in vitro and bone markers evaluated by reverse transcription polymerase chain reaction (RT-PCR). Scanning electron microscopy (SEM) was applied for detection of cells in the pores of scaffold. HA-TCP with differentiated cells as the test group and without cells as the cell-free group were implanted in separate defected sites of canine's tibia. After 8 weeks, specimens were evaluated by histological, immunohistochemical methods, and densitometry test. The data were analyzed using the SPSS 18 version software. RESULTS The expression of Type I collagen and osteocalcin genes in differentiated cells were indicated by RT-PCR. SEM results revealed the adhesion of cells in scaffold pores. Formation of trabecular bone confirmed by histological sections that revealed the thickness of bone trabecular was more in the test group. Production of osteopontin in extracellular matrix was indicated in both groups. Densitometry method indicated that strength in the test group was similar to cell-free group and natural bone (P > 0.05). CONCLUSIONS This research suggests that ADSCs-derived osteoblasts in HA-TCP could be used for bone tissue engineering and repairing.
Collapse
Affiliation(s)
- Batool Hashemibeni
- Department of Anatomical Sciences, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Leila Dehghani
- Department of Medical Sciences, Islamic Azad University, Najafabad Branch, Isfahan, Iran
| | - Farzaneh Sadeghi
- Department of Anatomical Sciences, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ebrahim Esfandiari
- Department of Anatomical Sciences, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Masood Gorbani
- Department of Tissue Engineering and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
- Applied Biotechnology Center, Pajooheshgah Baqiatallah University and Medical Sciences, Tehran, Iran
| | - Ali Akhavan
- Torabinegad Dental Research Center, Dental School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Soheil T. Tahani
- Department of Medical Sciences, Islamic Azad University, Najafabad Branch, Isfahan, Iran
- Isfahan Neurosciences Research Center, Al Zahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Bahramian
- Department of Anatomical Sciences, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vahid Goharian
- Department of Surgery, Amine Hospital, Isfahan, Iran
- Institute of Novin Tahlilgarane Nesfe-Jahan, Isfahan, Iran
| |
Collapse
|
42
|
Combellack EJ, Jessop ZM, Naderi N, Griffin M, Dobbs T, Ibrahim A, Evans S, Burnell S, Doak SH, Whitaker IS. Adipose regeneration and implications for breast reconstruction: update and the future. Gland Surg 2016; 5:227-41. [PMID: 27047789 PMCID: PMC4791352 DOI: 10.3978/j.issn.2227-684x.2016.01.01] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/17/2015] [Indexed: 12/20/2022]
Abstract
The evolution of breast reconstruction and management of breast cancer has evolved significantly since the earliest descriptions in the Edwin Smith Papyrus (3,000 BC). The development of surgical and scientific expertise has changed the way that women are managed, and plastic surgeons are now able to offer a wide range of reconstructive options to suit individual needs. Beyond the gold standard autologous flap based reconstructions, regenerative therapies promise the elimination of donor site morbidity whilst providing equivalent aesthetic and functional outcomes. Future research aims to address questions regarding ideal cell source, optimisation of scaffold composition and interaction of de novo adipose tissue in the microenvironment of breast cancer.
Collapse
|
43
|
Fotia C, Massa A, Boriani F, Baldini N, Granchi D. Prolonged exposure to hypoxic milieu improves the osteogenic potential of adipose derived stem cells. J Cell Biochem 2016; 116:1442-53. [PMID: 25648991 DOI: 10.1002/jcb.25106] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 01/27/2015] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cells (MSC) have been widely used in orthopedics for several applications. Conventionally, MSC are maintained under 21% O2 which does not reflect the real O2 tension in vivo. Recently, it was reported that different O2 conditions can give different cellular responses. Here, we investigated whether prolonged exposure to hypoxia affects the osteogenic differentiation of adipose-derived stem cells (ASC). ASC from six individuals were cultured under "low" (2-3%) or "air" (21%) oxygen tensions, either without or with osteogenic stimuli. The effect of the O2 tension was evaluated on cell proliferation, surface antigens, stemness and bone-related genes expression, alkaline phosphatase activity (ALP), mineralization activity, and release of osteogenic growth factors. Without differentiating stimuli, hypoxia favored ASC proliferation, reduced the number of CD184+ and CD34+ cells, and preserved the expression of NANOG and SOX2. The combination of hypoxia and osteogenic medium induced a high proliferation rate, a rapid and more pronounced mineralization activity, a higher expression of genes related to the MSC differentiation, a higher release of mitogenic growth factors (bFGF, PDGF-BB), and the decrease in TGF-β secretion, an inhibitor of the early stage of the osteoblast differentiation. We demonstrated that hypoxia acts dually, favoring ASC proliferation and the maintenance of the stemness in the absence of osteogenic stimuli, but inducing the differentiation in a bone-like microenvironment. In conclusion, prolonged cell culture in hypoxic microenvironment represents a proper method to modulate the stem cell function that may be used in several applications, for example, studies on bone pathophysiology or bone-tissue engineering.
Collapse
Affiliation(s)
- Caterina Fotia
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Annamaria Massa
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Filippo Boriani
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Nicola Baldini
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Rizzoli Orthopedic Institute, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Donatella Granchi
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Rizzoli Orthopedic Institute, Bologna, Italy
| |
Collapse
|
44
|
Guo X, Yu R, Xu Y, Lian R, Yu Y, Cui Z, Ji Q, Chen J, Li Z, Liu H, Chen J. PAC1R agonist maxadilan enhances hADSC viability and neural differentiation potential. J Cell Mol Med 2016; 20:874-90. [PMID: 26798992 PMCID: PMC4831362 DOI: 10.1111/jcmm.12772] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Accepted: 12/01/2015] [Indexed: 12/18/2022] Open
Abstract
Pituitary adenylate cyclase‐activating polypeptide (PACAP) is a structurally endogenous peptide with many biological roles. However, little is known about its presence or effects in human adipose‐derived stem cells (hADSCs). In this study, the expression of PACAP type I receptor (PAC1R) was first confirmed in hADSCs. Maxadilan, a specific agonist of PAC1R, could increase hADSC proliferation as determined by Cell Counting Kit‐8 and cell cycle analysis and promote migration as shown in wound‐healing assays. Maxadilan also showed anti‐apoptotic activity in hADSCs against serum withdrawal‐induced apoptosis based on Annexin V/propidium iodide analysis and mitochondrial membrane potential assays. The anti‐apoptotic effects of maxadilan correlated with the down‐regulation of Cleaved Caspase 3 and Caspase 9 as well as up‐regulation of Bcl‐2. The chemical neural differentiation potential could be enhanced by maxadilan as indicated through quantitative PCR, Western blot and cell morphology analysis. Moreover, cytokine neural redifferentiation of hADSCs treated with maxadilan acquired stronger neuron‐like functions with higher voltage‐dependent tetrodotoxin‐sensitive sodium currents, higher outward potassium currents and partial electrical impulses as determined using whole‐cell patch clamp recordings. Maxadilan up‐regulated the Wnt/β‐catenin signalling pathway associated with dimer‐dependent activity of PAC1R, promoting cell viability that was inhibited by XAV939, and it also activated the protein kinase A (PKA) signalling pathway associated with ligand‐dependent activity of PAC1R, enhancing cell viability and neural differentiation potential that was inhibited by H‐89. In summary, these results demonstrated that PAC1R is present in hADSCs, and maxadilan could enhance hADSC viability and neural differentiation potential in neural differentiation medium.
Collapse
Affiliation(s)
- Xiaoling Guo
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - Rongjie Yu
- Department of Cell Biology, Jinan University, Guangzhou, China
| | - Ying Xu
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Ruiling Lian
- Department of Ophthalmology, The First Clinical Medical College of Jinan University, Guangzhou, China
| | - Yankun Yu
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Zekai Cui
- Department of Cell Biology, Jinan University, Guangzhou, China
| | - Qingshan Ji
- Department of Ophthalmology, Affiliated Anhui Provincial Hospital of Anhui Medical University, Hefei, China
| | - Junhe Chen
- Department of Mathematics, South China University of Technology, Guangzhou, China
| | - Zhijie Li
- Eye Institute, Medical College of Jinan University, Guangzhou, China
| | - Hongwei Liu
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - Jiansu Chen
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China.,Department of Ophthalmology, The First Clinical Medical College of Jinan University, Guangzhou, China.,Eye Institute, Medical College of Jinan University, Guangzhou, China
| |
Collapse
|
45
|
Allogeneic Platelet Releasate Preparations Derived via a Novel Rapid Thrombin Activation Process Promote Rapid Growth and Increased BMP-2 and BMP-4 Expression in Human Adipose-Derived Stem Cells. Stem Cells Int 2015; 2016:7183734. [PMID: 26823671 PMCID: PMC4707349 DOI: 10.1155/2016/7183734] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/01/2015] [Indexed: 01/03/2023] Open
Abstract
The administration of human adipose-derived stem cells (ASCs) represents a promising regenerative therapy for the treatment of orthopedic injuries. While ASCs can be easily isolated from liposuction-derived adipose tissue, most clinical applications will likely require in vitro culture expansion of these cells using nonxenogeneic components. In this study, platelet releasate was generated using a novel rapid thrombin activation method (tPR). ASCs grown in media supplemented with tPR proliferated much faster than ASCs grown in media supplemented with 10% fetal bovine serum. The cells also retained the ability to differentiate along chondrogenic, adipogenic, and osteogenic lineages. The tPR cultured ASCs displayed elevated expression of BMP-4 (5.7 ± 0.97-fold increase) and BMP-2 (4.7 ± 1.3-fold increase) and decreased expression of PDGF-B (4.0 ± 1.4-fold decrease) and FGF-2 (33 ± 9.0-fold decrease). No significant changes in expression were seen with TGF-β and VEGF. This pattern of gene expression was consistent across different allogeneic tPR samples and different ASC lines. The use of allogeneic rapidly activated tPR to culture ASCs is associated with both an increased cell yield and a defined gene expression profile making it an attractive option for cell expansion prior to cell-based therapy for orthopedic applications.
Collapse
|
46
|
Potential Role of Activating Transcription Factor 5 during Osteogenesis. Stem Cells Int 2015; 2016:5282185. [PMID: 26770207 PMCID: PMC4684884 DOI: 10.1155/2016/5282185] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 07/30/2015] [Accepted: 08/02/2015] [Indexed: 01/22/2023] Open
Abstract
Human adipose-derived stem cells are an abundant population of stem cells readily isolated from human adipose tissue that can differentiate into connective tissue lineages including bone, cartilage, fat, and muscle. Activating transcription factor 5 is a transcription factor of the ATF/cAMP response element-binding protein (CREB) family. It is transcribed in two types of mRNAs (activating transcription factor 5 isoform 1 and activating transcription factor 5 isoform 2), encoding the same single 30-kDa protein. Although it is well demonstrated that it regulates the proliferation, differentiation, and apoptosis, little is known about its potential role in osteogenic differentiation. The aim of this study was to evaluate the expression levels of the two isoforms and protein during osteogenic differentiation of human adipose-derived stem cells. Our data indicate that activating transcription factor 5 is differentially expressed reaching a peak of expression at the stage of bone mineralization. These findings suggest that activating transcription factor 5 could play an interesting regulatory role during osteogenesis, which would provide a powerful tool to study bone physiology.
Collapse
|
47
|
Googe PB, Tidwell WJ, Rosenberg AE. Lipomatous metaplasia of superficial dermis. J Cutan Pathol 2015; 43:120-4. [DOI: 10.1111/cup.12631] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Revised: 08/15/2015] [Accepted: 08/27/2015] [Indexed: 01/03/2023]
Affiliation(s)
- Paul B. Googe
- Knoxville Dermatopathology Laboratory; Knoxville TN USA
- Department of Pathology; University of Tennessee Graduate School of Medicine; Knoxville TN USA
- Department of Pathology; Vanderbilt University; Nashville TN USA
| | - W. James Tidwell
- Division of Dermatology; University of Louisville; Louisville KY USA
| | | |
Collapse
|
48
|
Mattioli-Belmonte M, Teti G, Salvatore V, Focaroli S, Orciani M, Dicarlo M, Fini M, Orsini G, Di Primio R, Falconi M. Stem cell origin differently affects bone tissue engineering strategies. Front Physiol 2015; 6:266. [PMID: 26441682 PMCID: PMC4585109 DOI: 10.3389/fphys.2015.00266] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/09/2015] [Indexed: 12/22/2022] Open
Abstract
Bone tissue engineering approaches are encouraging for the improvement of conventional bone grafting technique drawbacks. Thanks to their self-renewal and multi-lineage differentiation ability, stem cells are one of the major actors in tissue engineering approaches, and among these adult mesenchymal stem cells (MSCs) hold a great promise for regenerative medicine strategies. Bone marrow MSCs (BM-MSCs) are the first- identified and well-recognized stem cell population used in bone tissue engineering. Nevertheless, several factors hamper BM-MSC clinical application and subsequently, new stem cell sources have been investigated for these purposes. The fruitful selection and combination of tissue engineered scaffold, progenitor cells, and physiologic signaling molecules allowed the surgeon to reconstruct the missing natural tissue. On the basis of these considerations, we analyzed the capability of two different scaffolds, planned for osteochondral tissue regeneration, to modulate differentiation of adult stem cells of dissimilar local sources (i.e., periodontal ligament, maxillary periosteum) as well as adipose-derived stem cells (ASCs), in view of possible craniofacial tissue engineering strategies. We demonstrated that cells are differently committed toward the osteoblastic phenotype and therefore, taking into account their specific features, they could be intriguing cell sources in different stem cell-based bone/periodontal tissue regeneration approaches.
Collapse
Affiliation(s)
- Monica Mattioli-Belmonte
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche Ancona, Italy
| | - Gabriella Teti
- Department of Biomedical and Neuromotor Sciences, University of Bologna Bologna, Italy
| | - Viviana Salvatore
- Department of Biomedical and Neuromotor Sciences, University of Bologna Bologna, Italy
| | - Stefano Focaroli
- Department of Biomedical and Neuromotor Sciences, University of Bologna Bologna, Italy
| | - Monia Orciani
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche Ancona, Italy
| | - Manuela Dicarlo
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche Ancona, Italy
| | - Milena Fini
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute Bologna, Italy
| | - Giovanna Orsini
- Department of Clinical Sciences and Stomatology, Università Politecnica delle Marche Ancona, Italy
| | - Roberto Di Primio
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche Ancona, Italy
| | - Mirella Falconi
- Department of Biomedical and Neuromotor Sciences, University of Bologna Bologna, Italy
| |
Collapse
|
49
|
Poloni A, Maurizi G, Mattiucci D, Busilacchi E, Mancini S, Discepoli G, Amici A, Falconi M, Cinti S, Leoni P. Biosafety evidence for human dedifferentiated adipocytes. J Cell Physiol 2015; 230:1525-1533. [PMID: 25641257 DOI: 10.1002/jcp.24898] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 12/16/2014] [Indexed: 01/10/2023]
Abstract
Mature adipocytes have shown dynamic plasticity to be converted into fibroblast-like and lipid-free cells. After the dedifferentiation process, these cells re-entered the cell cycle and acquired a high proliferation potential, becoming a valid source of stem cells. However, many aspects of the cellular biosafety about dedifferentiated fat cells remained unclear. This study aimed to elucidate their potential susceptibility to malignant transformation and to ascertain the safety of these cells for clinical use. To evaluate the genomic stability of dedifferentiated adipocytes, telomere length, hTERT gene transcription, the capacity of these cells to grow in an anchorage-independent manner and the presence of DNA damage by single cell gel electrophoresis assay were studied. Spontaneous chromosomal alterations were excluded by cytogenetic analysis and the expression level of c-myc and p53, tumor associated genes, were assessed, evaluating also p53 loss of function mutations. Despite the high proliferation capacity of dedifferentiated adipocytes, these cells showed stable telomere length compared with mature adipocytes, no hTERT transcriptions and consequently no telomerase activity, suggesting that both transformation and senescence were avoided. A constant expression level of c-myc and p53, the inability of dedifferentiated adipocytes to grow in an anchorage-independent manner, the absence of DNA damage suggested the safety of these cells. Moreover, a normal karyotype was preserved throughout the dedifferentiation process. Data in vivo showed that dedifferentiated adipocytes analyzed for tumorigenicity did not develop tumors. In conclusion, our data indicated that dedifferentiated adipocytes could be a relatively easily accessible resource for cell therapy and regenerative medicine.
Collapse
Affiliation(s)
- Antonella Poloni
- Clinica di Ematologia, Dipartimento Scienze Mediche e Chirurgiche, Università Politecnica delle Marche, Ancona, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Cai Z, Pan B, Jiang H, Zhang L. Chondrogenesis of Human Adipose-Derived Stem Cells by In Vivo Co-graft with Auricular Chondrocytes from Microtia. Aesthetic Plast Surg 2015; 39:431-9. [PMID: 25861768 DOI: 10.1007/s00266-015-0481-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 03/26/2015] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To evaluate the efficiency of chondrogenesis of human adipose-derived stem cells (ADSCs) induced by auricular chondrocytes from microtia via subcutaneous co-graft in nude mice. METHODS Human ADSCs and auricular chondrocytes were mixed at the ratio of 7:3 and suspended in 0.2 ml of Pluronic F-127 (5.0 × 10(7) cells/ml), and injected into Balb/c nude mice as the experimental group (Exp group). The same quantity of auricular chondrocytes (Ctr.1 group) or ADSCs (Ctr.2 group) in 0.2 ml of Pluronic F-127 was set as positive and negative control groups. The mixture of auricular chondrocytes (1.5 × 10(7) cells/ml) in 0.2 ml of Pluronic F-127 was set as the low concentration of chondrocyte control group (Ctr.3). At 8 weeks after grafting, the newly generated tissue pellets were isolated for morphological examination, haematoxylin and eosin staining, toluidine blue staining and safranin O staining of glycosaminoglycan (GAG), Masson's trichrome staining and immunohistochemical staining of type II collagen, and Verhoeff-iron-hematoxylin staining of elastic fibers. GAG content was determined by Alcian blue colorimetric method, and mRNA expression of type II collagen and aggrecan were examined by real-time PCR. RESULTS Cartilage-like tissue with a white translucent appearance and good elasticity was generated in the Exp and Ctr.1 groups. The tissue pellets in the Ctr.2 and Ctr.3 groups were much smaller than those in the Ctr.1 group. The mature cartilage lacunas could be observed in the Exp and Ctr.1 groups, while were rarely seen in the Ctr.3 group and not observed in the Ctr.2 group. The expression of cartilage-specific extracellular matrix such as type II collagen, GAG content, aggrecan, and elastic fibers in the Exp group was similar to that in the Ctr.1 group, whereas the expression of these extracellular matrix substances was significantly lower in the Ctr.2 and Ctr.3 groups (both P < 0.01). CONCLUSION Auricular chondrocytes from microtia can efficiently promote the chondrogenic differentiation and chondrogenesis of ADSCs by co-grafting in vivo. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors http://www.springer.com/00266 .
Collapse
Affiliation(s)
- Zhen Cai
- Department of Plastic Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, 610072, People's Republic of China
| | | | | | | |
Collapse
|