1
|
Uguten M, van der Sluis N, Vriend L, Coert JH, Harmsen MC, van der Lei B, van Dongen JA. Comparing mechanical and enzymatic isolation procedures to isolate adipose-derived stromal vascular fraction: A systematic review. Wound Repair Regen 2024. [PMID: 39444305 DOI: 10.1111/wrr.13228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/06/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
The stromal vascular fraction of adipose tissue has gained popularity as regenerative therapy for tissue repair. Both enzymatic and mechanical intraoperative SVF isolation procedures exist. To date, the quest for the preferred isolation procedure persists, due to the absence of standardised yield measurements and a defined clinical threshold. This systematic review is an update of the systematic review published in 2018, where guidelines were proposed to improve and standardise SVF isolation procedures. An elaborate data search in MEDLINE (PubMed), EMBASE (Ovid) and the Cochrane Central Register of Controlled Trials was conducted from September 2016 to date. A total of 26 full-text articles met inclusion criteria, evaluating 33 isolation procedures (11 enzymatic and 22 mechanical). In general, enzymatic and mechanical SVF isolation procedures yield comparable outcomes concerning cell yield (2.3-18.0 × 105 resp. 0.03-26.7 × 105 cells/ml), and cell viability (70%-99% resp. 46%-97.5%), while mechanical procedures are more time consuming (8-20 min vs. 50-210 min) and cost-efficient. However, as most studies used poorly validated outcome measures on SVF characterisation, it still remains unclear which intraoperative SVF isolation method is preferred. Future studies are recommended to implement standardised guidelines to standardise methods and improve comparability between studies.
Collapse
Affiliation(s)
- Mustafa Uguten
- Department of Plastic, Reconstructive and Hand Surgery, Medical Center Leeuwarden, Leeuwarden, The Netherlands
- Department of Plastic, Reconstructive and Hand Surgery, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Nanouk van der Sluis
- Department of Surgery, Erasmus University Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Linda Vriend
- Department of Pathology & Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - J H Coert
- Department of Plastic, Reconstructive and Hand Surgery, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Martin C Harmsen
- Department of Pathology & Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Berend van der Lei
- Department of Plastic Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Joris A van Dongen
- Department of Plastic, Reconstructive and Hand Surgery, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- Department of Pathology & Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
2
|
Moghtader F, Tabata Y, Karaöz E. Biohybrids for Combined Therapies of Skin Wounds: Agglomerates of Mesenchymal Stem Cells with Gelatin Hydrogel Beads Delivering Phages and Basic Fibroblast Growth Factor. Gels 2024; 10:493. [PMID: 39195022 DOI: 10.3390/gels10080493] [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: 05/31/2024] [Revised: 07/04/2024] [Accepted: 07/12/2024] [Indexed: 08/29/2024] Open
Abstract
There is great interest in developing effective therapies for the treatment of skin wounds accompanied by deep tissue losses and severe infections. We have attempted to prepare biohybrids formed of agglomerates of mesenchymal stem cells (MSCs) with gelatin hydrogel beads (GEL beads) delivering bacteriophages (phages) as antibacterial agents and/or basic fibroblast growth factor (bFGF) for faster and better healing, providing combined therapies for these types of skin wounds. The gelatin beads were produced through a two-step process using basic and/or acidic gelatins with different isoelectric points. Escherichia coli (E. coli) and its specific T4 phages were propagated. Phages and/or bFGF were loaded within the GELs and their release rates and modes were obtained. The phage release from the basic GEL beads was quite fast; in contrast, the bFGF release from the acidic GEL beads was sustained, as anticipated. MSCs were isolated from mouse adipose tissues and 2D-cultured. Agglomerates of these MSCs with GEL beads were formed and maturated in 3D cultures, and their time-dependent changes were followed. In these 3D culture experiments, it was observed that the agglomerates with GEL beads were very healthy and the MSCs formed tissue-like structures in 7 days, while the MSC agglomerates were not healthy and shrunk considerably as a result of cell death.
Collapse
Affiliation(s)
- Farzaneh Moghtader
- Nanobiyomedtek Biyomedikal ve Biyoteknoloji Sanayi ve Ticaret Limited Sirketi, Koycegiz 48800, Mugla, Turkey
- Laboratory of Biomaterials, Institute for Life and Medical Sciences, Department of Regeneration Science and Engineering, Kyoto University, Kyoto 606-850, Japan
- Institute of Health Sciences, Stem Cell and Tissue Engineering, Liv Hospital, İstinye University, Esenyurt, İstanbul 34517, Turkey
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Institute for Life and Medical Sciences, Department of Regeneration Science and Engineering, Kyoto University, Kyoto 606-850, Japan
| | - Erdal Karaöz
- Institute of Health Sciences, Stem Cell and Tissue Engineering, Liv Hospital, İstinye University, Esenyurt, İstanbul 34517, Turkey
| |
Collapse
|
3
|
Augustyniak K, Lesniak M, Latka H, Golan MP, Kubiak JZ, Zdanowski R, Malek K. Adipose-derived mesenchymal stem cells' adipogenesis chemistry analyzed by FTIR and Raman metrics. J Lipid Res 2024; 65:100573. [PMID: 38844049 PMCID: PMC11260339 DOI: 10.1016/j.jlr.2024.100573] [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: 03/22/2024] [Revised: 05/08/2024] [Accepted: 05/28/2024] [Indexed: 07/01/2024] Open
Abstract
The full understanding of molecular mechanisms of cell differentiation requires a holistic view. Here we combine label-free FTIR and Raman hyperspectral imaging with data mining to detect the molecular cell composition enabling noninvasive monitoring of cell differentiation and identifying biochemical heterogeneity. Mouse adipose-derived mesenchymal stem cells (AD-MSCs) undergoing adipogenesis were followed by Raman and FT-IR imaging, Oil Red, and immunofluorescence. A workflow of the data analysis (IRRSmetrics4stem) was designed to identify spectral predictors of adipogenesis and test machine-learning (ML) methods (hierarchical clustering, PCA, PLSR) for the control of the AD-MSCs differentiation degree. IRRSmetrics4stem provided insights into the chemism of adipogenesis. With single-cell tracking, we established IRRS metrics for lipids, proteins, and DNA variations during AD-MSCs differentiation. The over 90% predictive efficiency of the selected ML methods proved the high sensitivity of the IRRS metrics. Importantly, the IRRS metrics unequivocally recognize a switch from proliferation to differentiation. This study introduced a new bioassay identifying molecular markers indicating molecular transformations and delivering rapid and machine learning-based monitoring of adipogenesis that can be relevant to other differentiation processes. Thus, we introduce a novel, rapid, machine learning-based bioassay to identify molecular markers of adipogenesis. It can be relevant to identification of differentiation-related molecular processes in other cell types, and beyond the cell differentiation including progression of different cellular pathophysiologies reconstituted in vitro.
Collapse
Affiliation(s)
- Karolina Augustyniak
- Department of Chemical Physics, Faculty of Chemistry, Jagiellonian University in Krakow, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Krakow, Poland
| | - Monika Lesniak
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine - National Research Institute, Warszawa, Poland
| | - Hubert Latka
- Department of Chemical Physics, Faculty of Chemistry, Jagiellonian University in Krakow, Krakow, Poland
| | - Maciej P Golan
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine - National Research Institute, Warszawa, Poland; Institute of Psychology, The Maria Grzegorzewska University, Warsaw, Poland
| | - Jacek Z Kubiak
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine - National Research Institute, Warszawa, Poland; Dynamics and Mechanics of Epithelia Group, Institute of Genetics and Development of Rennes (IGDR), Faculty of Medicine, University of Rennes, CNRS, UMR 6290, Rennes, France.
| | - Robert Zdanowski
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine - National Research Institute, Warszawa, Poland.
| | - Kamilla Malek
- Department of Chemical Physics, Faculty of Chemistry, Jagiellonian University in Krakow, Krakow, Poland.
| |
Collapse
|
4
|
Chang CY, Aviña AE, Chang CJ, Lu LS, Chong YY, Ho TY, Yang TS. Exploring the biphasic dose-response effects of photobiomodulation on the viability, migration, and extracellular vesicle secretion of human adipose mesenchymal stem cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 256:112940. [PMID: 38776590 DOI: 10.1016/j.jphotobiol.2024.112940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
Photobiomodulation (PBM) is a well-established medical technology that employs diverse light sources like lasers or light-emitting diodes to generate diverse photochemical and photophysical reactions in cells, thereby producing beneficial clinical outcomes. In this study, we introduced an 830 nm near-infrared (NIR) laser irradiation system combined with a microscope objective to precisely and controllably investigate the impact of PBM on the migration and viability of human adipose mesenchymal stem cells (hADSCs). We observed a biphasic dose-response in hADSCs' viability and migration after PBM exposure (0-10 J/cm2), with the 5 J/cm2 group showing significantly higher cell viability and migration ability than other groups. Additionally, at the optimal dose of 5 J/cm2, we used nanoparticle tracking analysis (NTA) and found a 6.25-fold increase in the concentration of extracellular vesicles (EVs) derived from hADSCs (PBM/ADSC-EVs) compared to untreated cells (ADSC-EVs). Both PBM/ADSC-EVs and ADSC-EVs remained the same size, with an average diameter of 56 nm measured by the ExoView R200 system, which falls within the typical size range for exosomes. These findings demonstrate that PBM not only improves the viability and migration of hADSCs but also significantly increases the EV yield.
Collapse
Affiliation(s)
- Che-Yi Chang
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Ana Elena Aviña
- Graduate Institute of Biomedical Optomechatronics, Taipei Medical University, Taipei 110, Taiwan; Division of Plastic Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan; International PhD Program in Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Cheng-Jen Chang
- Graduate Institute of Biomedical Optomechatronics, Taipei Medical University, Taipei 110, Taiwan; Division of Plastic Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan; International PhD Program in Medicine, Taipei Medical University, Taipei 110, Taiwan; Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Long-Sheng Lu
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei 110, Taiwan; International PhD Program in Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan; Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110, Taiwan; Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Yi-Yong Chong
- Graduate Institute of Biomedical Optomechatronics, Taipei Medical University, Taipei 110, Taiwan
| | - Tzu Ying Ho
- Graduate Institute of Biomedical Optomechatronics, Taipei Medical University, Taipei 110, Taiwan
| | - Tzu-Sen Yang
- Graduate Institute of Biomedical Optomechatronics, Taipei Medical University, Taipei 110, Taiwan; International PhD Program in Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan; School of Dental Technology, Taipei Medical University, Taipei 110, Taiwan; Research Center of Biomedical Device, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
5
|
Adhikary P, Banerjee S, Dey BK, Gargari P, Chatterjee S, Chakraborty D, Chowdhury S. Association of adipocyte size and SREBP-1c in visceral and subcutaneous adipose tissue in non-obese type 2 diabetes mellitus. Endocrine 2024; 83:615-623. [PMID: 37733180 DOI: 10.1007/s12020-023-03535-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 09/12/2023] [Indexed: 09/22/2023]
Abstract
OBJECTIVE Non-obese type 2 diabetes seems to be common in India; hence the current study tried to understand the pathogenesis of diabetes in this group focusing on the role of adipocytes especially abdominal fat compartment. Comparison was made between non-obese subjects with newly detected diabetes and those without diabetes, in relation to levels of adipogenic factor and adipokines in pre-adipocytes and mature adipocytes respectively. RESEARCH DESIGN METHODS Non-obese subjects (BMI-18-25 Kg/m2) were consecutively selected of whom 15 had newly-detected, treatment naïve type 2 diabetes (HbA1% ≥6.5) while 25 were control (HbA1c% ≤5.6). We examined the expression of adipocyte differentiation factor - SREBP-1c from preadipocytes and adipocyte specific adipokines- HMW isoform and total adiponectin, leptin, FABP-4, TNF-α and IL-6 from adipocytesisolated from abdominal visceral and subcutaneous adipose tissues (VAT and SCAT) by RT-PCR and as well as from serum by ELISA. Size of cultured adipocytes was measured in a fully automated imaging system microscope. RESULT Both in SCAT and VAT, SREBP-1c and adiponectin had significantly lower expression along with increased mRNA level of inflammatory adipokinesdiabetes.Average adipocyte size and frequency of large(hypertrophied) adipocytes were comparatively higher in T2DM subjects and had significant negative correlation with SREBP-1c. HMW adiponectin level significantly reduced in the secretion from VAT and SCAT of T2DM subjects compared to control. CONCLUSION Reduced expression of SREBP-1c in preadipocytes may lead to increased number of hypertrophied adipocytes in T2DM. Therefore, these dysfunctional hypertrophied adipocytes could cause imbalanced expression of insulin resistant and insulin sensitive adipokines.
Collapse
Affiliation(s)
- Pieu Adhikary
- Department of Endocrinology & Metabolism, Institute of Post Graduate Medical Education & Research and Seth Sukhlal Karnani Memorial Hospital, 244, A.J.C. Bose Road, Kolkata, West Bengal, 700020, India
| | - Sudipta Banerjee
- Department of Endocrinology & Metabolism, Institute of Post Graduate Medical Education & Research and Seth Sukhlal Karnani Memorial Hospital, 244, A.J.C. Bose Road, Kolkata, West Bengal, 700020, India
| | - Bishal Kumar Dey
- Department of Endocrinology & Metabolism, Institute of Post Graduate Medical Education & Research and Seth Sukhlal Karnani Memorial Hospital, 244, A.J.C. Bose Road, Kolkata, West Bengal, 700020, India
| | - Piyas Gargari
- Department of Endocrinology & Metabolism, Institute of Post Graduate Medical Education & Research and Seth Sukhlal Karnani Memorial Hospital, 244, A.J.C. Bose Road, Kolkata, West Bengal, 700020, India
| | - Shamita Chatterjee
- Department of General Surgery, Institute of Post Graduate Medical Education & Research and Seth Sukhlal Karnani Memorial Hospital, 244, A.J.C. Bose Road, Kolkata, West Bengal, 700020, India
| | - Diya Chakraborty
- Department of Biochemistry, Ballygaunge Science College, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, India
| | - Subhankar Chowdhury
- Department of Endocrinology & Metabolism, Institute of Post Graduate Medical Education & Research and Seth Sukhlal Karnani Memorial Hospital, 244, A.J.C. Bose Road, Kolkata, West Bengal, 700020, India.
| |
Collapse
|
6
|
Lee CS, Lee M, Na K, Hwang HS. Stem Cell-Derived Extracellular Vesicles for Cancer Therapy and Tissue Engineering Applications. Mol Pharm 2023; 20:5278-5311. [PMID: 37867343 DOI: 10.1021/acs.molpharmaceut.3c00376] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Recently, stem cells and their secretomes have attracted great attention in biomedical applications, particularly extracellular vesicles (EVs). EVs are secretomes of cells for cell-to-cell communication. They play a role as intercellular messengers as they carry proteins, nucleic acids, lipids, and therapeutic agents. They have also been utilized as drug-delivery vehicles due to their biocompatibility, low immunogenicity, stability, targetability, and engineerable properties. The therapeutic potential of EVs can be further enhanced by surface engineering and modification using functional molecules such as aptamers, peptides, and antibodies. As a consequence, EVs hold great promise as effective delivery vehicles for enhancing treatment efficacy while avoiding side effects. Among various cell types that secrete EVs, stem cells are ideal sources of EVs because stem cells have unique properties such as self-renewal and regenerative potential for transplantation into damaged tissues that can facilitate their regeneration. However, challenges such as immune rejection and ethical considerations remain significant hurdles. Stem cell-derived EVs have been extensively explored as a cell-free approach that bypasses many challenges associated with cell-based therapy in cancer therapy and tissue regeneration. In this review, we summarize and discuss the current knowledge of various types of stem cells as a source of EVs, their engineering, and applications of EVs, focusing on cancer therapy and tissue engineering.
Collapse
Affiliation(s)
- Chung-Sung Lee
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Min Lee
- Division of Advanced Prosthodontics, University of California, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
| | - Kun Na
- Department of BioMedical-Chemical Engineering, The Catholic University of Korea, Bucheon 14662, Republic of Korea
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Hee Sook Hwang
- Department of Pharmaceutical Engineering, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
7
|
Mohd Nor NH, Mansor NI, Mohd Kashim MIA, Mokhtar MH, Mohd Hatta FA. From Teeth to Therapy: A Review of Therapeutic Potential within the Secretome of Stem Cells from Human Exfoliated Deciduous Teeth. Int J Mol Sci 2023; 24:11763. [PMID: 37511524 PMCID: PMC10380442 DOI: 10.3390/ijms241411763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Stem cells derived from human exfoliated deciduous teeth (SHED) have emerged as an alternative stem cell source for cell therapy and regenerative medicine because they are readily available, pose fewer ethical concerns, and have low immunogenicity and tumourigenicity. SHED offer a number of advantages over other dental stem cells, including a high proliferation rate with the potential to differentiate into multiple developmental lineages. The therapeutic effects of SHED are mediated by multiple mechanisms, including immunomodulation, angiogenesis, neurogenesis, osteogenesis, and adipogenesis. In recent years, there is ample evidence that the mechanism of action of SHED is mainly due to its paracrine action, releasing a wide range of soluble factors such as cytokines, chemokines, and trophic factors (also known as 'secretome') into the local tissue microenvironment to promote tissue survival and recovery. This review provides an overview of the secretome derived from SHED and highlights the bioactive molecules involved in tissue regeneration and their potential applications in regenerative medicine.
Collapse
Affiliation(s)
- Nurul Hafizah Mohd Nor
- Institute of Islamic Civilization, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor Darul Ehsan, Malaysia
| | - Nur Izzati Mansor
- Department of Nursing, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Kuala Lumpur, Malaysia
| | - Mohd Izhar Ariff Mohd Kashim
- Institute of Islamic Civilization, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor Darul Ehsan, Malaysia
- Faculty of Islamic Studies, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor Darul Ehsan, Malaysia
| | - Mohd Helmy Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Kuala Lumpur, Malaysia
| | - Farah Ayuni Mohd Hatta
- Institute of Islamic Civilization, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
8
|
Fu H, Dong S, Li K. Study on promoting the regeneration of grafted fat by cell-assisted lipotransfer. Regen Ther 2022; 22:7-18. [PMID: 36582606 PMCID: PMC9762074 DOI: 10.1016/j.reth.2022.11.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/10/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022] Open
Abstract
Background Cell-assisted lipotransfer (CAL), a modified adipose-derived stromal/stem cells (ADSCs)-based approach for autologous fat grafting that is an ideal option for soft tissue augmentation, has many shortcomings in terms of retention and adverse effects. The objective of our study was to improve the treatment efficacy of CAL by adding fibroblasts. Methods ADSCs and fibroblasts were isolated from human adipose and dermal tissues, with fibroblasts identified by immunofluorescence and ADSCs identified by the multilineage differentiation method. We performed cell proliferation, apoptosis, migration, adipogenic, and hemangioendothelial differentiation experiments, qPCR and Western blotting analysis in co-cultures of fibroblasts and ADSCs. Subsequently, we conducted animal experiments with BALB/c nude mice. Masson's staining, immunofluorescence staining and ultrasound were used to analyze the occurrence of adverse reactions of the grafted fat, and CT and three-dimensional reconstruction were used to accurately evaluate the volume of the grafted fat. Results We found that the co-culture of fibroblasts and ADSCs promoted their mutual proliferation, adipogenic differentiation, hemangioendothelial differentiation and proliferation and migration of HUVECs. Fibroblasts inhibit the apoptosis of ADSCs. Moreover, in animal experiments, the autografted adipose group combined with ADSCs and fibroblasts had the least occurrence of oily cysts, and fat had the best form of survival. Conclusions We enhanced adipocyte regeneration and angiogenesis in ADSCs and fibroblast cells after adding fibroblasts to conventional CAL autologous fat grafts. In turn, the volume retention rate of the grafted fat is improved, and the adverse reactions are reduced.
Collapse
Affiliation(s)
- Hongtao Fu
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Shanshan Dong
- Department of Medicine, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, China
| | - Kun Li
- Department of Emergency Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, NO. 161 Shaoshan South Road, Changsha 410004, Hunan, China,Corresponding author. The Affiliated Changsha Central Hospital, 161 Shaoshan South Road, Changsha 410004, China.
| |
Collapse
|
9
|
Pan LC, Hang NLT, Colley MM, Chang J, Hsiao YC, Lu LS, Li BS, Chang CJ, Yang TS. Single Cell Effects of Photobiomodulation on Mitochondrial Membrane Potential and Reactive Oxygen Species Production in Human Adipose Mesenchymal Stem Cells. Cells 2022; 11:cells11060972. [PMID: 35326423 PMCID: PMC8946980 DOI: 10.3390/cells11060972] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 12/10/2022] Open
Abstract
Photobiomodulation (PBM) has recently emerged in cellular therapy as a potent alternative in promoting cell proliferation, migration, and differentiation during tissue regeneration. Herein, a single-cell near-infrared (NIR) laser irradiation system (830 nm) and the image-based approaches were proposed for the investigation of the modulatory effects in mitochondrial membrane potential (ΔΨm), reactive oxygen species (ROS), and vesicle transport in single living human adipose mesenchymal stem cells (hADSCs). The irradiated-hADSCs were then stained with 2′,7′-dichlorodihydrofluorescein diacetate (H2DCFDA) and Rhodamine 123 (Rh123) to represent the ΔΨm and ROS production, respectively, with irradiation in the range of 2.5–10 (J/cm2), where time series of bright-field images were obtained to determine the vesicle transport phenomena. Present results showed that a fluence of 5 J/cm2 of PBM significantly enhanced the ΔΨm, ROS, and vesicle transport phenomena compared to the control group (0 J/cm2) after 30 min PBM treatment. These findings demonstrate the efficacy and use of PBM in regulating ΔΨm, ROS, and vesicle transport, which have potential in cell proliferation, migration, and differentiation in cell-based therapy.
Collapse
Affiliation(s)
- Li-Chern Pan
- Graduate Institute of Biomedical Optomechatronics, Taipei Medical University, Taipei 110, Taiwan; (L.-C.P.); (N.-L.-T.H.); (M.M.C.); (Y.-C.H.); (B.-S.L.)
| | - Nguyen-Le-Thanh Hang
- Graduate Institute of Biomedical Optomechatronics, Taipei Medical University, Taipei 110, Taiwan; (L.-C.P.); (N.-L.-T.H.); (M.M.C.); (Y.-C.H.); (B.-S.L.)
| | - Mamadi M.S Colley
- Graduate Institute of Biomedical Optomechatronics, Taipei Medical University, Taipei 110, Taiwan; (L.-C.P.); (N.-L.-T.H.); (M.M.C.); (Y.-C.H.); (B.-S.L.)
| | - Jungshan Chang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Yu-Cheng Hsiao
- Graduate Institute of Biomedical Optomechatronics, Taipei Medical University, Taipei 110, Taiwan; (L.-C.P.); (N.-L.-T.H.); (M.M.C.); (Y.-C.H.); (B.-S.L.)
| | - Long-Sheng Lu
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei 110, Taiwan;
- International Ph.D. Program in Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan
- Department of Medical Research, Taipei Medical University Hospital, Taipei 110, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
- Center for Cell Therapy, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan
| | - Bing-Sian Li
- Graduate Institute of Biomedical Optomechatronics, Taipei Medical University, Taipei 110, Taiwan; (L.-C.P.); (N.-L.-T.H.); (M.M.C.); (Y.-C.H.); (B.-S.L.)
| | - Cheng-Jen Chang
- Graduate Institute of Biomedical Optomechatronics, Taipei Medical University, Taipei 110, Taiwan; (L.-C.P.); (N.-L.-T.H.); (M.M.C.); (Y.-C.H.); (B.-S.L.)
- Department of Plastic Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Correspondence: (C.-J.C.); (T.-S.Y.); Tel.: +886-227-372-181 (ext. 3381) (C.-J.C.); +886-227-361-661 (ext. 5206) (T.-S.Y.)
| | - Tzu-Sen Yang
- Graduate Institute of Biomedical Optomechatronics, Taipei Medical University, Taipei 110, Taiwan; (L.-C.P.); (N.-L.-T.H.); (M.M.C.); (Y.-C.H.); (B.-S.L.)
- International Ph.D. Program in Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
- School of Dental Technology, Taipei Medical University, Taipei 110, Taiwan
- Research Center of Biomedical Device, Taipei Medical University, Taipei 110, Taiwan
- Correspondence: (C.-J.C.); (T.-S.Y.); Tel.: +886-227-372-181 (ext. 3381) (C.-J.C.); +886-227-361-661 (ext. 5206) (T.-S.Y.)
| |
Collapse
|
10
|
Rahmani-Moghadam E, Zarrin V, Mahmoodzadeh A, Owrang M, Talaei-Khozani T. Comparison of the Characteristics of Breast Milk-derived Stem Cells with the Stem Cells Derived from the Other Sources: A Comparative Review. Curr Stem Cell Res Ther 2021; 17:71-90. [PMID: 34161214 DOI: 10.2174/1574888x16666210622125309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/14/2021] [Accepted: 03/28/2021] [Indexed: 11/22/2022]
Abstract
Breast milk (BrM) not only supplies nutrition, but it also contains a diverse population of cells. It has been estimated that up to 6% of the cells in human milk possess the characteristics of mesenchymal stem cells (MSC). Available data also indicate that these cells are multipotent and capable of self-renewal and differentiation with other cells. In this review, we have compared different characteristics, such as CD markers, differentiation capacity, and morphology of stem cells, derived from human breast milk (hBr-MSC) with human bone marrow (hBMSC), Wharton's jelly (WJMSC), and human adipose tissue (hADMSC). Through the literature review, it was revealed that human breast milk-derived stem cells specifically express a group of cell surface markers, including CD14, CD31, CD45, and CD86. Importantly, a group of markers, CD13, CD29, CD44, CD105, CD106, CD146, and CD166, were identified, which were common in the four sources of stem cells. WJMSC, hBMSC, hADMSC, and hBr-MSC are potently able to differentiate into the mesoderm, ectoderm, and endoderm cell lineages. The ability of hBr-MSCs todifferentiate into the neural stem cells, neurons, adipocyte, hepatocyte, chondrocyte, osteocyte, and cardiomyocytes has made these cells a promising source of stem cells in regenerative medicine, while isolation of stem cells from the commonly used sources, such as bone marrow, requires invasive procedures. Although autologous breast milk-derived stem cells are an accessible source for women who are in the lactation period, breast milk can be considered as a source of stem cells with high differentiation potential without any ethical concern.
Collapse
Affiliation(s)
- Ebrahim Rahmani-Moghadam
- Department of Anatomical sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahideh Zarrin
- Laboratory for Stem Cell Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Mahmoodzadeh
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Marzieh Owrang
- Department of Anatomical sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tahereh Talaei-Khozani
- Department of Anatomical sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
11
|
Ghadimi D, Nielsen A, Hassan MFY, Fölster-Holst R, Ebsen M, Frahm SO, Röcken C, de Vrese M, Heller KJ. Modulation of Proinflammatory Bacteria- and Lipid-Coupled Intracellular Signaling Pathways in a Transwell Triple Co-Culture Model by Commensal Bifidobacterium Animalis R101-8. Antiinflamm Antiallergy Agents Med Chem 2021; 20:161-181. [PMID: 33135616 DOI: 10.2174/1871523019999201029115618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/22/2020] [Accepted: 09/30/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND AIMS Following a fat-rich diet, alterations in gut microbiota contribute to enhanced gut permeability, metabolic endotoxemia, and low grade inflammation-associated metabolic disorders. To better understand whether commensal bifidobacteria influence the expression of key metaflammation-related biomarkers (chemerin, MCP-1, PEDF) and modulate the pro-inflammatory bacteria- and lipid-coupled intracellular signaling pathways, we aimed at i) investigating the influence of the establishment of microbial signaling molecules-based cell-cell contacts on the involved intercellular communication between enterocytes, immune cells, and adipocytes, and ii) assessing their inflammatory mediators' expression profiles within an inflamed adipose tissue model. MATERIAL AND METHODS Bifidobacterium animalis R101-8 and Escherichia coli TG1, respectively, were added to the apical side of a triple co-culture model consisting of intestinal epithelial HT-29/B6 cell line, human monocyte-derived macrophage cells, and adipose-derived stem cell line in the absence or presence of LPS or palmitic acid. mRNA expression levels of key lipid metabolism genes HILPDA, MCP-1/CCL2, RARRES2, SCD, SFRP2 and TLR4 were determined using TaqMan qRT-PCR. Protein expression levels of cytokines (IL-1β, IL-6, and TNF-α), key metaflammation-related biomarkers including adipokines (chemerin and PEDF), chemokine (MCP- 1) as well as cellular triglycerides were assessed by cell-based ELISA, while those of p-ERK, p-JNK, p-p38, NF-κB, p-IκBα, pc-Fos, pc-Jun, and TLR4 were assessed by Western blotting. RESULTS B. animalis R101-8 inhibited LPS- and palmitic acid-induced protein expression of inflammatory cytokines IL-1β, IL-6, TNF-α concomitant with decreases in chemerin, MCP-1, PEDF, and cellular triglycerides, and blocked NF-kB and AP-1 activation pathway through inhibition of p- IκBα, pc-Jun, and pc-Fos phosphorylation. B. animalis R101-8 downregulated mRNA and protein levels of HILPDA, MCP-1/CCL2, RARRES2, SCD and SFRP2 and TLR4 following exposure to LPS and palmitic acid. CONCLUSION B. animalis R101-8 improves biomarkers of metaflammation through at least two molecular/signaling mechanisms triggered by pro-inflammatory bacteria/lipids. First, B. animalis R101-8 modulates the coupled intracellular signaling pathways via metabolizing saturated fatty acids and reducing available bioactive palmitic acid. Second, it inhibits NF-kB's and AP-1's transcriptional activities, resulting in the reduction of pro-inflammatory markers. Thus, the molecular basis may be formed by which commensal bifidobacteria improve intrinsic cellular tolerance against excess pro-inflammatory lipids and participate in homeostatic regulation of metabolic processes in vivo.
Collapse
Affiliation(s)
- Darab Ghadimi
- Department of Microbiology and Biotechnology, Max Rubner-Institute, Hermann-Weigmann-Str 1, D-24103 Kiel, Germany
| | - Annegret Nielsen
- Department of Microbiology and Biotechnology, Max Rubner-Institute, Hermann-Weigmann-Str 1, D-24103 Kiel, Germany
| | | | - Regina Fölster-Holst
- Clinic of Dermatology, University Hospital Schleswig-Holstein, Schittenhelmstr. 7, D-24105 Kiel, Germany
| | - Michael Ebsen
- Department of Pathology, Städtisches MVZ Kiel GmbH (Kiel City Hospital), Chemnitzstr.33, 24116 Kiel, Germany
| | - Sven Olaf Frahm
- Medizinisches Versorgungszentrum (MVZ), Pathology and Laboratory Medicine Dr. Rabenhorst, Prüner Gang 7, 24103 Kiel, Germany
| | - Christoph Röcken
- Institute of Pathology, Kiel University, University Hospital, Schleswig-Holstein, Arnold-Heller-Straße 3/14, D-24105 Kiel, Germany
| | - Michael de Vrese
- Department of Microbiology and Biotechnology, Max Rubner-Institute, Hermann-Weigmann-Str 1, D-24103 Kiel, Germany
| | - Knut J Heller
- Department of Microbiology and Biotechnology, Max Rubner-Institute, Hermann-Weigmann-Str 1, D-24103 Kiel, Germany
| |
Collapse
|
12
|
Fu G, Lu L, Pan Z, Fan A, Yin F. Adipose-derived stem cell exosomes facilitate rotator cuff repair by mediating tendon-derived stem cells. Regen Med 2021; 16:359-372. [PMID: 33871287 DOI: 10.2217/rme-2021-0004] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: To evaluate the potential capability of adipose-derived stem cell exosomes (ADSC-exos) on rotator cuff repair by mediating the tendon-derived stem cells (TDSCs) and explored the mechanism. Methods: First, we investigated the growth, survival and migration of TDSCs in the presence of ADSC-exos in vitro. Using a rat rotator cuff injury model to analyze the ability of the ADSC-exos to promote rotator cuff healing in vivo. Results: The hydrogel with ADSC-exos significantly improved the osteogenic and adipogenesis differentiation and enhanced the expression of RUNX2, Sox-9, TNMD, TNC and Scx and the mechanical properties of the articular portion. Conclusion: The ADSC-exos have the potential to promote the rotator cuff repair by mediating the TDSCs.
Collapse
Affiliation(s)
- Guojian Fu
- Department of Joint Surgery, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai, 200120, PR China.,Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University, Shanghai, 200120, PR China.,Department of Joint Surgery, Nanjing Jiangbei Hospital, Nantong University, Nanjing, 210048, PR China
| | - Liangyu Lu
- Department of Joint Surgery, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai, 200120, PR China.,Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University, Shanghai, 200120, PR China
| | - Zhangyi Pan
- Department of Joint Surgery, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai, 200120, PR China.,Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University, Shanghai, 200120, PR China
| | - Aoyuan Fan
- Department of Joint Surgery, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai, 200120, PR China.,Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University, Shanghai, 200120, PR China
| | - Feng Yin
- Department of Joint Surgery, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai, 200120, PR China.,Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University, Shanghai, 200120, PR China
| |
Collapse
|
13
|
Zhang X, Cai Z, Wu M, Huangfu X, Li J, Liu X. Adipose Stem Cell-Derived Exosomes Recover Impaired Matrix Metabolism of Torn Human Rotator Cuff Tendons by Maintaining Tissue Homeostasis. Am J Sports Med 2021; 49:899-908. [PMID: 33719604 DOI: 10.1177/0363546521992469] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Adipose stem cell-derived exosomes (ASC-Exos) are reported to effectively prevent muscle atrophy and degeneration of torn rat rotator cuff, but their influence on human samples and their potential mechanism are still unclear. PURPOSE We aimed to investigate the effects of ASC-Exos on the metabolic activities of torn human rotator cuff tendons and explore the potential mechanism behind it. STUDY DESIGN Controlled laboratory study. METHODS Diseased supraspinatus tendons were harvested from 15 patients with a mean ± SD age of 65.8 ± 3.2 years who underwent reverse shoulder arthroplasty for chronic rotator cuff tears associated with glenohumeral pathological changes. Each tendon was dissected into 3 × 4 × 4-mm explants: the ones derived from the same tendon were placed into 12-well plates and cultured in complete culture media (control) or in complete culture media supplemented with ASC-Exos for 72 hours. Afterward, the concentrations of cytokines secreted into the culture media-including interleukin 1β (IL-1β), IL-6, IL-8, and matrix metalloproteinase 9 (MMP-9)-were measured using enzyme-linked immunosorbent assay (ELISA). Tendons were stained with hematoxylin and eosin and immunohistochemistry (type I and III collagens) for histological analyses. Moreover, the expression of anabolic genes (TIMP-1 and TIMP-3; type I and III collagen encoding) and catabolic genes (MMP-9 and MMP-13) in tendons were measured using real-time quantitative polymerase chain reaction. Phosphorylated AMPKα and Wnt/β-catenin pathways were assayed by western blotting to explore the potential mechanism of action of ASC-Exos. RESULTS Secretion of proinflammatory cytokines, including IL-1β, IL-6, and MMP-9, was significantly reduced in the ASC-Exos group as compared with the control group. Supraspinatus tendons in the ASC-Exos group exhibited superior histological properties, as demonstrated by higher tendon maturing scores and more type I collagen content, but there was no significant difference in type III collagen content between groups. Expression of MMP-9 and MMP-13 genes was decreased in the ASC-Exos group versus the control group. Increased expression of type I and III collagens and an elevated type I/III ratio were found in the ASC-Exos group when compared with the control group. There was no significant difference in the secretion of IL-8 and expression of TIMP-1 and TIMP-3 genes between the ASC-Exos and control groups. Western blotting revealed that ASC-Exos enhanced phosphorylated AMPKα and decreased β-catenin levels to prevent tendon degeneration. CONCLUSION ASC-Exos maintained metabolic homeostasis of torn human rotator cuff tendons to improve their histological properties, which might be achieved by enhancing AMPK signaling to suppress Wnt/β-catenin activity. CLINICAL RELEVANCE ASC-Exos could be used as an effective biological tool to promote healing in torn human rotator cuff tendons.
Collapse
Affiliation(s)
- Xuancheng Zhang
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zhuochang Cai
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Minghu Wu
- Department of Orthopaedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqiao Huangfu
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Juehong Li
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xudong Liu
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
14
|
Valipour F, Valipour F, Rahbarghazi R, Navali AM, Rashidi MR, Davaran S. Novel hybrid polyester-polyacrylate hydrogels enriched with platelet-derived growth factor for chondrogenic differentiation of adipose-derived mesenchymal stem cells in vitro. J Biol Eng 2021; 15:6. [PMID: 33588910 PMCID: PMC7885552 DOI: 10.1186/s13036-021-00257-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/31/2021] [Indexed: 12/13/2022] Open
Abstract
Background The goal of the present study was to create a new biodegradable hybrid PCL-P (HEMA-NIPAAm) thermosensitive hydrogel scaffold by grafting PNIPAAm-based copolymers with biodegradable polyesters to promote the chondrogenic differentiation of human progenitor cells (adipose-derived stem cells-hASCs) in the presence of the platelet-derived growth factor (PDGF-BB). Different mixture ratios including 50 mmol ε-caprolactone and 10 mmol HEMA (S-1), 30 mmol ε-caprolactone and 10 mmol HEMA (S-2), 10 mmol ε-caprolactone and 30 mmol HEMA (S-3) were copolymerized followed by the addition of NIPAAm. Results A mild to moderate swelling and wettability rates were found in S-2 group copmpared to the S-1 ans S-3 samples. After 7 weeks, S-2 degradation rate reached ~ 43.78%. According to the LCST values, S-2, reaching 37 °C, was selected for different in vitro assays. SEM imaging showed nanoparticulate structure of the scaffold with particle size dimensions of about 62–85 nm. Compressive strength, Young’s modulus, and compressive strain (%) of S-2 were 44.8 MPa, 0.7 MPa, and 75.5%. An evaluation of total proteins showed that the scaffold had the potential to gradually release PDGF-BB. When hASCs were cultured on PCL-P (HEMA-NIPAAm) in the presence of PDGF-BB, the cells effectively attached and flattened on the scaffold surface for a period of at least 14 days, the longest time point evaluated, with increased cell viability rates as measured by performing an MTT assay (p < 0.05). Finally, a real-time RT-PCR analysis demonstrated that the combination of PCL-P (HEMA-NIPAAm) and PDGF-BB promoted the chondrogenesis of hASCs over a period of 14 days by up-regulating the expression of aggrecan, type-II collagen, SOX9, and integrin β1 compared with the non-treated control group (p < 0.05). Conclusion These results demonstrate that the PCL-P(HEMA-NIPAAm) hydrogel scaffold carrying PDGF-BB as a matrix for hASC cell seeding is a valuable system that may be used in the future as a three-dimensional construct for implantation in cartilage injuries.
Collapse
Affiliation(s)
- Fereshteh Valipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farzaneh Valipour
- Department of Molecular Biology, Faculty of Science, Hacettepe University, Ankara, Turkey
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Reza Rashidi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soodabeh Davaran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran. .,Applied Drug Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
15
|
Fazal N, Khawaja H, Naseer N, Khan AJ, Latief N. Daphne mucronata enhances cell proliferation and protects human adipose stem cells against monosodium iodoacetate induced oxidative stress in vitro. Adipocyte 2020; 9:495-508. [PMID: 32867575 PMCID: PMC7714443 DOI: 10.1080/21623945.2020.1812242] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/11/2020] [Accepted: 08/15/2020] [Indexed: 01/01/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are being used to treat many diseases as they exhibit great regenerative potential. However, MSC's transplantation sometimes does not yield the maximum regenerative outcome as they are unable to survive in inflammatory conditions. Several approaches including preconditioning are used to improve the survival rate of mesenchymal stem cells. One such recently reported approach is preconditioning MSCs with plant extracts. The present study was designed to evaluate the effect of Daphne mucronata extract on stressed human adipose-derived mesenchymal stem cells (hADMSCs). Isolated hADMSCs were preconditioned with different concentrations of Daphne muconata extract and the protective, proliferative, antioxidant and anti-inflammatory effect was assessed through various assays and expression analysis of inflammatory markers regulated through NF-κB pathway. Results suggest that preconditioning hADMSCs with Daphne mucronata increased the cell viability, proliferative and protective potential of hADMSCs with a concomitant reduction in LDH, ROS and elevation in SOD activity. Moreover, both the ELISA and gene expression analysis demonstrated down regulations of inflammatory markers (IL1-β, TNF-α, p65, p50, MMP13) in Daphne mucronata preconditioned hADMSCs as compared to stress. This is the first study to report the use of MIA induced oxidative stress against hADMSC's and effect of Daphne mucronata on stressed hADMSCs. Results of these studies provided evidence that Daphne mucronata protects the hADMSCs during stress conditions by down regulating the inflammatory markers and hence increase the viability and proliferative potential of hADMSCs that is crucial for transplantation purposes.
Collapse
Affiliation(s)
- Numan Fazal
- National Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Hamzah Khawaja
- National Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Nadia Naseer
- National Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Azim Jahangir Khan
- Allama Iqbal Medical College, University of Health Sciences, Lahore, Pakistan
| | - Noreen Latief
- National Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
16
|
Hong Y, Sun Y, Rong X, Li D, Lu Y, Ji Y. Exosomes from adipose-derived stem cells attenuate UVB-induced apoptosis, ROS, and the Ca 2+ level in HLEC cells. Exp Cell Res 2020; 396:112321. [PMID: 33045215 DOI: 10.1016/j.yexcr.2020.112321] [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: 08/10/2020] [Revised: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 12/22/2022]
Abstract
Cartilage acid protein 1 (CRTAC1) encodes a protein containing the Ca2+binding domain, which can promote apoptosis of human lens epithelial cells (HLECs) induced by ultraviolet B radiation. Exosomes secreted from adipose-derived stem cells (ASC-exo) have been used to treat many diseases, but the effect of ASC-exo on cataracts has not been established. We hypothesized that ASC-exo has a therapeutic effect on cataracts by regulating CRTAC1. We established the UVB-induced injured HLECs model to test the interactions between CRTAC1 and miR-10a-5p, and the effect on the Ca2+ level and reactive oxygen species (ROS) generation in apoptotic HLECs. We found that UVB significantly increased the level of CRTAC1 expression and induced HLEC apoptosis, while ASC-exo inhibited the induction of UVB and exosome inhibitor reduced the inhibition of ASC-exo. The qRT-PCR results showed that miR-10a-5p had a low level of expression in cataract lesions, whereas CRTAC1 was highly expressed. There was a negative correlation between the expression of CRTAC1 and miR-10a-5p. ASC-exo reversed UVB-inhibited miR-10a-5p expression and miR-10a-5p negatively regulated CRTAC1. In vitro data showed that miR-10a-5p reversed UVB-induced ROS, apoptosis, and the Ca2+ level in HLECs. Overexpression of CRTAC1 reversed the induction of ASC-exo in UVB-injured HLECs, and low expression of CRTAC1 reversed the induction of miR-10a-5p inhibitor. By upregulating the level of miR-10a-5p expression and downregulating the level of CRTAC1 expression, exosomes from ASCs attenuated UVB-induced apoptosis, ROS generation, and the Ca2+ level in HLECs. Our research provides novel insight into the treatment methods and associated mechanisms underlying cataracts.
Collapse
Affiliation(s)
- Yingying Hong
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, 200031, China
| | - Yang Sun
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, 200031, China
| | - Xianfang Rong
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, 200031, China
| | - Dan Li
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, 200031, China
| | - Yi Lu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, 200031, China
| | - Yinghong Ji
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, 200031, China.
| |
Collapse
|
17
|
Eslami A, Dehbashi M, Ashja-Arvan M, Salehi H, Azimzadeh M, Ganjalikhani-Hakemi M. Assessment of ability of human adipose derived stem cells for long term overexpression of IL-11 and IL-13 as therapeutic cytokines. Cytotechnology 2020; 72:773-784. [PMID: 32935166 PMCID: PMC7547926 DOI: 10.1007/s10616-020-00421-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/02/2020] [Indexed: 02/05/2023] Open
Abstract
Adipose-derived stem cells (ADSCs) are a type of mesenchymal stem cells with the therapeutic effects that make them one of the best sources for cell therapy. In this study, we aimed to assess the ability of human ADSCs for constant expression of IL-11 and IL-13, simultaneously. In this study, the characterized hADSCs were transduced with a lentiviral vector (PCDH-513B) containing IL-11 and IL-13 genes, and the ability of long-term expression of the transgenes was evaluated by ELISA technique on days 15, 45 and 75 after transduction. Our results indicated a high rate of transduction (more than 90%) in the isolated hADSCs. Our data showed the highest rate of expression on days 75 after transduction which was 242.67 pg/ml for IL-11 and 303.6 pg/ml for IL-13 compared with 35.2 pg/ml and 35.6 pg/ml in untreated cells, respectively (p = 0.001). Besides, MTT assay showed transduction of hADSCs with lentiviral viruses containing IL-11 and IL-13 had no adverse effect on hADSCs proliferation (p-value = 0.89). Finally, we successfully constructed a hADSC population stably overexpressing IL-11 as the neurotrophic cytokine and IL-13 as the anti-inflammatory cytokine and this transduced cells can be used for further studies in EAE mice model.
Collapse
Affiliation(s)
- Asma Eslami
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Moein Dehbashi
- Division of Genetics, Department of Cell and Molecular Biology, Faculty of Biological Sciences and Technologies, University of Isfahan, 81746-73441 Isfahan, Iran
| | - Mehnoosh Ashja-Arvan
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Salehi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Azimzadeh
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | |
Collapse
|
18
|
Teimourinejad A, Hashemibeni B, Salehi H, Mostafavi FS, Kazemi M, Bahramian H. Chondrogenic activity of two herbal products; pomegranate fruit extract and avocado/soybean unsaponifiable. Res Pharm Sci 2020; 15:358-366. [PMID: 33312214 PMCID: PMC7714020 DOI: 10.4103/1735-5362.293514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 06/16/2020] [Accepted: 08/22/2020] [Indexed: 01/22/2023] Open
Abstract
Background and purpose Articular cartilage defects aren't repaired by itself. Numerous studies have been conducted in the area of cartilage tissue engineering and some of them considered herbal products. An attempt was made in this study to compare the effects of pomegranate fruit extract (PFE), avocado/soybean unsaponifiable (ASU), and their equal proportional mixture on the chondrogenesis of human adipose-derived stem cells (hADSCs). Experimental approach PFE was prepared through the percolation method. ASU powder was dissolved in ethanol at 10 μg/mL concentration and was sterilized. The hADSCs first were isolated, expanded in monolayer culture and identified, and next seeded on fibrin scaffolds. The hADSCs/fibrin scaffolds were divided into 4 groups of control, ASU, PFE, and PFE+ ASU and subjected to in vitro induction for 2 weeks. The control group received chondrogenic medium, other groups received chondrogenic medium plus ASU, PFE, or PFE + ASU, respectively. The MTT assay was performed for cell viability evaluation, real-time polymerase chain reaction for expression of cartilage genes, and the toluidine blue, safranin-O, and immunohistochemistry for staining of the constructs. Findings / Results Cell viability, cartilage genes expression, matrix staining density, and collagen II protein levels in PFE samples were significantly higher than those of the other groups (P < 0.05). Histological assessments revealed more chondrogenic centers (P < 0.05) in the PFE group compared to the other groups. Conclusion and implications In this study, it was revealed that PFE can be considered as an induction factor for future chondrogenic studies.
Collapse
Affiliation(s)
- Ahmad Teimourinejad
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Batool Hashemibeni
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Hossein Salehi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Fatemeh Sadat Mostafavi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Mohammad Kazemi
- Genetic and Molecular Biology Department, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Hamid Bahramian
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| |
Collapse
|
19
|
Bucan A, Dhumale P, Jørgensen MG, Dalaei F, Wiinholt A, Hansen CR, Hvidsten S, Baun C, Hejbøl EK, Schrøder HD, Sørensen JA. Comparison between stromal vascular fraction and adipose derived stem cells in a mouse lymphedema model. J Plast Surg Hand Surg 2020; 54:302-311. [PMID: 32520635 DOI: 10.1080/2000656x.2020.1772799] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background: Lymphedema is one of the most common complications following breast cancer. Axillary lymph node dissection and radiotherapy are two well-known risk factors resulting in either removal or damage to the lymph nodes. As stem cells are known for their regenerative capabilities, they could theoretically repair/restore the damaged lymph vessels leading to a decrease in lymphedema.Methods: We evaluated the treatment of SVF and ASC on a mouse lymphedema model. Forty-five mice were allocated into three groups containing 15 mice each. The SVF group was injected with 100 μl containing 1 × 106 SVF, the ASC group with 100 μl ml containing 1 × 106 ASC and the NS with 100 μl ml of NS. Volumes of the mice were assessed weekly by μCT hindlimb volumetry for a total of 8 weeks. Lymph vessel morphometry was assessed by cross-sections of both hindlimbs stained for anti-LYVE1. Lymphatic function was assessed by lymphatic clearance.Results: The volume change between the groups was non-significant throughout all 8 weeks. The immunohistochemistry showed a statistically significant difference between the hindlimbs in ASC vs. NS group p = 0.032, 95% CI [-2121, -103].Conclusion: The volume of the hindlimbs showed that treatment with SVF or ASC yielded very similar results compared to the control group when assessed after 8 weeks. In week two the biggest difference between ASC and NS was seen but the difference diminished during the 8 weeks. The secondary outcomes showed that the lymph vessel lumen decreased when treated with ASC compared to the control group. Lymphoscintigraphy yielded non-significant results.
Collapse
Affiliation(s)
- Amar Bucan
- Research Unit for Plastic Surgery, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Pratibha Dhumale
- Department of Clinical Biochemistry and Pharmacology, University of Southern Denmark, Odense, Denmark
| | - Mads Gustaf Jørgensen
- Research Unit for Plastic Surgery, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Farima Dalaei
- Research Unit for Plastic Surgery, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Alexander Wiinholt
- Research Unit for Plastic Surgery, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Christian Rønn Hansen
- Laboratory of Radiation Physics, Odense University Hospital, Odense, Denmark.,Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Svend Hvidsten
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | - Christina Baun
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | | | | | - Jens Ahm Sørensen
- Research Unit for Plastic Surgery, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
20
|
Ashja-Arvan M, Dehbashi M, Eslami A, Salehi H, Yoosefi M, Ganjalikhani-Hakemi M. Impact of IFN-β and LIF overexpression on human adipose-derived stem cells properties. J Cell Physiol 2020; 235:8736-8746. [PMID: 32324266 DOI: 10.1002/jcp.29717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 03/11/2020] [Accepted: 04/05/2020] [Indexed: 02/05/2023]
Abstract
Adipose-derived stem cells (ADSCs) are a subset of mesenchymal stem cells that their therapeutic effects in various diseases make them an interesting tool in cell therapy. In the current study, we aimed to overexpress interferon-β (IFN-β) and leukemia inhibitory factor (LIF) cytokines in human ADSCs to evaluate the impact of this overexpression on human ADSCs properties. Here, we designed a construct containing IFN-β and LIF and then, transduced human adipose-derived stem cells (hADSCs) by this construct via a lentiviral vector (PCDH-513B). We assessed the ability of long-term expression of the transgene in transduced cells by western blot analysis and enzyme-linked immunosorbent assay techniques on Days 15, 45, and 75 after transduction. For the evaluation of stem cell properties, flow cytometry and differentiation assays were performed. Finally, the MTT assay was done to assess the proliferation of transduced cells compares to controls. Our results showed high-efficiency transduction with highest expression rates on Day 75 after transduction which were 70 pg/ml for IFN-β and 77.9 pg/ml for LIF in comparison with 25.60 pg/ml and 27.63 pg/ml, respectively, in untransduced cells (p = .0001). Also, transduced cells expressed a high level of ADSCs surface markers and successfully differentiated into adipocytes, chondrocytes, neural cells, and osteocytes besides the preservation rate of proliferation near untreated cells (p = .88). All in all, we successfully constructed an hADSC population stably overexpressed IFN-β and LIF cytokines. Considering the IFN-β and LIF anti-inflammatory and neuroprotective effects as well as immune-regulatory properties of hADSCs, the obtained cells of this study could be subjected for further evaluations in experimental autoimmune encephalomyelitis mice model.
Collapse
Affiliation(s)
- Mehnoosh Ashja-Arvan
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Moein Dehbashi
- Division of Genetics, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Asma Eslami
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Salehi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdiyeh Yoosefi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | | |
Collapse
|
21
|
Luo T, Yang X, Sun Y, Huang X, Zou L, Liu J. Effect of MicroRNA-20a on Osteogenic Differentiation of Human Adipose Tissue-Derived Stem Cells. Cells Tissues Organs 2020; 208:148-157. [PMID: 32097913 DOI: 10.1159/000506304] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 02/02/2020] [Indexed: 02/05/2023] Open
Abstract
Osteogenic differentiation of human adipose tissue-derived stem cells (hASCs) is a complex process that is regulated by multiple factors, including microRNAs (miRNAs). The miRNA miR-20a was shown to promote bone formation from bone marrow-derived mesenchymal stem cells. However, the role of miR-20a in osteogenic differentiation of hASCs remains unclear. In this study, we systematically evaluated the function of miR-20a in regulating hASC osteogenesis in vitro. hASCs were transduced with miR-20a-overexpressing and miR-20a-sponge lentiviral vectors, with green fluorescent protein (GFP) as a control. The results showed that miR-20a transcription was upregulated after hASC mineralization. Compared with the miR-20a-sponge, GFP, and hASC groups, the miR-20a-overexpressing group showed higher alkaline phosphatase (ALP) activity on days 7 and 14. Moreover, the mRNA level of ALP increased significantly in the miR-20a-overexpressing group on day 14. Furthermore, the protein of the target gene PPARγ was decreased, and the osteogenic differentiation-associated proteins ALP, osteocalcin, and RUNX2 were upregulated. hASCs anchored to HA/β-TCP revealed a healthy polygonal morphology and developed cytoplasmic extensions. miR-20a promoted osteogenic differentiation of the cell scaffold. Taken together, these data -confirm that miRNA-20a promotes the osteogenesis of hASCs in vitro, and its essential role in vivo needs further -investigation.
Collapse
Affiliation(s)
- Tao Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Stomatology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xueqin Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yan Sun
- Department of Conservative Dentistry and Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Xinqi Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Zou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China,
| | - Jun Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Lee SJ, Lee CR, Kim KJ, Ryu YH, Kim E, Han YN, Moon SH, Rhie JW. Optimal Condition of Isolation from an Adipose Tissue-Derived Stromal Vascular Fraction for the Development of Automated Systems. Tissue Eng Regen Med 2020; 17:203-208. [PMID: 31997256 DOI: 10.1007/s13770-019-00238-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/20/2019] [Accepted: 12/25/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The stromal vascular fraction (SVF) isolated from adipose tissue, which contains stem cells as well as other cell types, has been applied in various research fields. Although different enzymatic concentrations and treatment durations have been applied to isolate the SVF, optimal conditions have not been established. Thus, we aimed to establish the optimal conditions for isolation of the SVF from adipose tissue by automated systems. METHODS The SVF was collected from removed adipose tissues of five donors during surgery. The SVF was treated with 0.1% or 0.2% collagenase type I for 20, 40, or 60 min. Then, colony forming unit (CFU) assays and flow cytometry were performed to characterize the adipose stem cells (ASCs). A cytokine array was used to investigate the correlation between colony-formation ability and the secretion of isolated ASCs. RESULTS Treatment with 0.1% collagenase type I for 60 min resulted in a higher SVF yield, whereas treatment with 0.1% collagenase for 40 min resulted in higher CFU values. In addition, expression of interleukin (IL)-6, IL-8, and monocyte chemoattractant protein-1 in the SVF was higher in the high-CFU group than in the low-CFU group. CONCLUSION The optimal conditions for isolation of the SVF from adipose tissue were treatment with 0.1% collagenase type I for 40 min. We identified the conditions required for efficient SVF isolation based on high CFU values, and our results will facilitate the development of automated systems.
Collapse
Affiliation(s)
- Su Jin Lee
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Department of Plastic and Reconstructive Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Chae Rim Lee
- Department of Plastic and Reconstructive Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Ki Joo Kim
- Department of Plastic and Reconstructive Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Yeon Hee Ryu
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Department of Plastic and Reconstructive Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Eunjin Kim
- Department of Plastic and Reconstructive Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Yu Na Han
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Department of Plastic and Reconstructive Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Suk-Ho Moon
- Department of Plastic and Reconstructive Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Jong-Won Rhie
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea. .,Department of Plastic and Reconstructive Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| |
Collapse
|
23
|
Goudarzi N, Shabani R, Ebrahimi M, Baghestani A, Dehdashtian E, Vahabzadeh G, Soleimani M, Moradi F, Katebi M. Comparative phenotypic characterization of human colostrum and breast milk-derived stem cells. Hum Cell 2020; 33:308-317. [PMID: 31975030 DOI: 10.1007/s13577-019-00320-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/20/2019] [Indexed: 01/14/2023]
Abstract
There is a diverse population of stem cells in human breast milk that can be employed for therapeutic purposes as a reservoir of cells. The current study mainly aimed to determine the nature markers expressing on stem cells. For this aim, the expression of embryonic stem cell markers, as well as the expression of endothelial, mesenchymal, neural, and hematopoietic markers were evaluated by the flow cytometry analysis in fresh colostrum, breast milk, and cultured colostrum samples. The results showed that the embryonic (OCT4, SOX2, HLA-DR), hematopoietic (CD33, CD45, CD117), neural (CD133, Nestin), and mesenchymal (CD44, SCA1) stem cell markers present in colostrum had higher expression in comparison with their counterpart markers in fresh breast milk. The expression markers of stem cells in colostrum following a 2-week culture period were significantly increased compared with their counterpart markers in colostrum before the culture process. In the culture of breastmilk, cells were not observed adherent cells and colonies. Our findings form flow cytometry and cell culture suggest that the lactation stage could be one of the factors influencing the stem cell population and, consequently, the cultivation of breastmilk cells. The present study indicates that colostrum is a tremendous source of stem cells that could be applied in cell-based research.
Collapse
Affiliation(s)
- Nasim Goudarzi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, East Hemmat Highway, Tehran, Iran.,Department of Anatomy, Faculty of Medical Sciences, Iran University of Medical Sciences, East Hemmat Highway, Tehran, Iran
| | - Ronak Shabani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, East Hemmat Highway, Tehran, Iran.,Department of Anatomy, Faculty of Medical Sciences, Iran University of Medical Sciences, East Hemmat Highway, Tehran, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, End of North Banihashem, West Resalat Highway, Tehran, Iran
| | - Amir Baghestani
- Department of Anatomy, Scholar Medicine, University of Medical Sciences, East Hemmat Highway, Tehran, Iran
| | - Ehsan Dehdashtian
- Department of Anatomy, Scholar Medicine, University of Medical Sciences, East Hemmat Highway, Tehran, Iran
| | - Gelareh Vahabzadeh
- Department of Pharmacology, Faculty of Medical Sciences, Iran University of Medical Sciences, East Hemmat Highway, Tehran, Iran
| | - Mansoure Soleimani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, East Hemmat Highway, Tehran, Iran.,Department of Anatomy, Faculty of Medical Sciences, Iran University of Medical Sciences, East Hemmat Highway, Tehran, Iran
| | - Fatemeh Moradi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, East Hemmat Highway, Tehran, Iran. .,Department of Anatomy, Faculty of Medical Sciences, Iran University of Medical Sciences, East Hemmat Highway, Tehran, Iran.
| | - Majid Katebi
- Department of Anatomy, Faculty of Medical Science, Hormozgan University of Medical Sciences, Shahid Chamran Boulevard, Bandar Abbas, Hormozgan, Iran.
| |
Collapse
|
24
|
Wang C, Song W, Chen B, Liu X, He Y. Exosomes Isolated From Adipose-Derived Stem Cells: A New Cell-Free Approach to Prevent the Muscle Degeneration Associated With Torn Rotator Cuffs. Am J Sports Med 2019; 47:3247-3255. [PMID: 31560856 DOI: 10.1177/0363546519876323] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Fatty infiltration, inflammation, and apoptosis are common degenerative changes in patients with chronic rotator cuff tears that can lead to muscle atrophy and can even result in massive irreparable rotator cuff tears. Some data have demonstrated the proregenerative, anti-inflammatory, and anti-apoptotic properties of stem cell-derived exosomes in some orthopaedic disorders, but their effect on torn rotator cuff muscles has never been investigated. PURPOSE To study the effect of exosomes isolated from human adipose-derived stem cells (ASCs-Exos) on muscle degeneration, regeneration, and biomechanical properties in a rat model of a massive rotator cuff tear (MRCT). STUDY DESIGN Controlled laboratory study. METHODS A bilateral supraspinatus and infraspinatus tenotomy was performed on rats to create an MRCT model. Forty-two rats were randomly assigned to 3 groups: the sham surgery group, the saline group (lesions treated with a saline injection), and the ASCs-Exos group (lesions treated with an ASCs-Exos injection). Wet muscle weight, fatty infiltration, inflammation, vascularization, regeneration, and biomechanical properties were evaluated at 8 and 16 weeks after surgery. RESULTS The results revealed that the ASCs-Exos treatment could prevent the atrophy, fatty infiltration, inflammation, and vascularization of muscles in the MRCT model (P < .001). Additionally, the myofiber regeneration and biomechanical properties of ASCs-Exos-treated rotator cuffs were significantly elevated compared with those in the saline-treated group (P < .001). CONCLUSION This study demonstrates that ASCs-Exos can effectively decrease atrophy and degeneration and improve muscle regeneration and biomechanical properties in torn rotator cuff muscles. CLINICAL RELEVANCE ASCs-Exos can be used as a new cell-free approach to prevent the muscle degeneration associated with torn rotator cuffs and may be helpful to repair torn rotator cuffs. Nevertheless, further work needs to be done in a large animal model owing to the inherent regenerative potential possessed by rodents.
Collapse
Affiliation(s)
- Chongyang Wang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wei Song
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Bi Chen
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xudong Liu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yaohua He
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Orthopedics, Shanghai Sixth People's Hospital, Jinshan Branch, Shanghai, China
| |
Collapse
|
25
|
Pöttler M, Fliedner A, Bergmann J, Bui LK, Mühlberger M, Braun C, Graw M, Janko C, Friedrich O, Alexiou C, Lyer S. Magnetic Tissue Engineering of the Vocal Fold Using Superparamagnetic Iron Oxide Nanoparticles. Tissue Eng Part A 2019; 25:1470-1477. [DOI: 10.1089/ten.tea.2019.0009] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Marina Pöttler
- Section of Experimental Oncology and Nanomedicine, Head and Neck Surgery, Department of Otorhinolaryngology, Else Kröner-Fresenius-Foundation-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Anna Fliedner
- Section of Experimental Oncology and Nanomedicine, Head and Neck Surgery, Department of Otorhinolaryngology, Else Kröner-Fresenius-Foundation-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Julia Bergmann
- Section of Experimental Oncology and Nanomedicine, Head and Neck Surgery, Department of Otorhinolaryngology, Else Kröner-Fresenius-Foundation-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Linh Katrin Bui
- Section of Experimental Oncology and Nanomedicine, Head and Neck Surgery, Department of Otorhinolaryngology, Else Kröner-Fresenius-Foundation-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Marina Mühlberger
- Section of Experimental Oncology and Nanomedicine, Head and Neck Surgery, Department of Otorhinolaryngology, Else Kröner-Fresenius-Foundation-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christian Braun
- Institute of Legal Medicine, Ludwig-Maximilians-Universität München, Munchen, Germany
| | - Matthias Graw
- Institute of Legal Medicine, Ludwig-Maximilians-Universität München, Munchen, Germany
| | - Christina Janko
- Section of Experimental Oncology and Nanomedicine, Head and Neck Surgery, Department of Otorhinolaryngology, Else Kröner-Fresenius-Foundation-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Friedrich-Alexander Universität Erlangen–Nürnberg, Erlangen, Germany
| | - Christoph Alexiou
- Section of Experimental Oncology and Nanomedicine, Head and Neck Surgery, Department of Otorhinolaryngology, Else Kröner-Fresenius-Foundation-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Stefan Lyer
- Section of Experimental Oncology and Nanomedicine, Head and Neck Surgery, Department of Otorhinolaryngology, Else Kröner-Fresenius-Foundation-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
26
|
Izadyari Aghmiuni A, Heidari Keshel S, Sefat F, Akbarzadeh Khiyavi A. Quince seed mucilage-based scaffold as a smart biological substrate to mimic mechanobiological behavior of skin and promote fibroblasts proliferation and h-ASCs differentiation into keratinocytes. Int J Biol Macromol 2019; 142:668-679. [PMID: 31622718 DOI: 10.1016/j.ijbiomac.2019.10.008] [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: 08/18/2019] [Revised: 09/24/2019] [Accepted: 10/01/2019] [Indexed: 10/25/2022]
Abstract
The use of biological macromolecules like quince seed mucilage (QSM), as the common curative practice has a long history in traditional folk medicine to cure wounds and burns. However, this gel cannot be applied on exudative wounds because of the high water content and non-absorption of infection of open wounds. It also limits cell-to-cell interactions and leads to the slow wound healing process. In this study to overcome these problems, a novel QSM-based hybrid scaffold modified by PCL/PEG copolymer was designed and characterized. The properties of this scaffold (PCL/QSM/PEG) were also compared with four scaffolds of PCL/PEG, PCL/Chitosan/PEG, chitosan, and QSM, to assess the role of QSM and the combined effect of polymers in improving the function of skin tissue-engineered scaffolds. It was found, the physicochemical properties play a crucial role in regulating cell behaviors so that, PCL/QSM/PEG as a smart/stimuli-responsive bio-matrix promotes not only human-adipose stem cells (h-ASCs) adhesion but also supports fibroblasts growth, via providing a porous-network. PCL/QSM/PEG could also induce keratinocytes at a desirable level for wound healing, by increasing the mechanobiological signals. Immunocytochemistry analysis confirmed keratinocytes differentiation pattern and their normal phenotype on PCL/QSM/PEG. Our study demonstrates, QSM as a differentiation/growth-promoting biological factor can be a proper candidate for design of wound dressings and skin tissue-engineered substrates containing cell.
Collapse
Affiliation(s)
- Azadeh Izadyari Aghmiuni
- Department of Chemical Engineering, Ayatollah Amoli Branch, Islamic Azad University, Amol, Iran; Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran.
| | - Saeed Heidari Keshel
- Medical Nanotechnology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Farshid Sefat
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford, UK; Interdisciplinary Research Centre in Polymer Science & Technology (IRC Polymer), University of Bradford, Bradford, UK
| | | |
Collapse
|
27
|
Zhang H, Chen F, Liang ZH, Wu Y, Pi JS. Isolation, culture, and identification of duck intestinal epithelial cells and oxidative stress model constructed. In Vitro Cell Dev Biol Anim 2019; 55:733-740. [PMID: 31385166 DOI: 10.1007/s11626-019-00388-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/18/2019] [Indexed: 01/20/2023]
Abstract
Intestinal epithelial cells (IECs) not only have an absorption function but also act as a physical barrier between the body and the intestinal bacterial flora. Damage to IECs leads to the breakdown of this barrier and has negative effects on animal health. Intestinal epithelial damage is frequently associated with long-term acute stress, such as increased temperature and new stress management models. The intestinal epithelial damage caused by environmental stress has been linked to oxidative stress. Until now, the effects of intestinal epithelial antioxidant activity from feed additives and treatments could be tested in ducks only in vivo because of the lack of in vitro cell culture systems. In this study, we describe our protocol for the easy isolation and culture of IECs from the small intestine of duck embryos. Immunofluorescence was used for the cytological identification of IECs. In addition, IEC marker genes (IAP and CDH1) could also be detected in cultured cells. And cell status assessments were performed, and cell proliferation viability was analyzed by CCK-8 assay. Furthermore, we constructed an oxidative stress model to be used to research the oxidative stress response mechanism, and drugs acting on the cell signal transduction pathway. In conclusion, we have developed an effective and rapid protocol for obtaining duck primary IECs and constructed an oxidative stress model. These IECs exhibit features consistent with epithelial cells and could be used to explore the physiological mechanisms of oxidative stress ex vivo.
Collapse
Affiliation(s)
- Hao Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Hubei Academy of Agricultural sciences, Wuhan, 430064, China.
| | - Fang Chen
- Institute of Animal Husbandry and Veterinary Medicine, Hubei Academy of Agricultural sciences, Wuhan, 430064, China
| | - Zhen-Hua Liang
- Institute of Animal Husbandry and Veterinary Medicine, Hubei Academy of Agricultural sciences, Wuhan, 430064, China
| | - Yan Wu
- Institute of Animal Husbandry and Veterinary Medicine, Hubei Academy of Agricultural sciences, Wuhan, 430064, China
| | - Jin-Song Pi
- Institute of Animal Husbandry and Veterinary Medicine, Hubei Academy of Agricultural sciences, Wuhan, 430064, China
| |
Collapse
|
28
|
CRISPR/Cas9-Mediated BRCA1 Knockdown Adipose Stem Cells Promote Breast Cancer Progression. Plast Reconstr Surg 2019; 143:747-756. [PMID: 30817646 DOI: 10.1097/prs.0000000000005316] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND The tumor microenvironment within the breast is rich in adipose elements. The interaction between adipose cells and breast cancer is poorly understood, particularly as it pertains to patients with genetic susceptibility to breast cancer. This study focuses on the phenotype of human adipose-derived stem cells with the BRCA1 mutation and the effect they may have on breast cancer cell behavior. METHODS CRISPR/Cas9 was used to generate de novo BRCA1-knockdown human adipose-derived stem cells. The effect of the BRCA1 knockdown on the adipose-derived stem cell phenotype was compared to wild-type adipose-derived stem cells and patient-derived breast adipose-derived stem cells with known BRCA1 mutations. Interactions between adipose-derived stem cells and the MDA-MB-231 breast cancer cell line were evaluated. RESULTS BRCA1-knockdown adipose-derived stem cells stimulated MDA-MB-231 proliferation (1.4-fold increase on day 4; p = 0.0074) and invasion (2.3-fold increase on day 2; p = 0.0171) compared to wild-type cells. Immunofluorescence staining revealed higher levels of phosphorylated ataxia telangiectasia-mutated activation in BRCA1-knockdown cells (72.9 ± 5.32 percent versus 42.9 ± 4.97 percent; p = 0.0147), indicating higher levels of DNA damage. Beta-galactosidase staining demonstrated a significantly higher level of senescence in BRCA1-knockdown cells compared with wild-type cells (7.9 ± 0.25 percent versus 0.17 ± 0.17 percent; p < 0.0001). Using quantitative enzyme-linked immunosorbent assay to evaluate conditioned media, the authors found significantly higher levels of interleukin-8 in BRCA1-knockdown cells (2.57 ± 0.32-fold; p = 0.0049). CONCLUSIONS The authors show for the first time that the BRCA1 mutation affects the adipose-derived stem cell phenotype. Moreover, CRISPR/Cas9-generated BRCA1-knockdown adipose-derived stem cells stimulate a more aggressive behavior in breast cancer cells than wild-type adipose-derived stem cells. This appears to be related to increased inflammatory cytokine production by means of a DNA damage-mediated cell senescence pathway.
Collapse
|
29
|
Akdere ÖE, Shikhaliyeva İ, Gümüşderelioğlu M. Boron mediated 2D and 3D cultures of adipose derived mesenchymal stem cells. Cytotechnology 2019; 71:611-622. [PMID: 30905012 DOI: 10.1007/s10616-019-00310-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/19/2019] [Indexed: 01/17/2023] Open
Abstract
Boron (B), which is a beneficial bioactive element for human, has an increasing interest in tissue engineering for the last 5 years. However, the effective B concentration in cell culture is still unknown. The aim of the present study is to investigate in vitro osteogenic potential of mesenchymal stem cells, isolated from adipose tissue (AdMSCs), on boron containing 2D and 3D cell cultures. At first, the effects of B concentrations between 1 and 20 μg/mL were evaluated on the survival and osteogenic differentiation of AdMSCs cultured on 2D cell cultures. The 3D cultures were established by using chitosan (Ch) scaffolds prepared by freeze-drying and Ch scaffolds combined with hydroxyapatite (HAp) and B containing hydroxyapatite (B-HAp) that are produced by microwave-induced biomimetic method. The proliferation and osteogenic differentiation of AdMSCs on Ch, HAp/Ch and B-HAp/Ch scaffolds were investigated by in vitro cell culture studies. The results were evaluated with respect to cell viability, bone related ECM gene expressions, and cellular morphology. It was demonstrated that cellular functions of AdMSCs were enhanced by boron in both 2D and 3D cultures. Especially, B-HAp/Ch scaffolds, which have both osteoinductive and osteoconductive properties based on presence of B and HAp in its structure, promoted adhesion, proliferation and osteogenic differentiation of AdMSCs.
Collapse
Affiliation(s)
- Özge Ekin Akdere
- Department of Bioengineering, Hacettepe University, Ankara, Turkey
| | | | - Menemşe Gümüşderelioğlu
- Department of Bioengineering, Hacettepe University, Ankara, Turkey.
- Department of Chemical Engineering, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
30
|
SIRT1 reverses senescence via enhancing autophagy and attenuates oxidative stress-induced apoptosis through promoting p53 degradation. Int J Biol Macromol 2018; 117:225-234. [DOI: 10.1016/j.ijbiomac.2018.05.174] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 02/06/2023]
|
31
|
Liu Q, Chen F, Wang L, Zhang Y. [Research progress of the donor factors and experimental factors affecting adipogenic differentiation of adipose derived stem cells]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2017; 31:1390-1395. [PMID: 29798597 PMCID: PMC8632588 DOI: 10.7507/1002-1892.201704057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 08/31/2017] [Indexed: 11/03/2022]
Abstract
Objective To summarize the donor factors and experimental factors that affect adipogenic differentiation of adipose derived stem cells, so as to provide reference for adipogenic differentiation of adipose derived stem cells. Methods The related research literature about donor factors and experimental factors affecting adipogenic differentiation of adipose derived stem cells in recent years was extensively reviewed and summarized. Results There are a lot of donor factors and experimental factors affecting adipogenic differentiation of adipose derived stem cells, but some of the factors are still controversial, such as donor age, health status, adipose tissue of different parts, and so on. These factors need to be further studied. Conclusion The donor factors and experimental factors that affect adipogenic differentiation of adipose derived stem cells should be deeply studied and the controversial issues should be clarified to lay a solid foundation for the application of adipose derived stem cells in adipose tissue engineering.
Collapse
Affiliation(s)
- Qin Liu
- Department of Medical Experiments, Wuhan General Hospital of Chinese PLA, Wuhan Hubei, 430070, P.R.China
| | - Fang Chen
- Department of Medical Experiments, Wuhan General Hospital of Chinese PLA, Wuhan Hubei, 430070, P.R.China
| | - Liping Wang
- Department of Medical Experiments, Wuhan General Hospital of Chinese PLA, Wuhan Hubei, 430070, P.R.China
| | - Yi Zhang
- Department of Medical Experiments, Wuhan General Hospital of Chinese PLA, Wuhan Hubei, 430070,
| |
Collapse
|