1
|
Wang B, Pi Q, Mohsin A, Gao WQ, Guo M, Xu H. Exosomal miR-1246 of adipose stem cells attenuates obesity by polarizing M2 macrophages, reducing fat mass, and beiging of white adipose tissue. J Adv Res 2025:S2090-1232(25)00369-8. [PMID: 40419017 DOI: 10.1016/j.jare.2025.05.049] [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: 01/29/2025] [Revised: 03/28/2025] [Accepted: 05/23/2025] [Indexed: 05/28/2025] Open
Abstract
INTRODUCTION Adipose stem cells (ADSC) have demonstrated therapeutic potential in ameliorating obesity and metabolic disorders, with their exosomes showing comparable therapeutic effects. However, the underlying molecular mechanism remains incompletely understood. Furthermore, the limited availability and inherent heterogeneity of primary ADSC present substantial challenges for consistent therapeutic outcomes. OBJECTIVE This study aimed to investigate the molecular mechanism underlying the unique biological effects mediated by microRNAs (miRNAs) in exosomes derived from immortalized adipose stem cells (iADSC). METHODS AND RESULTS We first established stable iADSC and isolated their exosomes (iADSC-EXO), which exhibited both high yield and stability. In high fat diet (HFD)-fed obese mice, iADSC-EXO administration significantly attenuated obesity, reduced blood glucose and lipid levels, and alleviated hepatic steatosis. miRNA chips analysis revealed that miR-1246 is highly enriched in exosomes derived from iADSC and ADSC. Administration of miR-1246 to HFD-fed obese mice produced beneficial effects on obesity and metabolic disorders comparable to those with iADSC-EXO. Mechanistic studies revealed that miR-1246 exerts its beneficial effects through multiple pathways: First, reducing fat mass by downregulating of the expression of fat mass and obesity-associate protein (FTO) and subsequent inhibition of adipogenesis and lipogenesis. Second, enhancing white adipose tissue (WAT) beiging by suppressing Runt-related transcription factor 1 (RUNX1T1) and FTO expression. Third, promoting M2 macrophage polarization and suppressing WAT inflammation via inhibition of TNF receptor-associated factor 6 (TRAF6)-mediated inflammatory pathway. CONCLUSION Our study elucidates a novel molecular mechanism through which ADSC regulates adipose tissue homeostasis and metabolism and presents a potential therapeutic approach for treating obesity and related metabolic disorders via iADSC-EXO or miR-1246-based interventions.
Collapse
Affiliation(s)
- Bo Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, RenJi Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Qingmeng Pi
- Department of Plastic Surgery, RenJi Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Ali Mohsin
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Wei-Qiang Gao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, RenJi Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Meijin Guo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Huiming Xu
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, RenJi Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| |
Collapse
|
2
|
Wang B, Zhang G, Hu Y, Mohsin A, Chen Z, Hao W, Li Z, Gao WQ, Guo M, Xu H. Uncovering impaired mitochondrial and lysosomal function in adipose-derived stem cells from obese individuals with altered biological activity. Stem Cell Res Ther 2024; 15:12. [PMID: 38185703 PMCID: PMC10773039 DOI: 10.1186/s13287-023-03625-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 12/22/2023] [Indexed: 01/09/2024] Open
Abstract
BACKGROUND Adipose-derived stem cells (ADSCs) have been extensively used in preclinical and clinical trials for treating various diseases. However, the differences between ADSCs from lean individuals (L-ADSCs) and those from obese individuals (O-ADSCs) have not been thoroughly investigated, particularly regarding their mitochondrial and lysosomal functions. Therefore, this study aims to evaluate the differences between L-ADSCs and O-ADSCs in terms of cell biological activity, mitochondria, and lysosomes. METHODS We first isolated and cultured L-ADSCs and O-ADSCs. We then compared the differences between the two groups in terms of biological activity, including cell proliferation, differentiation potential, and their effect on the polarization of macrophages. Additionally, we observed the mitochondrial and lysosomal morphology of ADSCs using an electronic microscope, MitoTracker Red, and lysotracker Red dyes. We assessed mitochondrial function by examining mitochondrial membrane potential and membrane fluidity, antioxidative ability, and cell energy metabolism. Lysosomal function was evaluated by measuring autophagy and phagocytosis. Finally, we performed transcriptome analysis of the ADSCs using RNA sequencing. RESULTS The biological activities of O-ADSCs were decreased, including cell immunophenotypic profiles, cell proliferation, and differentiation potential. Furthermore, compared to L-ADSCs, O-ADSCs promoted M1-type macrophage polarization and inhibited M2-type macrophage polarization. Additionally, the mitochondrial morphology of O-ADSCs was altered, with the size of the cells becoming smaller and mitochondrial fragments increasing. O-ADSCs also exhibited decreased mitochondrial membrane potential and membrane fluidity, antioxidative ability, and energy metabolism. With respect to lysosomes, O-ADSCs contained ungraded materials in their lysosomes, enhanced lysosomal permeability, and reduced autophagy and phagocytosis ability. RNA sequence analysis indicated that the signalling pathways related to cell senescence, cancer, and inflammation were upregulated, whereas the signalling pathways associated with stemness, cell differentiation, metabolism, and response to stress and stimuli were downregulated. CONCLUSIONS This study indicates that ADSCs from individuals (BMI > 30 kg/m2) exhibit impaired mitochondrial and lysosomal function with decreased biological activity.
Collapse
Affiliation(s)
- Bo Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Rd., P.O. Box 329#, Shanghai, 200237, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, and Renji-MedX Clinical Stem Cell Research Center RenJi Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China
| | - Ge Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Rd., P.O. Box 329#, Shanghai, 200237, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, and Renji-MedX Clinical Stem Cell Research Center RenJi Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China
| | - Yuwen Hu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Rd., P.O. Box 329#, Shanghai, 200237, People's Republic of China
| | - Ali Mohsin
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Rd., P.O. Box 329#, Shanghai, 200237, People's Republic of China
| | - Zhimin Chen
- State Key Laboratory of Oncogenes and Related Genes, and Renji-MedX Clinical Stem Cell Research Center RenJi Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China
| | - Weijie Hao
- State Key Laboratory of Oncogenes and Related Genes, and Renji-MedX Clinical Stem Cell Research Center RenJi Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China
| | - Zhanxia Li
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Xuhui District, Shanghai, 200235, People's Republic of China.
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, and Renji-MedX Clinical Stem Cell Research Center RenJi Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China.
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, People's Republic of China.
| | - Meijin Guo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Rd., P.O. Box 329#, Shanghai, 200237, People's Republic of China.
| | - Huiming Xu
- State Key Laboratory of Oncogenes and Related Genes, and Renji-MedX Clinical Stem Cell Research Center RenJi Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China.
| |
Collapse
|
3
|
Shen X, Tang J, Huang Y, Lan X, Lei C, Chen H. CircRNF111 Contributes to Adipocyte Differentiation by Elevating PPARγ Expression via miR-27a-3p. Epigenetics 2023; 18:2145058. [PMID: 36377797 PMCID: PMC9980459 DOI: 10.1080/15592294.2022.2145058] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The content and distribution of adipocytes is an important factor that affects meat quality. Previous studies showed that circRNAs are involved in various physiological processes. Nevertheless, more research is needed to investigate the function of circRNAs in adipogenesis. The present study examines the effects of circRNF111 on adipogenesis of bovine preadipocyte and aims to elucidate the underlying molecular mechanisms. In our study, the sequence signature of circRNF111 was identified using bioinformatics, RNA-FISH, and sequencing. Mechanistically, knockdown or exogenous expression of circRNF111 in preadipocytes was done to prove the functional significance of circRNF111. Combined with bioinformatics, a dual fluorescein reporter system, and immunoprecipitation, the interaction between circRNF111, miR-27a-3p, and the target gene PPARγ was verified. The results reveal that circRNF111 is positively correlated with adipocyte differentiation. The newly identified bovine circRNF111 functions as a miR-27a-3p sponge to rescue the inhibitory effect of miR-27a-3p on the PPARγ gene, thereby promoting adipogenesis.
Collapse
Affiliation(s)
- Xuemei Shen
- Engineering Research Center of Sichuan Province Higher School of Local Chicken Breeds Industrialization in Southern Sichuan, College of Life Science, Leshan Normal University, Leshan, China,Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Jia Tang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Yongzhen Huang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xianyong Lan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Hong Chen
- College of Animal Science, Xinjiang Agricultural University, Urumqi, China,CONTACT Hong Chen College of Animal Science, Xinjiang Agricultural University, Urumqi, China
| |
Collapse
|
4
|
Kowalczuk A, Bourebaba N, Panchuk J, Marycz K, Bourebaba L. Calystegines Improve the Metabolic Activity of Human Adipose Derived Stromal Stem Cells (ASCs) under Hyperglycaemic Condition through the Reduction of Oxidative/ER Stress, Inflammation, and the Promotion of the AKT/PI3K/mTOR Pathway. Biomolecules 2022; 12:460. [PMID: 35327652 PMCID: PMC8946193 DOI: 10.3390/biom12030460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 02/06/2023] Open
Abstract
Hyperglycaemia and its resulting glucotoxicity are among the most prominent hallmarks of diabetes mellitus (DM) development. Persistent hyperglycaemia further leads to oxidative stress via mitochondrial dysfunction and subsequent ER stress onset, while associated hyperlipidaemia triggers the adipose tissue to secrete pro-inflammatory cytokines. In this study, the effect of calystegines has been investigated in an experimental model of hyperglycaemia induced on human ASCs cells. Different cellular pathways including apoptosis, oxidative and ER stress, inflammation as well as Pi3K/AKT/mTOR metabolic-associated axis have been evaluated by means on RT-qPCR, western blot, and flow cytometry techniques. Treatment of HuASCs cells with calystegines strongly promoted the hyperglycaemic cells survival and significantly diminished oxidative stress, mitochondrial dynamics failure and ER stress, while improving the endogenous cellular antioxidant defenses. Interestingly, nortropane alkaloids efficiently prevented the hyperglycaemia-mediated inflammatory response, as evidenced by the regulation of the pro- and anti-inflammatory response in HuASCs cells. Finally, we evidenced that calystegines may exert their protective effect on HuASCs cells metabolic functions through the restoration of the defective PI3K/AKT/mTOR pathway. Overall, the present investigation demonstrated that calystegines possess important abilities to protect HuASCs against hyperglycaemia-induced cellular dysfunction, and it evidenced that the observed effects are associated to the promotion of PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
| | - Nabila Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, 50-375 Wrocław, Poland; (N.B.); (J.P.); (K.M.)
| | - Juliia Panchuk
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, 50-375 Wrocław, Poland; (N.B.); (J.P.); (K.M.)
| | - Krzysztof Marycz
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, 50-375 Wrocław, Poland; (N.B.); (J.P.); (K.M.)
- International Institute of Translational Medicine, 55-114 Wisznia Mała, Poland
| | - Lynda Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, 50-375 Wrocław, Poland; (N.B.); (J.P.); (K.M.)
- International Institute of Translational Medicine, 55-114 Wisznia Mała, Poland
| |
Collapse
|
5
|
Porro S, Genchi VA, Cignarelli A, Natalicchio A, Laviola L, Giorgino F, Perrini S. Dysmetabolic adipose tissue in obesity: morphological and functional characteristics of adipose stem cells and mature adipocytes in healthy and unhealthy obese subjects. J Endocrinol Invest 2021; 44:921-941. [PMID: 33145726 DOI: 10.1007/s40618-020-01446-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022]
Abstract
The way by which subcutaneous adipose tissue (SAT) expands and undergoes remodeling by storing excess lipids through expansion of adipocytes (hypertrophy) or recruitment of new precursor cells (hyperplasia) impacts the risk of developing cardiometabolic and respiratory diseases. In unhealthy obese subjects, insulin resistance, type 2 diabetes, hypertension, and obstructive sleep apnoea are typically associated with pathologic SAT remodeling characterized by adipocyte hypertrophy, as well as chronic inflammation, hypoxia, increased visceral adipose tissue (VAT), and fatty liver. In contrast, metabolically healthy obese individuals are generally associated with SAT development characterized by the presence of smaller and numerous mature adipocytes, and a lower degree of VAT inflammation and ectopic fat accumulation. The remodeling of SAT and VAT is under genetic regulation and influenced by inherent depot-specific differences of adipose tissue-derived stem cells (ASCs). ASCs have multiple functions such as cell renewal, adipogenic capacity, and angiogenic properties, and secrete a variety of bioactive molecules involved in vascular and extracellular matrix remodeling. Understanding the mechanisms regulating the proliferative and adipogenic capacity of ASCs from SAT and VAT in response to excess calorie intake has become a focus of interest over recent decades. Here, we summarize current knowledge about the biological mechanisms able to foster or impair the recruitment and adipogenic differentiation of ASCs during SAT and VAT development, which regulate body fat distribution and favorable or unfavorable metabolic responses.
Collapse
Affiliation(s)
- S Porro
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - V A Genchi
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - A Cignarelli
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - A Natalicchio
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - L Laviola
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - F Giorgino
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy.
| | - S Perrini
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| |
Collapse
|
6
|
Increase in Leptin and PPAR-γ Gene Expression in Lipedema Adipocytes Differentiated in vitro from Adipose-Derived Stem Cells. Cells 2020; 9:cells9020430. [PMID: 32059474 PMCID: PMC7072543 DOI: 10.3390/cells9020430] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/23/2022] Open
Abstract
Lipedema is a painful loose connective tissue disorder characterized by a bilaterally symmetrical fat deposition in the lower extremities. The goal of this study was to characterize the adipose-derived stem cells (ASCs) of healthy and lipedema patients by the expression of stemness markers and the adipogenic and osteogenic differentiation potential. Forty patients, 20 healthy and 20 with lipedema, participated in this study. The stromal vascular fraction (SVF) was obtained from subcutaneous thigh (SVF-T) and abdomen (SVF-A) fat and plated for ASCs characterization. The data show a similar expression of mesenchymal markers, a significant increase in colonies (p < 0.05) and no change in the proliferation rate in ASCs isolated from the SVF-T or SVF-A of lipedema patients compared with healthy patients. The leptin gene expression was significantly increased in lipedema adipocytes differentiated from ASCs-T (p = 0.04) and the PPAR-γ expression was significantly increased in lipedema adipocytes differentiated from ASCs-A (p = 0.03) compared to the corresponding cells from healthy patients. No significant changes in the expression of genes associated with inflammation were detected in lipedema ASCs or differentiated adipocytes. These results suggest that lipedema ASCs isolated from SVF-T and SVF-A have a higher adipogenic differentiation potential compared to healthy ASCs.
Collapse
|
7
|
Ogrodnik M, Zhu Y, Langhi LGP, Tchkonia T, Krüger P, Fielder E, Victorelli S, Ruswhandi RA, Giorgadze N, Pirtskhalava T, Podgorni O, Enikolopov G, Johnson KO, Xu M, Inman C, Palmer AK, Schafer M, Weigl M, Ikeno Y, Burns TC, Passos JF, von Zglinicki T, Kirkland JL, Jurk D. Obesity-Induced Cellular Senescence Drives Anxiety and Impairs Neurogenesis. Cell Metab 2019; 29:1061-1077.e8. [PMID: 30612898 PMCID: PMC6509403 DOI: 10.1016/j.cmet.2018.12.008] [Citation(s) in RCA: 314] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 10/23/2018] [Accepted: 12/05/2018] [Indexed: 12/25/2022]
Abstract
Cellular senescence entails a stable cell-cycle arrest and a pro-inflammatory secretory phenotype, which contributes to aging and age-related diseases. Obesity is associated with increased senescent cell burden and neuropsychiatric disorders, including anxiety and depression. To investigate the role of senescence in obesity-related neuropsychiatric dysfunction, we used the INK-ATTAC mouse model, from which p16Ink4a-expressing senescent cells can be eliminated, and senolytic drugs dasatinib and quercetin. We found that obesity results in the accumulation of senescent glial cells in proximity to the lateral ventricle, a region in which adult neurogenesis occurs. Furthermore, senescent glial cells exhibit excessive fat deposits, a phenotype we termed "accumulation of lipids in senescence." Clearing senescent cells from high fat-fed or leptin receptor-deficient obese mice restored neurogenesis and alleviated anxiety-related behavior. Our study provides proof-of-concept evidence that senescent cells are major contributors to obesity-induced anxiety and that senolytics are a potential new therapeutic avenue for treating neuropsychiatric disorders.
Collapse
Affiliation(s)
- Mikolaj Ogrodnik
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK; Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Yi Zhu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Larissa G P Langhi
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Patrick Krüger
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Edward Fielder
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Stella Victorelli
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Rifqha A Ruswhandi
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Nino Giorgadze
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Tamar Pirtskhalava
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Oleg Podgorni
- Department of Anesthesiology, Stony Brook School of Medicine, 101 Nicolls Road, Stony Brook, New York, NY 11794, USA; Center for Developmental Genetics, Stony Brook University, 100 Nicolls Road, Stony Brook, New York, NY 11794, USA
| | - Grigori Enikolopov
- Department of Anesthesiology, Stony Brook School of Medicine, 101 Nicolls Road, Stony Brook, New York, NY 11794, USA; Center for Developmental Genetics, Stony Brook University, 100 Nicolls Road, Stony Brook, New York, NY 11794, USA; Department of Nano-, Bio-, Information Technology and Cognitive Science, Moscow Institute of Physics and Technology, Moscow, Russia; Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Kurt O Johnson
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Ming Xu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Christine Inman
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Allyson K Palmer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Marissa Schafer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Moritz Weigl
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Yuji Ikeno
- The Barshop Institute for Longevity and Aging Studies, San Antonio, Department of Pathology, The University of Texas Health Science Center at San Antonio, Research Service, Audie L. Murphy VA Hospital (STVHCS), San Antonio, TX 78229, USA
| | - Terry C Burns
- Departments of Neurologic Surgery and Neuroscience, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - João F Passos
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Thomas von Zglinicki
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK; Near East University, Arts and Sciences Faculty, Molecular Biology and Genetics, Nicosia, North Cyprus POB 99138 Mersin 10, Turkey
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| | - Diana Jurk
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK; Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
8
|
Silva KR, Baptista LS. Adipose-derived stromal/stem cells from different adipose depots in obesity development. World J Stem Cells 2019; 11:147-166. [PMID: 30949294 PMCID: PMC6441940 DOI: 10.4252/wjsc.v11.i3.147] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 01/27/2019] [Accepted: 02/28/2019] [Indexed: 02/06/2023] Open
Abstract
The increasing prevalence of obesity is alarming because it is a risk factor for cardiovascular and metabolic diseases (such as type 2 diabetes). The occurrence of these comorbidities in obese patients can arise from white adipose tissue (WAT) dysfunctions, which affect metabolism, insulin sensitivity and promote local and systemic inflammation. In mammals, WAT depots at different anatomical locations (subcutaneous, preperitoneal and visceral) are highly heterogeneous in their morpho-phenotypic profiles and contribute differently to homeostasis and obesity development, depending on their ability to trigger and modulate WAT inflammation. This heterogeneity is likely due to the differential behavior of cells from each depot. Numerous studies suggest that adipose-derived stem/stromal cells (ASC; referred to as adipose progenitor cells, in vivo) with depot-specific gene expression profiles and adipogenic and immunomodulatory potentials are keys for the establishment of the morpho-functional heterogeneity between WAT depots, as well as for the development of depot-specific responses to metabolic challenges. In this review, we discuss depot-specific ASC properties and how they can contribute to the pathophysiology of obesity and metabolic disorders, to provide guidance for researchers and clinicians in the development of ASC-based therapeutic approaches.
Collapse
Affiliation(s)
- Karina Ribeiro Silva
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology, Duque de Caxias, RJ 25250-020, Brazil
- Post-Graduation Program of Biotechnology, National Institute of Metrology, Quality and Technology, Duque de Caxias, RJ 25250-020, Brazil
| | - Leandra Santos Baptista
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology, Duque de Caxias, RJ 25250-020, Brazil
- Post-Graduation Program of Biotechnology, National Institute of Metrology, Quality and Technology, Duque de Caxias, RJ 25250-020, Brazil
- Multidisciplinary Center for Biological Research (Numpex-Bio), Federal University of Rio de Janeiro Campus Duque de Caxias, Duque de Caxias, RJ 25245-390, Brazil
| |
Collapse
|
9
|
Substrate elasticity regulates adipose-derived stromal cell differentiation towards osteogenesis and adipogenesis through β-catenin transduction. Acta Biomater 2018; 79:83-95. [PMID: 30134207 DOI: 10.1016/j.actbio.2018.08.018] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 08/03/2018] [Accepted: 08/17/2018] [Indexed: 02/05/2023]
Abstract
It is generally recognised that mesenchymal stem cells (MSCs) can differentiate into multiple lineages through guidance from the biophysical properties of the substrates. However, the precise biophysical mechanism that enables MSCs to respond to substrate properties remains unclear. In the current study, polydimethylsiloxane (PDMS) substrates with different stiffnesses were fabricated and the way in which the elastic modulus of the substrate regulated differentiation towards osteogenesis and adipogenesis in adipose-derived stromal cells (ASCs) was explored. Initially, a cell morphology change by SEM was observed between the stiff and soft substrates. The cytoskeleton stains including microfilament by F-actin and microtubule by α- and β-tubulin further showed a larger cell spreading area on the stiff substrate. Then the expression of vinculin, in charge for the linkage of adhesion molecules to the actin cytoskeleton, was enhanced on the stiff substrate. This change in focal adhesion plaque further triggered intracellular β-catenin signaling and promoted its nuclear translocation especially on the stiff substrate. The influence of β-catenin signaling on direct differentiation to osteogenic lineages was through direct binding between its downstream protein, Lef-1, and the osteogenic transcriptional factors, Runx2 and Osx, while on differentiation to adipogenic lineages was through modulating the expression of PPARγ. The imbalance of stiffness-induced β-catenin signaling finally induced a stronger osteogenesis and a weaker adipogenesis on the stiff substrate relative to those on the soft substrate. This study indicates the importance of stiffness on ASC differentiation and could help to increase understanding of the mechanism underlying molecular signal transduction from mechanosensing, mechanotransducing to stem cell differentiation. STATEMENT OF SIGNIFICANCE Mesenchymal stem cells can differentiate into multiple lineages, such as adipogenesis, myogenesis, neurogenesis, angiogenesis and osteogenesis, through influence of biophysical properties of the extracellular matrix. However, the precise bio-mechanism that triggers stem cell differentiation in response to matrix biophysical properties remains unclear. In the current study, we provide a series of experiments involving the characterization of cell morphology, microfilament, microtubule and adhesion capacity of adipose-derived stromal cells (ASCs) in response to substrate stiffness, and further elucidation of cytoplasmic β-catenin-dependent signal transduction, nuclear translocation and resultant promoter activation of transcriptional factors for osteogenesis and adipogenesis. This study provides an explanation on deeper understanding of bio-mechanism underlying substrate stiffness-triggered β-catenin signal transduction from active mechanosensing, mechanotransducing to stem cell differentiation.
Collapse
|
10
|
Sadie-Van Gijsen H. Adipocyte biology: It is time to upgrade to a new model. J Cell Physiol 2018; 234:2399-2425. [PMID: 30192004 DOI: 10.1002/jcp.27266] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 07/25/2018] [Indexed: 12/15/2022]
Abstract
Globally, the obesity pandemic is profoundly affecting quality of life and economic productivity, but efforts to address this, especially on a pharmacological level, have generally proven unsuccessful to date, serving as a stark demonstration that our understanding of adipocyte biology and pathophysiology is incomplete. To deliver better insight into adipocyte function and obesity, we need improved adipocyte models with a high degree of fidelity in representing the in vivo state and with a diverse range of experimental applications. Adipocyte cell lines, especially 3T3-L1 cells, have been used extensively over many years, but these are limited in terms of relevance and versatility. In this review, I propose that primary adipose-derived stromal/stem cells (ASCs) present a superior model with which to study adipocyte biology ex vivo. In particular, ASCs afford us the opportunity to study adipocytes from different, functionally distinct, adipose depots and to investigate, by means of in vivo/ex vivo studies, the effects of many different physiological and pathophysiological factors, such as age, body weight, hormonal status, diet and nutraceuticals, as well as disease and pharmacological treatments, on the biology of adipocytes and their precursors. This study will give an overview of the characteristics of ASCs and published studies utilizing ASCs, to highlight the areas where our knowledge is lacking. More comprehensive studies in primary ASCs will contribute to an improved understanding of adipose tissue, in healthy and dysfunctional states, which will enhance our efforts to more successfully manage and treat obesity.
Collapse
Affiliation(s)
- Hanél Sadie-Van Gijsen
- Division of Endocrinology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Parow, South Africa.,Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Parow, South Africa
| |
Collapse
|
11
|
Xie J, Zhang D, Ling Y, Yuan Q, Chenchen Z, Wei D, Zhou X. Substrate elasticity regulates vascular endothelial growth factor A (VEGFA) expression in adipose-derived stromal cells: Implications for potential angiogenesis. Colloids Surf B Biointerfaces 2018; 175:576-585. [PMID: 30580148 DOI: 10.1016/j.colsurfb.2018.08.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/22/2018] [Accepted: 08/16/2018] [Indexed: 02/05/2023]
Abstract
Adipose-derived stromal cells (ASCs) have potential in bioengineering angiogenesis due to their paracrine role in supporting endothelial tubulogenesis and vascular network formation. However, the precise mechanism of the inner angiogenic capacity of ASCs determined by the biophysical properties of the extracellular matrix needs to be further elucidated. In the current study, we fabricated two silicon-based elastomer polydimethylsiloxane (PDMS) substrates with different stiffnesses (stiff substrate, E = 195 kPa and soft substrate, E = 15 kPa) and found there were cytoskeletal changes in ASCs in response to different substrate stiffnesses. We then showed the expression of vinculin in focal adhesion plaques was enhanced and the nuclear translocation of β-catenin signaling was increased in ASCs on the stiff substrate relative to those on the soft substrate. We next used bioinformatics and found the downstream proteins of β-catenin signaling had binding sites in the promoter of vascular endothelial growth factor A (VEGFA), which is responsible for angiogenesis; then, we further confirmed the enhanced endogenous VEGFA expression in ASCs on the stiff substrate relative to that on the soft substrate. Finally, by using ectogenic VEGFA, we showed the stiff substrate could promote angiogenesis of ASCs in the form of more ring-like formations in 2D and vessel-like structure formations in 3D under VEGFA induction compared to that of the soft substrate. This study not only indicates the inner angiogenic capacity of ASCs but also elucidates the influence of substrate elasticity on ASC differentiation in bioengineering angiogenesis.
Collapse
Affiliation(s)
- Jing Xie
- State Key Laboratory of Oral Diseases, Endodontic Department West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, Endodontic Department West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ye Ling
- State Key Laboratory of Oral Diseases, Endodontic Department West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases, Endodontic Department West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhou Chenchen
- State Key Laboratory of Oral Diseases, Endodontic Department West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Du Wei
- State Key Laboratory of Oral Diseases, Endodontic Department West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, Endodontic Department West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
12
|
Altay C, Seçil M, Demir T, Atik T, Akıncı G, Özdemir Kutbay N, Keskin Temeloğlu E, Yıldırım Şimşir I, Özışık S, Demir L, Eren E, Tuna EB, Aytaç H, Onay H, Akıncı B. Determining residual adipose tissue characteristics with MRI in patients with various subtypes of lipodystrophy. Diagn Interv Radiol 2018; 23:428-434. [PMID: 29044029 DOI: 10.5152/dir.2017.17019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
PURPOSE We aimed to investigate residual adipose tissue with whole-body magnetic resonance imaging to differentiate between subtypes of lipodystrophy. METHODS A total of 32 patients (12 with congenital generalized lipodystrophy [CGL], 1 with acquired generalized lipodystrophy [AGL], 12 with familial partial lipodystrophy [FPLD], and 7 with acquired partial lipodystrophy [APL]) were included. RESULTS Despite generalized loss of metabolically active adipose tissue, patients with CGL1 caused by AGPAT2 mutations had a significant amount of residual adipose tissue in the scalp, earlobes, retro-orbital region, and palms and soles. No residual adipose tissue was noted particularly in the head and neck, palms and soles in CGL2 caused by BSCL2 mutations. CGL4 caused by mutations in the PTRF gene was characterized with well-preserved retro-orbital and bone marrow fat in the absence of any visible residual adipose tissue in other areas. No residual adipose tissue was observed in AGL. Despite loss of subcutaneous fat, periarticular adipose tissue was preserved in the lower limbs of patients with FPLD. Retro-orbital adipose tissue was surprisingly preserved in APL, although they lacked head and neck fat. CONCLUSION Lipodystrophies are a heterogeneous group of disorders characterized by generalized or partial loss of adipose tissue, which can be congenital or acquired. Our results suggest that residual adipose tissue characteristics can help distinguish different subtypes of lipodystrophy.
Collapse
Affiliation(s)
- Canan Altay
- Department of Radiology, Dokuz Eylül University School of Medicine, İzmir, Turkey.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Chitosan-assisted differentiation of porcine adipose tissue-derived stem cells into glucose-responsive insulin-secreting clusters. PLoS One 2017; 12:e0172922. [PMID: 28253305 PMCID: PMC5333835 DOI: 10.1371/journal.pone.0172922] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 02/11/2017] [Indexed: 12/25/2022] Open
Abstract
The unique advantage of easy access and abundance make the adipose-derived stem cells (ADSCs) a promising system of multipotent cells for transplantation and regenerative medicine. Among the available sources, porcine ADSCs (pADSCs) deserve especial attention due to the close resemblance of human and porcine physiology, as well as for the upcoming availability of humanized porcine models. Here, we report on the isolation and conversion of pADSCs into glucose-responsive insulin-secreting cells. We used the stromal-vascular fraction of the dorsal subcutaneous adipose from 9-day-old male piglets to isolate pADSCs, and subjected the cells to an induction scheme for differentiation on chitosan-coated plates. This one-step procedure promoted differentiation of pADSCs into pancreatic islet-like clusters (PILC) that are characterized by the expression of a repertoire of pancreatic proteins, including pancreatic and duodenal homeobox (Pdx-1), insulin gene enhancer protein (ISL-1) and insulin. Upon glucose challenge, these PILC secreted high amounts of insulin in a dose-dependent manner. Our data also suggest that chitosan plays roles not only to enhance the differentiation potential of pADSCs, but also to increase the glucose responsiveness of PILCs. Our novel approach is, therefore, of great potential for transplantation-based amelioration of type 1 diabetes.
Collapse
|
14
|
Rotondo F, Romero MDM, Ho-Palma AC, Remesar X, Fernández-López JA, Alemany M. Quantitative analysis of rat adipose tissue cell recovery, and non-fat cell volume, in primary cell cultures. PeerJ 2016; 4:e2725. [PMID: 27917316 PMCID: PMC5131620 DOI: 10.7717/peerj.2725] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/26/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND White adipose tissue (WAT) is a complex, diffuse, multifunctional organ which contains adipocytes, and a large proportion of fat, but also other cell types, active in defense, regeneration and signalling functions. Studies with adipocytes often require their isolation from WAT by breaking up the matrix of collagen fibres; however, it is unclear to what extent adipocyte number in primary cultures correlates with their number in intact WAT, since recovery and viability are often unknown. EXPERIMENTAL DESIGN Epididymal WAT of four young adult rats was used to isolate adipocytes with collagenase. Careful recording of lipid content of tissue, and all fraction volumes and weights, allowed us to trace the amount of initial WAT fat remaining in the cell preparation. Functionality was estimated by incubation with glucose and measurement of glucose uptake and lactate, glycerol and NEFA excretion rates up to 48 h. Non-adipocyte cells were also recovered and their sizes (and those of adipocytes) were measured. The presence of non-nucleated cells (erythrocytes) was also estimated. RESULTS Cell numbers and sizes were correlated from all fractions to intact WAT. Tracing the lipid content, the recovery of adipocytes in the final, metabolically active, preparation was in the range of 70-75%. Cells showed even higher metabolic activity in the second than in the first day of incubation. Adipocytes were 7%, erythrocytes 66% and other stromal (nucleated cells) 27% of total WAT cells. However, their overall volumes were 90%, 0.05%, and 0.2% of WAT. Non-fat volume of adipocytes was 1.3% of WAT. CONCLUSIONS The methodology presented here allows for a direct quantitative reference to the original tissue of studies using isolated cells. We have also found that the "live cell mass" of adipose tissue is very small: about 13 µL/g for adipocytes and 2 µL/g stromal, plus about 1 µL/g blood (the rats were killed by exsanguination). These data translate (with respect to the actual "live cytoplasm" size) into an extremely high metabolic activity, which make WAT an even more significant agent in the control of energy metabolism.
Collapse
Affiliation(s)
- Floriana Rotondo
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine, University of Barcelona, Barcelona, Spain
| | - María del Mar Romero
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine, University of Barcelona, Barcelona, Spain
- CIBER OBN, Barcelona, Spain
| | - Ana Cecilia Ho-Palma
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Xavier Remesar
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine, University of Barcelona, Barcelona, Spain
- CIBER OBN, Barcelona, Spain
| | - José Antonio Fernández-López
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine, University of Barcelona, Barcelona, Spain
- CIBER OBN, Barcelona, Spain
| | - Marià Alemany
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine, University of Barcelona, Barcelona, Spain
- CIBER OBN, Barcelona, Spain
| |
Collapse
|
15
|
Casado-Díaz A, Anter J, Müller S, Winter P, Quesada-Gómez JM, Dorado G. Transcriptomic Analyses of Adipocyte Differentiation From Human Mesenchymal Stromal-Cells (MSC). J Cell Physiol 2016; 232:771-784. [PMID: 27349923 DOI: 10.1002/jcp.25472] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 06/27/2016] [Indexed: 12/20/2022]
Abstract
Adipogenesis is a physiological process required for fat-tissue development, mainly involved in regulating the organism energetic-state. Abnormal distribution-changes and dysfunctions in such tissue are associated to different pathologies. Adipocytes are generated from progenitor cells, via a complex differentiating process not yet well understood. Therefore, we investigated differential mRNA and miRNA expression patterns of human mesenchymal stromal-cells (MSC) induced and not induced to differentiate into adipocytes by next (second)-generation sequencing. A total of 2,866 differentially expressed genes (101 encoding miRNA) were identified, with 705 (46 encoding miRNA) being upregulated in adipogenesis. They were related to different pathways, including PPARG, lipid, carbohydrate and energy metabolism, redox, membrane-organelle biosynthesis, and endocrine system. Downregulated genes were related to extracellular matrix and cell migration, proliferation, and differentiation. Analyses of mRNA-miRNA interaction showed that repressed miRNA-encoding genes can act downregulating PPARG-related genes; mostly the PPARG activator (PPARGC1A). Induced miRNA-encoding genes regulate downregulated genes related to TGFB1. These results shed new light to understand adipose-tissue differentiation and physiology, increasing our knowledge about pathologies like obesity, type-2 diabetes and osteoporosis. J. Cell. Physiol. 232: 771-784, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Antonio Casado-Díaz
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Jaouad Anter
- Dep. Genética, Universidad de Córdoba, Córdoba, Spain
| | | | | | - José Manuel Quesada-Gómez
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Gabriel Dorado
- Dep. Bioquímica y Biología Molecular, Campus de Rabanales C6-1-E17, Campus de Excelencia Internacional Agroalimentario (ceiA3), Universidad de Córdoba, Córdoba, Spain
| |
Collapse
|
16
|
Chen YJ, Liu HY, Chang YT, Cheng YH, Mersmann HJ, Kuo WH, Ding ST. Isolation and Differentiation of Adipose-Derived Stem Cells from Porcine Subcutaneous Adipose Tissues. J Vis Exp 2016:e53886. [PMID: 27077225 PMCID: PMC4841321 DOI: 10.3791/53886] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Obesity is an unconstrained worldwide epidemic. Unraveling molecular controls in adipose tissue development holds promise to treat obesity or diabetes. Although numerous immortalized adipogenic cell lines have been established, adipose-derived stem cells from the stromal vascular fraction of subcutaneous white adipose tissues provide a reliable cellular system ex vivo much closer to adipose development in vivo. Pig adipose-derived stem cells (pADSC) are isolated from 7- to 9-day old piglets. The dorsal white fat depot of porcine subcutaneous adipose tissues is sliced, minced and collagenase digested. These pADSC exhibit strong potential to differentiate into adipocytes. Moreover, the pADSC also possess multipotency, assessed by selective stem cell markers, to differentiate into various mesenchymal cell types including adipocytes, osteocytes, and chondrocytes. These pADSC can be used for clarification of molecular switches in regulating classical adipocyte differentiation or in direction to other mesenchymal cell types of mesodermal origin. Furthermore, extended lineages into cells of ectodermal and endodermal origin have recently been achieved. Therefore, pADSC derived in this protocol provide an abundant and assessable source of adult mesenchymal stem cells with full multipotency for studying adipose development and application to tissue engineering of regenerative medicine.
Collapse
Affiliation(s)
- Yu-Jen Chen
- Institute of Biotechnology, National Taiwan University; Department of Animal Science and Technology, National Taiwan University
| | - Hui-Yu Liu
- Department of Animal Science and Technology, National Taiwan University
| | - Yun-Tsui Chang
- Department of Animal Science and Technology, National Taiwan University
| | - Ying-Hung Cheng
- Department of Animal Science and Technology, National Taiwan University
| | - Harry J Mersmann
- Department of Animal Science and Technology, National Taiwan University
| | - Wen-Hung Kuo
- Department of Surgery, National Taiwan University Hospital and College of Medicine
| | - Shih-Torng Ding
- Institute of Biotechnology, National Taiwan University; Department of Animal Science and Technology, National Taiwan University;
| |
Collapse
|
17
|
Xie Q, Xie J, Zhong J, Cun X, Lin S, Lin Y, Cai X. Hypoxia enhances angiogenesis in an adipose-derived stromal cell/endothelial cell co-culture 3D gel model. Cell Prolif 2016; 49:236-45. [PMID: 26997164 DOI: 10.1111/cpr.12244] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 12/19/2015] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES This study aimed to investigate the influence of hypoxia on angiogenesis in a 3D gel, with co-culturing adipose-derived stromal cells (ASCs) and endothelial cells (ECs). MATERIALS AND METHODS ASCs from green fluorescent protein-labeled mice and ECs from red fluorescent protein-labeled mice were co-cultured in 3D collagen gels at 1:1 ratio, in normal and hypoxic oxygen conditions, and morphology of angiogenesis was observed using confocal laser scanning microscopy. To discover changes in growth factors between monoculture ASCs and ECs, transwell co-cultures of ASCs and ECs were applied. Semi-quantitative PCR was performed to explore mRNA expression of growth factors. RESULTS Enhanced angiogenesis was observed in 3D gels implanted with 1:1 mixture of ASCs and ECs after 7 days hypoxia. Genes including VEGFA/B, EGF-1, HIF-1a, IGF-1, PDGF, TGF-β1 and BMP-2/4 in ECs, both monoculture and co-culture, were significantly enhanced after being cultured under hypoxia. In comparison, genes VEGFA/B, EGF-1, HIF-1a, TGF-β1 and BMP-2 in ASCs increased. In all, factors VEGFA/B, EGF-1, HIF-1a, TGF-β1 and BMP-2 increased in both ASCs and ECs after being cultured in hypoxia no matter whether as monoculture or co-culture. CONCLUSIONS Co-culture of ASCs and ECs at 1:1 ratio in a 3D gel under hypoxia promoted angiogenesis. Those growth factors which were increased in both ASCs and ECs, indicate that VEGFA/B, EGF-1, HIF-1a, TGF-β1 and BMP-2 might be responsible for enhancement in angiogenesis triggered by hypoxia.
Collapse
Affiliation(s)
- Qiang Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Juan Zhong
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Xiangzhu Cun
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Shiyu Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, 610041, China
| |
Collapse
|
18
|
Li C, Zhou L. Inhibitory effect 6-gingerol on adipogenesis through activation of the Wnt/β-catenin signaling pathway in 3T3-L1 adipocytes. Toxicol In Vitro 2015; 30:394-401. [PMID: 26498061 DOI: 10.1016/j.tiv.2015.09.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 09/19/2015] [Accepted: 09/24/2015] [Indexed: 11/26/2022]
Abstract
6-Gingerol has been reported to inhibit adipogenesis and lipid content accumulation. However, the mechanism of its anti-adipogenic effect remains unclear. Our aim is to investigate the molecular mechanism of the anti-adipogenic effect of 6-gingerol. The lipid content in adipocytes was measured by Oil Red O staining and cell viability was analyzed by MTT assay. The extent of suppression of differentiation by 6-gingerol was characterized by measuring the triglyceride content and GPDH activity. The regulation of adipogenic markers and the components of the Wnt/β-catenin pathway were analyzed by real-time PCR and Western blotting. The nuclear location of β-catenin was identified using immunofluorescence assay. Small interfering RNA transfection was conducted to elucidate the crucial role of β-catenin in anti-adipogenic effect of 6-gingerol. Our results showed that 6-gingerol inhibited the adipogenesis and lowered the mRNA expression levels of transcription factors and the key lipogenic enzymes in 3T3-L1 cells. The effect of 6-gingerol on adipogenic differentiation was accompanied by stimulating the activation of the Wnt/β-catenin signaling. In addition, we found that 6-gingerol induced phosphorylations of glycogen synthase kinase-3β(GSK-3β), and promoted the nuclear accumulation of β-catenin. Importantly, the inhibitory effect of 6-gingerol on adipogenic differentiation was reversed after the siRNA knockdown of β-catenin was added. Our findings demonstrated that 6-gingerol inhibits the adipogenic differentiation of 3T3-L1 cells through activating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Chunbo Li
- Department of Gynaecology and Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, China.
| | - Lin Zhou
- Department of urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
19
|
Lee HY, Lee HL, Yun Y, Kim JS, Ha Y, Yoon DH, Lee SH, Shin DA. Human Adipose Stem Cells Improve Mechanical Allodynia and Enhance Functional Recovery in a Rat Model of Neuropathic Pain. Tissue Eng Part A 2015; 21:2044-52. [DOI: 10.1089/ten.tea.2014.0713] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Hye Yeong Lee
- Department of Neurosurgery, Spine & Spinal Cord Institute, Yonsei University, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Hye-Lan Lee
- Department of Neurosurgery, Spine & Spinal Cord Institute, Yonsei University, Seoul, Republic of Korea
| | - Yeomin Yun
- Department of Neurosurgery, Spine & Spinal Cord Institute, Yonsei University, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Jin-Su Kim
- Department of Biomedical Science, CHA University, Seongnam, Republic of Korea
| | - Yoon Ha
- Department of Neurosurgery, Spine & Spinal Cord Institute, Yonsei University, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Do Heum Yoon
- Department of Neurosurgery, Spine & Spinal Cord Institute, Yonsei University, Seoul, Republic of Korea
| | - Soo-Hong Lee
- Department of Biomedical Science, CHA University, Seongnam, Republic of Korea
| | - Dong Ah Shin
- Department of Neurosurgery, Spine & Spinal Cord Institute, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
20
|
Cun X, Xie J, Lin S, Fu N, Deng S, Xie Q, Zhong J, Lin Y. Gene profile of soluble growth factors involved in angiogenesis, in an adipose-derived stromal cell/endothelial cell co-culture, 3D gel model. Cell Prolif 2015; 48:405-12. [PMID: 26037311 DOI: 10.1111/cpr.12193] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 03/05/2015] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES The aim of this study was to investigate gene expressions of growth factors for angiogenesis, in a three-dimensional (3D) gel populated with adipose-derived stromal cells (ASCs) and endothelial cells (ECs) in co-culture. MATERIALS AND METHODS The 3D gel, mixed with green fluorescent protein (GFP)-positive ASCs and DsRed-Express-positive ECs, 1:1 ratio, was established in vitro. The phenomenon of angiogenesis was observed using confocal microscopy. To detect gene expressions for growth factor proteins in both ASCs and ECs, transwell co-culture was used, and cell lysate samples were collected at 1, 3, 5 and 7 days. Semi-quantitative polymerase chain reaction (PCR) was conducted to quantify mRNA expressions of the growth factors. RESULTS Angiogenesis was first observed in the gels by 7 days post-implantation. Over this time in ECs, genes coding for VEGFA/B, IGF-1, HIF-1α, FGF-1/-2 and BMP-5/-7 significantly increased. Meanwhile in ASCs, genes coding for VEGFA/B, IGF-1, HIF-1α, FGF-1/-2 and BMP-6 also were significantly enhanced. In particular, increased amounts of IGF-1 and HIF-1α in both ECs and ASCs were prominent relative to other factors. CONCLUSIONS Contact co-culture with ASCs and ECs at 1:1 ratio, in the 3D gel promoted angiogenesis; non-contact co-culture further confirmed gene expressions for growth factors, VEGFA/B, IGF-1, HIF-1α and FGF-1/-2 in both ASCs and ECs; BMP-5/-7 in ECs and BMP-6 in ASCs were also confirmed. This establishment of growth factor expression seemed to be responsible for enhancement of angiogenesis. This indicates that these factors could be utilized as targets for engineered angiogenesis.
Collapse
Affiliation(s)
- Xiangzhu Cun
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Shiyu Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Na Fu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Shuwen Deng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Qiang Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Juan Zhong
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
21
|
Iwen KA, Priewe AC, Winnefeld M, Rose C, Siemers F, Rohwedel J, Cakiroglu F, Lehnert H, Schepky A, Klein J, Kramer J. Gluteal and abdominal subcutaneous adipose tissue depots as stroma cell source: gluteal cells display increased adipogenic and osteogenic differentiation potentials. Exp Dermatol 2015; 23:395-400. [PMID: 24689514 DOI: 10.1111/exd.12406] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2014] [Indexed: 02/06/2023]
Abstract
Human adipose-derived stroma cells (ADSCs) have successfully been employed in explorative therapeutic studies. Current evidence suggests that ADSCs are unevenly distributed in subcutaneous adipose tissue; therefore, the anatomical origin of ADSCs may influence clinical outcomes. This study was designed to investigate proliferation and differentiation capacities of ADSCs from the gluteal and abdominal depot of 8 females. All had normal BMI (22.01 ± 0.39 kg/m(2) ) and waist circumference (81.13 ± 2.33 cm). Examination by physicians and analysis of 31 laboratory parameters did not reveal possibly confounding medical disorders. Gluteal and abdominal adipose tissue was sampled by en bloc resection on day 7 (±1) after the last menses. Histological examination did not reveal significant depot-specific differences. As assessed by BrdU assay, proliferation of cells from both depots was similar after 24 h and analysis of 15 cell surface markers by flow cytometry identified the isolated cells as ADSCs, again without depot-specific differences. ADSCs from both depots differentiated poorly to chondroblasts. Gluteal ADSCs displayed significantly higher adipogenic differentiation potential than abdominal cells. Osteogenic differentiation was most pronounced in gluteal cells, whereas differentiation of abdominal ADSCs was severely impaired. Our data demonstrate a depot-specific difference in ADSC differentiation potential with abdominal cells failing to meet the criteria of multipotent ADSCs. This finding should be taken into account in future explorations of ADSC-derived therapeutic strategies.
Collapse
|
22
|
Baptista LS, Silva KR, Borojevic R. Obesity and weight loss could alter the properties of adipose stem cells? World J Stem Cells 2015; 7:165-173. [PMID: 25621116 PMCID: PMC4300927 DOI: 10.4252/wjsc.v7.i1.165] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 09/22/2014] [Accepted: 10/27/2014] [Indexed: 02/06/2023] Open
Abstract
The discovery that adipose tissue represents an interesting source of multipotent stem cells has led to many studies exploring the clinical potential of these cells in cell-based therapies. Recent advances in understanding the secretory capacity of adipose tissue and the role of adipokines in the development of obesity and associated disorders have added a new dimension to the study of adipose tissue biology in normal and diseased states. Subcutaneous adipose tissue forms the interface between the clinical application of regenerative medicine and the establishment of the pathological condition of obesity. These two facets of adipose tissue should be understood as potentially related phenomena. Because of the functional characteristics of adipose stem cells, these cells represent a fundamental tool for understanding how these two facets are interconnected and could be important for therapeutic applications. In fact, adipose tissue stem cells have multiple functions in obesity related to adipogenic, angiogenic and secretory capacities. In addition, we have also previously described a predominance of larger blood vessels and an adipogenic memory in the subcutaneous adipose tissue after massive weight loss subsequent to bariatric surgery (ex-obese patients). Understanding the reversibility of the behavior of adipose stem cells in obeses and in weight loss is relevant to both physiological studies and the potential use of these cells in regenerative medicine.
Collapse
|
23
|
Cho JS, Park JH, Kang JH, Kim SE, Park IH, Lee HM. Isolation and characterization of multipotent mesenchymal stem cells in nasal polyps. Exp Biol Med (Maywood) 2014; 240:185-93. [PMID: 25294891 DOI: 10.1177/1535370214553898] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent progenitor cells in adult tissues. This study aimed to investigate nasal polyp (NP) tissues as a potential new source of multipotent MSCs that maintain their stemness and differentiation potential following multiple rounds of passaging. NP tissues were obtained from 10 patients during endoscopic sinus surgery. After isolating and culturing NP-derived MSCs (npMSCs), the expression levels of the surface markers CD34, CD44, CD45, CD73, CD90, CD105, CD106, CD146 and human leukocyte antigens-class II DR antigen (HLA-DR) were estimated by flow cytometry. NpMSCs were cultured in chondrogenic, osteogenic, adipogenic, or neurogenic differentiation medium. The differentiation potential of npMSCs was analyzed by Alcian blue, alizarin red S, oil red O, and immunocytochemical staining and reverse transcription-polymerase chain reaction. The clonogenic potential of npMSCs was measured using a colony-forming unit assay. Cell proliferation of npMSCs was measured using the 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT) assay. Flow cytometry analysis revealed that npMSCs were negative for hematopoietic lineage markers (CD34, CD45, and HLA-DR) and positive for MSC markers (CD44, CD73, CD90, and CD105). The npMSCs differentiated into osteogenic, adipogenic, chondrogenic, and neurogenic lineages, respectively. Chondrogenically differentiated npMSCs were stained with Alcian blue, osteogenically differentiated npMSCs were stained with alizarin red S, and adipogenically differentiated npMSCs were stained with oil red O. Real-time polymerase chain reaction results showed that the differentiated npMSCs expressed the respective differentiation markers (Sox 9 and Col2A for chondrogenesis, Runx2 and osteocalcin for osteogenesis, fatty acid-binding protein 4 and peroxisome proliferator-activated receptor γ for adipogenesis, TuJ1, neurofilament light chain, and neurofilament heavy chain for neurogenesis). There were no significant differences in the clonogenic potential and proliferation rate between early and late passage npMSCs. These results show that npMSCs possess the characteristics of MSCs in terms of morphology, multipotent differentiation capacity, cell surface marker expression, and clonogenicity. Thus, npMSCs may represent an alternative source of MSCs.
Collapse
Affiliation(s)
- Jung-Sun Cho
- Biomedical Science, College of Medicine, Korea University, Seoul 152-703, Korea College of Medicine, Institute for Medical Devices Clinical Trial Center, Guro Hospital, Korea University, Seoul 152-703, Korea
| | - Joo-Hoo Park
- Biomedical Science, College of Medicine, Korea University, Seoul 152-703, Korea
| | - Ju-Hyung Kang
- Biomedical Science, College of Medicine, Korea University, Seoul 152-703, Korea
| | - Sung Eun Kim
- Department of Orthopedic Surgery and Rare Diseases Institute, College of Medicine, Korea University, Seoul 152-703, Korea
| | - Il-Ho Park
- College of Medicine, Institute for Medical Devices Clinical Trial Center, Guro Hospital, Korea University, Seoul 152-703, Korea Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Korea University, College of Medicine, Seoul 152-703, Korea
| | - Heung-Man Lee
- Biomedical Science, College of Medicine, Korea University, Seoul 152-703, Korea College of Medicine, Institute for Medical Devices Clinical Trial Center, Guro Hospital, Korea University, Seoul 152-703, Korea Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Korea University, College of Medicine, Seoul 152-703, Korea
| |
Collapse
|
24
|
Busser H, De Bruyn C, Urbain F, Najar M, Pieters K, Raicevic G, Meuleman N, Bron D, Lagneaux L. Isolation of adipose-derived stromal cells without enzymatic treatment: expansion, phenotypical, and functional characterization. Stem Cells Dev 2014; 23:2390-400. [PMID: 24805167 DOI: 10.1089/scd.2014.0071] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Stem cell therapy is a potential method for the treatment of numerous diseases. The most frequent cellular source is bone-marrow-derived mesenchymal stromal cells (BM-MSCs). Human adipose-derived stromal cells (ADSCs) share similar properties with BM-MSCs as they support hematopoiesis, modulate ongoing immune responses, and differentiate into cells of mesodermal origin. On the other hand, ADSCs have higher frequency in situ, higher availability, and very few ethical issues compared with BM-MSCs, giving them an advantage over BM-MSCs for clinical use. Most of the methods used to isolate ADSCs contain a collagenase digestion step, but the type of collagenase and time of sample digestion vary among studies and these differences could have an impact on the cell properties and thus in result comparison. To overcome this obstacle, we propose a new method to isolate ADSCs from lipoaspirate without collagenase digestion step. We compared ADSCs obtained with our method versus classical protocol using collagenase digestion. Cells obtained with our method are equivalent but they have a better long-term hematopoietic support than those obtained with classical method. Moreover, our method has an advantage over the classical one as it is easier, safer, faster, less expensive, and more consistent with good manufacturing practices to obtain large number of ADSCs ex vivo.
Collapse
Affiliation(s)
- Hélène Busser
- 1 Laboratory of Clinical Cell Therapy, Jules Bordet Institut, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Watson JE, Patel NA, Carter G, Moor A, Patel R, Ghansah T, Mathur A, Murr MM, Bickford P, Gould LJ, Cooper DR. Comparison of Markers and Functional Attributes of Human Adipose-Derived Stem Cells and Dedifferentiated Adipocyte Cells from Subcutaneous Fat of an Obese Diabetic Donor. Adv Wound Care (New Rochelle) 2014; 3:219-228. [PMID: 24669358 DOI: 10.1089/wound.2013.0452] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 05/22/2013] [Indexed: 12/12/2022] Open
Abstract
Objective: Adipose tissue is a robust source of adipose-derived stem cells (ADSCs) that may be able to provide secreted factors that promote the ability of wounded tissue to heal. However, adipocytes also have the potential to dedifferentiate in culture to cells with stem cell-like properties that may improve their behavior and functionality for certain applications. Approach: ADSCs are adult mesenchymal stem cells that are cultured from the stromal vascular fraction of adipose tissue. However, adipocytes are capable of dedifferentiating into cells with stem cell properties. In this case study, we compare ADSC and dedifferentiated fat (DFAT) cells from the same patient and fat depot for mesenchymal cell markers, embryonic stem cell markers, ability to differentiate to adipocytes and osteoblasts, senescence and telomerase levels, and ability of conditioned media (CM) to stimulate migration of human dermal fibroblasts (HDFs). Innovation and Conclusions: ADSCs and DFAT cells displayed identical levels of CD90, CD44, CD105, and were CD34- and CD45-negative. They also expressed similar levels of Oct4, BMI1, KLF4, and SALL4. DFAT cells, however, showed higher efficiency in adipogenic and osteogenic capacity. Telomerase levels of DFAT cells were double those of ADSCs, and senescence declined in DFAT cells. CM from both cell types altered the migration of fibroblasts. Despite reports of ADSCs from a number of human depots, there have been no comparisons of the ability of dedifferentiated DFAT cells from the same donor and depot to differentiate or modulate migration of HDFs. Since ADSCs were from an obese diabetic donor, reprogramming of DFAT cells may help improve a patient's cells for regenerative medicine applications.
Collapse
Affiliation(s)
- James E. Watson
- Research Service, James A. Haley Veterans Hospital, Tampa, Florida
| | - Niketa A. Patel
- Research Service, James A. Haley Veterans Hospital, Tampa, Florida
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Gay Carter
- Research Service, James A. Haley Veterans Hospital, Tampa, Florida
| | - Andrea Moor
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Rekha Patel
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Tomar Ghansah
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Abhishek Mathur
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Michel M. Murr
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Paula Bickford
- Research Service, James A. Haley Veterans Hospital, Tampa, Florida
- Department of Neurosurgery, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Lisa J. Gould
- Research Service, James A. Haley Veterans Hospital, Tampa, Florida
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Denise R. Cooper
- Research Service, James A. Haley Veterans Hospital, Tampa, Florida
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| |
Collapse
|
26
|
Hart DA. Is Adipocyte Differentiation the Default Lineage for Mesenchymal Stem/Progenitor Cells after Loss of Mechanical Loading? A Perspective from Space Flight and Model Systems. ACTA ACUST UNITED AC 2014. [DOI: 10.4236/jbise.2014.710079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
27
|
Makridakis M, Roubelakis MG, Vlahou A. Stem cells: Insights into the secretome. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2380-4. [DOI: 10.1016/j.bbapap.2013.01.032] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 01/19/2013] [Accepted: 01/23/2013] [Indexed: 01/06/2023]
|