1
|
Wolf M, Brochhausen C, Ramakrishnan V, Iberl S, Roth J, Seitz S, Burkhardt R, Stadler SC. Histologic Characterization of Tumor-Adjacent Mammary Adipose Tissue in Normal-Weight and Overweight/Obese Patients with Triple-Negative Breast Cancer. Cancers (Basel) 2024; 16:3515. [PMID: 39456610 PMCID: PMC11506523 DOI: 10.3390/cancers16203515] [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: 09/03/2024] [Revised: 10/01/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Obesity is a risk factor of several types of cancer, including breast cancer. In this study, we aimed to histologically characterize the adipose tissue of the tumor microenvironment (TME) of triple-negative breast cancer (TNBC) in overweight/obese versus normal-weight patients. Methods: TNBC tissue sections from normal-weight (BMI<25) and overweight/obese patients (BMI≥25) were stained with antibodies against CD68, CD163, CD31, CD34, and vimentin. At the invasive tumor front, positive cells were counted in tumor adjacent adipose tissue (AT) and within cancer tissue (CT). Further, the size of the tumor-adjacent and distant mammary adipocytes was determined in perilipin stained sections. Expression of ANGPTL4, CD36 and FABP4, proteins involved in fatty acid metabolism, was analyzed in marginal tumor cells using an immune reactive score. Results: Overweight/obese TNBC patients had significantly larger adipocytes, higher numbers of CD163+ macrophages (BMI<25: 2.80 vs. BMI≥25: 10.45; p = 0.011) and lower numbers of CD31+ (BMI<25: 4.20 vs. BMI≥25: 2.40; p = 0.018) and CD34+ (BMI<25: 14.60 vs. BMI≥25: 5.20; p = 0.045) cells as markers of angiogenesis in the AT as well as a higher frequency of cancer-associated-fibroblast-like cells in the AT and CT (BMI<25: 7.60 vs. BMI≥25: 25.39 in total; p = 0.001). Moreover, expression of CD36 (BMI<25: 2.15 vs. BMI≥25: 2.60; p = 0.041) and ANGPTL4 (BMI<25: 6.00 vs. BMI≥25: 9.80; p = 0.026) was elevated in the TNBC cells of overweight/obese patients. Conclusions: Our data suggest BMI-related changes in the TME of overweight/obese TNBC patients, including hypertrophied adipocytes, reduced vascularization, more M2-like macrophages and CAF-like cells, and an increase in the expression of fatty acid metabolizing proteins in marginal tumor cells, all contributing to a more tumor-promoting, immunosuppressive environment.
Collapse
Affiliation(s)
- Marietta Wolf
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany (R.B.)
- Department of Operative Dentistry and Periodontology, Center for Dental Medicine, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany
| | - Christoph Brochhausen
- Institute of Pathology, Medical Faculty Mannheim, University Heidelberg, 69120 Mannheim, Germany
- Institute of Pathology, Regensburg University, 93053 Regensburg, Germany
| | | | - Sabine Iberl
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany (R.B.)
| | - Jonas Roth
- Department of Gynecology and Obstetrics, University Medical Centre Regensburg, 93053 Regensburg, Germany
| | - Stephan Seitz
- Department of Gynecology and Obstetrics, University Medical Centre Regensburg, 93053 Regensburg, Germany
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany (R.B.)
| | - Sonja C. Stadler
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany (R.B.)
| |
Collapse
|
2
|
Kostecka A, Kalamon N, Skoniecka A, Koczkowska M, Skowron PM, Piotrowski A, Pikuła M. Adipose-derived mesenchymal stromal cells in clinical trials: Insights from single-cell studies. Life Sci 2024; 351:122761. [PMID: 38866216 DOI: 10.1016/j.lfs.2024.122761] [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: 04/02/2024] [Revised: 05/15/2024] [Accepted: 05/27/2024] [Indexed: 06/14/2024]
Abstract
Mesenchymal Stromal Cells (MSCs) offer tremendous potential for the treatment of various diseases and their healing properties have been explored in hundreds of clinical trials. These trails primarily focus on immunological and neurological disorders, as well as regenerative medicine. Adipose tissue is a rich source of mesenchymal stromal cells and methods to obtain and culture adipose-derived MSCs (AD-MSCs) have been well established. Promising results from pre-clinical testing of AD-MSCs activity prompted clinical trials that further led to the approval of AD-MSCs for the treatment of complex perianal fistulas in Crohn's disease and subcutaneous tissue defects. However, AD-MSC heterogeneity along with various manufacturing protocols or different strategies to boost their activity create the need for standardized quality control procedures and safety assessment of the intended cell product. High-resolution transcriptomic methods have been recently gaining attention, as they deliver insight into gene expression profiles of individual cells, helping to deconstruct cellular hierarchy and differentiation trajectories, and to understand cell-cell interactions within tissues. This article presents a comprehensive overview of completed clinical trials evaluating the safety and efficacy of AD-MSC treatment, together with current single-cell studies of human AD-MSC. Furthermore, our work emphasizes the increasing significance of single-cell research in elucidating the mechanisms of cellular action and predicting their therapeutic effects.
Collapse
Affiliation(s)
- Anna Kostecka
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland; 3P - Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland.
| | - Natalia Kalamon
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland.
| | - Aneta Skoniecka
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Faculty of Medicine, Medical University of Gdansk, Dębinki 1, 80-211 Gdańsk, Poland.
| | - Magdalena Koczkowska
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland; 3P - Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland.
| | - Piotr M Skowron
- Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland.
| | - Arkadiusz Piotrowski
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland; 3P - Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland.
| | - Michał Pikuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Faculty of Medicine, Medical University of Gdansk, Dębinki 1, 80-211 Gdańsk, Poland.
| |
Collapse
|
3
|
Giacobbi AS, Meyer L, Ribot M, Yvinec R, Soula H, Audebert C. Mathematical modeling of adipocyte size distributions: Identifiability and parameter estimation from rat data. J Theor Biol 2024; 581:111747. [PMID: 38278344 DOI: 10.1016/j.jtbi.2024.111747] [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: 05/30/2023] [Revised: 12/05/2023] [Accepted: 01/20/2024] [Indexed: 01/28/2024]
Abstract
Fat cells, called adipocytes, are designed to regulate energy homeostasis by storing energy in the form of lipids. Adipocyte size distribution is assumed to play a role in the development of obesity-related diseases. These cells that do not have a characteristic size, indeed a bimodal size distribution is observed in adipose tissue. We propose a model based on a partial differential equation to describe adipocyte size distribution. The model includes a description of the lipid fluxes and the cell size fluctuations and using a formulation of a stationary solution fast computation of bimodal distribution is achieved. We investigate the parameter identifiability and estimate parameter values with CMA-ES algorithm. We first validate the procedure on synthetic data, then we estimate parameter values with experimental data of 32 rats. We discuss the estimated parameter values and their variability within the population, as well as the relation between estimated values and their biological significance. Finally, a sensitivity analysis is performed to specify the influence of parameters on cell size distribution and explain the differences between the model and the measurements. The proposed framework enables the characterization of adipocyte size distribution with four parameters and can be easily adapted to measurements of cell size distribution in different health conditions.
Collapse
Affiliation(s)
- Anne-Sophie Giacobbi
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Laboratory of Computational and Quantitative Biology UMR 7238, 75005 Paris, France.
| | - Leo Meyer
- Institut Denis Poisson, Université d'Orléans, CNRS, Université de Tours, 45067 Orléans, France
| | - Magali Ribot
- Institut Denis Poisson, Université d'Orléans, CNRS, Université de Tours, 45067 Orléans, France
| | - Romain Yvinec
- PRC, INRAE, CNRS, Université de Tours, 37380 Nouzilly, France; Université Paris-Saclay, Inria, Centre Inria de Saclay, 91120 Palaiseau, France
| | - Hedi Soula
- Nutriomics, La Pitié-Salpétrière, Sorbonne Université, CNRS, 75013 Paris, France
| | - Chloe Audebert
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Laboratory of Computational and Quantitative Biology UMR 7238, 75005 Paris, France; Sorbonne Université, CNRS, Université de Paris, Laboratoire Jacques-Louis Lions UMR 7598, 75005 Paris, France.
| |
Collapse
|
4
|
Šiklová M, Šrámková V, Koc M, Krauzová E, Čížková T, Ondrůjová B, Wilhelm M, Varaliová Z, Kuda O, Neubert J, Lambert L, Elkalaf M, Gojda J, Rossmeislová L. The role of adipogenic capacity and dysfunctional subcutaneous adipose tissue in the inheritance of type 2 diabetes mellitus: cross-sectional study. Obesity (Silver Spring) 2024; 32:547-559. [PMID: 38221680 DOI: 10.1002/oby.23969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 10/13/2023] [Accepted: 11/09/2023] [Indexed: 01/16/2024]
Abstract
OBJECTIVE This study tested the hypothesis that limited subcutaneous adipose tissue (SAT) expansion represents a primary predisposition to the development of type 2 diabetes mellitus (T2DM), independent of obesity, and identified novel markers of SAT dysfunction in the inheritance of T2DM. METHODS First-degree relatives (FDR) of T2DM patients (n = 19) and control individuals (n = 19) without obesity (fat mass < 25%) were cross-sectionally compared. Body composition (bioimpedance, computed tomography) and insulin sensitivity (IS; oral glucose tolerance test, clamp) were measured. SAT obtained by needle biopsy was used to analyze adipocyte size, lipidome, mRNA expression, and inflammatory markers. Primary cultures of adipose precursors were analyzed for adipogenic capacity and metabolism. RESULTS Compared with control individuals, FDR individuals had lower IS and a higher amount of visceral fat. However, SAT-derived adipose precursors did not differ in their ability to proliferate and differentiate or in metabolic parameters (lipolysis, mitochondrial oxidation). In SAT of FDR individuals, lipidomic and mRNA expression analysis revealed accumulation of triglycerides containing polyunsaturated fatty acids and increased mRNA expression of lysyl oxidase (LOX). These parameters correlated with IS, visceral fat accumulation, and mRNA expression of inflammatory and cellular stress genes. CONCLUSIONS The intrinsic adipogenic potential of SAT is not affected by a family history of T2DM. However, alterations in LOX mRNA and polyunsaturated fatty acids in triacylglycerols are likely related to the risk of developing T2DM independent of obesity.
Collapse
Affiliation(s)
- Michaela Šiklová
- Department of Pathophysiology, Center for Research on Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic
- Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Veronika Šrámková
- Department of Pathophysiology, Center for Research on Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic
- Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Michal Koc
- Department of Pathophysiology, Center for Research on Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic
- Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Eva Krauzová
- Department of Pathophysiology, Center for Research on Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic
- Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Internal Medicine, Královské Vinohrady University Hospital, Prague, Czech Republic
| | - Terezie Čížková
- Department of Pathophysiology, Center for Research on Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Barbora Ondrůjová
- Department of Pathophysiology, Center for Research on Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marek Wilhelm
- Department of Pathophysiology, Center for Research on Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Zuzana Varaliová
- Department of Pathophysiology, Center for Research on Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Ondrej Kuda
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Jana Neubert
- Department of Pathophysiology, Center for Research on Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Lukáš Lambert
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Moustafa Elkalaf
- Department of Pathophysiology, Center for Research on Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Gojda
- Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Internal Medicine, Královské Vinohrady University Hospital, Prague, Czech Republic
| | - Lenka Rossmeislová
- Department of Pathophysiology, Center for Research on Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic
- Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
5
|
Ding H, Liu J, Chen Z, Huang S, Yan C, Kwek E, He Z, Zhu H, Chen ZY. Protocatechuic acid alleviates TMAO-aggravated atherosclerosis via mitigating inflammation, regulating lipid metabolism, and reshaping gut microbiota. Food Funct 2024; 15:881-893. [PMID: 38165856 DOI: 10.1039/d3fo04396g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
Trimethylamine-N-oxide (TMAO) is a risk factor for atherosclerosis. As a natural phenolic acid, protocatechuic acid (PCA) is abundant in various plant foods. The present study investigated the effect of PCA on TMAO-aggravated atherosclerosis in ApoE-/- mice. The mice were randomly divided into five groups and fed one of the following five diets for 12 weeks: namely a low-fat diet (LFD), a western diet (WD), a WD + 0.2% TMAO diet (WDT), a WDT + 0.5% PCA diet (WDT + LPCA), and a WDT + 1.0% PCA diet (WDT + HPCA). Results demonstrated that dietary TMAO exacerbated the development of atherosclerosis by eliciting inflammation and disturbing lipid metabolism. The diet with PCA at 1% reduced TMAO-induced aortic plaque by 30% and decreased the levels of plasma pro-inflammatory cytokines. PCA also improved lipid metabolism by up-regulating the hepatic gene expression of peroxisome proliferator-activated receptor alpha (PPARα). In addition, PCA supplementation enhanced fecal excretion of fatty acids and decreased hepatic fat accumulation. PCA supplementation favorably modulated gut microbiota by increasing the α-diversity with an increase in the abundance of beneficial genera (Rikenella, Turicibacter, Clostridium_sensu_stricto and Bifidobacterium) and a decrease in the abundance of the harmful Helicobacter genus. In summary, PCA could alleviate the TMAO-exacerbated atherosclerosis and inflammation, improve the lipid metabolism, and modulate gut microbiota.
Collapse
Affiliation(s)
- Huafang Ding
- Food & Nutritional Sciences Programme, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China.
| | - Jianhui Liu
- Food & Nutritional Sciences Programme, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China.
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, Nanjing 210023, China
| | - Zixing Chen
- Food & Nutritional Sciences Programme, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China.
| | - Shouhe Huang
- Food & Nutritional Sciences Programme, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China.
| | - Chi Yan
- Food & Nutritional Sciences Programme, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China.
| | - Erika Kwek
- Food & Nutritional Sciences Programme, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China.
| | - Zouyan He
- Food & Nutritional Sciences Programme, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China.
| | - Hanyue Zhu
- Food & Nutritional Sciences Programme, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China.
| | - Zhen-Yu Chen
- Food & Nutritional Sciences Programme, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China.
| |
Collapse
|
6
|
Fukui M, Lai F, Hihara M, Mitsui T, Matsuoka Y, Sun Z, Kunieda S, Taketani S, Odaka T, Okuma K, Kakudo N. Activation of cell adhesion and migration is an early event of platelet-rich plasma (PRP)-dependent stimulation of human adipose-derived stem/stromal cells. Hum Cell 2024; 37:181-192. [PMID: 37787969 DOI: 10.1007/s13577-023-00989-1] [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: 05/17/2023] [Accepted: 09/14/2023] [Indexed: 10/04/2023]
Abstract
Stem cell therapy is a promising treatment in regenerative medicine. Human adipose-derived stem/stromal cells (hASCs), a type of mesenchymal stem cell, are easy to harvest. In plastic and aesthetic surgery, hASC may be applied in the treatment of fat grafting, wound healing, and scar remodeling. Platelet-rich plasma (PRP) contains various growth factors, including platelet-derived growth factor (PDGF), which accelerates wound healing. We previously reported that PRP promotes the proliferation of hASC via multiple signaling pathways, and we evaluated the effect of PRP on the stimulation of hASC adhesion and migration, leading to the proliferation of these cells. When hASCs were treated with PRP, AKT, ERK1/2, paxillin and RhoA were rapidly activated. PRP treatment led to the formation of F-actin stress fibers. Strong signals for integrin β1, paxillin and RhoA at the cell periphery of RPR-treated cells indicated focal adhesion. PRP promoted cell adhesion and movement of hASC, compared with the control group. Imatinib, an inhibitor of the PDGF receptor tyrosine kinase, inhibited the promotion of PRP-dependent cell migration. PDGF treatment of hASCs also stimulated cell adhesion and migration but to a lesser extent than PRP treatment. PRP promoted the adhesion and the migration of hASC, mediated by the activation of AKT in the integrin signaling pathway. PRP treatment was more effective than PDGF treatment in enhancing cell migration. Thus, the ability of PRPs to promote migration of hASC to enhance cell growth is evident.
Collapse
Affiliation(s)
- Michika Fukui
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1, Hirakata, Osaka, 573-1010, Japan.
| | - Fangyuan Lai
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1, Hirakata, Osaka, 573-1010, Japan
| | - Masakatsu Hihara
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1, Hirakata, Osaka, 573-1010, Japan
| | - Toshihito Mitsui
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1, Hirakata, Osaka, 573-1010, Japan
| | - Yuki Matsuoka
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1, Hirakata, Osaka, 573-1010, Japan
| | - Zhongxin Sun
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1, Hirakata, Osaka, 573-1010, Japan
| | - Sakurako Kunieda
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1, Hirakata, Osaka, 573-1010, Japan
| | - Shigeru Taketani
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1, Hirakata, Osaka, 573-1010, Japan
| | - Tokifumi Odaka
- Department of Microbiology, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan
| | - Kazu Okuma
- Department of Microbiology, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan
| | - Natsuko Kakudo
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1, Hirakata, Osaka, 573-1010, Japan
| |
Collapse
|
7
|
Ernst AM, Steiner M, Kainz V, Tempfer H, Spitzer G, Plank T, Bauer HC, Bresgen N, Habenbacher A, Bauer H, Lipp AT. Lipedema: The Use of Cultured Adipocytes for Identification of Diagnostic Markers. Plast Reconstr Surg 2023; 152:1036-1046. [PMID: 36912938 DOI: 10.1097/prs.0000000000010392] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
BACKGROUND Lipedema, diagnosed most often in women, is a progressive disease characterized by the disproportionate and symmetrical distribution of adipose tissue, primarily in the extremities. Although numerous results from in vitro and in vivo studies have been published, many questions regarding the pathology and genetic background of lipedema remain unanswered. METHODS In this study, adipose tissue-derived stromal/stem cells were isolated from lipoaspirates derived from nonobese and obese donors with or without lipedema. Growth and morphology, metabolic activity, differentiation potential, and gene expression were evaluated using quantification of lipid accumulation, metabolic activity assay, live-cell imaging, reverse transcription polymerase chain reaction, quantitative polymerase chain reaction, and immunocytochemical staining. RESULTS The adipogenic potential of lipedema and nonlipedema adipose tissue-derived stromal/stem cells did not rise in parallel with the donors' body mass index and did not differ significantly between groups. However, in vitro differentiated adipocytes from nonobese lipedema donors showed significant upregulation of adipogenic gene expression compared with nonobese controls. All other genes tested were expressed equally in lipedema and nonlipedema adipocytes. The adiponectin/leptin ratio was significantly reduced in adipocytes from obese lipedema donors compared with their nonobese lipedema counterparts. Increased stress fiber-integrated smooth muscle actin was visible in lipedema adipocytes compared with nonlipedema controls and appeared enhanced in adipocytes from obese lipedema donors. CONCLUSIONS Not only lipedema per se but also body mass index of donors affect adipogenic gene expression substantially in vitro. The significantly reduced adiponectin/leptin ratio and the increased occurrence of myofibroblast-like cells in obese lipedema adipocyte cultures underscores the importance of attention to the co-occurrence of lipedema and obesity. These are important findings toward accurate diagnosis of lipedema. CLINICAL RELEVANCE STATEMENT Our study highlights not only the difficulty in lipedema diagnostics but also the tremendous need for further studies on lipedema tissue. Although lipedema might seem to be an underestimated field in plastic and reconstructive surgery, the power it holds to provide better treatment to future patients can not be promoted enough.
Collapse
Affiliation(s)
- Anna M Ernst
- From the Department of Biosciences, Paris Lodron University of Salzburg
| | - Marianne Steiner
- From the Department of Biosciences, Paris Lodron University of Salzburg
| | - Verena Kainz
- From the Department of Biosciences, Paris Lodron University of Salzburg
| | - Herbert Tempfer
- Institute of Tendon and Bone Regeneration, Spinal Cord Injury and Tissue Regeneration Centre Salzburg, Paracelsus Medical University Salzburg
- Austrian Cluster for Tissue Regeneration
| | - Gabriel Spitzer
- Institute of Tendon and Bone Regeneration, Spinal Cord Injury and Tissue Regeneration Centre Salzburg, Paracelsus Medical University Salzburg
- Austrian Cluster for Tissue Regeneration
| | - Tanja Plank
- From the Department of Biosciences, Paris Lodron University of Salzburg
| | - Hans-Christian Bauer
- From the Department of Biosciences, Paris Lodron University of Salzburg
- Institute of Tendon and Bone Regeneration, Spinal Cord Injury and Tissue Regeneration Centre Salzburg, Paracelsus Medical University Salzburg
- Austrian Cluster for Tissue Regeneration
| | - Nikolaus Bresgen
- From the Department of Biosciences, Paris Lodron University of Salzburg
| | - Andreas Habenbacher
- Department of Plastic Surgery and Hand Surgery, Klinikum rechts der Isar, Technical University of Munich
| | - Hannelore Bauer
- From the Department of Biosciences, Paris Lodron University of Salzburg
| | - Anna-Theresa Lipp
- Department of Plastic Surgery and Hand Surgery, Klinikum rechts der Isar, Technical University of Munich
| |
Collapse
|
8
|
Jovic D, Preradovic L, Kremenovic M, Jovic F, Antonic M, Aleksic Z, Ljubojevic V. Effect of Donor Site Selection for Fat Grafting on the Yield and Viability of the Stromal Vascular Fraction. Aesthet Surg J 2023; 43:NP704-NP712. [PMID: 37289983 DOI: 10.1093/asj/sjad184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/10/2023] Open
Abstract
BACKGROUND The efficacy of stromal vascular fraction (SVF) treatment, or stem cell treatment, directly depends on the SVF cell count and the cells' viability. The SVF cell count and viability are in direct correlation with the adipose tissue harvesting site that yields SVF cells, making this research a contribution to developing tissue guidance. OBJECTIVES The aim of this study was to investigate the importance of harvesting subcutaneous adipose tissue-derived SVF cells on the concentration and viability of SVF. METHODS Adipose tissue was collected by vibration-assisted liposuction from the regions of the upper and lower abdomen, lumbar region, and inner thigh region. With the semiautomatic UNISTATION 2nd Version system, the obtained fat was chemically processed (with collagenase enzyme) and a concentrate of SVF cells was obtained by centrifugation. These samples were then analyzed with the Luna-Stem Counter device to measure the number and viability of SVF cells. RESULTS When comparing the regions of the upper abdomen, lower abdomen, lumbar region, and inner thigh, the highest concentration of SVF was found in the lumbar region, specifically at an average of 97,498.00 per 1.0 mL of concentrate. The lowest concentration was found in the upper abdominal region. When ranking the viability values, the highest cell viability of SVF was observed in the lumbar region, measuring 36.6200%. The lowest viability was found in the upper abdominal region, measuring 24.4967%. CONCLUSIONS By comparing the upper and lower abdominal, lumbar, and inner thigh regions, the authors have come to the conclusion that, on average, the largest number of cells with the highest viability was obtained from the lumbar region.
Collapse
|
9
|
Lee YR, Lee HB, Kim Y, Shin KS, Park HY. Prebiotic and Anti-Adipogenic Effects of Radish Green Polysaccharide. Microorganisms 2023; 11:1862. [PMID: 37513035 PMCID: PMC10385334 DOI: 10.3390/microorganisms11071862] [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: 07/03/2023] [Revised: 07/20/2023] [Accepted: 07/22/2023] [Indexed: 07/30/2023] Open
Abstract
Radish (Raphanus sativus L.) greens are consumed as a source of nutrition, and their polysaccharides such as rhamnogalacturonan-I possess certain beneficial properties. This study investigated the prebiotic effects of a radish green polysaccharide (RGP) on gut health and obesity. The prebiotic activity of RGP was evaluated based on the pH changes and short-chain fatty acids (SCFAs) concentration. The results showed that 0.5% RGP had a higher prebiotic activity score than inulin and increased SCFAs production in all five prebiotic strains. Moreover, RGP inhibited fat accumulation in 3T3-L1 adipocytes, indicating its potential to reduce obesity. Overall, these findings suggested that the polysaccharide of radish greens has prebiotic effects and may serve as a beneficial prebiotic for gut health and obesity.
Collapse
Affiliation(s)
- Yu Ra Lee
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea
| | - Hye-Bin Lee
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea
| | - Yoonsook Kim
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea
| | - Kwang-Soon Shin
- Department of Food Science and Biotechnology, Kyonggi University, Suwon 16227, Republic of Korea
| | - Ho-Young Park
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea
- Department of Food Biotechnology, University of Science and Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
10
|
Abstract
SUMMARY Over the past 30 years, there has been a dramatic increase in the use of autologous fat grafting for soft-tissue augmentation and to improve facial skin quality. Several studies have highlighted the impact of aging on adipose tissue, leading to a decrease of adipose tissue volume and preadipocyte proliferation and increase of fibrosis. Recently, there has been a rising interest in adipose tissue components, including adipose-derived stem/stromal cells (ASCs) because of their regenerative potential, including inflammation, fibrosis, and vascularization modulation. Because of their differentiation potential and paracrine function, ASCs have been largely used for fat grafting procedures, as they are described to be a key component in fat graft survival. However, many parameters as surgical procedures or adipose tissue biology could change clinical outcomes. Variation on fat grafting methods have led to numerous inconsistent clinical outcomes. Donor-to-donor variation could also be imputed to ASCs, tissue inflammatory state, or tissue origin. In this review, the authors aim to analyze (1) the parameters involved in graft survival, and (2) the effect of aging on adipose tissue components, especially ASCs, that could lead to a decrease of skin regeneration and fat graft retention. CLINICAL RELEVANCE STATEMENT This review aims to enlighten surgeons about known parameters that could play a role in fat graft survival. ASCs and their potential mechanism of action in regenerative medicine are more specifically described.
Collapse
|
11
|
Pathogenic Role of Adipose Tissue-Derived Mesenchymal Stem Cells in Obesity and Obesity-Related Inflammatory Diseases. Cells 2023; 12:cells12030348. [PMID: 36766689 PMCID: PMC9913687 DOI: 10.3390/cells12030348] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/12/2023] [Accepted: 01/14/2023] [Indexed: 01/19/2023] Open
Abstract
Adipose tissue-derived mesenchymal stem cells (ASCs) are adult stem cells, endowed with self-renewal, multipotent capacities, and immunomodulatory properties, as mesenchymal stem cells (MSCs) from other origins. However, in a pathological context, ASCs like MSCs can exhibit pro-inflammatory properties and attract inflammatory immune cells at their neighborhood. Subsequently, this creates an inflammatory microenvironment leading to ASCs' or MSCs' dysfunctions. One such example is given by obesity where adipogenesis is impaired and insulin resistance is initiated. These opposite properties have led to the classification of MSCs into two categories defined as pro-inflammatory ASC1 or anti-inflammatory ASC2, in which plasticity depends on the micro-environmental stimuli. The aim of this review is to (i) highlight the pathogenic role of ASCs during obesity and obesity-related inflammatory diseases, such as rheumatoid arthritis, multiple sclerosis, psoriasis, inflammatory bowel disease, and cancer; and (ii) describe some of the mechanisms leading to ASCs dysfunctions. Thus, the role of soluble factors, adhesion molecules; TLRs, Th17, and Th22 cells; γδ T cells; and immune checkpoint overexpression will be addressed.
Collapse
|
12
|
Amir Levy Y, P Ciaraldi T, R. Mudaliar S, A. Phillips S, R. Henry R. Adipose tissue from subjects with type 2 diabetes exhibits impaired capillary formation in response to GROα: involvement of MMPs-2 and -9. Adipocyte 2022; 11:276-286. [PMID: 35481427 PMCID: PMC9116416 DOI: 10.1080/21623945.2022.2070949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 12/03/2022] Open
Abstract
Type 2 Diabetes (T2D) is associated with impaired vascularization of adipose tissue (AT) . IL8, GROα and IL15 are pro-angiogenic myokines, secreted at elevated levels by T2D myotubes. We explored the direct impact of these myokines on AT vascularization. AT explants from subjects with T2D and without diabetes (non-diabetic, ND) were treated with rIL8, rGROα and rIL15 in concentrations equal to those in conditioned media (CM) from T2D and ND myotubes, and sprout formation evaluated. Endothelial cells (EC) were isolated from T2D and ND-AT, treated with rGROα and tube formation evaluated. Finally, we investigated the involvement of MMP-2 and -9 in vascularization. ND and T2D concentrations of IL8 or IL15 caused similar stimulation of sprout formation in ND- and T2D-AT. GROα exerted a similar effect in ND-AT. When T2D-AT explants were exposed to GROα, sprout formation in response to T2D concentrations was reduced compared to ND. Exposure of EC from T2D-AT to GROα at T2D concentrations resulted in reduced tube formation. Reduced responses to GROα in T2D-AT and EC were also seen for secretion of MMP-2 and -9. The data indicate that skeletal muscle can potentially regulate AT vascularization, with T2D-AT having impairments in sensitivity to GROα, while responding normally to IL8 and IL15.
Collapse
Affiliation(s)
- Yifat Amir Levy
- Center for Metabolic Research, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Departments of Medicine, University of California, La Jolla, CA, USA
| | - Theodore P Ciaraldi
- Center for Metabolic Research, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Departments of Medicine, University of California, La Jolla, CA, USA
| | - Sunder R. Mudaliar
- Center for Metabolic Research, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Departments of Medicine, University of California, La Jolla, CA, USA
| | - Susan A. Phillips
- Center for Metabolic Research, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Departments of Pediatrics, University of California, La Jolla, CA, USA
| | - Robert R. Henry
- Center for Metabolic Research, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Departments of Medicine, University of California, La Jolla, CA, USA
| |
Collapse
|
13
|
Wang X, Guan J, Chen Y, Li Y, Lu F, Dong Z. Density-Based High-Quality Fat: Characterization and Correlation with Different Body Fat Ratio. Aesthetic Plast Surg 2022; 46:3003-3012. [PMID: 35764812 DOI: 10.1007/s00266-022-02973-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/02/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Lipoaspirate can be divided into high-quality fat and low-quality fat using Coleman's centrifugation by adding 0.935 g/ml marker float; the ratio obtained by different individuals is different. OBJECTIVES This study aimed to examine the HQF obtained from different individuals and establish the relationship between individual body data and HQF. METHODS We used Coleman's centrifugation method (1200 g, 3 min) with 0.935 g/ml density float to process lipoaspirate and collect HQF from different individuals for the analysis of fat characteristics and in vivo grafting. RESULTS The HQF obtained from different individuals had similar stromal vascular fraction cell numbers and extracellular matrix content. In animal experiments at different time points (especially 12 weeks), the appearance, retention rate, hematoxylin and eosin staining, and immunohistochemistry results of HQF grafts were similar, while being different from those of Coleman fat. The HQF obtained from individuals with higher body fat ratio was less than those with lower body fat ratio. Following the establishment of the relationship between high-quality fat percentage and the body fat ratio of the donors, we proposed an innovative calculation formula model for the required lipoaspirate. CONCLUSIONS HQF obtained from different individuals has similar fat characteristics, transplantation process, and outcome. The HQF percentage obtained from different individuals is negatively correlated with the body fat ratio. The amount of liposuction can be predicted using the proposed formula and improve the predictability of fat transplantation. LEVEL OF EVIDENCE IV This journal requires that authors assign a level of evidence to each article. For a full description of these evidence-based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266.
Collapse
Affiliation(s)
- Xinhui Wang
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Jingyan Guan
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Yunzi Chen
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Ye Li
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Feng Lu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China.
| | - Ziqing Dong
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
14
|
Iyoshi S, Sumi A, Yoshihara M, Kitami K, Mogi K, Uno K, Fujimoto H, Miyamoto E, Tano S, Yoshikawa N, Emoto R, Matsui S, Kajiyama H. Obesity contributes to the stealth peritoneal dissemination of ovarian cancer: a multi-institutional retrospective cohort study. Obesity (Silver Spring) 2022; 30:1599-1607. [PMID: 35851756 DOI: 10.1002/oby.23497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/06/2022] [Accepted: 04/28/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE The clinical significance of a higher BMI on the prognosis of ovarian cancer remains controversial; therefore, a more detailed analysis is demanded. This study investigated the impact of BMI on peritoneum-specific recurrence to clarify the involvement of adipose tissue in the proliferation of cancer cells at sites of peritoneal dissemination. METHODS Among 4,730 patients with malignant ovarian tumors, 280 diagnosed with International Federation of Gynecology and Obstetrics (FIGO) stage IIB to IIIC epithelial ovarian cancer and who underwent complete resection in the primary surgery were included in the present study. RESULTS There were 42, 201, and 37 women in the low, normal, and high BMI groups, respectively. Peritoneum-specific recurrence-free survival and overall survival were both significantly shorter in patients with a high BMI than in those with a normal BMI (p = 0.028 and 0.018, respectively). No significant differences were observed in the distribution of sites of recurrence between these two groups. A multivariate analysis identified obesity as an independent prognostic factor in addition to pT3 tumor staging and positive ascites cytology. CONCLUSIONS Patients with a high BMI had a significantly worse prognosis than those with a normal BMI, and peritoneal adipose tissue may have contributed to this difference.
Collapse
Affiliation(s)
- Shohei Iyoshi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Asami Sumi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Obstetrics and Gynecology, Tosei General Hospital, Seto, Japan
| | - Masato Yoshihara
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuhisa Kitami
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Obstetrics and Gynecology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kazumasa Mogi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kaname Uno
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University Graduate School of Medicine, Lund, Sweden
| | - Hiroki Fujimoto
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Emiri Miyamoto
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sho Tano
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nobuhisa Yoshikawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryo Emoto
- Department of Biostatistics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shigeyuki Matsui
- Department of Biostatistics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
15
|
Schmitz D, Robering JW, Weisbach V, Arkudas A, Ludolph I, Horch RE, Boos AM, Kengelbach-Weigand A. Specific features of ex-obese patients significantly influence the functional cell properties of adipose-derived stromal cells. J Cell Mol Med 2022; 26:4463-4478. [PMID: 35818175 PMCID: PMC9357603 DOI: 10.1111/jcmm.17471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 03/17/2022] [Accepted: 05/27/2022] [Indexed: 11/29/2022] Open
Abstract
Adipose-derived stromal cells (ADSC) are increasingly used in clinical applications due to their regenerative capabilities. However, ADSC therapies show variable results. This study analysed the effects of specific factors of ex-obese patients on ADSC functions. ADSC were harvested from abdominal tissues (N = 20) after massive weight loss. Patients were grouped according to age, sex, current and maximum body mass index (BMI), BMI difference, weight loss method, smoking and infection at the surgical site. ADSC surface markers, viability, migration, transmigration, sprouting, differentiation potential, cytokine secretion, telomere length and mtDNA copy number were analysed. All ADSC expressed CD73, CD90, CD105, while functional properties differed significantly among patients. A high BMI difference due to massive weight loss was negatively correlated with ADSC proliferation, migration and transmigration, while age, sex or weight loss method had a smaller effect. ADSC from female and younger donors and individuals after weight loss by increase of exercise and diet change had a higher activity. Telomere length, mtDNA copy number, differentiation potential and the secretome did not correlate with patient factors or cell function. Therefore, we suggest that factors such as age, sex, increase of exercise and especially weight loss should be considered for patient selection and planning of regenerative therapies.
Collapse
Affiliation(s)
- Deborah Schmitz
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Plastic Surgery, Hand Surgery, Burns Center, University Hospital RWTH Aachen University, University Hospital Aachen, Germany
| | - Jan W Robering
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Plastic Surgery, Hand Surgery, Burns Center, University Hospital RWTH Aachen University, University Hospital Aachen, Germany
| | - Volker Weisbach
- Department of Transfusion Medicine and Hemostaseology, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Arkudas
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ingo Ludolph
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Raymund E Horch
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Anja M Boos
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Plastic Surgery, Hand Surgery, Burns Center, University Hospital RWTH Aachen University, University Hospital Aachen, Germany
| | - Annika Kengelbach-Weigand
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
16
|
Stefano ABD, Cammarata E, Trapani M, Pirrello R, Montesano L, Meraviglia S, Moschella F, Cordova A, Toia F. Correlation between tissue-harvesting method and donor-site with the yield of spheroids from adipose-derived stem cells. J Plast Reconstr Aesthet Surg 2022; 75:3628-3651. [DOI: 10.1016/j.bjps.2022.06.091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 11/29/2022]
|
17
|
Fernández-Santos ME, Garcia-Arranz M, Andreu EJ, García-Hernández AM, López-Parra M, Villarón E, Sepúlveda P, Fernández-Avilés F, García-Olmo D, Prosper F, Sánchez-Guijo F, Moraleda JM, Zapata AG. Optimization of Mesenchymal Stromal Cell (MSC) Manufacturing Processes for a Better Therapeutic Outcome. Front Immunol 2022; 13:918565. [PMID: 35812460 PMCID: PMC9261977 DOI: 10.3389/fimmu.2022.918565] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/10/2022] [Indexed: 12/20/2022] Open
Abstract
MSCs products as well as their derived extracellular vesicles, are currently being explored as advanced biologics in cell-based therapies with high expectations for their clinical use in the next few years. In recent years, various strategies designed for improving the therapeutic potential of mesenchymal stromal cells (MSCs), including pre-conditioning for enhanced cytokine production, improved cell homing and strengthening of immunomodulatory properties, have been developed but the manufacture and handling of these cells for their use as advanced therapy medicinal products (ATMPs) remains insufficiently studied, and available data are mainly related to non-industrial processes. In the present article, we will review this topic, analyzing current information on the specific regulations, the selection of living donors as well as MSCs from different sources (bone marrow, adipose tissue, umbilical cord, etc.), in-process quality controls for ensuring cell efficiency and safety during all stages of the manual and automatic (bioreactors) manufacturing process, including cryopreservation, the use of cell banks, handling medicines, transport systems of ATMPs, among other related aspects, according to European and US legislation. Our aim is to provide a guide for a better, homogeneous manufacturing of therapeutic cellular products with special reference to MSCs.
Collapse
Affiliation(s)
- Maria Eugenia Fernández-Santos
- Cardiology Department, HGU Gregorio Marañón. GMP-ATMPs Production Unit, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM). Complutense University, CIBER Cardiovascular (CIBERCV), ISCIII, Madrid, Spain
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
| | - Mariano Garcia-Arranz
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD). Surgery Department, Autonoma University of Madrid, Madrid, Spain
| | - Enrique J. Andreu
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- Hematology Department and Cell Therapy Area, Clínica Universidad de Navarra. CIBEROC and IDISNA, Pamplona, Spain
| | - Ana Maria García-Hernández
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Miriam López-Parra
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- Cell Therapy Area and Hematology Department, IBSAL-University Hospital of Salamanca, University of Salamanca, Salamanca, Spain
| | - Eva Villarón
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- Cell Therapy Area and Hematology Department, IBSAL-University Hospital of Salamanca, University of Salamanca, Salamanca, Spain
| | - Pilar Sepúlveda
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Francisco Fernández-Avilés
- Cardiology Department, HGU Gregorio Marañón. GMP-ATMPs Production Unit, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM). Complutense University, CIBER Cardiovascular (CIBERCV), ISCIII, Madrid, Spain
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
| | - Damian García-Olmo
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD). Surgery Department, Autonoma University of Madrid, Madrid, Spain
| | - Felipe Prosper
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- Hematology Department and Cell Therapy Area, Clínica Universidad de Navarra. CIBEROC and IDISNA, Pamplona, Spain
| | - Fermin Sánchez-Guijo
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- Cell Therapy Area and Hematology Department, IBSAL-University Hospital of Salamanca, University of Salamanca, Salamanca, Spain
| | - Jose M. Moraleda
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Agustin G. Zapata
- Platform GMP Units from TerCel and TERAV Networks. RETIC TerCel & RICORS TERAV, ISCIII, Madrid, Spain
- Department of Cell Biology, Complutense University, Madrid, Spain
- *Correspondence: Maria Eugenia Fernández-Santos, ; Agustin G. Zapata,
| |
Collapse
|
18
|
Abstract
Recent technological developments have allowed determination of the age of fat cells and their lipids in adult humans. In contrast to prior views, this has demonstrated a high turnover rate of the fat cells (10%/year) and their unilocular lipid droplets (six times/10 years). Fat cell turnover is increased in obesity and when adipose tissue is composed of many but small adipocytes (hyperplasia, a benign adipose phenotype). While fat mass gain increases adipocyte number and size, only the latter is altered (decreased) after weight loss, which may facilitate weight regain. Fat cell lipid turnover is attenuated in subjects with excess body fat. In the subcutaneous region, this dysregulation occurs already in the overweight state while in the visceral depot, it is only observed in severe obesity. This may explain why the latter depot is particularly pernicious in the overweight/obese state as it allows for more rapid lipid fluxes between visceral fat and the liver. Adipose lipid turnover decreases during ageing due to impaired breakdown (lipolysis) of stored triglycerides. If this decline is not compensated by reduced adipocyte lipid uptake, bodyweight will increase over time. In concordance with this, low lipolysis rates are a risk factor for future weight gain and glucose intolerance. Adipose lipid turnover is also decreased in insulin resistance and certain forms of dyslipidemia. Altogether, adult human adipose tissue is in a highly dynamic state. Alterations in the turnover of fat cells and their lipids are therefore novel factors to consider in the pathophysiology of common metabolic disorders.
Collapse
Affiliation(s)
- Peter Arner
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Mikael Rydén
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
19
|
Hunt C, Mun CJ, Owens M, Lerman S, Kunatharaju S, Tennen H, Buenaver L, Campbell C, Haythornthwaite J, Smith M, Finan PH. Sleep, Positive Affect, and Circulating Interleukin-6 in Women With Temporomandibular Joint Disorder. Psychosom Med 2022; 84:383-392. [PMID: 35067649 PMCID: PMC8976725 DOI: 10.1097/psy.0000000000001047] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Systemic inflammation is commonly observed in idiopathic chronic pain conditions, including temporomandibular joint disorder (TMD). Trait positive affect (PA) is associated with lower inflammation in healthy controls, but those effects may be threatened by poor sleep. The associations between PA with proinflammatory cytokine activity and potential moderation by sleep in chronic pain are not known. We thus investigated the association between PA and circulating interleukin-6 (IL-6) and moderation of that association by sleep in a sample of women with TMD and sleep difficulties. METHODS Participants (n = 110) completed the insomnia severity index and provided blood samples at five intervals throughout an evoked pain testing session. They then completed a 14-day diary assessing sleep and affect, along with wrist actigraphy. RESULTS There was not a significant main effect of PA on resting or pain-evoked IL-6 (b = 0.04, p = .33). Diary total sleep time (b = -0.002, p = .008), sleep efficiency (b = -0.01, p = .005), sleep onset latency (b = 0.006, p = .010), and wake after sleep onset (b = 0.003, p = .033) interacted with PA to predict IL-6, such that PA inversely predicted IL-6 at higher levels of total sleep time and sleep efficiency and at lower levels of sleep onset latency and wake after sleep onset. Surprisingly, when sleep was poor, PA predicted greater IL-6. CONCLUSIONS The potential salutary effects of PA on resting IL-6 erode when sleep is poor, underscoring the importance of considering sleep in conceptual and intervention models of TMD.
Collapse
Affiliation(s)
- Carly Hunt
- Johns Hopkins University School of Medicine, Department of Psychiatry, Division of Behavioral Medicine
| | - Chung Jung Mun
- Johns Hopkins University School of Medicine, Department of Psychiatry, Division of Behavioral Medicine
| | - Michael Owens
- Johns Hopkins University School of Medicine, Department of Psychiatry, Division of Behavioral Medicine
| | - Sheera Lerman
- Johns Hopkins University School of Medicine, Department of Psychiatry, Division of Behavioral Medicine
| | - Shriya Kunatharaju
- Johns Hopkins University School of Medicine, Department of Psychiatry, Division of Behavioral Medicine
| | | | - Luis Buenaver
- Johns Hopkins University School of Medicine, Department of Psychiatry, Division of Behavioral Medicine
| | - Claudia Campbell
- Johns Hopkins University School of Medicine, Department of Psychiatry, Division of Behavioral Medicine
| | - Jennifer Haythornthwaite
- Johns Hopkins University School of Medicine, Department of Psychiatry, Division of Behavioral Medicine
| | - Michael Smith
- Johns Hopkins University School of Medicine, Department of Psychiatry, Division of Behavioral Medicine
| | - Patrick H. Finan
- Johns Hopkins University School of Medicine, Department of Psychiatry, Division of Behavioral Medicine
| |
Collapse
|
20
|
Frasca D. Obesity Accelerates Age Defects in Human B Cells and Induces Autoimmunity. IMMUNOMETABOLISM 2022; 4:e220010. [PMID: 35433040 PMCID: PMC9012215 DOI: 10.20900/immunometab20220010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/17/2022] [Indexed: 11/24/2022]
Abstract
Aging is associated with systemic inflammation and decreased production of protective antibodies while the production of autoimmune antibodies is increased. Our results have shown that the human obese adipose tissue (AT), which increases in size with aging, contributes to systemic and B cell intrinsic inflammation, reduced protective and increased pathogenic B cell responses leading to increased secretion of autoimmune antibodies. With this R56 funding, we have been able to investigate the cellular and molecular mechanisms by which the human obese AT induces intrinsic B cell inflammation and dysfunctional B cell responses, stimulates the secretion of autoimmune antibodies, whose specificity has been characterized, and engages different AT cell types in antigen presentation pathways to allow secretion of these autoimmune antibodies. Briefly, immune cells are recruited to the AT by chemokines released by both non-immune (adipocytes) and by resident and infiltrating immune cells. We have identified several mechanisms responsible for the release of "self" antigens, and we have shown that reduced oxygen availability and hypoxia, cell cytotoxicity and DNA damage induce cell death and lead to further release of pro-inflammatory cytokines, "self" protein antigens, cell-free DNA and lipids. We have also identified different antigen presenting cells in the AT, responsible for the activation of pathogenic B cells, class switch and secretion of autoimmune IgG antibodies. The experiments performed have allowed the discovery of novel mechanisms for pathogenic responses and the identification of pathways to target in order to promote better humoral immunity during aging.
Collapse
Affiliation(s)
- Daniela Frasca
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
21
|
Patil PJ, Usman M, Zhang C, Mehmood A, Zhou M, Teng C, Li X. An updated review on food-derived bioactive peptides: Focus on the regulatory requirements, safety, and bioavailability. Compr Rev Food Sci Food Saf 2022; 21:1732-1776. [PMID: 35142435 DOI: 10.1111/1541-4337.12911] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/07/2021] [Accepted: 12/22/2021] [Indexed: 02/06/2023]
Abstract
Food-derived bioactive peptides (BAPs) are recently utilized as functional food raw materials owing to their potential health benefits. Although there is a huge amount of scientific research about BAPs' identification, purification, characterization, and physiological functions, and subsequently, many BAPs have been marketed, there is a paucity of review on the regulatory requirements, bioavailability, and safety of BAPs. Thus, this review focuses on the toxic peptides that could arise from their primary proteins throughout protein extraction, protein pretreatment, and BAPs' formulation. Also, the influences of BAPs' length and administration dosage on safety are summarized. Lastly, the challenges and possibilities in BAPs' bioavailability and regulatory requirements in different countries were also presented. Results revealed that the human studies of BAPs are essential for approvals as healthy food and to prevent the consumers from misinformation and false promises. The BAPs that escape the gastrointestinal tract epithelium and move to the stomach are considered good peptides and get circulated into the blood using different pathways. In addition, the hydrophobicity, net charge, molecular size, length, amino acids composition/sequences, and structural characteristics of BAPs are critical for bioavailability, and appropriate food-grade carriers can enhance it. The abovementioned features are also vital to optimize the solubility, water holding capacity, emulsifying ability, and foaming property of BAPs in food products. In the case of safety, the possible allergenic and toxic peptides often exhibit physiological functions and could be produced during the hydrolysis of food proteins. It was also noted that the production of iso-peptides bonds and undesirable Maillard reaction might occur during protein extraction, sample pretreatments, and peptide synthesis.
Collapse
Affiliation(s)
- Prasanna J Patil
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China.,School of Food and Health, Beijing Technology and Business University, Beijing, China
| | - Muhammad Usman
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China.,School of Food and Health, Beijing Technology and Business University, Beijing, China
| | - Chengnan Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China.,School of Food and Health, Beijing Technology and Business University, Beijing, China
| | - Arshad Mehmood
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China.,School of Food and Health, Beijing Technology and Business University, Beijing, China
| | - Mingchun Zhou
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China.,School of Food and Health, Beijing Technology and Business University, Beijing, China
| | - Chao Teng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China.,School of Food and Health, Beijing Technology and Business University, Beijing, China.,Beijing Engineering and Technology Research Center of Food Additives, School of Food and Chemical Technology, Beijing Technology and Business University, Beijing, China
| | - Xiuting Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China.,School of Food and Health, Beijing Technology and Business University, Beijing, China.,Beijing Engineering and Technology Research Center of Food Additives, School of Food and Chemical Technology, Beijing Technology and Business University, Beijing, China
| |
Collapse
|
22
|
Zuo Q, Band S, Kesavadas M, Madak Erdogan Z. Obesity and Postmenopausal Hormone Receptor-positive Breast Cancer: Epidemiology and Mechanisms. Endocrinology 2021; 162:6370080. [PMID: 34519778 DOI: 10.1210/endocr/bqab195] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Indexed: 12/11/2022]
Abstract
Obesity is a potential risk for several cancers, including postmenopausal, hormone dependent breast cancers. In this review, we summarize recent studies on the impact of obesity on postmenopausal women's health and discuss several mechanisms that were proposed to increase the risk of breast carcinogenesis.
Collapse
Affiliation(s)
- Qianying Zuo
- Department of Food Science and Human Nutrition, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Shoham Band
- Department of Food Science and Human Nutrition, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Mrinali Kesavadas
- Department of Food Science and Human Nutrition, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Zeynep Madak Erdogan
- Department of Food Science and Human Nutrition, University of Illinois, Urbana-Champaign, Urbana, IL, USA
- Cancer Center at Illinois, University of Illinois, Urbana-Champaign, Urbana, IL, USA
- Institute for Genomic Biology, University of Illinois, Urbana-Champaign, Urbana, IL, USA
- Division of Nutritional Sciences, University of Illinois, Urbana-Champaign, Urbana, IL, USA
- Department of Biomedical and Translational Sciences, Carle Illinois, College of Medicine, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
23
|
Cha HG, Kim DG, Chang J, Song Y, Jeong S, Nam SM, Wee SY, Cho KW, Choi CY. "Fasting: An Effective Preconditioning Method to Increase Fat Graft Survival". Aesthetic Plast Surg 2021; 46:1439-1449. [PMID: 34676429 DOI: 10.1007/s00266-021-02630-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/10/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Most preconditioning techniques before fat grafting require external manipulation. Since nutrition is the main factor maintaining the balance of lipogenesis and lipolysis, we hypothesized that fasting before undergoing autologous fat grafting may increase lipolysis and reduce adipocyte size, thereby improving the fat graft survival rate. METHODS C57BL/6 mice were divided into 24 h starved or fed groups. Adipose tissue lipolysis, adipogenesis, and angiogenesis-related gene expression, in fat from both groups, were analyzed. The volume and weight of the grafted fat at 4-8 weeks postoperatively were measured using micro-computed tomography. Immunohistochemistry staining and mRNA expression analysis were also performed to evaluate the effect of fasting on fat graft survival. RESULTS Fasting decreased adipocyte size by inducing adipose tissue lipolysis. Adipogenesis-related genes were remarkably downregulated while lipolysis-related genes and angiogenesis inducer genes were significantly upregulated in the starved adipose tissue. The mice grafted with the fat from the 24 h starved group had approximately 20% larger volumes and considerably heavier weights than those from the fed group. Increased viable adipocytes and vessels, and reduced macrophages in the fat grafts obtained from the 24 h starved group were also observed. CONCLUSIONS Fasting for 24 h before harvesting fat increased the retention volume of fat graft by increasing angiogenesis via VEGF induction. Therefore, fasting would be a novel and reliable preconditioning strategy to improve graft survival in autologous fat grafting. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266.
Collapse
Affiliation(s)
- Han Gyu Cha
- Department of Plastic and Reconstructive Surgery, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, 170 Jomaru-ro, Bucheon-si, Gyeonggi-do, Korea
| | - Dong Gyu Kim
- Department of Plastic and Reconstructive Surgery, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, 170 Jomaru-ro, Bucheon-si, Gyeonggi-do, Korea
| | - Jiyeon Chang
- Department of Integrated of Biomedical Science, Soonchunhyang University, Cheonan, Korea
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan, Korea
| | - Yuri Song
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan, Korea
| | - Seongfeel Jeong
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan, Korea
| | - Seung Min Nam
- Department of Plastic and Reconstructive Surgery, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, 170 Jomaru-ro, Bucheon-si, Gyeonggi-do, Korea
| | - Syeo Young Wee
- Department of Plastic and Reconstructive Surgery, Soonchunhyang University Gumi Hospital, Soonchunhyang University College of Medicine, Gumi, Korea
| | - Kae Won Cho
- Department of Integrated of Biomedical Science, Soonchunhyang University, Cheonan, Korea.
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan, Korea.
| | - Chang Yong Choi
- Department of Plastic and Reconstructive Surgery, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, 170 Jomaru-ro, Bucheon-si, Gyeonggi-do, Korea.
| |
Collapse
|
24
|
Li Q, Hagberg CE, Silva Cascales H, Lang S, Hyvönen MT, Salehzadeh F, Chen P, Alexandersson I, Terezaki E, Harms MJ, Kutschke M, Arifen N, Krämer N, Aouadi M, Knibbe C, Boucher J, Thorell A, Spalding KL. Obesity and hyperinsulinemia drive adipocytes to activate a cell cycle program and senesce. Nat Med 2021; 27:1941-1953. [PMID: 34608330 DOI: 10.1038/s41591-021-01501-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 08/12/2021] [Indexed: 01/10/2023]
Abstract
Obesity is considered an important factor for many chronic diseases, including diabetes, cardiovascular disease and cancer. The expansion of adipose tissue in obesity is due to an increase in both adipocyte progenitor differentiation and mature adipocyte cell size. Adipocytes, however, are thought to be unable to divide or enter the cell cycle. We demonstrate that mature human adipocytes unexpectedly display a gene and protein signature indicative of an active cell cycle program. Adipocyte cell cycle progression associates with obesity and hyperinsulinemia, with a concomitant increase in cell size, nuclear size and nuclear DNA content. Chronic hyperinsulinemia in vitro or in humans, however, is associated with subsequent cell cycle exit, leading to a premature senescent transcriptomic and secretory profile in adipocytes. Premature senescence is rapidly becoming recognized as an important mediator of stress-induced tissue dysfunction. By demonstrating that adipocytes can activate a cell cycle program, we define a mechanism whereby mature human adipocytes senesce. We further show that by targeting the adipocyte cell cycle program using metformin, it is possible to influence adipocyte senescence and obesity-associated adipose tissue inflammation.
Collapse
Affiliation(s)
- Qian Li
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Carolina E Hagberg
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.,Cardiovascular Medicine Division, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Helena Silva Cascales
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Shuai Lang
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Mervi T Hyvönen
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.,School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Firoozeh Salehzadeh
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Ping Chen
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.,Center for Infectious Medicine, Department of Medicine, Karolinska Institute, Stockolm, Sweden
| | - Ida Alexandersson
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Eleni Terezaki
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Matthew J Harms
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Maria Kutschke
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.,Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Nahida Arifen
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Niels Krämer
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Myriam Aouadi
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.,Center for Infectious Medicine, Department of Medicine, Karolinska Institute, Stockolm, Sweden
| | - Carole Knibbe
- CarMeN Laboratory, Lyon University, INRIA, INSA Lyon, Lyon, France
| | - Jeremie Boucher
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.,The Lundberg Laboratory for Diabetes Research, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Anders Thorell
- Department of Clinical Science, Danderyds Hospital, Karolinska Institutet and Department of Surgery, Ersta Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Kirsty L Spalding
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden. .,Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
25
|
Molitor M, Trávníčková M, Měšťák O, Christodoulou P, Sedlář A, Bačáková L, Lucchina S. The Influence of High and Low Negative Pressure Liposuction and Various Harvesting Techniques on the Viability and Function of Harvested Cells-a Systematic Review of Animal and Human Studies. Aesthetic Plast Surg 2021; 45:2379-2394. [PMID: 33876289 DOI: 10.1007/s00266-021-02249-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 03/16/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND An understanding of fat grafting methodology, techniques and patient-related factors is crucial when considering fat grafting. Multiple factors can influence the success of a fat graft and consequently the outcome of the procedure. The aim of this systematic review is to elucidate the influence of negative pressure and various techniques of fat harvesting on the viability and function of cells, particularly adipocytes and adipose-derived stem cells. METHODS We conducted a literature search from 1975 to 2020 using the PubMed bibliography, ScienceDirect, SCOPUS and the Google Scholar databases which produced 168,628 articles on the first pass. After applying all the exclusion criteria by two independent reviewers, we were left with 21 articles (level IV of Oxford Centre for Evidence-Based Studies and Grade C of Grade Practice Recommendation from the American Society of Plastic Surgeons) on which this review is based. RESULTS From 11 studies focused on different negative pressures, no one found using high negative pressure advantageous. Summarising 13 studies focused on various harvesting techniques (excision, syringe, and pump-machine), most often equal results were reported, followed by excision being better than either syringe or liposuction. CONCLUSION From our systematic review, we can conclude that the low negative pressure seems to yield better results and that the excision seems to be the most sparing method for fat graft harvesting. However, we have to point out that this conclusion is based on a very limited number of statistically challengeable articles and we recommend well-conducted further research. LEVEL OF EVIDENCE III This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Martin Molitor
- Department of Plastic Surgery, First Faculty of Medicine, Charles University and Na Bulovce Hospital, Budinova 67/2, 180 81, Prague 8-Liben, Czech Republic.
| | - Martina Trávníčková
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague 4-Krc, Czech Republic
| | - Ondřej Měšťák
- Department of Plastic Surgery, First Faculty of Medicine, Charles University and Na Bulovce Hospital, Budinova 67/2, 180 81, Prague 8-Liben, Czech Republic
| | - Petros Christodoulou
- Department of Plastic Surgery, First Faculty of Medicine, Charles University and Na Bulovce Hospital, Budinova 67/2, 180 81, Prague 8-Liben, Czech Republic
| | - Antonín Sedlář
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague 4-Krc, Czech Republic
| | - Lucie Bačáková
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague 4-Krc, Czech Republic
| | - Stefano Lucchina
- Hand Unit, General Surgery Department, Locarno's Regional Hospital, Via Ospedale 1, 6600, Locarno, Switzerland
| |
Collapse
|
26
|
Abstract
This Review focuses on the mechanistic evidence for a link between obesity, dysregulated cellular metabolism and breast cancer. Strong evidence now links obesity with the development of 13 different types of cancer, including oestrogen receptor-positive breast cancer in postmenopausal women. A number of local and systemic changes are hypothesized to support this relationship, including increased circulating levels of insulin and glucose as well as adipose tissue-derived oestrogens, adipokines and inflammatory mediators. Metabolic pathways of energy production and utilization are dysregulated in tumour cells and this dysregulation is a newly accepted hallmark of cancer. Dysregulated metabolism is also hypothesized to be a feature of non-neoplastic cells in the tumour microenvironment. Obesity-associated factors regulate metabolic pathways in both breast cancer cells and cells in the breast microenvironment, which provides a molecular link between obesity and breast cancer. Consequently, interventions that target these pathways might provide a benefit in postmenopausal women and individuals with obesity, a population at high risk of breast cancer.
Collapse
Affiliation(s)
- Kristy A Brown
- Sandra and Edward Meyer Cancer Center and Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
27
|
Prantl L, Eigenberger A, Brix E, Kempa S, Baringer M, Felthaus O. Adipose Tissue-Derived Stem Cell Yield Depends on Isolation Protocol and Cell Counting Method. Cells 2021; 10:cells10051113. [PMID: 34063138 PMCID: PMC8148142 DOI: 10.3390/cells10051113] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 12/15/2022] Open
Abstract
In plastic surgery, lipofilling is a frequent procedure. Unsatisfactory vascularization and impaired cell vitality can lead to unpredictable take rates in the fat graft. The proliferation and neovascularization inducing properties of adipose tissue-derived stem cells may contribute to solve this problem. Therefore, the enrichment of fat grafts with stem cells is studied intensively. However, it is difficult to compare these studies because many factors—often not precisely described—are influencing the results. Our study summarizes some factors which influence the cell yield like harvesting, isolation procedure and quantification. Stem cells were isolated after liposuction. Quantification was done using a cell chamber, colony counting, or flow cytometry with changes to one parameter, only, for each comparison. Quantification of cells isolated after liposuction at the same harvesting site from the same patient can vary greatly depending on the details of the isolation protocol and the method of quantification. Cell yield can be influenced strongly by many factors. Therefore, a comparison of different studies should be handled with care.
Collapse
|
28
|
Majumdar A, Giambartolomei C, Cai N, Haldar T, Schwarz T, Gandal M, Flint J, Pasaniuc B. Leveraging eQTLs to identify individual-level tissue of interest for a complex trait. PLoS Comput Biol 2021; 17:e1008915. [PMID: 34019542 PMCID: PMC8174686 DOI: 10.1371/journal.pcbi.1008915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 06/03/2021] [Accepted: 03/26/2021] [Indexed: 12/26/2022] Open
Abstract
Genetic predisposition for complex traits often acts through multiple tissues at different time points during development. As a simple example, the genetic predisposition for obesity could be manifested either through inherited variants that control metabolism through regulation of genes expressed in the brain, or that control fat storage through dysregulation of genes expressed in adipose tissue, or both. Here we describe a statistical approach that leverages tissue-specific expression quantitative trait loci (eQTLs) corresponding to tissue-specific genes to prioritize a relevant tissue underlying the genetic predisposition of a given individual for a complex trait. Unlike existing approaches that prioritize relevant tissues for the trait in the population, our approach probabilistically quantifies the tissue-wise genetic contribution to the trait for a given individual. We hypothesize that for a subgroup of individuals the genetic contribution to the trait can be mediated primarily through a specific tissue. Through simulations using the UK Biobank, we show that our approach can predict the relevant tissue accurately and can cluster individuals according to their tissue-specific genetic architecture. We analyze body mass index (BMI) and waist to hip ratio adjusted for BMI (WHRadjBMI) in the UK Biobank to identify subgroups of individuals whose genetic predisposition act primarily through brain versus adipose tissue, and adipose versus muscle tissue, respectively. Notably, we find that these individuals have specific phenotypic features beyond BMI and WHRadjBMI that distinguish them from random individuals in the data, suggesting biological effects of tissue-specific genetic contribution for these traits.
Collapse
Affiliation(s)
- Arunabha Majumdar
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
- Department of Mathematics, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Claudia Giambartolomei
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Na Cai
- Wellcome Sanger Institute, Wellcome genome campus, Hinxton, United Kingdom
- European Bioinformatics Institute (EMBL-EBI), Wellcome genome campus, Hinxton, United Kingdom
| | - Tanushree Haldar
- Institute for Human Genetics, University of California, San Francisco, California, United States of America
| | - Tommer Schwarz
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, California, United States of America
| | - Michael Gandal
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Jonathan Flint
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Bogdan Pasaniuc
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, California, United States of America
| |
Collapse
|
29
|
Andjelkov K, Conde-Green A, Mosahebi A. Smoking and Physical Activity Significantly Influence Stromal Vascular Fraction Cell Yield and Viability. Aesthetic Plast Surg 2021; 45:315-321. [PMID: 33083844 DOI: 10.1007/s00266-020-02008-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 10/04/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Subcutaneous tissue is an abundant source of adipose-derived regenerative cells. It is readily available and easy to extract by means of liposuction, making it one of the most popular sources for tissue engineering and regenerative medical applications. METHODS The stromal vascular fraction (SVF) cell yield and viability of the lipoaspirate obtained from 43 patients undergoing elective liposuction were examined in correlation with their age, gender, body mass index, smoking status, and physical activity. The lipoaspirate was processed with the Celution® 800/CRS system to isolate the SVF and a few drops of the obtained pellet were used for cell counting with NecleoCounter® NC-100TM. RESULTS Twenty-eight (65.1%) were men and 15 (34.9%) were women with an average age of 40.7 ± 10.4 years (women) and 38.9 ± 11.8 years (men). Viable SVF cells/g fat was significantly correlated with smoking level (negative correlation, ρ= - 0.312, P < 0.05) and with marginal significance with female gender. Cell viability showed a significant negative correlation with physical activity level (ρ = - 0.432, P < 0.01); borderline significance for correlation of this parameter with smoking level should not be neglected. Other parameters did not influence the cell yield nor the viability of the stromal vascular fraction. CONCLUSION Many factors may influence SVF cell yield and viability. Our findings indicate that age and smoking significantly influenced SVF cell yield, age positively while smoking negatively. Increased physical activity had a negative correlation with SVF cell viability. LEVEL OF EVIDENCE N/A This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Katarina Andjelkov
- Faculty of Medicine, University of Belgrade, BelPrime Clinic, 16 Brane Crncevica, 11000, Belgrade, Serbia.
| | | | | |
Collapse
|
30
|
Dufau J, Shen JX, Couchet M, De Castro Barbosa T, Mejhert N, Massier L, Griseti E, Mouisel E, Amri EZ, Lauschke VM, Rydén M, Langin D. In vitro and ex vivo models of adipocytes. Am J Physiol Cell Physiol 2021; 320:C822-C841. [PMID: 33439778 DOI: 10.1152/ajpcell.00519.2020] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adipocytes are specialized cells with pleiotropic roles in physiology and pathology. Several types of fat cells with distinct metabolic properties coexist in various anatomically defined fat depots in mammals. White, beige, and brown adipocytes differ in their handling of lipids and thermogenic capacity, promoting differences in size and morphology. Moreover, adipocytes release lipids and proteins with paracrine and endocrine functions. The intrinsic properties of adipocytes pose specific challenges in culture. Mature adipocytes float in suspension culture due to high triacylglycerol content and are fragile. Moreover, a fully differentiated state, notably acquirement of the unilocular lipid droplet of white adipocyte, has so far not been reached in two-dimensional culture. Cultures of mouse and human-differentiated preadipocyte cell lines and primary cells have been established to mimic white, beige, and brown adipocytes. Here, we survey various models of differentiated preadipocyte cells and primary mature adipocyte survival describing main characteristics, culture conditions, advantages, and limitations. An important development is the advent of three-dimensional culture, notably of adipose spheroids that recapitulate in vivo adipocyte function and morphology in fat depots. Challenges for the future include isolation and culture of adipose-derived stem cells from different anatomic location in animal models and humans differing in sex, age, fat mass, and pathophysiological conditions. Further understanding of fat cell physiology and dysfunction will be achieved through genetic manipulation, notably CRISPR-mediated gene editing. Capturing adipocyte heterogeneity at the single-cell level within a single fat depot will be key to understanding diversities in cardiometabolic parameters among lean and obese individuals.
Collapse
Affiliation(s)
- Jérémy Dufau
- Inserm, Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR1297, Toulouse, France.,Faculté de Médecine, I2MC, UMR1297, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Joanne X Shen
- Karolinska Institutet, Department of Physiology and Pharmacology, Stockholm, Sweden
| | - Morgane Couchet
- Karolinska Institutet, Department of Medicine (H7), Stockholm, Sweden
| | | | - Niklas Mejhert
- Karolinska Institutet, Department of Medicine (H7), Stockholm, Sweden
| | - Lucas Massier
- Karolinska Institutet, Department of Medicine (H7), Stockholm, Sweden
| | - Elena Griseti
- Inserm, Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR1297, Toulouse, France.,Faculté de Médecine, I2MC, UMR1297, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Etienne Mouisel
- Inserm, Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR1297, Toulouse, France.,Faculté de Médecine, I2MC, UMR1297, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | | | - Volker M Lauschke
- Karolinska Institutet, Department of Physiology and Pharmacology, Stockholm, Sweden
| | - Mikael Rydén
- Karolinska Institutet, Department of Medicine (H7), Stockholm, Sweden
| | - Dominique Langin
- Inserm, Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR1297, Toulouse, France.,Faculté de Médecine, I2MC, UMR1297, Université de Toulouse, Université Paul Sabatier, Toulouse, France.,Toulouse University Hospitals, Department of Biochemistry, Toulouse, France
| |
Collapse
|
31
|
A distribution-centered approach for analyzing human adipocyte size estimates and their association with obesity-related traits and mitochondrial function. Int J Obes (Lond) 2021; 45:2108-2117. [PMID: 34172828 PMCID: PMC8380540 DOI: 10.1038/s41366-021-00883-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/10/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Cell diameter, area, and volume are established quantitative measures of adipocyte size. However, these different adipocyte sizing parameters have not yet been directly compared regarding their distributions. Therefore, the study aimed to investigate how these adipocyte size measures differ in their distribution and assessed their correlation with anthropometry and laboratory chemistry. In addition, we were interested to investigate the relationship between fat cell size and adipocyte mitochondrial respiratory chain capacity. METHODS Subcutaneous and visceral histology-based adipocyte size estimates from 188 individuals were analyzed by applying a panel of parameters to describe the underlying cell population. Histology-based adipocyte diameter distributions were compared with adipocyte diameter distributions from collagenase digestion. Associations of mean adipocyte size with body mass index (BMI), glucose, HbA1C, blood lipids as well as mature adipocyte mitochondrial respiration were investigated. RESULTS All adipocyte area estimates derived from adipose tissue histology were not normally distributed, but rather characterized by positive skewness. The shape of the size distribution depends on the adipocyte sizing parameter and on the method used to determine adipocyte size. Despite different distribution shapes histology-derived adipocyte area, diameter, volume, and surface area consistently showed positive correlations with BMI. Furthermore, associations between adipocyte sizing parameters and glucose, HbA1C, or HDL specifically in the visceral adipose depot were revealed. Increasing subcutaneous adipocyte diameter was negatively correlated with adipocyte mitochondrial respiration. CONCLUSIONS Despite different underlying size distributions, the correlation with obesity-related traits was consistent across adipocyte sizing parameters. Decreased mitochondrial respiratory capacity with increasing subcutaneous adipocyte diameter could display a novel link between adipocyte hypertrophy and adipose tissue function.
Collapse
|
32
|
Adipose-Derived Stem Cells from Obese Donors Polarize Macrophages and Microglia toward a Pro-Inflammatory Phenotype. Cells 2020; 10:cells10010026. [PMID: 33375695 PMCID: PMC7823699 DOI: 10.3390/cells10010026] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/20/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
Macrophages and microglia represent the primary phagocytes and first line of defense in the peripheral and central immune systems. They activate and polarize into a spectrum of pro- and anti-inflammatory phenotypes in response to various stimuli. This activation is tightly regulated to balance the appropriate immune response with tissue repair and homeostasis. Disruption of this balance results in inflammatory disease states and tissue damage. Adipose stem cells (ASCs) have great therapeutic potential because of the potent immunomodulatory capabilities which induce the polarization of microglia and macrophages to the anti-inflammatory, M2, phenotype. In this study, we examined the effects of donor heterogeneity on ASC function. Specifically, we investigated the impact of donor obesity on ASC stemness and immunomodulatory abilities. Our findings revealed that ASCs from obese donors (ObASCs) exhibited reduced stem cell characteristics when compared to ASCs from lean donors (LnASCs). We also found that ObASCs promote a pro-inflammatory phenotype in murine macrophage and microglial cells, as indicated by the upregulated expression of pro-inflammatory genes, increased nitric oxide pathway activity, and impaired phagocytosis and migration. These findings highlight the importance of considering individual donor characteristics such as obesity when selecting donors and cells for use in ASC therapeutic applications and regenerative medicine.
Collapse
|
33
|
Miao Z, Alvarez M, Ko A, Bhagat Y, Rahmani E, Jew B, Heinonen S, Muñoz-Hernandez LL, Herrera-Hernandez M, Aguilar-Salinas C, Tusie-Luna T, Mohlke KL, Laakso M, Pietiläinen KH, Halperin E, Pajukanta P. The causal effect of obesity on prediabetes and insulin resistance reveals the important role of adipose tissue in insulin resistance. PLoS Genet 2020; 16:e1009018. [PMID: 32925908 PMCID: PMC7515203 DOI: 10.1371/journal.pgen.1009018] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 09/24/2020] [Accepted: 07/29/2020] [Indexed: 01/06/2023] Open
Abstract
Reverse causality has made it difficult to establish the causal directions between obesity and prediabetes and obesity and insulin resistance. To disentangle whether obesity causally drives prediabetes and insulin resistance already in non-diabetic individuals, we utilized the UK Biobank and METSIM cohort to perform a Mendelian randomization (MR) analyses in the non-diabetic individuals. Our results suggest that both prediabetes and systemic insulin resistance are caused by obesity (p = 1.2×10-3 and p = 3.1×10-24). As obesity reflects the amount of body fat, we next studied how adipose tissue affects insulin resistance. We performed both bulk RNA-sequencing and single nucleus RNA sequencing on frozen human subcutaneous adipose biopsies to assess adipose cell-type heterogeneity and mitochondrial (MT) gene expression in insulin resistance. We discovered that the adipose MT gene expression and body fat percent are both independently associated with insulin resistance (p≤0.05 for each) when adjusting for the decomposed adipose cell-type proportions. Next, we showed that these 3 factors, adipose MT gene expression, body fat percent, and adipose cell types, explain a substantial amount (44.39%) of variance in insulin resistance and can be used to predict it (p≤2.64×10-5 in 3 independent human cohorts). In summary, we demonstrated that obesity is a strong determinant of both prediabetes and insulin resistance, and discovered that individuals' adipose cell-type composition, adipose MT gene expression, and body fat percent predict their insulin resistance, emphasizing the critical role of adipose tissue in systemic insulin resistance.
Collapse
Affiliation(s)
- Zong Miao
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, California, United States of America
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Arthur Ko
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Yash Bhagat
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Elior Rahmani
- Computer Science Department in the School of Engineering, UCLA, Los Angeles, California, United States of America
| | - Brandon Jew
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, California, United States of America
- Computer Science Department in the School of Engineering, UCLA, Los Angeles, California, United States of America
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Linda Liliana Muñoz-Hernandez
- Unidad de Investigación en Enfermedades Metabólicas, Dirección de Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Departamento de Endocrinología y Metabolismo, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo Leon, México
| | - Miguel Herrera-Hernandez
- Departamento de Cirugía, Instituto Nacional de Ciencias Médicas y Nutrición, Mexico City, Mexico
| | - Carlos Aguilar-Salinas
- Unidad de Investigación en Enfermedades Metabólicas, Dirección de Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Departamento de Endocrinología y Metabolismo, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo Leon, México
| | - Teresa Tusie-Luna
- Unidad de Biología Molecular y Medicina Genómica Instituto de Investigaciones Biomédicas UNAM / Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, Mexico City, Mexico
| | - Karen L. Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Kirsi H. Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Obesity Center, Endocrinology, Abdominal Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Eran Halperin
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Computer Science Department in the School of Engineering, UCLA, Los Angeles, California, United States of America
- Department of Computational Medicine, UCLA, Los Angeles, California, United States of America
- Department of Anesthesiology and Perioperative Medicine, UCLA, Los Angeles, California, United States of America
- Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, California, United States of America
- Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
34
|
Effect of Breast Cancer and Adjuvant Therapy on Adipose-Derived Stromal Cells: Implications for the Role of ADSCs in Regenerative Strategies for Breast Reconstruction. Stem Cell Rev Rep 2020; 17:523-538. [PMID: 32929604 DOI: 10.1007/s12015-020-10038-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2020] [Indexed: 12/14/2022]
Abstract
Tissue engineering using Adipose Derived Stromal Cells (ADSCs) has emerged as a novel regenerative medicine approach to replace and reconstruct soft tissue damaged or lost as a result of disease process or therapeutic surgical resection. ADSCs are an attractive cell source for soft tissue regeneration due to the fact that they are easily accessible, multipotent, non-immunogenic and pro-angiogenic. ADSC based regenerative strategies have been successfully translated to the clinical setting for the treatment of Crohn's fistulae, musculoskeletal pathologies, wound healing, and cosmetic breast augmentation (fat grafting). ADSCs are particularly attractive as a source for adipose tissue engineering as they exhibit preferential differentiation to adipocytes and support maintenance of mature adipose graft volume. The potential for reconstruction with an autologous tissue sources and a natural appearance and texture is particularly appealing in the setting of breast cancer; up to 40% of patients require mastectomy for locoregional control and current approaches to post-mastectomy breast reconstruction (PMBR) are limited by the potential for complications at the donor and reconstruction sites. Despite their potential, the use of ADSCs in breast cancer patients is controversial due to concerns regarding oncological safety. These concerns relate to the regeneration of tissue at a site where a malignancy has been treated and the impact this may have on stimulating local disease recurrence or dissemination. Pre-clinical data suggest that ADSCs exhibit pro-oncogenic characteristics and are involved in stimulating progression, and growth of tumour cells. However, there have been conflicting reports on the oncologic outcome, in terms of locoregional recurrence, for breast cancer patients in whom ADSC enhanced fat grafting was utilised as an alternative to reconstruction for small volume defects. A further consideration which may impact the successful translation of ADSC based regenerative strategies for post cancer reconstruction is the potential effects of cancer therapy. This review aims to address the effect of malignant cells, adjuvant therapies and patient-specific factors that may influence the success of regenerative strategies using ADSCs for post cancer tissue regeneration.
Collapse
|
35
|
Yu JS, Sahar NE, Bi YR, Jung K, Pang C, Huh JY, Kim KH. The Effects of Triterpenoid Saponins from the Seeds of Momordica cochinchinensis on Adipocyte Differentiation and Mature Adipocyte Inflammation. PLANTS 2020; 9:plants9080984. [PMID: 32756459 PMCID: PMC7466054 DOI: 10.3390/plants9080984] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/27/2020] [Accepted: 07/31/2020] [Indexed: 01/20/2023]
Abstract
Obesity is a medical condition in which abnormal or excessive fat accumulates to an extent that is associated with various diseases. In our ongoing research to figure out natural products with anti-obesity effects, a phytochemical investigation of the EtOH extract of the seeds of Momordica cochinchinensis was carried out, which resulted in the isolation of two major triterpenoid saponins: gypsogenin 3-O-β-d-galactopyranosyl(1→2)-[α-l-rhamnopyranosyl (1→3)]-β-d-glucuronopyranoside (1) and quillaic acid 3-O-β-d-galactopyranosyl(1→2)-[α-l-rhamnopyranosyl(1→3)]-β-d-glucuronopyranoside (2). Then, the effects of the isolated triterpenoid saponins (1 and 2) on adipocyte differentiation were evaluated, and it was demonstrated that the isolated saponin (1) showed inhibitory effects on adipogenesis. In mature adipocytes, the isolated saponin (1) reversed tumor necrosis factor α (TNFα)-induced proinflammatory cytokine gene expression. Additionally, the isolated saponin (1) reduced lipolytic gene expression leading to decreased basal lipolysis activity. Collectively, these findings suggest that saponin (1) of M. cochinchinensis exerts beneficial effects in the regulation of adipogenesis and adipocyte inflammation and could be a potential therapeutic alternative in the treatment of obesity-induced metabolic diseases.
Collapse
Affiliation(s)
- Jae Sik Yu
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea;
| | - Namood E. Sahar
- College of Pharmacy, Chonnam National University, Gwangju 61186, Korea; (N.E.S.); (Y.-R.B.)
| | - Yan-Ran Bi
- College of Pharmacy, Chonnam National University, Gwangju 61186, Korea; (N.E.S.); (Y.-R.B.)
| | - Kiwon Jung
- Institute of Pharmaceutical Sciences, College of Pharmacy, CHA University, Seongnam 13488, Korea;
| | - Changhyun Pang
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea;
| | - Joo Young Huh
- College of Pharmacy, Chonnam National University, Gwangju 61186, Korea; (N.E.S.); (Y.-R.B.)
- Correspondence: (J.Y.H.); (K.H.K.); Tel.: +(82)-62-530-2938 (J.Y.H.); +(82)-31-290-7700 (K.H.K.)
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea;
- Correspondence: (J.Y.H.); (K.H.K.); Tel.: +(82)-62-530-2938 (J.Y.H.); +(82)-31-290-7700 (K.H.K.)
| |
Collapse
|
36
|
Yeo CR, Agrawal M, Hoon S, Shabbir A, Shrivastava MK, Huang S, Khoo CM, Chhay V, Shabeer M, Shyong Tai E, Vidal-Puig A, Toh SA. Reply to: 'Browning capabilities of human primary adipose-derived stromal cells compared to SGBS cells'. Sci Rep 2020; 10:9634. [PMID: 32541770 PMCID: PMC7295789 DOI: 10.1038/s41598-020-64706-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 04/16/2020] [Indexed: 11/09/2022] Open
Affiliation(s)
- Chia Rou Yeo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore
| | - Madhur Agrawal
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore
| | - Shawn Hoon
- Molecular Engineering Laboratory, Biomedical Sciences Institutes, A*Star, 138668, Singapore, Singapore
| | - Asim Shabbir
- Department of Surgery, National University Hospital, 119074, Singapore, Singapore
| | - Manu Kunaal Shrivastava
- Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QC, UK
| | - Shiqi Huang
- Food Science and Technology Program, Department of Chemistry, National University of Singapore, 117542, Singapore, Singapore
| | - Chin Meng Khoo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore
| | - Vanna Chhay
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore
| | - Muhammad Shabeer
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore
| | - E Shyong Tai
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore
| | - Antonio Vidal-Puig
- Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QC, UK
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Sue-Anne Toh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore.
| |
Collapse
|
37
|
Zhang L, Virgous C, Si H. How Does Ginsenoside Rh2 Mitigate Adipogenesis in Cultured Cells and Obese Mice? Molecules 2020; 25:E2412. [PMID: 32455850 PMCID: PMC7287807 DOI: 10.3390/molecules25102412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/02/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023] Open
Abstract
Ginsenoside Rh2, an intermediate metabolite of ginseng, but not naturally occurring, has recently drawn attention because of its anticancer effect. However, it is not clear if and how Rh2 inhibits preadipocytes differentiation. In the present study, we hypothesized that ginsenoside Rh2 attenuates adipogenesis through regulating the peroxisome proliferator-activated receptor gamma (PPAR-γ) pathway both in cells and obese mice. Different concentrations of Rh2 were applied both in 3T3-L1 cells and human primary preadipocytes to determine if Rh2 inhibits cell differentiation. Dietary Rh2 was administered to obese mice to determine if Rh2 prevents obesity in vivo. The mRNA and protein expression of PPAR-γ pathway molecules in cells and tissues were measured by real-time polymerase chain reaction (RT-PCR) and Western blot, respectively. Our results show that Rh2 dose-dependently (30-60 μM) inhibited cell differentiation in 3T3-L1 cells (44.5% ± 7.8% of control at 60 μM). This inhibitory effect is accompanied by the attenuation of the protein and/or mRNA expression of adipogenic markers including PPAR-γ and CCAAT/enhancer binding protein alpha, fatty acid synthase, fatty acid binding protein 4, and perilipin significantly (p < 0.05). Moreover, Rh2 significantly (p < 0.05) inhibited differentiation in human primary preadipocytes at much lower concentrations (5-15 μM). Furthermore, dietary intake of Rh2 (0.1 g Rh2/kg diet, w/w for eight weeks) significantly (p < 0.05) reduced protein PPAR-γ expression in liver and hepatic glutathione reductase and lowered fasting blood glucose. These results suggest that ginsenoside Rh2 dose-dependently inhibits adipogenesis through down-regulating the PPAR-γ pathway, and Rh2 may be a potential agent in preventing obesity in vivo.
Collapse
Affiliation(s)
- Longyun Zhang
- Department of Human Sciences, Tennessee State University, Nashville, TN 37209, USA;
| | - Carlos Virgous
- Animal Care Facility, Meharry Medical College, Nashville, TN 37208, USA;
| | - Hongwei Si
- Department of Human Sciences, Tennessee State University, Nashville, TN 37209, USA;
| |
Collapse
|
38
|
Sabol RA, Villela VA, Denys A, Freeman BT, Hartono AB, Wise RM, Harrison MAA, Sandler MB, Hossain F, Miele L, Bunnell BA. Obesity-Altered Adipose Stem Cells Promote Radiation Resistance of Estrogen Receptor Positive Breast Cancer through Paracrine Signaling. Int J Mol Sci 2020; 21:ijms21082722. [PMID: 32326381 PMCID: PMC7216284 DOI: 10.3390/ijms21082722] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/10/2020] [Accepted: 04/10/2020] [Indexed: 12/17/2022] Open
Abstract
Obesity is associated with poorer responses to chemo- and radiation therapy for breast cancer, which leads to higher mortality rates for obese women who develop breast cancer. Adipose stem cells (ASCs) are an integral stromal component of the tumor microenvironment (TME). In this study, the effects of obesity-altered ASCs (obASCs) on estrogen receptor positive breast cancer cell’s (ER+BCCs) response to radiotherapy (RT) were evaluated. We determined that BCCs had a decreased apoptotic index and increased surviving fraction following RT when co-cultured with obASCs compared to lnASCs or non-co-cultured cells. Further, obASCs reduced oxidative stress and induced IL-6 expression in co-cultured BCCs after radiation. obASCs produce increased levels of leptin relative to ASCs from normal-weight individuals (lnASCs). obASCs upregulate the expression of IL-6 compared to non-co-cultured BCCs, but BCCs co-cultured with leptin knockdown obASCs did not upregulate IL-6. The impact of shLeptin obASCs on radiation resistance of ER+BCCs demonstrate a decreased radioprotective ability compared to shControl obASCs. Key NOTCH signaling players were enhanced in ER+BBCs following co-culture with shCtrl obASCs but not shLep obASCs. This work demonstrates that obesity-altered ASCs, via enhanced secretion of leptin, promote IL-6 and NOTCH signaling pathways in ER+BCCs leading to radiation resistance.
Collapse
Affiliation(s)
- Rachel A. Sabol
- Center for Stem Cell Research, Tulane University School of Medicine, New Orleans, LA 70112, USA; (R.A.S.); (V.A.V.); (A.D.); (R.M.W.); (M.A.A.H.); (M.B.S.)
| | - Vidal A. Villela
- Center for Stem Cell Research, Tulane University School of Medicine, New Orleans, LA 70112, USA; (R.A.S.); (V.A.V.); (A.D.); (R.M.W.); (M.A.A.H.); (M.B.S.)
| | - Alexandra Denys
- Center for Stem Cell Research, Tulane University School of Medicine, New Orleans, LA 70112, USA; (R.A.S.); (V.A.V.); (A.D.); (R.M.W.); (M.A.A.H.); (M.B.S.)
| | - Benjamin T. Freeman
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA 70112, USA;
| | - Alifiani B. Hartono
- Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA;
| | - Rachel M. Wise
- Center for Stem Cell Research, Tulane University School of Medicine, New Orleans, LA 70112, USA; (R.A.S.); (V.A.V.); (A.D.); (R.M.W.); (M.A.A.H.); (M.B.S.)
| | - Mark A. A. Harrison
- Center for Stem Cell Research, Tulane University School of Medicine, New Orleans, LA 70112, USA; (R.A.S.); (V.A.V.); (A.D.); (R.M.W.); (M.A.A.H.); (M.B.S.)
| | - Maxwell B. Sandler
- Center for Stem Cell Research, Tulane University School of Medicine, New Orleans, LA 70112, USA; (R.A.S.); (V.A.V.); (A.D.); (R.M.W.); (M.A.A.H.); (M.B.S.)
| | - Fokhrul Hossain
- Louisiana State University Health Sciences Center (LSUHSC), Department of Genetics, New Orleans, LA 70112, USA; (F.H.); (L.M.)
- Louisiana Cancer Research Center (LCRC), Stanley S. Scott Cancer Center, LSUSHC, New Orleans, LA 70112, USA
| | - Lucio Miele
- Louisiana State University Health Sciences Center (LSUHSC), Department of Genetics, New Orleans, LA 70112, USA; (F.H.); (L.M.)
- Louisiana Cancer Research Center (LCRC), Stanley S. Scott Cancer Center, LSUSHC, New Orleans, LA 70112, USA
| | - Bruce A. Bunnell
- Center for Stem Cell Research, Tulane University School of Medicine, New Orleans, LA 70112, USA; (R.A.S.); (V.A.V.); (A.D.); (R.M.W.); (M.A.A.H.); (M.B.S.)
- Department of Pharmacology, Tulane University, New Orleans, LA 70112, USA
- Division of Regenerative Medicine, Tulane National Primate Research Center, Covington, LA 70433, USA
- Correspondence: ; Tel.: +1-504-988-7071
| |
Collapse
|
39
|
Zhang J, Liu Y, Yin W, Hu X. Adipose-derived stromal cells in regulation of hematopoiesis. Cell Mol Biol Lett 2020; 25:16. [PMID: 32161623 PMCID: PMC7059705 DOI: 10.1186/s11658-020-00209-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 02/25/2020] [Indexed: 12/21/2022] Open
Abstract
Over the past decade, mesenchymal stromal cells (MSCs) found in the bone marrow microenvironment have been considered to be important candidates in cellular therapy. However, the application of MSCs in clinical settings is limited by the difficulty and low efficiency associated with the separation of MSCs from the bone marrow. Therefore, distinct sources of MSCs have been extensively explored. Adipose-derived stromal cells (ASCs), a cell line similar to MSCs, have been identified as a promising source. ASCs have become increasingly popular in many fields, as they can be conveniently extracted from fat tissue. This review focuses on the properties of ASCs in hematopoietic regulation and the underlying mechanisms, as well as the current applications and future perspectives in ASC-based therapy.
Collapse
Affiliation(s)
- Jing Zhang
- 1Department of Transfusion Medicine, Xijing Hospital, Xi'an, 710032 China
| | - Yunsheng Liu
- 2Department of Rocket Force Medicine, Third Military Medical University, Chongqing, 400038 China
| | - Wen Yin
- 1Department of Transfusion Medicine, Xijing Hospital, Xi'an, 710032 China
| | - Xingbin Hu
- 1Department of Transfusion Medicine, Xijing Hospital, Xi'an, 710032 China
| |
Collapse
|
40
|
Sato T, Vargas D, Miyazaki K, Uchida K, Ariyani W, Miyazaki M, Okada J, Lizcano F, Koibuchi N, Shimokawa N. EID1 suppresses lipid accumulation by inhibiting the expression of GPDH in 3T3-L1 preadipocytes. J Cell Physiol 2020; 235:6725-6735. [PMID: 32056205 DOI: 10.1002/jcp.29567] [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: 04/01/2019] [Accepted: 01/13/2020] [Indexed: 01/23/2023]
Abstract
The imbalance between food intake and energy expenditure causes high accumulation of triglycerides in adipocytes. Obesity is related with the increased lipid accumulation in white adipose tissue, which is a major risk factor for the development of metabolic disorders, such as type 2 diabetes and cardiovascular disease. This study highlights the role of E1A-like inhibitor of differentiation 1 (EID1) in the modulation of adipogenesis through the downregulation of glycerol-3-phosphate dehydrogenase (GPDH), which is a key enzyme in the synthesis of triglycerides and is considered to be a marker of adipogenesis. By analyzing DNA microarray data, we found that when EID1 is overexpressed in preadipocytes (3T3-L1 cells) during adipocyte differentiation, EID1 inhibits lipid accumulation through the downregulation of GPDH. In contrast, EID1 is not involved in the regulation of intracellular glucose via the translocation of glucose transporter. A confocal image analysis showed that EID1 is located in the nucleus of preadipocytes in the form of speckles, which could be involved as a regulator of the transcriptional process. We further confirmed that EID1 is able to bind to the promoter sequence of GPDH in the nucleus. These findings provide a molecular explanation for the inhibitory effect of EID1 on lipid accumulation in adipocytes.
Collapse
Affiliation(s)
- Tomohiko Sato
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan.,Department of Nutrition, Takasaki University of Health and Welfare, Takasaki, Gunma, Japan.,Department of Physical Therapy, Ota College of Medical Technology, Ota, Gunma, Japan
| | - Diana Vargas
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan.,Department of Nutrition, Takasaki University of Health and Welfare, Takasaki, Gunma, Japan.,Center of Biomedical Research, Universidad de La Sabana, Chia, Colombia
| | - Kakushin Miyazaki
- Department of Nutrition, Takasaki University of Health and Welfare, Takasaki, Gunma, Japan
| | - Kaoru Uchida
- Department of Nutrition, Takasaki University of Health and Welfare, Takasaki, Gunma, Japan
| | - Winda Ariyani
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Mitsue Miyazaki
- Department of Nutrition, Takasaki University of Health and Welfare, Takasaki, Gunma, Japan
| | - Junichi Okada
- Department of Physical Therapy, Ota College of Medical Technology, Ota, Gunma, Japan
| | - Fernando Lizcano
- Center of Biomedical Research, Universidad de La Sabana, Chia, Colombia
| | - Noriyuki Koibuchi
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Noriaki Shimokawa
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan.,Department of Nutrition, Takasaki University of Health and Welfare, Takasaki, Gunma, Japan
| |
Collapse
|
41
|
Vyas KS, Bole M, Vasconez HC, Banuelos JM, Martinez-Jorge J, Tran N, Lemaine V, Mardini S, Bakri K. Profile of Adipose-Derived Stem Cells in Obese and Lean Environments. Aesthetic Plast Surg 2019; 43:1635-1645. [PMID: 31267153 DOI: 10.1007/s00266-019-01397-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 05/04/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND With the demand for stem cells in regenerative medicine, new methods of isolating stem cells are highly sought. Adipose tissue is a readily available and non-controversial source of multipotent stem cells that carries a low risk for potential donors. However, elevated donor body mass index has been associated with an altered cellular microenvironment and thus has implications for stem cell efficacy in recipients. This review explored the literature on adipose-derived stem cells (ASCs) and the effect of donor obesity on cellular function. METHODS A review of published articles on obesity and ASCs was conducted with the PubMed database and the following search terms: obesity, overweight, adipose-derived stem cells and ASCs. Two investigators screened and reviewed the relevant abstracts. RESULTS There is agreement on reduced ASC function in response to obesity in terms of angiogenic differentiation, proliferation, migration, viability, and an altered and inflammatory transcriptome. Osteogenic differentiation and cell yield do not show reasonable agreement. Weight loss partially rescues some of the aforementioned features. CONCLUSIONS Generally, obesity reduces ASC qualities and may have an effect on the therapeutic value of ASCs. Because weight loss and some biomolecules have been shown to rescue these qualities, further research should be conducted on methods to return obese-derived ASCs to baseline. LEVEL V This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors- www.springer.com/00266.
Collapse
Affiliation(s)
- Krishna S Vyas
- Division of Plastic Surgery, Department of Surgery, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA.
| | - Madhav Bole
- Division of Orthopaedic Surgery, London Health Sciences Centre, University Hospital, 339 Windermere Rd., London, ON, N6A 5A5, Canada
| | - Henry C Vasconez
- Division of Plastic Surgery, University of Kentucky, Lexington, KY, USA
| | - Joseph M Banuelos
- Division of Plastic Surgery, Department of Surgery, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Jorys Martinez-Jorge
- Division of Plastic Surgery, Department of Surgery, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Nho Tran
- Division of Plastic Surgery, Department of Surgery, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Valerie Lemaine
- Division of Plastic Surgery, Department of Surgery, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Samir Mardini
- Division of Plastic Surgery, Department of Surgery, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Karim Bakri
- Division of Plastic Surgery, Department of Surgery, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| |
Collapse
|
42
|
Frasca D, Diaz A, Romero M, Blomberg BB. Leptin induces immunosenescence in human B cells. Cell Immunol 2019; 348:103994. [PMID: 31831137 DOI: 10.1016/j.cellimm.2019.103994] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 11/16/2022]
Abstract
Leptin is an adipokine secreted primarily by the adipocytes. Leptin has endocrine and immune functions and increases the secretion of pro-inflammatory cytokines by immune cells. Here we show that incubation of B cells from young lean individuals with leptin increases the frequencies of pro-inflammatory B cells and induces intrinsic B cell inflammation, characterized by mRNA expression of pro-inflammatory cytokines (TNF-α and IL-6), chemokines (IL-8), micro-RNAs (miR-155 and miR-16), TLR4 and p16, a cell cycle regulator associated with immunosenescence. We have previously shown that the expression of these pro-inflammatory markers in unstimulated B cells is negatively associated with the response of the same B cells after in vivo or in vitro stimulation. B cells from young lean individuals, after in vitro incubation with leptin, show reduced class switch and influenza vaccine-specific IgG production. Our results altogether show that leptin makes B cells from youn lean individuals similar to those from young obese and elderly lean individuals, suggesting that leptin may be a mechanisms of immunosenescence in human B cells.
Collapse
Affiliation(s)
- Daniela Frasca
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Alain Diaz
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Maria Romero
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Bonnie B Blomberg
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
43
|
Abstract
Adipose stem cells (ASCs) are the basis of procedures intended for tissue regeneration. These cells are heterogeneous, owing to various factors, including the donor age, sex, body mass index, and clinical condition; the isolation procedure (liposuction or fat excision); the place from where the cells were sampled (body site and depth of each adipose depot); culture surface; type of medium (whether supplemented with fetal bovine serum or xeno-free), that affect the principal phenotypic features of ASCs. The features related to ASCs heterogeneity are relevant for the success of therapeutic procedures; these features include proliferation capacity, differentiation potential, immunophenotype, and the secretome. These are important characteristics for the success of regenerative tissue engineering, not only because of their effects upon the reconstruction and healing exerted by ASCs themselves, but also because of the paracrine signaling of ASCs and its impact on recipient tissues. Knowledge of sources of heterogeneity will be helpful in the standardization of ASCs-based procedures. New avenues of research could include evaluation of the effects of the use of more homo1geneous ASCs for specific purposes, the study of ASCs-recipient interactions in heterologous cell transplantation, and the characterization of epigenetic changes in ASCs, as well as investigations of the effect of the metabolome upon ASCs behavior in culture.
Collapse
|
44
|
Sabol RA, Giacomelli P, Beighley A, Bunnell BA. Adipose Stem Cells and Cancer: Concise Review. Stem Cells 2019; 37:1261-1266. [DOI: 10.1002/stem.3050] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/11/2019] [Accepted: 06/04/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Rachel A. Sabol
- Center for Stem Cell Research; Tulane University School of Medicine; New Orleans Louisiana USA
| | - Paulina Giacomelli
- Center for Stem Cell Research; Tulane University School of Medicine; New Orleans Louisiana USA
| | - Adam Beighley
- Center for Stem Cell Research; Tulane University School of Medicine; New Orleans Louisiana USA
| | - Bruce A. Bunnell
- Center for Stem Cell Research; Tulane University School of Medicine; New Orleans Louisiana USA
- Department of Pharmacology; Tulane University; New Orleans Louisiana USA
- Division of Regenerative Medicine; Tulane National Primate Research Center; Covington Louisiana USA
| |
Collapse
|
45
|
Human adipocytes and CD34 + cells from the stromal vascular fraction of the same adipose tissue differ in their energy metabolic enzyme configuration. Exp Cell Res 2019; 380:47-54. [PMID: 31002814 DOI: 10.1016/j.yexcr.2019.04.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/26/2019] [Accepted: 04/15/2019] [Indexed: 01/24/2023]
Abstract
Adipose tissue plays a role in energy storage and metabolic balance and is composed of different cell types. The metabolic activity of the tissue itself has been a matter of research for a long time, but comparative data about the energy metabolism of different cell types of human subcutaneous adipose tissue are sparse. Therefore, we compared the activity of major energy metabolic pathways of adipocytes and CD34+ cells from the stromal vascular fraction (SVF) separated from the same tissue. This CD34+ cell fraction is enriched with adipose tissue-derived mesenchymal progenitors, as they account for the largest proportion of CD34+ cells of the SVF. Adipocytes displayed significantly higher mitochondrial enzyme capacities compared to CD34+ SVF-cells, as shown by the higher activities of isocitrate dehydrogenase and ß-hydroxyacyl-CoA dehydrogenase. Inversely, the CD34+ SVF-cells showed higher capacities for cytosolic carbohydrate metabolism, represented by the activity of glycolysis and the pentose phosphate pathway. Thus, the CD34+ SVF-cells may ensure the provision of pentose phosphates and reduction equivalents for the replication of DNA during proliferation. The data indicate that these two cell fractions of the human adipose tissue vary in their metabolic configuration adapted to their physiological demands regarding proliferation and differentiation in vivo.
Collapse
|
46
|
Glastonbury CA, Couto Alves A, El-Sayed Moustafa JS, Small KS. Cell-Type Heterogeneity in Adipose Tissue Is Associated with Complex Traits and Reveals Disease-Relevant Cell-Specific eQTLs. Am J Hum Genet 2019; 104:1013-1024. [PMID: 31130283 PMCID: PMC6556877 DOI: 10.1016/j.ajhg.2019.03.025] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 03/26/2019] [Indexed: 12/31/2022] Open
Abstract
Adipose tissue is an important endocrine organ with a role in many cardiometabolic diseases. It is comprised of a heterogeneous collection of cell types that can differentially impact disease phenotypes. Cellular heterogeneity can also confound -omic analyses but is rarely taken into account in analysis of solid-tissue transcriptomes. Here, we investigate cell-type heterogeneity in two population-level subcutaneous adipose-tissue RNA-seq datasets (TwinsUK, n = 766 and the Genotype-Tissue Expression project [GTEx], n = 326) by estimating the relative proportions of four distinct cell types (adipocytes, macrophages, CD4+ T cells, and micro-vascular endothelial cells). We find significant cellular heterogeneity within and between the TwinsUK and GTEx adipose datasets. We find that adipose cell-type composition is heritable and confirm the positive association between adipose-resident macrophage proportion and obesity (high BMI), but we find a stronger BMI-independent association with dual-energy X-ray absorptiometry (DXA) derived body-fat distribution traits. We benchmark the impact of adipose-tissue cell composition on a range of standard analyses, including phenotype-gene expression association, co-expression networks, and cis-eQTL discovery. Our results indicate that it is critical to account for cell-type composition when combining adipose transcriptome datasets in co-expression analysis and in differential expression analysis with obesity-related traits. We applied gene expression by cell-type proportion interaction models (G × Cell) to identify 26 cell-type-specific expression quantitative trait loci (eQTLs) in 20 genes, including four autoimmune disease genome-wide association study (GWAS) loci. These results identify cell-specific eQTLs and demonstrate the potential of in silico deconvolution of bulk tissue to identify cell-type-restricted regulatory variants.
Collapse
Affiliation(s)
- Craig A Glastonbury
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK.
| | | | | | - Kerrin S Small
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK.
| |
Collapse
|
47
|
Keuper M. On the role of macrophages in the control of adipocyte energy metabolism. Endocr Connect 2019; 8:R105-R121. [PMID: 31085768 PMCID: PMC6590200 DOI: 10.1530/ec-19-0016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 05/14/2019] [Indexed: 12/11/2022]
Abstract
The crosstalk between macrophages (MΦ) and adipocytes within white adipose tissue (WAT) influences obesity-associated insulin resistance and other associated metabolic disorders, such as atherosclerosis, hypertension and type 2 diabetes. MΦ infiltration is increased in WAT during obesity, which is linked to decreased mitochondrial content and activity. The mechanistic interplay between MΦ and mitochondrial function of adipocytes is under intense investigation, as MΦ and inflammatory pathways exhibit a pivotal role in the reprogramming of WAT metabolism in physiological responses during cold, fasting and exercise. Thus, the underlying immunometabolic pathways may offer therapeutic targets to correct obesity and metabolic disease. Here, I review the current knowledge on the quantity and the quality of human adipose tissue macrophages (ATMΦ) and their impact on the bioenergetics of human adipocytes. The effects of ATMΦ and their secreted factors on mitochondrial function of white adipocytes are discussed, including recent research on MΦ as part of an immune signaling cascade involved in the 'browning' of WAT, which is defined as the conversion from white, energy-storing adipocytes into brown, energy-dissipating adipocytes.
Collapse
Affiliation(s)
- Michaela Keuper
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
48
|
García-Vázquez MD, Herrero de la Parte B, García-Alonso I, Morales MC. [Analysis of Biological Properties of Human Adult Mesenchymal Stem Cells and Their Effect on Mouse Hind Limb Ischemia]. J Vasc Res 2019; 56:77-91. [PMID: 31079101 DOI: 10.1159/000498919] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 02/13/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Due to their self-renewal, proliferation, differentiation, and angiogenesis-inducing capacity, human adipose mesenchymal stem cells (AMSC) have potential clinical applications in the treatment of limb ischemia. AMSC from healthy donors have been shown to induce neovascularization in animal models. However, when cells were obtained from donors suffering from any pathology, their autologous application showed limited effectiveness. We studied whether liposuction niche and obesity could determine the regenerative properties of cells meaning that not all cell batches are suitable for clinical practice. METHODS AMSC obtained from 10 donors, obese and healthy, were expanded in vitro following a good manufacturing practice-like production protocol. Cell viability, proliferation kinetics, morphological analysis, phenotype characterization, and stemness potency were assessed over the course of the expansion process. AMSC selected for having the most suitable biological properties were used as an experimental treatment in a preclinical mouse model of hind limb ischemia. RESULT All cell batches were positively characterized as mesenchymal stem cells, but not all of them showed the same properties or were successfully expanded in vitro, depending on the characteristics of the donor and the extraction area. Notably, AMSC from the abdomen of obese donors showed undesirable biological properties. AMSC with low duplication times and multilineage differentiation potential and forming large densely packed colonies, were able, following expansion in vitro, to increase neovascularization and repair when implanted in the ischemic tissue of mice. CONCLUSION An extensive AMSC biological properties study could be useful to predict the potential clinical efficacy of cells before in vivo transplantation. Thus, peripheral ischemia and possibly other pathologies could benefit from stem cell treatments as shown in our preclinical model in terms of tissue damage repair and regeneration after ischemic injury.
Collapse
Affiliation(s)
| | - Borja Herrero de la Parte
- Department of Surgery and Radiology and Physical Medicine, University of the Basque Country, Leioa, Spain
| | - Ignacio García-Alonso
- Department of Surgery and Radiology and Physical Medicine, University of the Basque Country, Leioa, Spain
| | - María-Celia Morales
- Department of Cell Biology and Histology, University of the Basque Country, Leioa, Spain,
| |
Collapse
|
49
|
Fisch SC, Nikou AF, Wright EA, Phan JD, Leung KL, Grogan TR, Abbott DH, Chazenbalk GD, Dumesic DA. Precocious subcutaneous abdominal stem cell development to adipocytes in normal-weight women with polycystic ovary syndrome. Fertil Steril 2019; 110:1367-1376. [PMID: 30503136 DOI: 10.1016/j.fertnstert.2018.08.042] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/31/2018] [Accepted: 08/15/2018] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To examine whether abnormal subcutaneous (SC) abdominal adipose stem cell (ASC) development to adipocytes in polycystic ovary syndrome (PCOS) correlates with hyperandrogenism. DESIGN Prospective cohort study. SETTING Academic medical center. PATIENT(S) Eight normal-weight women with PCOS and eight normoandrogenic ovulatory (control) women matched for age and body mass index. INTERVENTION(S) Circulating hormone and metabolic measurements, intravenous glucose tolerance testing, total body dual-energy X-ray absorptiometry, and SC abdominal fat biopsy. MAIN OUTCOME MEASURE(S) In vitro ASC commitment to preadipocytes (ZFP423 protein expression, day 0.5), preadipocyte differentiation to adipocytes (PPARγ gene expression, day 3) and adipocyte lipid content (Oil-Red-O fluorescence, day 12) comparisons correlated with clinical outcomes. RESULT(S) In women with PCOS, SC abdominal ASCs compared with those of control women showed exaggerated commitment to preadipocytes and had greater lipid content in newly formed adipocytes after in vitro maturation. In all women combined, ZFP423 protein expression negatively correlated with fasting plasma glucose levels whereas the lipid content of newly formed adipocytes positively correlated with both PPARγ gene expression and serum free testosterone levels. CONCLUSION(S) In normal-weight women with PCOS compared with the control group, exaggerated SC abdominal ASC commitment to preadipocytes and enhanced adipocyte lipid content during maturation in vitro negatively and positively correlate with circulating fasting glucose and androgen levels, respectively, as a possible mechanism to maintain glucose-insulin homeostasis when fat accretion is accelerated.
Collapse
Affiliation(s)
- Samantha C Fisch
- Department of Obstetrics and Gynecology, University of California-Los Angeles, Los Angeles, California
| | - Ariella Farzan Nikou
- Department of Obstetrics and Gynecology, University of California-Los Angeles, Los Angeles, California
| | - Elizabeth A Wright
- Department of Obstetrics and Gynecology, University of California-Los Angeles, Los Angeles, California
| | - Julia D Phan
- Department of Obstetrics and Gynecology, University of California-Los Angeles, Los Angeles, California
| | - Karen L Leung
- Department of Obstetrics and Gynecology, University of California-Los Angeles, Los Angeles, California
| | - Tristan R Grogan
- Department of Medicine Statistics Core, University of California-Los Angeles, Los Angeles, California
| | - David H Abbott
- Department of Obstetrics and Gynecology, Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin
| | - Gregorio D Chazenbalk
- Department of Obstetrics and Gynecology, University of California-Los Angeles, Los Angeles, California
| | - Daniel A Dumesic
- Department of Obstetrics and Gynecology, University of California-Los Angeles, Los Angeles, California.
| |
Collapse
|
50
|
Takahashi H, Sakata N, Yoshimatsu G, Hasegawa S, Kodama S. Regenerative and Transplantation Medicine: Cellular Therapy Using Adipose Tissue-Derived Mesenchymal Stromal Cells for Type 1 Diabetes Mellitus. J Clin Med 2019; 8:jcm8020249. [PMID: 30781427 PMCID: PMC6406504 DOI: 10.3390/jcm8020249] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/09/2019] [Accepted: 02/13/2019] [Indexed: 02/06/2023] Open
Abstract
Type 1 diabetes mellitus (T1DM) is caused by the autoimmune targeting of pancreatic β-cells, and, in the advanced stage, severe hypoinsulinemia due to islet destruction. In patients with T1DM, continuous exogenous insulin therapy cannot be avoided. However, an insufficient dose of insulin easily induces extreme hyperglycemia or diabetic ketoacidosis, and intensive insulin therapy may cause hypoglycemic symptoms including hypoglycemic shock. While these insulin therapies are efficacious in most patients, some additional therapies are warranted to support the control of blood glucose levels and reduce the risk of hypoglycemia in patients who respond poorly despite receiving appropriate treatment. There has been a recent gain in the popularity of cellular therapies using mesenchymal stromal cells (MSCs) in various clinical fields, owing to their multipotentiality, capacity for self-renewal, and regenerative and immunomodulatory potential. In particular, adipose tissue-derived MSCs (ADMSCs) have become a focus in the clinical setting due to the abundance and easy isolation of these cells. In this review, we outline the possible therapeutic benefits of ADMSC for the treatment of T1DM.
Collapse
Affiliation(s)
- Hiroyuki Takahashi
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Department of Gastroenterological Surgery, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Naoaki Sakata
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Gumpei Yoshimatsu
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Suguru Hasegawa
- Department of Gastroenterological Surgery, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Shohta Kodama
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| |
Collapse
|