1
|
Suchanecka M, Grzelak J, Farzaneh M, Azizidoost S, Dari MAG, Józkowiak M, Data K, Domagała D, Niebora J, Kotrych K, Czerny B, Kamiński A, Torlińska-Walkowiak N, Bieniek A, Szepietowski J, Piotrowska-Kempisty H, Dzięgiel P, Mozdziak P, Kempisty B. Adipose derived stem cells - Sources, differentiation capacity and a new target for reconstructive and regenerative medicine. Biomed Pharmacother 2025; 186:118036. [PMID: 40194335 DOI: 10.1016/j.biopha.2025.118036] [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: 01/27/2025] [Revised: 03/28/2025] [Accepted: 04/03/2025] [Indexed: 04/09/2025] Open
Abstract
Adipose-derived stem cells (ADSCs) are mesenchymal stem cells (MSCs) derived from adipose tissue with mesenchymal lineage differentiation potential and remarkable potential in regenerative medicine. ADSCs are easily sourced from adipose tissue, share regenerative characteristics akin to other MSCs. Their convenient adherence to plastic culture flasks, coupled with their capacity for in vitro expansion and multi-lineage differentiation, underscores their promise as a robust tool for tissue repair and enhancement. The accessibility of human adipose tissue and the development of minimally invasive isolation protocols have further propelled the autologous use of ADSCs, fueling excitement in both organ repair and regenerative medicine. Consequently, research in ADSCsis experiencing rapid growth. A detailed overview of the current landscape of ADSCs isolation and differentiation capacity including the latest advancements in ADSCs usage, encompassing ongoing clinical investigations are important considerations to understand their potential to shape the landscape of regenerative medicine.
Collapse
Affiliation(s)
- Małgorzata Suchanecka
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw 50-368, Poland
| | - Joanna Grzelak
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw 50-368, Poland
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shirin Azizidoost
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahrokh Abouali Gale Dari
- Department of Obstetrics and Gynecology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Małgorzata Józkowiak
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw 50-368, Poland; Department of Toxicology, Poznan University of Medical Sciences, Poznań 61-631, Poland
| | - Krzysztof Data
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw 50-368, Poland
| | - Dominika Domagała
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw 50-368, Poland
| | - Julia Niebora
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw 50-368, Poland
| | - Katarzyna Kotrych
- Department of General and Dental Radiology, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, Szczecin 70-111, Poland
| | - Bogusław Czerny
- Department of General Pharmacology and Pharmacoeconomics, Pomeranian Medical University in Szczecin, Żołnierska 48, Szczecin 71-230, Poland; Department of Stem Cells and Regenerative Medicine, Institute of Natural Fibres and Medicinal Plants, Kolejowa 2, Plewiska 62-064, Poland
| | - Adam Kamiński
- Department of Pediatric Orthopedics and Musculosceletal Oncology, Pomeranian Medical University
| | | | - Andrzej Bieniek
- University Center for General and Oncological Dermatology, Wroclaw Medical University, Wroclaw 50-367, Poland
| | - Jacek Szepietowski
- Faculty of Medicine, Wroclaw University of Science and Technology, Wroclaw, Poland; Department of Dermato-Venereology, 4th Military Hospital, Wroclaw, Poland
| | - Hanna Piotrowska-Kempisty
- Department of Toxicology, Poznan University of Medical Sciences, Poznań 61-631, Poland; Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun 87-100, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Wroclaw 50-368, Poland
| | - Paul Mozdziak
- Graduate Physiology Program, North Carolina State University, Raleigh, NC 27695, USA
| | - Bartosz Kempisty
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw 50-368, Poland; Center of Assisted Reproduction, Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Brno 625 00, Czech Republic; Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun 87-100, Poland; North Carolina State University College of Agriculture and Life Sciences, Raleigh, NC 27695, USA.
| |
Collapse
|
2
|
Halurkar MS, Inoue O, Singh A, Mukherjee R, Ginugu M, Ahn C, Bonatto Paese CL, Duszynski M, Brugmann SA, Lim HW, Sanchez-Gurmaches J. The widely used Ucp1-Cre transgene elicits complex developmental and metabolic phenotypes. Nat Commun 2025; 16:770. [PMID: 39824816 PMCID: PMC11742029 DOI: 10.1038/s41467-024-54763-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 11/20/2024] [Indexed: 01/20/2025] Open
Abstract
Bacterial artificial chromosome transgenic models, including most Cre-recombinases, enable potent interrogation of gene function in vivo but require rigorous validation as limitations emerge. Due to its high relevance to metabolic studies, we perform comprehensive analysis of the Ucp1-CreEvdr line which is widely used for brown fat research. Hemizygotes exhibit major brown and white fat transcriptomic dysregulation, indicating potential altered tissue function. Ucp1-CreEvdr homozygotes also show high mortality, tissue specific growth defects, and craniofacial abnormalities. Mapping the transgene insertion site reveals insertion in chromosome 1 accompanied by large genomic alterations disrupting several genes expressed in a range of tissues. Notably, Ucp1-CreEvdr transgene retains an extra Ucp1 gene copy that may be highly expressed under high thermogenic burden. Our multi-faceted analysis highlights a complex phenotype arising from the presence of the Ucp1-CreEvdr transgene independently of intended genetic manipulations. Overall, comprehensive validation of transgenic mice is imperative to maximize discovery while mitigating unexpected, off-target effects.
Collapse
Affiliation(s)
- Manasi Suchit Halurkar
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Oto Inoue
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Archana Singh
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Rajib Mukherjee
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Pioneering Medicines, 140 First St., Suite 302, Cambridge, MA, USA
| | - Meghana Ginugu
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Christopher Ahn
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Christian Louis Bonatto Paese
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Surgery, Division of Plastic Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Molly Duszynski
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Samantha A Brugmann
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Surgery, Division of Plastic Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Hee-Woong Lim
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Joan Sanchez-Gurmaches
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
3
|
Singh M, Sachdeva M, Kumar N. Assessment of the Anti-adipogenic Effect of Crateva religiosa Bark Extract for Molecular Regulation of Adipogenesis: In Silico and In Vitro Approaches for Management of Hyperlipidemia Through the 3T3-L1 Cell Line. Curr Pharm Biotechnol 2025; 26:778-794. [PMID: 39206484 DOI: 10.2174/0113892010314594240816050240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/16/2024] [Accepted: 07/02/2024] [Indexed: 09/04/2024]
Abstract
AIMS This study aimed to determine the phytoconstituents of Crateva religiosa bark (CRB) and evaluate the hypolipidemic effect of bioactive CRB extract by preventing adipocyte differentiation and lipogenesis. BACKGROUND After performing the preliminary phytochemicals screening, the antioxidant activity of CRB extracts was determined through a DPPH (2, 2-diphenyl-1-picrylhydrazyl) assay. Ethyl acetate extract (CREAE) and ethanol extract (CRETE) of CRB were selected for chromatographic evaluation. METHODS The antihyperlipidemic potential was analyzed by molecular docking through the PKCMS software platform. Further, a 3T3-L1 cell line study via in vitro sulforhodamine B assay and western blotting was performed to confirm the prevention of adipocyte differentiation and lipogenesis. RESULTS The total phenolic contents in CREAE and CRETE were estimated as 29.47 and 81.19 μg/mg equivalent to gallic acid, respectively. The total flavonoid content was found to be 8.78 and 49.08 μg/mg, equivalent to quercetin in CREAE and CRETE, respectively. CRETE exhibited greater scavenging activity with the IC50 value of 61.05 μg/ mL. GC-MS analysis confirmed the presence of three bioactive molecules, stigmasterol, gamma sitosterol, and lupeol, in CRETE. Molecular docking studies predicted that the bioactive molecules interact with HMG-CoA reductase, PPARγ, and CCAAT/EBP, which are responsible for lipid metabolism. In vitro, Sulforhodamine B assays revealed that CRETE dose-dependently reduced cell differentiation and viability. Cellular staining using 'Oil Red O' revealed a decreased lipid content in the CRETE-treated cell lines. CRETE significantly inhibited the induction of PPARγ and CCAAT/EBP expression, as determined through protein expression via western blotting. CONCLUSION The influence of CRETE on lipid metabolism in 3T3-L1 cells is potentially suggesting a new approach to managing hyperlipidemia.
Collapse
Affiliation(s)
- Monika Singh
- Department of Pharmacology, I.T.S. College of Pharmacy, Ghaziabad, U.P., Affiliated with Dr. A.P.J. Abdul Kalam Technical University, Lucknow, India
| | - Monika Sachdeva
- Department of Pharmacy, Raj Kumar Goel Institute of Technology, Ghaziabad U.P., Affiliated with Dr. A.P.J. Abdul Kalam Technical University, Lucknow, India
| | - Nitin Kumar
- Department of Pharmacy, Meerut Institute of Technology, Meerut, Affiliated with Dr. A.P.J. Abdul Kalam Technical University, Lucknow, India
| |
Collapse
|
4
|
Park SY, Shin D, Yoon YS, Park S, Im SS, Kim Y, Kim YS, Choi C, Hur MW. TAT38 and TAT38 mimics potently inhibit adipogenesis by repressing C/EBPα, PPARγ, Pi-PPARγ, and SREBP1 expression. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2024; 1867:195030. [PMID: 38670485 DOI: 10.1016/j.bbagrm.2024.195030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024]
Abstract
Antiretroviral therapy-naive people living with HIV possess less fat than people without HIV. Previously, we found that HIV-1 transactivator of transcription (TAT) decreases fat in ob/ob mice. The TAT38 (a.a. 20-57) is important in the inhibition of adipogenesis and contains three functional domains: Cys-ZF domain (a.a. 20-35 TACTNCYCAKCCFQVC), core-domain (a.a. 36-46, FITKALGISYG), and protein transduction domain (PTD)(a.a. 47-57, RAKRRQRRR). Interestingly, the TAT38 region interacts with the Cyclin T1 of the P-TEFb complex, of which expression increases during adipogenesis. The X-ray crystallographic structure of the complex showed that the Cys-ZF and the core domain bind to the Cyclin T1 via hydrophobic interactions. To prepare TAT38 mimics with structural and functional similarities to TAT38, we replaced the core domain with a hydrophobic aliphatic amino acid (from carbon numbers 5 to 8). The TAT38 mimics with 6-hexanoic amino acid (TAT38 Ahx (C6)) and 7-heptanoic amino acid (TAT38 Ahp (C7)) inhibited adipogenesis of 3T3-L1 potently, reduced cellular triglyceride content, and decreased body weight of diet-induced obese (DIO) mice by 10.4-11 % in two weeks. The TAT38 and the TAT38 mimics potently repressed the adipogenic transcription factors genes, C/EBPα, PPARγ, and SREBP1. Also, they inhibit the phosphorylation of PPARγ. The TAT peptides may be promising candidates for development into a drug against obesity or diabetes.
Collapse
Affiliation(s)
- Sun-Young Park
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, 134, ShinChon-Dong, SeoDaeMoon-Ku, Seoul 120-752, Republic of Korea
| | - Dongyoon Shin
- Proteomics Research Team, CHA Institute of Future Medicine, Seongnam, Republic of Korea; Department of Medical Science, School of Medicine, CHA University, Seongnam, Republic of Korea
| | - Young So Yoon
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, 134, ShinChon-Dong, SeoDaeMoon-Ku, Seoul 120-752, Republic of Korea
| | - Sujin Park
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, 134, ShinChon-Dong, SeoDaeMoon-Ku, Seoul 120-752, Republic of Korea
| | - Seung-Soon Im
- Department of Physiology, Keimyung University School of Medicine, 1095 Dalgubeol-daero, Dalseo-gu, Daegu 42601, Republic of Korea
| | - Yeongshin Kim
- Proteomics Research Team, CHA Institute of Future Medicine, Seongnam, Republic of Korea; Department of Medical Science, School of Medicine, CHA University, Seongnam, Republic of Korea
| | - Young-Soo Kim
- Proteomics Research Team, CHA Institute of Future Medicine, Seongnam, Republic of Korea; Department of Medical Science, School of Medicine, CHA University, Seongnam, Republic of Korea
| | - CheolSoo Choi
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea; Division of Endocrinology & Metabolism, Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon 21565, Republic of Korea
| | - Man-Wook Hur
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, 134, ShinChon-Dong, SeoDaeMoon-Ku, Seoul 120-752, Republic of Korea.
| |
Collapse
|
5
|
Shi L, Yang J, Tao Z, Zheng L, Bui T, Alonso R, Yue F, Cheng Z. Loss of FoxO1 activates an alternate mechanism of mitochondrial quality control for healthy adipose browning. Clin Sci (Lond) 2024; 138:371-385. [PMID: 38469619 PMCID: PMC10932742 DOI: 10.1042/cs20230973] [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: 08/25/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/13/2024]
Abstract
Browning of white adipose tissue is hallmarked by increased mitochondrial density and metabolic improvements. However, it remains largely unknown how mitochondrial turnover and quality control are regulated during adipose browning. In the present study, we found that mice lacking adipocyte FoxO1, a transcription factor that regulates autophagy, adopted an alternate mechanism of mitophagy to maintain mitochondrial turnover and quality control during adipose browning. Post-developmental deletion of adipocyte FoxO1 (adO1KO) suppressed Bnip3 but activated Fundc1/Drp1/OPA1 cascade, concurrent with up-regulation of Atg7 and CTSL. In addition, mitochondrial biogenesis was stimulated via the Pgc1α/Tfam pathway in adO1KO mice. These changes were associated with enhanced mitochondrial homeostasis and metabolic health (e.g., improved glucose tolerance and insulin sensitivity). By contrast, silencing Fundc1 or Pgc1α reversed the changes induced by silencing FoxO1, which impaired mitochondrial quality control and function. Ablation of Atg7 suppressed mitochondrial turnover and function, causing metabolic disorder (e.g., impaired glucose tolerance and insulin sensitivity), regardless of elevated markers of adipose browning. Consistently, suppression of autophagy via CTSL by high-fat diet was associated with a reversal of adO1KO-induced benefits. Our data reveal a unique role of FoxO1 in coordinating mitophagy receptors (Bnip3 and Fundc1) for a fine-tuned mitochondrial turnover and quality control, underscoring autophagic clearance of mitochondria as a prerequisite for healthy browning of adipose tissue.
Collapse
Affiliation(s)
- Limin Shi
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL 32611, U.S.A
- Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL 32611, U.S.A
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL 32610, U.S.A
| | - Jinying Yang
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL 32611, U.S.A
- Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL 32611, U.S.A
| | - Zhipeng Tao
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, U.S.A
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, U.S.A
| | - Louise Zheng
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, U.S.A
| | - Tyler F. Bui
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL 32611, U.S.A
| | - Ramon L. Alonso
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL 32611, U.S.A
| | - Feng Yue
- Department of Animal Sciences, University of Florida, Gainesville, FL 32611, U.S.A
| | - Zhiyong Cheng
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL 32611, U.S.A
- Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL 32611, U.S.A
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL 32610, U.S.A
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, U.S.A
| |
Collapse
|
6
|
Halurkar MS, Inoue O, Mukherjee R, Paese CLB, Duszynski M, Brugmann SA, Lim HW, Sanchez-Gurmaches J. The widely used Ucp1-CreEvdr transgene elicits complex developmental and metabolic phenotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.20.563165. [PMID: 37904917 PMCID: PMC10614962 DOI: 10.1101/2023.10.20.563165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Bacterial artificial chromosome transgenic models, including most Cre-recombinases, enable potent interrogation of gene function in vivo but require rigorous validation as limitations emerge. Due to its high relevance to metabolic studies, we performed comprehensive analysis of the Ucp1-CreEvdr line which is widely used for brown fat research. Hemizygotes exhibited major brown and white fat transcriptomic dysregulation, indicating potential altered tissue function. Ucp1-CreEvdr homozygotes also show high mortality, growth defects, and craniofacial abnormalities. Mapping the transgene insertion site revealed insertion in chromosome 1 accompanied by large genomic alterations disrupting several genes expressed in a range of tissues. Notably, Ucp1-CreEvdr transgene retains an extra Ucp1 gene copy that may be highly expressed under high thermogenic burden. Our multi-faceted analysis highlights a complex phenotype arising from the presence of the Ucp1-CreEvdr transgene independently of the intended genetic manipulations. Overall, comprehensive validation of transgenic mice is imperative to maximize discovery while mitigating unexpected, off-target effects.
Collapse
Affiliation(s)
- Manasi Suchit Halurkar
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center Cincinnati, OH 45229, USA
| | - Oto Inoue
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center Cincinnati, OH 45229, USA
| | - Rajib Mukherjee
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center Cincinnati, OH 45229, USA
| | | | - Molly Duszynski
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center Cincinnati, OH 45229, USA
| | - Samantha A. Brugmann
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Department of Surgery, Division of Plastic Surgery, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Hee-Woong Lim
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Joan Sanchez-Gurmaches
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center Cincinnati, OH 45229, USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| |
Collapse
|
7
|
Anantha P, Liu Z, Raj P, Barman I. Optical diffraction tomography and Raman spectroscopy reveal distinct cellular phenotypes during white and brown adipocyte differentiation. Biosens Bioelectron 2023; 235:115388. [PMID: 37207582 PMCID: PMC10626559 DOI: 10.1016/j.bios.2023.115388] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/06/2023] [Accepted: 05/10/2023] [Indexed: 05/21/2023]
Abstract
White adipose tissue (WAT) and brown adipose tissue (BAT) are the primary types of fats in humans, and they play prominent roles in energy storage and thermogenesis, respectively. While the mechanisms of terminal adipogenesis are well understood, much remains unknown about the early stages of adipogenic differentiation. Label-free approaches, such as optical diffraction tomography (ODT) and Raman spectroscopy, offer the ability to retrieve morphological and molecular information at the single cell level without the negative effects of photobleaching and system perturbation due to introduction of fluorophores. In this study, we employed 3D ODT and Raman spectroscopy to gain deeper insights into the early stages of differentiation of human white preadipocytes (HWPs) and human brown preadipocytes (HBPs). We utilized ODT to retrieve morphological information, including cell dry mass and lipid mass, and Raman spectroscopy to obtain molecular information about lipids. Our findings reveal that HWPs and HBPs undergo dynamic and differential changes during the differentiation process. Notably, we found that HBPs accumulated lipids more rapidly and had a higher lipid mass than HWPs. Additionally, both cell types experienced an increase and subsequent decrease in cell dry mass during the first seven days, followed by an increase after day 7, which we attribute to the transformation of adipogenic precursors in the early stages. Finally, HBPs had higher lipid unsaturation levels than HWPs for the same differentiation timepoints. The insights gained from our study provide crucial contributions towards the advancement of new therapies for obesity and related diseases.
Collapse
Affiliation(s)
- Pooja Anantha
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Zhenhui Liu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Piyush Raj
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Ishan Barman
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA; Department of Oncology, Johns Hopkins University, Baltimore, MD, 21287, USA; The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
8
|
Ye J, Gao C, Liang Y, Hou Z, Shi Y, Wang Y. Characteristic and fate determination of adipose precursors during adipose tissue remodeling. CELL REGENERATION (LONDON, ENGLAND) 2023; 12:13. [PMID: 37138165 PMCID: PMC10156890 DOI: 10.1186/s13619-023-00157-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 12/30/2022] [Indexed: 05/05/2023]
Abstract
Adipose tissues are essential for actively regulating systemic energy balance, glucose homeostasis, immune responses, reproduction, and longevity. Adipocytes maintain dynamic metabolic needs and possess heterogeneity in energy storage and supply. Overexpansion of adipose tissue, especially the visceral type, is a high risk for diabetes and other metabolic diseases. Changes in adipocytes, hypertrophy or hyperplasia, contribute to the remodeling of obese adipose tissues, accompanied by abundant immune cell accumulation, decreased angiogenesis, and aberrant extracellular matrix deposition. The process and mechanism of adipogenesis are well known, however, adipose precursors and their fate decision are only being defined with recent information available to decipher how adipose tissues generate, maintain, and remodel. Here, we discuss the key findings that identify adipose precursors phenotypically, with special emphasis on the intrinsic and extrinsic signals in instructing and regulating the fate of adipose precursors under pathophysiological conditions. We hope that the information in this review lead to novel therapeutic strategies to combat obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Jiayin Ye
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Cheng Gao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Yong Liang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Zongliu Hou
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, 650000, Yunnan, China
| | - Yufang Shi
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.
- The Third Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, 199 Renai Road, Suzhou, 215123, Jiangsu, China.
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.
| |
Collapse
|
9
|
Shi L, Tao Z, Zheng L, Yang J, Hu X, Scott K, de Kloet A, Krause E, Collins JF, Cheng Z. FoxO1 regulates adipose transdifferentiation and iron influx by mediating Tgfβ1 signaling pathway. Redox Biol 2023; 63:102727. [PMID: 37156218 DOI: 10.1016/j.redox.2023.102727] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/24/2023] [Accepted: 04/30/2023] [Indexed: 05/10/2023] Open
Abstract
Adipose plasticity is critical for metabolic homeostasis. Adipocyte transdifferentiation plays an important role in adipose plasticity, but the molecular mechanism of transdifferentiation remains incompletely understood. Here we show that the transcription factor FoxO1 regulates adipose transdifferentiation by mediating Tgfβ1 signaling pathway. Tgfβ1 treatment induced whitening phenotype in beige adipocytes, reducing UCP1 and mitochondrial capacity and enlarging lipid droplets. Deletion of adipose FoxO1 (adO1KO) dampened Tgfβ1 signaling by downregulating Tgfbr2 and Smad3 and induced browning of adipose tissue in mice, increasing UCP1 and mitochondrial content and activating metabolic pathways. Silencing FoxO1 also abolished the whitening effect of Tgfβ1 on beige adipocytes. The adO1KO mice exhibited a significantly higher energy expenditure, lower fat mass, and smaller adipocytes than the control mice. The browning phenotype in adO1KO mice was associated with an increased iron content in adipose tissue, concurrent with upregulation of proteins that facilitate iron uptake (DMT1 and TfR1) and iron import into mitochondria (Mfrn1). Analysis of hepatic and serum iron along with hepatic iron-regulatory proteins (ferritin and ferroportin) in the adO1KO mice revealed an adipose tissue-liver crosstalk that meets the increased iron requirement for adipose browning. The FoxO1-Tgfβ1 signaling cascade also underlay adipose browning induced by β3-AR agonist CL316243. Our study provides the first evidence of a FoxO1-Tgfβ1 axis in the regulation of adipose browning-whitening transdifferentiation and iron influx, which sheds light on the compromised adipose plasticity in conditions of dysregulated FoxO1 and Tgfβ1 signaling.
Collapse
Affiliation(s)
- Limin Shi
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, 32611, USA; Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL, 32611, USA; Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, 32610, USA
| | - Zhipeng Tao
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, 24061, USA; Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Louise Zheng
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Jinying Yang
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, 32611, USA; Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL, 32611, USA
| | - Xinran Hu
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, 32611, USA
| | - Karen Scott
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, 32610, USA; Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL32610, USA
| | - Annette de Kloet
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, 32610, USA; Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Eric Krause
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, 32610, USA; Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL32610, USA
| | - James F Collins
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, 32611, USA; Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL, 32611, USA
| | - Zhiyong Cheng
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, 32611, USA; Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL, 32611, USA; Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, 32610, USA; Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
10
|
Yoon H, Seo JK, Park TE. Microphysiological system recapitulating the pathophysiology of adipose tissue in obesity. Acta Biomater 2023; 159:188-200. [PMID: 36724863 DOI: 10.1016/j.actbio.2023.01.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 01/05/2023] [Accepted: 01/17/2023] [Indexed: 01/30/2023]
Abstract
A growing body of evidence has indicated that white adipose tissue (AT) remodeling is a major trigger for obesity-associated metabolic complications. However, the scarcity of translational models is an obstacle to the development of medicines that act on adipose restoration. Here, we describe a microphysiological system (MPS) that emulates the unique features of reprogrammed AT as a new in vitro tool for studying AT pathophysiology in obesity. The AT MPS contained mature adipocytes embedded in an extracellular matrix (ECM) hydrogel interfaced with AT microvascular endothelium, which was constantly perfused with fresh media. The unique biochemical signals due to the remodeled ECM in obesity were recapitulated using a decellularized AT ECM (AT dECM) hydrogel, which preserves the features of altered ECM composition in obesity. The mature adipocytes embedded in the AT dECM hydrogel maintained their function and morphology for a week without dedifferentiation. Using the AT MPS, we successfully modeled inflammation-induced AT microvascular dysfunction, the recruitment of immune cells due to the upregulation of cell adhesion molecules, and higher cancer cell adhesion as an indicator of metastasis, which are observed in obese individuals. The AT MPS may therefore represent a promising platform for understanding the dynamic cellular interplay in obesity-induced AT remodeling and validating the efficacy of drugs targeting AT in obesity. STATEMENT OF SIGNIFICANCE: The lack of translational in vitro white adipose tissue (AT) models is one of the main obstacles for understanding the obesity-induced reprogramming and the development of medicines. We report herein the AT microphysiological system (MPS), which recapitulates obesity and normal conditions and yields cell- and AT dECM-derived signals, thereby allowing accurate comparative in vitro analyses. Using the AT MPS, we successfully modeled reprogrammed AT in obesity conditions, including inflammation-induced AT vascular dysfunction, the recruitment of immune cells, and higher cancer cell metastasis, which are observed in obese individuals. Our proposed adipose tissue model providing physiological relevance and complexity may therefore enhance the understanding of obesity-associated disorders and be used to investigate their underlying molecular mechanisms to develop pharmacologic treatment strategies.
Collapse
Affiliation(s)
- Heejeong Yoon
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Jeong Kon Seo
- UNIST Central Research Facilities (UCRF), Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Tae-Eun Park
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
| |
Collapse
|
11
|
Wu K, Li B, Ma Y, Tu T, Lin Q, Zhu J, Zhou Y, Liu N, Liu Q. Nicotinamide mononucleotide attenuates HIF-1α activation and fibrosis in hypoxic adipose tissue via NAD +/SIRT1 axis. Front Endocrinol (Lausanne) 2023; 14:1099134. [PMID: 36777361 PMCID: PMC9909340 DOI: 10.3389/fendo.2023.1099134] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/13/2023] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Fibrosis is increasingly considered as a major contributor in adipose tissue dysfunction. Hypoxic activation of hypoxia-inducible factor 1α (HIF-1α) induces a profibrotic transcription, leading to adipose fibrosis. Nicotinamide mononucleotide (NMN), a member of the vitamin B3 family, has been shown to relieve hepatic and cardiac fibrosis, but its effects on hypoxic adipose fibrosis and the underlying mechanism remain unclear. We aimed to elucidate the roles of NMN in regulating HIF-1α and fibrosis in hypoxic adipose tissue. METHODS Mice were placed in a hypobaric chamber for four weeks to induce adipose fibrosis. NMN (500 mg/kg, every three days) was administered by intraperitoneal injection. In vitro, Stromal vascular fractions (SVF) cells were treated by hypoxia with or without NMN (200μM), sirtinol (25μM, a SIRT1 inhibitor) and CoCl2 (100μM, a HIF1α enhancer). The effects of NMN on hypoxia-associated adipose fibrosis, inflammation, NAD+/SIRT1 axis alteration, and HIF-1α activation were evaluated by real-time polymerase chain reaction (PCR), western blots, immunohistochemistry staining, immunoprecipitation, and assay kits. RESULTS Mice placed in a hypoxic chamber for four weeks showed obvious adipose fibrosis and inflammation, which were attenuated by NMN. NMN also restore the compromised NAD+/SIRT1 axis and inhibited the activation of HIF-1α induced by hypoxia. In hypoxia-induced SVFs, the SIRT1 inhibitor sirtinol blocked the anti-fibrotic and anti-inflammatory effects of NMN, upregulated the HIF-1α and its acetylation level. The HIF1α stabilizer CoCl2 showed similar effects as sirtinol. CONCLUSION NMN effectively attenuated HIF-1α activation-induced adipose fibrosis and inflammation by restoring the compromised NAD+/SIRT1 axis.
Collapse
Affiliation(s)
- Keke Wu
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Biao Li
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Cardiology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Yingxu Ma
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tao Tu
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qiuzhen Lin
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiayi Zhu
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yong Zhou
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Na Liu
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Qiming Liu, ; Na Liu,
| | - Qiming Liu
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Qiming Liu, ; Na Liu,
| |
Collapse
|
12
|
Kfoury S, Michl P, Roth L. Modeling Obesity-Driven Pancreatic Carcinogenesis-A Review of Current In Vivo and In Vitro Models of Obesity and Pancreatic Carcinogenesis. Cells 2022; 11:3170. [PMID: 36231132 PMCID: PMC9563584 DOI: 10.3390/cells11193170] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/01/2022] [Accepted: 10/06/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common pancreatic malignancy with a 5-year survival rate below 10%, thereby exhibiting the worst prognosis of all solid tumors. Increasing incidence together with a continued lack of targeted treatment options will cause PDAC to be the second leading cause of cancer-related deaths in the western world by 2030. Obesity belongs to the predominant risk factors for pancreatic cancer. To improve our understanding of the impact of obesity on pancreatic cancer development and progression, novel laboratory techniques have been developed. In this review, we summarize current in vitro and in vivo models of PDAC and obesity as well as an overview of a variety of models to investigate obesity-driven pancreatic carcinogenesis. We start by giving an overview on different methods to cultivate adipocytes in vitro as well as various in vivo mouse models of obesity. Moreover, established murine and human PDAC cell lines as well as organoids are summarized and the genetically engineered models of PCAC compared to xenograft models are introduced. Finally, we review published in vitro and in vivo models studying the impact of obesity on PDAC, enabling us to decipher the molecular basis of obesity-driven pancreatic carcinogenesis.
Collapse
Affiliation(s)
- Sally Kfoury
- Department of Internal Medicine I, Martin-Luther University Halle/Wittenberg, Ernst-Grube-Strasse 40, D-06120 Halle (Saale), Germany
| | - Patrick Michl
- Department of Internal Medicine I, Martin-Luther University Halle/Wittenberg, Ernst-Grube-Strasse 40, D-06120 Halle (Saale), Germany
- Department of Medicine, Internal Medicine IV, University Hospital Heidelberg, Im Neuenheimer Feld 410, D-69120 Heidelberg, Germany
| | - Laura Roth
- Department of Internal Medicine I, Martin-Luther University Halle/Wittenberg, Ernst-Grube-Strasse 40, D-06120 Halle (Saale), Germany
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
13
|
Men X, Han X, Lee SJ, Oh G, Park KT, Han JK, Choi SI, Lee OH. Anti-Obesogenic Effects of Sulforaphane-Rich Broccoli (Brassica oleracea var. italica) Sprouts and Myrosinase-Rich Mustard (Sinapis alba L.) Seeds in Vitro and in Vivo. Nutrients 2022; 14:nu14183814. [PMID: 36145190 PMCID: PMC9505190 DOI: 10.3390/nu14183814] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 11/30/2022] Open
Abstract
Glucoraphanin (GRA), a glucosinolate particularly abundant in broccoli (Brassica oleracea var. italica) sprouts, can be converted to sulforaphane (SFN) by the enzyme myrosinase. Herein, we investigated the anti-obesogenic effects of broccoli sprout powder (BSP), mustard (Sinapis alba L.) seed powder (MSP), and sulforaphane-rich MSP-BSP mixture powder (MBP) in bisphenol A (BPA)-induced 3T3-L1 cells and obese C57BL/6J mice. In vitro experiments showed that MBP, BSP, and MSP have no cytotoxic effects. Moreover, MBP and BSP inhibited the lipid accumulation in BPA-induced 3T3-L1 cells. In BPA-induced obese mice, BSP and MBP treatment inhibited body weight gain and ameliorated dyslipidemia. Furthermore, our results showed that BSP and MBP could activate AMPK, which increases ACC phosphorylation, accompanied by the upregulation of lipolysis-associated proteins (UCP-1 and CPT-1) and downregulation of adipogenesis-related proteins (C/EBP-α, FAS, aP2, PPAR-γ, and SREBP-1c), both in vitro and in vivo. Interestingly, MBP exerted a greater anti-obesogenic effect than BSP. Taken together, these findings indicate that BSP and MBP could inhibit BPA-induced adipocyte differentiation and adipogenesis by increasing the expression of the proteins related to lipid metabolism and lipolysis, effectively treating BPA-induced obesity. Thus, BSP and MBP can be developed as effective anti-obesogenic drugs.
Collapse
Affiliation(s)
- Xiao Men
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Korea
| | - Xionggao Han
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Korea
| | - Se-Jeong Lee
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Korea
| | - Geon Oh
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Korea
| | - Keun-Tae Park
- Research and Development Center, Milae Bioresourece Co., Ltd., Seoul 05542, Korea
| | - Jong-Kwon Han
- Research and Development Center, Milae Bioresourece Co., Ltd., Seoul 05542, Korea
| | - Sun-Il Choi
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Korea
- Correspondence: (S.-I.C.); (O.-H.L.); Tel.: +82-33-250-6454 (S.-I.C.); +82-33-250-6454 (O.-H.L.); Fax: +82-33-259-5561 (S.-I.C.); +82-33-259-5561 (O.-H.L.)
| | - Ok-Hwan Lee
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Korea
- Correspondence: (S.-I.C.); (O.-H.L.); Tel.: +82-33-250-6454 (S.-I.C.); +82-33-250-6454 (O.-H.L.); Fax: +82-33-259-5561 (S.-I.C.); +82-33-259-5561 (O.-H.L.)
| |
Collapse
|
14
|
Stanek E, Pacia MZ, Kaczor A, Czamara K. The distinct phenotype of primary adipocytes and adipocytes derived from stem cells of white adipose tissue as assessed by Raman and fluorescence imaging. Cell Mol Life Sci 2022; 79:383. [PMID: 35752714 PMCID: PMC9233632 DOI: 10.1007/s00018-022-04391-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/09/2022] [Accepted: 05/23/2022] [Indexed: 11/25/2022]
Abstract
Spectroscopy-based analysis of chemical composition of cells is a tool still scarcely used in biological sciences, although it provides unique information about the cell identity accessible in vivo and in situ. Through time-lapse spectroscopic monitoring of adipogenesis in brown and white adipose tissue-derived stem cells we have demonstrated that considerable chemical and functional changes occur along with cells differentiation and maturation, yet yielding mature adipocytes with a similar chemical composition, independent of the cellular origin (white or brown adipose tissue). However, in essence, these stem cell-derived adipocytes have a markedly different chemical composition compared to mature primary adipocytes. The consequences of this different chemical (and, hence, functional) identity have great importance in the context of selecting a suitable methodology for adipogenesis studies, particularly in obesity-related research.
Collapse
Affiliation(s)
- Ewa Stanek
- Jagiellonian Centre of Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348, Kraków, Poland
| | - Marta Z Pacia
- Jagiellonian Centre of Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348, Kraków, Poland
| | - Agnieszka Kaczor
- Jagiellonian Centre of Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348, Kraków, Poland
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387, Kraków, Poland
| | - Krzysztof Czamara
- Jagiellonian Centre of Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348, Kraków, Poland.
| |
Collapse
|
15
|
Acosta FM, Stojkova K, Zhang J, Garcia Huitron EI, Jiang JX, Rathbone CR, Brey EM. Engineering Functional Vascularized Beige Adipose Tissue from Microvascular Fragments of Models of Healthy and Type II Diabetes Conditions. J Tissue Eng 2022; 13:20417314221109337. [PMID: 35782994 PMCID: PMC9248044 DOI: 10.1177/20417314221109337] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 06/08/2022] [Indexed: 01/10/2023] Open
Abstract
Engineered beige adipose tissues could be used for screening therapeutic strategies or as a direct treatment for obesity and metabolic disease. Microvascular fragments are vessel structures that can be directly isolated from adipose tissue and may contain cells capable of differentiation into thermogenic, or beige, adipocytes. In this study, culture conditions were investigated to engineer three-dimensional, vascularized functional beige adipose tissue using microvascular fragments isolated from both healthy animals and a model of type II diabetes (T2D). Vascularized beige adipose tissues were engineered and exhibited increased expression of beige adipose markers, enhanced function, and improved cellular respiration. While microvascular fragments isolated from both lean and diabetic models were able to generate functional tissues, differences were observed in regard to vessel assembly and tissue function. This study introduces an approach that could be employed to engineer vascularized beige adipose tissues from a single, potentially autologous source of cells.
Collapse
Affiliation(s)
- Francisca M. Acosta
- Department of Biomedical Engineering
and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX,
USA
- UTSA-UTHSCSA Joint Graduate Program in
Biomedical Engineering, San Antonio, TX, USA
- Department of Biochemistry and
Structural Biology, University of Texas Health Science Center, San Antonio, TX,
USA
| | - Katerina Stojkova
- Department of Biomedical Engineering
and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX,
USA
| | - Jingruo Zhang
- Department of Biochemistry and
Structural Biology, University of Texas Health Science Center, San Antonio, TX,
USA
| | - Eric Ivan Garcia Huitron
- Department of Biomedical Engineering
and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX,
USA
| | - Jean X. Jiang
- Department of Biochemistry and
Structural Biology, University of Texas Health Science Center, San Antonio, TX,
USA
| | - Christopher R. Rathbone
- Department of Biomedical Engineering
and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX,
USA
- UTSA-UTHSCSA Joint Graduate Program in
Biomedical Engineering, San Antonio, TX, USA
| | - Eric M. Brey
- Department of Biomedical Engineering
and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX,
USA
- UTSA-UTHSCSA Joint Graduate Program in
Biomedical Engineering, San Antonio, TX, USA
| |
Collapse
|
16
|
Lee DK, Jang HD. Carnosic Acid Attenuates an Early Increase in ROS Levels during Adipocyte Differentiation by Suppressing Translation of Nox4 and Inducing Translation of Antioxidant Enzymes. Int J Mol Sci 2021; 22:ijms22116096. [PMID: 34198827 PMCID: PMC8201016 DOI: 10.3390/ijms22116096] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/27/2021] [Accepted: 06/01/2021] [Indexed: 12/19/2022] Open
Abstract
The objective of this study was to investigate molecular mechanisms underlying the ability of carnosic acid to attenuate an early increase in reactive oxygen species (ROS) levels during MDI-induced adipocyte differentiation. The levels of superoxide anion and ROS were determined using dihydroethidium (DHE) and 2′-7′-dichlorofluorescin diacetate (DCFH-DA), respectively. Both superoxide anion and ROS levels peaked on the second day of differentiation. They were suppressed by carnosic acid. Carnosic acid attenuates the translation of NADPH (nicotinamide adenine dinucleotide phosphate) oxidase 4 (Nox4), p47phox, and p22phox, and the phosphorylation of nuclear factor-kappa B (NF-κB) and NF-κB inhibitor (IkBa). The translocation of NF-κB into the nucleus was also decreased by carnosic acid. In addition, carnosic acid increased the translation of heme oxygenase-1 (HO-1), γ–glutamylcysteine synthetase (γ-GCSc), and glutathione S-transferase (GST) and both the translation and nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2). Taken together, these results indicate that carnosic acid could down-regulate ROS level in an early stage of MPI-induced adipocyte differentiation by attenuating ROS generation through suppression of NF-κB-mediated translation of Nox4 enzyme and increasing ROS neutralization through induction of Nrf2-mediated translation of phase II antioxidant enzymes such as HO-1, γ-GCS, and GST, leading to its anti-adipogenetic effect.
Collapse
|
17
|
Poret JM, Molina PE, Simon L. Isolation, Proliferation and Differentiation of Rhesus Macaque Adipose-Derived Stem Cells. J Vis Exp 2021:10.3791/61732. [PMID: 34125096 PMCID: PMC8210449 DOI: 10.3791/61732] [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] [Indexed: 10/31/2022] Open
Abstract
Adipose tissue provides a rich and accessible source of multipotent stem cells, which are able to self-renew. These adipose-derived stem cells (ADSCs) provide a consistent ex vivo cellular system that are functionally like that of in vivo adipocytes. Use of ADSCs in biomedical research allows for cellular investigation of adipose tissue metabolic regulation and function. ADSC differentiation is necessary for adequate adipocyte expansion, and suboptimal differentiation is a major mechanism of adipose dysfunction. Understanding changes in ADSC differentiation is crucial to understanding the development of metabolic dysfunction and disease. The protocols described in this manuscript, when followed, will yield mature adipocytes that can be used for several in vitro functional tests to assess ADSC metabolic function, including but not limited to assays measuring glucose uptake, lipolysis, lipogenesis, and secretion. Rhesus macaques (Macaca mulatta) are physiologically, anatomically, and evolutionarily similar to humans and as such, their tissues and cells have been used extensively in biomedical research and for development of treatments. Here, we describe ADSC isolation using fresh subcutaneous and omental adipose tissue obtained from 4-9-year old rhesus macaques. Adipose tissue samples are enzymatically digested in collagenase followed by filtration and centrifugation to isolate ADSCs from the stromal vascular fraction. Isolated ADSCs are proliferated in stromal media followed by approximately 14-21 days of differentiation using a cocktail of 0.5 μg/mL dexamethasone, 0.5 mM isobutyl methylxanthine, and 50 μM indomethacin in stromal media. Mature adipocytes are observed at approximately 14 days of differentiation. In this manuscript, we describe protocols for ADSC isolation, proliferation, and differentiation in vitro. Although, we have focused on ADSCs from rhesus macaque adipose tissue, these protocols can be utilized for adipose tissue obtained from other animals with minimal adjustments.
Collapse
Affiliation(s)
- Jonquil M Poret
- Department of Physiology, Louisiana State University Health Sciences Center - New Orleans; Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center - New Orleans
| | - Patricia E Molina
- Department of Physiology, Louisiana State University Health Sciences Center - New Orleans; Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center - New Orleans
| | - Liz Simon
- Department of Physiology, Louisiana State University Health Sciences Center - New Orleans; Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center - New Orleans;
| |
Collapse
|
18
|
Gómez-Serrano M, Ponath V, Preußer C, Pogge von Strandmann E. Beyond the Extracellular Vesicles: Technical Hurdles, Achieved Goals and Current Challenges When Working on Adipose Cells. Int J Mol Sci 2021; 22:ijms22073362. [PMID: 33805982 PMCID: PMC8036456 DOI: 10.3390/ijms22073362] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 12/17/2022] Open
Abstract
Adipose tissue and its crosstalk with other organs plays an essential role in the metabolic homeostasis of the entire body. Alteration of this communication (i.e., due to obesity) is related to the development of several comorbidities including type 2 diabetes, cardiovascular diseases, or cancer. Within the adipose depot, adipocytes are the main cell type and thus the main source of secreted molecules, which exert modulating effects not only at a local but also at a systemic level. Extracellular vesicles (EVs) have recently emerged as important mediators in cell–cell communication and account for part of the cellular secretome. In recent years, there has been a growing body of research on adipocyte-derived extracellular vesicles (Ad-EVs). However, there is still a lack of standardized methodological approaches, especially regarding primary adipocytes. In this review, we will provide an outline of crucial aspects when working on adipose-derived material, with a special focus on primary adipocytes. In parallel, we will point out current methodological challenges in the EV field and how they impact the transcriptomic, proteomic and functional evaluations of Ad-EVs.
Collapse
|
19
|
Lee JS, Hyun IK, Seo HJ, Song D, Kim MY, Kang SS. Biotransformation of whey by Weissella cibaria suppresses 3T3-L1 adipocyte differentiation. J Dairy Sci 2021; 104:3876-3887. [PMID: 33612219 DOI: 10.3168/jds.2020-19677] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/21/2020] [Indexed: 11/19/2022]
Abstract
Biotransformation, the structural modification of chemical compounds, has proved to be an indispensable tool in providing beneficial health effects. Although the health benefits of biotransformation using plant sources has been widely studied, the anti-adipogenic effect of biotransformed dairy products, such as whey, have not yet been demonstrated. Here, we investigated the anti-adipogenic effect of whey biotransformed by Weissella cibaria in 3T3-L1 adipocytes. Weissella cibaria-biotransformed whey considerably reduced the accumulation of lipid droplets and intracellular triglycerides in 3T3-L1 cells. In the presence of W. cibaria-biotransformed whey, the mRNA and protein expression of a key transcription factor, peroxisome proliferator-activated receptor γ (PPARγ), for adipogenesis was markedly suppressed in 3T3-L1 cells. Additionally, W. cibaria-biotransformed whey also decreased the mRNA and protein expressions of lipoprotein lipase and adipocyte fatty acid-binding protein, which are regulated by PPARγ. Moreover, W. cibaria-biotransformed whey inhibited the expression of adipokines, resistin, and leptin. Collectively, these results suggest that whey biotransformed by W. cibaria has the potential to exert anti-adipogenic effects by inhibiting intracellular signaling events of adipogenic-related transcription factors and target genes.
Collapse
Affiliation(s)
- Ji Soo Lee
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - In Kyung Hyun
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Hye-Jin Seo
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Dahyun Song
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Min Young Kim
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Seok-Seong Kang
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University-Seoul, Goyang 10326, Republic of Korea.
| |
Collapse
|
20
|
Pope BD, Warren CR, Dahl MO, Pizza CV, Henze DE, Sinatra NR, Gonzalez GM, Chang H, Liu Q, Glieberman AL, Ferrier JP, Cowan CA, Parker KK. Fattening chips: hypertrophy, feeding, and fasting of human white adipocytes in vitro. LAB ON A CHIP 2020; 20:4152-4165. [PMID: 33034335 PMCID: PMC7818847 DOI: 10.1039/d0lc00508h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Adipose is a distributed organ that performs vital endocrine and energy homeostatic functions. Hypertrophy of white adipocytes is a primary mode of both adaptive and maladaptive weight gain in animals and predicts metabolic syndrome independent of obesity. Due to the failure of conventional culture to recapitulate adipocyte hypertrophy, technology for production of adult-size adipocytes would enable applications such as in vitro testing of weight loss therapeutics. To model adaptive adipocyte hypertrophy in vitro, we designed and built fat-on-a-chip using fiber networks inspired by extracellular matrix in adipose tissue. Fiber networks extended the lifespan of differentiated adipocytes, enabling growth to adult sizes. By micropatterning preadipocytes in a native cytoarchitecture and by adjusting cell-to-cell spacing, rates of hypertrophy were controlled independent of culture time or differentiation efficiency. In vitro hypertrophy followed a nonlinear, nonexponential growth model similar to human development and elicited transcriptomic changes that increased overall similarity with primary tissue. Cells on the chip responded to simulated meals and starvation, which potentiated some adipocyte endocrine and metabolic functions. To test the utility of the platform for therapeutic development, transcriptional network analysis was performed, and retinoic acid receptors were identified as candidate drug targets. Regulation by retinoid signaling was suggested further by pharmacological modulation, where activation accelerated and inhibition slowed hypertrophy. Altogether, this work presents technology for mature adipocyte engineering, addresses the regulation of cell growth, and informs broader applications for synthetic adipose in pharmaceutical development, regenerative medicine, and cellular agriculture.
Collapse
Affiliation(s)
- Benjamin D Pope
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA. and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Curtis R Warren
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Madeleine O Dahl
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Christina V Pizza
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Douglas E Henze
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Nina R Sinatra
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Grant M Gonzalez
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Huibin Chang
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Qihan Liu
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Aaron L Glieberman
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - John P Ferrier
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Chad A Cowan
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA and Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Kevin Kit Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA. and Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
21
|
Bahmad HF, Daouk R, Azar J, Sapudom J, Teo JCM, Abou-Kheir W, Al-Sayegh M. Modeling Adipogenesis: Current and Future Perspective. Cells 2020; 9:2326. [PMID: 33092038 PMCID: PMC7590203 DOI: 10.3390/cells9102326] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/07/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue is contemplated as a dynamic organ that plays key roles in the human body. Adipogenesis is the process by which adipocytes develop from adipose-derived stem cells to form the adipose tissue. Adipose-derived stem cells' differentiation serves well beyond the simple goal of producing new adipocytes. Indeed, with the current immense biotechnological advances, the most critical role of adipose-derived stem cells remains their tremendous potential in the field of regenerative medicine. This review focuses on examining the physiological importance of adipogenesis, the current approaches that are employed to model this tightly controlled phenomenon, and the crucial role of adipogenesis in elucidating the pathophysiology and potential treatment modalities of human diseases. The future of adipogenesis is centered around its crucial role in regenerative and personalized medicine.
Collapse
Affiliation(s)
- Hisham F. Bahmad
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, 1107 2260 Beirut, Lebanon; (H.F.B.); (R.D.); (J.A.)
| | - Reem Daouk
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, 1107 2260 Beirut, Lebanon; (H.F.B.); (R.D.); (J.A.)
| | - Joseph Azar
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, 1107 2260 Beirut, Lebanon; (H.F.B.); (R.D.); (J.A.)
| | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, 2460 Abu Dhabi, UAE;
| | - Jeremy C. M. Teo
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, 2460 Abu Dhabi, UAE;
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, 1107 2260 Beirut, Lebanon; (H.F.B.); (R.D.); (J.A.)
| | - Mohamed Al-Sayegh
- Biology Division, New York University Abu Dhabi, 2460 Abu Dhabi, UAE
| |
Collapse
|
22
|
Qin S, Jiang J, Lu Y, Nice EC, Huang C, Zhang J, He W. Emerging role of tumor cell plasticity in modifying therapeutic response. Signal Transduct Target Ther 2020; 5:228. [PMID: 33028808 PMCID: PMC7541492 DOI: 10.1038/s41392-020-00313-5] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/25/2020] [Accepted: 08/30/2020] [Indexed: 02/07/2023] Open
Abstract
Resistance to cancer therapy is a major barrier to cancer management. Conventional views have proposed that acquisition of resistance may result from genetic mutations. However, accumulating evidence implicates a key role of non-mutational resistance mechanisms underlying drug tolerance, the latter of which is the focus that will be discussed here. Such non-mutational processes are largely driven by tumor cell plasticity, which renders tumor cells insusceptible to the drug-targeted pathway, thereby facilitating the tumor cell survival and growth. The concept of tumor cell plasticity highlights the significance of re-activation of developmental programs that are closely correlated with epithelial-mesenchymal transition, acquisition properties of cancer stem cells, and trans-differentiation potential during drug exposure. From observations in various cancers, this concept provides an opportunity for investigating the nature of anticancer drug resistance. Over the years, our understanding of the emerging role of phenotype switching in modifying therapeutic response has considerably increased. This expanded knowledge of tumor cell plasticity contributes to developing novel therapeutic strategies or combination therapy regimens using available anticancer drugs, which are likely to improve patient outcomes in clinical practice.
Collapse
Affiliation(s)
- Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, People's Republic of China
| | - Jingwen Jiang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, People's Republic of China
| | - Yi Lu
- School of Medicine, Southern University of Science and Technology Shenzhen, Shenzhen, Guangdong, 518055, People's Republic of China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen, Guangdong, People's Republic of China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, People's Republic of China.
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Road, 611137, Chengdu, People's Republic of China.
| | - Jian Zhang
- School of Medicine, Southern University of Science and Technology Shenzhen, Shenzhen, Guangdong, 518055, People's Republic of China.
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen, Guangdong, People's Republic of China.
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China.
- Chongqing Key Laboratory for Disease Proteomics, Chongqing, People's Republic of China.
| |
Collapse
|
23
|
Asante EC, Pallegar NK, Hoffmann AJ, Viloria-Petit AM, Christian SL. Adipose Tissue from Lean and Obese Mice Induces a Mesenchymal to Epithelial Transition-Like Effect in Triple Negative Breast Cancers Cells Grown in 3-Dimensional Culture. Int J Mol Sci 2020; 21:E6439. [PMID: 32899433 PMCID: PMC7503907 DOI: 10.3390/ijms21176439] [Citation(s) in RCA: 5] [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: 07/22/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 01/01/2023] Open
Abstract
Breast cancer is the second leading cause of cancer-related mortality among women globally with obesity being one risk factor. Obese breast cancer patients have at least a 30% increased risk of death from breast cancer compared to non-obese breast cancer patients because they present with larger tumors and generally have increased rates of metastasis. Moreover, obese breast cancer patients respond more poorly to treatment compared to non-obese patients, particularly pre-menopausal women diagnosed with triple negative breast cancer (TNBC). To help understand the molecular mechanisms underlying the increased metastasis associated with obesity, we previously established a three-dimensional culture system that permits the co-culture of adipocytes and TNBC cells in a manner that mimics an in vivo milieu. Using this system, we demonstrate that white adipose tissue from both lean and obese mice can induce a partial mesenchymal-to-epithelial transition (MET). Triple negative breast cancer cells adopt an epithelial morphology and have an increased expression of some epithelial markers, but they maintain the expression of mesenchymal markers, furnishing the breast cancer cells with hybrid properties that are associated with more aggressive tumors. Thus, these data suggest that adipose tissue has the potential to promote secondary tumor formation in lean and obese women. Further work is needed to determine if targeting the partial MET induced by adipose tissue could reduce metastasis.
Collapse
Affiliation(s)
- Emmanuel C. Asante
- Department of Biochemistry, Memorial University of Newfoundland, St. John’s, NL A1B 3X9, Canada; (E.C.A.); (N.K.P.); (A.J.H.)
| | - Nikitha K. Pallegar
- Department of Biochemistry, Memorial University of Newfoundland, St. John’s, NL A1B 3X9, Canada; (E.C.A.); (N.K.P.); (A.J.H.)
| | - Alica J. Hoffmann
- Department of Biochemistry, Memorial University of Newfoundland, St. John’s, NL A1B 3X9, Canada; (E.C.A.); (N.K.P.); (A.J.H.)
| | - Alicia M. Viloria-Petit
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Sherri L. Christian
- Department of Biochemistry, Memorial University of Newfoundland, St. John’s, NL A1B 3X9, Canada; (E.C.A.); (N.K.P.); (A.J.H.)
| |
Collapse
|
24
|
Vohra MS, Ahmad B, Serpell CJ, Parhar IS, Wong EH. Murine in vitro cellular models to better understand adipogenesis and its potential applications. Differentiation 2020; 115:62-84. [PMID: 32891960 DOI: 10.1016/j.diff.2020.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/08/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023]
Abstract
Adipogenesis has been extensively studied using in vitro models of cellular differentiation, enabling long-term regulation of fat cell metabolism in human adipose tissue (AT) material. Many studies promote the idea that manipulation of this process could potentially reduce the prevalence of obesity and its related diseases. It has now become essential to understand the molecular basis of fat cell development to tackle this pandemic disease, by identifying therapeutic targets and new biomarkers. This review explores murine cell models and their applications for study of the adipogenic differentiation process in vitro. We focus on the benefits and limitations of different cell line models to aid in interpreting data and selecting a good cell line model for successful understanding of adipose biology.
Collapse
Affiliation(s)
- Muhammad Sufyan Vohra
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Bilal Ahmad
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Christopher J Serpell
- School of Physical Sciences, Ingram Building, University of Kent, Canterbury, Kent, CT2 7NH, United Kingdom.
| | - Ishwar S Parhar
- Brain Research Institute, Jeffery Cheah School of Medicine and Health Sciences, Monash University, Bandar Sunway, PJ 47500, Selangor, Malaysia.
| | - Eng Hwa Wong
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
25
|
Vishvanath L, Gupta RK. Contribution of adipogenesis to healthy adipose tissue expansion in obesity. J Clin Invest 2020; 129:4022-4031. [PMID: 31573549 DOI: 10.1172/jci129191] [Citation(s) in RCA: 370] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The manner in which white adipose tissue (WAT) expands and remodels directly impacts the risk of developing metabolic syndrome in obesity. Preferential accumulation of visceral WAT is associated with increased risk for insulin resistance, whereas subcutaneous WAT expansion is protective. Moreover, pathologic WAT remodeling, typically characterized by adipocyte hypertrophy, chronic inflammation, and fibrosis, is associated with insulin resistance. Healthy WAT expansion, observed in the "metabolically healthy" obese, is generally associated with the presence of smaller and more numerous adipocytes, along with lower degrees of inflammation and fibrosis. Here, we highlight recent human and rodent studies that support the notion that the ability to recruit new fat cells through adipogenesis is a critical determinant of healthy adipose tissue distribution and remodeling in obesity. Furthermore, we discuss recent advances in our understanding of the identity of tissue-resident progenitor populations in WAT made possible through single-cell RNA sequencing analysis. A better understanding of adipose stem cell biology and adipogenesis may lead to novel strategies to uncouple obesity from metabolic disease.
Collapse
|
26
|
Harms MJ, Li Q, Lee S, Zhang C, Kull B, Hallen S, Thorell A, Alexandersson I, Hagberg CE, Peng XR, Mardinoglu A, Spalding KL, Boucher J. Mature Human White Adipocytes Cultured under Membranes Maintain Identity, Function, and Can Transdifferentiate into Brown-like Adipocytes. Cell Rep 2020; 27:213-225.e5. [PMID: 30943403 DOI: 10.1016/j.celrep.2019.03.026] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 01/23/2019] [Accepted: 03/06/2019] [Indexed: 12/24/2022] Open
Abstract
White adipose tissue (WAT) is a central factor in the development of type 2 diabetes, but there is a paucity of translational models to study mature adipocytes. We describe a method for the culture of mature white adipocytes under a permeable membrane. Compared to existing culture methods, MAAC (membrane mature adipocyte aggregate cultures) better maintain adipogenic gene expression, do not dedifferentiate, display reduced hypoxia, and remain functional after long-term culture. Subcutaneous and visceral adipocytes cultured as MAAC retain depot-specific gene expression, and adipocytes from both lean and obese patients can be cultured. Importantly, we show that rosiglitazone treatment or PGC1α overexpression in mature white adipocytes induces a brown fat transcriptional program, providing direct evidence that human adipocytes can transdifferentiate into brown-like adipocytes. Together, these data show that MAAC are a versatile tool for studying phenotypic changes of mature adipocytes and provide an improved translational model for drug development.
Collapse
Affiliation(s)
- Matthew J Harms
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Qian Li
- Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Stockholm 17177, Sweden
| | - Sunjae Lee
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm 17121, Sweden
| | - Cheng Zhang
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm 17121, Sweden
| | - Bengt Kull
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Stefan Hallen
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Anders Thorell
- Department of Clinical Sciences, Danderyds Hospital, Karolinska Institutet and Department of Surgery, Ersta Hospital, Stockholm 11691, Sweden
| | - Ida Alexandersson
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Carolina E Hagberg
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine, Karolinska Institutet, Stockholm 17176, Sweden
| | - Xiao-Rong Peng
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm 17121, Sweden; Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 9RT, United Kingdom
| | - Kirsty L Spalding
- Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Stockholm 17177, Sweden; Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine, Karolinska Institutet, Stockholm 17176, Sweden
| | - Jeremie Boucher
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden; The Lundberg Laboratory for Diabetes Research, University of Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Sweden.
| |
Collapse
|
27
|
Zohora FT, Aldebs AI, Nosoudi N, Singh SP, Ramirez-Vick JE. Gene Expression Profiling of Human Adipose Tissue Stem Cells during 2D versus 3D Adipogenesis. Cells Tissues Organs 2020; 208:113-133. [PMID: 32464628 DOI: 10.1159/000507187] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/10/2020] [Indexed: 11/19/2022] Open
Abstract
Much of the current understanding on molecular and cellular events of adipose developmental biology comes from monolayer cell culture models using preadipocyte cell lines, although in vivo adipose tissue consists of a much more complex three-dimensional microenvironment of diverse cell types, extracellular network, and tissue-specific morphological and functional features. Added to this fact, the preadipocytes, on which the adipogenesis mechanisms are mostly explored, possess some serious limitations (e.g., time of initial subculture and adipogenic differentiation time), which, perhaps, can efficiently be replaced with progenitor cells such as adipose tissue-derived stem cells (ASCs). With the objective of developing a better in vitro model for adipose developmental biology, this project involves gene expression profiling of human ASCs (hASCs) during their differentiation to adipocytes in a 2D versus 3D culture model. This transcriptional-level analysis revealed that gene expression patterns of adipogenesis-induced hASCs in a 3D self-assembled polypeptide hydrogel are relatively different from the 2D monolayered cells on plastic hard substrate. Moreover, analysis of adipogenic lineage progression 9 days after adipogenic induction shows earlier differentiation of hASCs in 2D over their 3D counterparts. However, differentiation in 2D shows some unexpected behavior in terms of gene expression, which does not seem to be related to adipogenic lineage specification. Since hASCs are already being used in clinical trials due to their therapeutic potential, it is important to have a clear understanding of the molecular mechanisms in an in vivo model microenvironment like the one presented here.
Collapse
Affiliation(s)
- Fatema Tuj Zohora
- Department of Biomedical, Industrial, and Human Factors Engineering, Wright State University, Dayton, Ohio, USA
| | - Alyaa Isam Aldebs
- Department of Biomedical, Industrial, and Human Factors Engineering, Wright State University, Dayton, Ohio, USA
| | - Nasim Nosoudi
- Biomedical Engineering Program,Marshall University, Huntington, West Virginia, USA
| | - Surinder Pal Singh
- CSIR-National Physical Laboratory, Dr. K.S. Krishanan Marg, New Delhi, India
| | - Jaime Eduardo Ramirez-Vick
- Department of Biomedical, Industrial, and Human Factors Engineering, Wright State University, Dayton, Ohio, USA,
| |
Collapse
|
28
|
Schopow N, Kallendrusch S, Gong S, Rapp F, Körfer J, Gericke M, Spindler N, Josten C, Langer S, Bechmann I. Examination of ex-vivo viability of human adipose tissue slice culture. PLoS One 2020; 15:e0233152. [PMID: 32453755 PMCID: PMC7250419 DOI: 10.1371/journal.pone.0233152] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 04/29/2020] [Indexed: 01/29/2023] Open
Abstract
Obesity is associated with significantly higher mortality rates, and excess adipose tissue is involved in respective pathologies. Here we established a human adipose tissue slice cultures (HATSC) model ex vivo. HATSC match the in vivo cell composition of human adipose tissue with, among others, mature adipocytes, mesenchymal stem cells as well as stroma tissue and immune cells. This is a new method, optimized for live imaging, to study adipose tissue and cell-based mechanisms of obesity in particular. HATSC survival was tested by means of conventional and immunofluorescence histological techniques, functional analyses and live imaging. Surgery-derived tissue was cut with a tissue chopper in 500 μm sections and transferred onto membranes building an air-liquid interface. HATSC were cultured in six-well plates filled with Dulbecco’s Modified Eagle’s Medium (DMEM), insulin, transferrin, and selenium, both with and without serum. After 0, 1, 7 and 14 days in vitro, slices were fixated and analyzed by morphology and Perilipin A for tissue viability. Immunofluorescent staining against IBA1, CD68 and Ki67 was performed to determine macrophage survival and proliferation. These experiments showed preservation of adipose tissue as well as survival and proliferation of monocytes and stroma tissue for at least 14 days in vitro even in the absence of serum. The physiological capabilities of adipocytes were functionally tested by insulin stimulation and measurement of Phospho-Akt on day 7 and 14 in vitro. Viability was further confirmed by live imaging using Calcein-AM (viable cells) and propidium iodide (apoptosis/necrosis). In conclusion, HATSC have been successfully established by preserving the monovacuolar form of adipocytes and surrounding macrophages and connective tissue. This model allows further analysis of mature human adipose tissue biology ex vivo.
Collapse
Affiliation(s)
- Nikolas Schopow
- Institute of Anatomy, University Leipzig, Leipzig, Germany
- Department for Orthopedics, Trauma Surgery, and Reconstructive Surgery, University Hospital Leipzig, Leipzig, Germany
- * E-mail:
| | | | - Siming Gong
- Institute of Anatomy, University Leipzig, Leipzig, Germany
- Department for Orthopedics, Trauma Surgery, and Reconstructive Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Felicitas Rapp
- Institute of Anatomy, University Leipzig, Leipzig, Germany
- GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
| | - Justus Körfer
- Institute of Anatomy, University Leipzig, Leipzig, Germany
- University Cancer Center Leipzig (UCCL), University Hospital Leipzig, Leipzig, Germany
| | - Martin Gericke
- Institute of Anatomy, University Leipzig, Leipzig, Germany
| | - Nick Spindler
- Department for Orthopedics, Trauma Surgery, and Reconstructive Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Christoph Josten
- Department for Orthopedics, Trauma Surgery, and Reconstructive Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Stefan Langer
- Department for Orthopedics, Trauma Surgery, and Reconstructive Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Ingo Bechmann
- Institute of Anatomy, University Leipzig, Leipzig, Germany
| |
Collapse
|
29
|
Shahin-Shamsabadi A, Selvaganapathy PR. A 3D Self-Assembled In Vitro Model to Simulate Direct and Indirect Interactions between Adipocytes and Skeletal Muscle Cells. ACTA ACUST UNITED AC 2020; 4:e2000034. [PMID: 32390329 DOI: 10.1002/adbi.202000034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/07/2020] [Accepted: 04/23/2020] [Indexed: 02/06/2023]
Abstract
The molecular mechanisms of the development and progression of diabetes and obesity involve complex interactions between adipocytes and skeletal muscle cells. Although 2D in-vitro models are the gold standard for the mechanistic study of such behaviors, they do not recreate the complexity and dynamics of the interactions between the cell types involved. Alternatively, animal models are used but are expensive, difficult to visualize or analyze, are not completely representative of human physiology or genetic background, and have associated ethical considerations. 3D co-culture systems can be complementary to these approaches. Here, using a newly developed 3D biofabrication method, adipocytes and myoblasts are positioned precisely either in direct physical contact or in close proximity such that the paracrine effects could be systematically studied. Suitable protocols for growth and differentiation of both cells in the co-culture system is also developed. Cells show more restrained lipid and protein production in 3D systems compared to 2D ones and adipocytes show more lipolysis in indirect contact with myoblasts as response to drug treatment. These findings emphasize importance of physical contact between cells that have been overlooked in co-culture systems using transwell inserts and can be used in studies for the development of anti-obesity drugs.
Collapse
Affiliation(s)
- Alireza Shahin-Shamsabadi
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L7, Canada
| | - Ponnambalam Ravi Selvaganapathy
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L7, Canada.,Department of Mechanical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L7, Canada
| |
Collapse
|
30
|
E2F1 Regulates Adipocyte Differentiation and Adipogenesis by Activating ICAT. Cells 2020; 9:cells9041024. [PMID: 32326181 PMCID: PMC7225968 DOI: 10.3390/cells9041024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 03/25/2020] [Accepted: 04/07/2020] [Indexed: 02/06/2023] Open
Abstract
Wnt/β-catenin is a crucial repressor of adipogenesis. We have shown that E2 promoter binding factor 1 (E2F1) suppresses Wnt/β-catenin activity through transactivation of β-catenin interacting protein 1 (CTNNBIP1), also known as inhibitor of β-catenin and TCF4 (ICAT) in human colorectal cancers. However, it remains unknown whether ICAT is required for E2F1 to promote differentiation by inhibiting β-catenin activity in pre-adipocytes. In the present study, we found that 1-methyl-3-isobutylxanthine, dexamethasone, and insulin (MDI)-induced differentiation and lipid accumulation in 3T3-L1 pre-adipocytes was reversed by activation of β-catenin triggered by CHIR99021, a GSK3β inhibitor. Intriguingly, we observed a reduced protein level of E2F1 and ICAT at a later stage of pre-adipocytes differentiation. Importantly, overexpression of ICAT in 3T3-L1 pre-adipocytes markedly promote the adipogenesis and partially reversed the inhibitory effect of CHIR99021 on MDI-induced adipogenesis and lipid accumulation by regulating adipogenic regulators and Wnt/β-catenin targets. Moreover, pre-adipocytes differentiation induced by MDI were markedly inhibited in siE2F1 or siICAT transfected 3T3-L1 cells. Gene silencing of ICAT in the E2F1 overexpressed adipocytes also inhibited the adipogenesis. These data indicated that E2F1 is a metabolic regulator with an ability to promote pre-adipocyte differentiation by activating ICAT, therefore represses Wnt/β-catenin activity in 3T3-L1 cells. We also demonstrated that ICAT overexpression did not affect oleic acid-induced lipid accumulation at the surface of Hela and HepG2 cells. In conclusion, we show that E2F1 is a critical regulator with an ability to promote differentiation and adipogenesis by activating ICAT in pre-adipocytes.
Collapse
|
31
|
Feraco A, Marzolla V, Scuteri A, Armani A, Caprio M. Mineralocorticoid Receptors in Metabolic Syndrome: From Physiology to Disease. Trends Endocrinol Metab 2020; 31:205-217. [PMID: 31843490 DOI: 10.1016/j.tem.2019.11.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/07/2019] [Accepted: 11/12/2019] [Indexed: 01/28/2023]
Abstract
Over the past decade, several studies have shown that activity of extra-renal mineralocorticoid receptors (MR) regulates vascular tone, adipogenesis, adipose tissue function, and cardiomyocyte contraction. In mice, abnormal activation of MR in the vasculature and in adipose tissue favors the occurrence of several components of the metabolic syndrome (MetS), such as hypertension, obesity, and glucose intolerance. Accordingly, high levels of aldosterone are associated with obesity and MetS in humans, suggesting that altered activation of aldosterone-MR system in extra-renal tissues leads to profound metabolic dysfunctions. In this context, in addition to the classical indications for heart failure and hypertension, MR antagonists (MRAs) nowadays represent a promising approach to tackle cardiovascular and metabolic disorders occurring in the MetS.
Collapse
Affiliation(s)
- Alessandra Feraco
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Vincenzo Marzolla
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Angelo Scuteri
- Department of Medical, Surgical, and Experimental Science, University of Sassari, Sassari, Italy
| | - Andrea Armani
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy; Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy.
| |
Collapse
|
32
|
Haylett WL, Ferris WF. Adipocyte-progenitor cell communication that influences adipogenesis. Cell Mol Life Sci 2020; 77:115-128. [PMID: 31352534 PMCID: PMC11104918 DOI: 10.1007/s00018-019-03256-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/05/2019] [Accepted: 07/24/2019] [Indexed: 12/12/2022]
Abstract
Adipose tissue is located in discrete depots that are differentially associated with elevated risk of metabolic complications, with fat accretion in visceral depots being most detrimental to metabolic health. Currently, the regulation of specific adipose depot expansion, by adipocyte hypertrophy and hyperplasia and consequently fat distribution, is not well understood. However, a growing body of evidence from in vitro investigations indicates that mature adipocytes secrete factors that modulate the proliferation and differentiation of progenitor, adipose-derived stem cells (ADSCs). It is therefore plausible that endocrine communication between adipocytes and ADSCs located in different depots influences fat distribution, and may therefore contribute to the adverse health outcomes associated with visceral adiposity. This review will explore the available evidence of paracrine and endocrine crosstalk between mature adipocytes and ADSCs that affects adipogenesis, as a better understanding of the regulatory roles of the extracellular signalling mechanisms within- and between adipose depots may profoundly change the way we view adipose tissue growth in obesity and related comorbidities.
Collapse
Affiliation(s)
- William Lloyd Haylett
- Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - William Frank Ferris
- Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
| |
Collapse
|
33
|
Hagberg CE, Li Q, Kutschke M, Bhowmick D, Kiss E, Shabalina IG, Harms MJ, Shilkova O, Kozina V, Nedergaard J, Boucher J, Thorell A, Spalding KL. Flow Cytometry of Mouse and Human Adipocytes for the Analysis of Browning and Cellular Heterogeneity. Cell Rep 2019; 24:2746-2756.e5. [PMID: 30184507 PMCID: PMC6137819 DOI: 10.1016/j.celrep.2018.08.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 06/29/2018] [Accepted: 08/02/2018] [Indexed: 01/07/2023] Open
Abstract
Adipocytes, once considered simple lipid-storing cells, are rapidly emerging as complex cells with many biologically diverse functions. A powerful high-throughput method for analyzing single cells is flow cytometry. Several groups have attempted to analyze and sort freshly isolated adipocytes; however, using an adipocyte-specific reporter mouse, we demonstrate that these studies fail to detect the majority of white adipocytes. We define critical settings required for adipocyte flow cytometry and provide a rigid strategy for analyzing and sorting white and brown adipocyte populations. The applicability of our protocol is shown by sorting mouse adipocytes based on size or UCP1 expression and demonstrating that a subset of human adipocytes lacks the β2-adrenergic receptor, particularly in the insulin-resistant state. In conclusion, the present study confers key technological insights for analyzing and sorting mature adipocytes, opening up numerous downstream research applications.
Collapse
Affiliation(s)
- Carolina E Hagberg
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine, Karolinska Institutet, Stockholm 14157, Sweden.
| | - Qian Li
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine, Karolinska Institutet, Stockholm 14157, Sweden; Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Maria Kutschke
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine, Karolinska Institutet, Stockholm 14157, Sweden
| | - Debajit Bhowmick
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine, Karolinska Institutet, Stockholm 14157, Sweden
| | - Endre Kiss
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Irina G Shabalina
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm 10691, Sweden
| | - Matthew J Harms
- Cardiovascular, Renal, and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg 43150, Sweden
| | - Olga Shilkova
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Viviana Kozina
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm 10691, Sweden
| | - Jeremie Boucher
- Cardiovascular, Renal, and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg 43150, Sweden; The Lundberg Laboratory for Diabetes Research, University of Gothenburg, Gothenburg 41345, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg 41345, Sweden
| | - Anders Thorell
- Karolinska Institutet, Department of Clinical Science, Danderyds Hospital, Stockholm 18288, Sweden; Department of Surgery, Ersta Hospital, Stockholm 11691, Sweden
| | - Kirsty L Spalding
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine, Karolinska Institutet, Stockholm 14157, Sweden; Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm 17177, Sweden.
| |
Collapse
|
34
|
Khor NWM, Swarbrick MM, Gunton JE. Inducible UCP1 silencing: A lentiviral RNA-interference approach to quantify the contribution of beige fat to energy homeostasis. PLoS One 2019; 14:e0223987. [PMID: 31751350 PMCID: PMC6872148 DOI: 10.1371/journal.pone.0223987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 10/02/2019] [Indexed: 11/17/2022] Open
Abstract
Energy consuming, heat-producing beige adipocytes, located in classic white adipose tissue (WAT), hold promise for the treatment of obesity. Few reports have quantitatively assessed the contribution of browned 'WAT' to energy expenditure. There is a need for methods to examine beige-fat thermogenesis, independently of classical brown fat. The aim of this study is to optimize an inducible lentiviral shRNA to conditionally knock-down Ucp1 and assess the effects on 'browned' WAT. Primary adipocytes from mouse inguinal WAT converted into thermogenic adipocytes when stimulated with β-adrenergic agonist and thiazolidinedione. There was increased UCP1 protein and importantly increases in various indicators of mitochondrial bioenergetics. Next, we determined optimal transfection conditions for the UCP1-shRNA lentiviral system and subsequently applied this to 'browned' WAT. UCP1 knockdown decreased the brown/beige-fat gene profile and decreased mitochondrial respiration. In summary, this study optimizes lentiviral UCP1-shRNA technology in vitro. This technique could be applied to inguinal fat depots in vivo. This would allow investigation of contribution of depots to whole-body metabolism to help elucidate the physiological relevance of beige fat.
Collapse
Affiliation(s)
- Nicole Wen Mun Khor
- The Westmead Institute for Medical Research, Westmead, Sydney, Australia.,Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia.,Faculty of Medicine, University of New South Wales, Sydney, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Michael M Swarbrick
- The Westmead Institute for Medical Research, Westmead, Sydney, Australia.,Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia.,Faculty of Medicine, University of New South Wales, Sydney, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Jenny E Gunton
- The Westmead Institute for Medical Research, Westmead, Sydney, Australia.,Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia.,Faculty of Medicine, University of New South Wales, Sydney, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| |
Collapse
|
35
|
Anti-obesity effect of cocoa proteins (Theobroma cacao L.) variety “Criollo” and the expression of genes related to the dysfunction of white adipose tissue in high-fat diet-induced obese rats. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103519] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
36
|
Analysis of Tks4 Knockout Mice Suggests a Role for Tks4 in Adipose Tissue Homeostasis in the Context of Beigeing. Cells 2019; 8:cells8080831. [PMID: 31387265 PMCID: PMC6721678 DOI: 10.3390/cells8080831] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/22/2019] [Accepted: 07/30/2019] [Indexed: 12/28/2022] Open
Abstract
Obesity and adipocyte malfunction are related to and arise as consequences of disturbances in signaling pathways. Tyrosine kinase substrate with four Src homology 3 domains (Tks4) is a scaffold protein that establishes a platform for signaling cascade molecules during podosome formation and epidermal growth factor receptor (EGFR) signaling. Several lines of evidence have also suggested that Tks4 has a role in adipocyte biology; however, its roles in the various types of adipocytes at the cellular level and in transcriptional regulation have not been studied. Therefore, we hypothesized that Tks4 functions as an organizing molecule in signaling networks that regulate adipocyte homeostasis. Our aims were to study the white and brown adipose depots of Tks4 knockout (KO) mice using immunohistology and western blotting and to analyze gene expression changes regulated by the white, brown, and beige adipocyte-related transcription factors via a PCR array. Based on morphological differences in the Tks4-KO adipocytes and increased uncoupling protein 1 (UCP1) expression in the white adipose tissue (WAT) of Tks4-KO mice, we concluded that the beigeing process was more robust in the WAT of Tks4-KO mice compared to the wild-type animals. Furthermore, in the Tks4-KO WAT, the expression profile of peroxisome proliferator-activated receptor gamma (PPARγ)-regulated adipogenesis-related genes was shifted in favor of the appearance of beige-like cells. These results suggest that Tks4 and its downstream signaling partners are novel regulators of adipocyte functions and PPARγ-directed white to beige adipose tissue conversion.
Collapse
|
37
|
Ishay-Ronen D, Christofori G. Targeting Cancer Cell Metastasis by Converting Cancer Cells into Fat. Cancer Res 2019; 79:5471-5475. [PMID: 31331908 DOI: 10.1158/0008-5472.can-19-1242] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/18/2019] [Accepted: 07/09/2019] [Indexed: 11/16/2022]
Abstract
Cancer is a systemic heterogeneous disease that can undergo several rounds of latency and activation. Malignant tumors evolve through dynamic responses to microenvironmental signals and development of resistance following therapeutic interventions. Cancer cell adaptation is required for cell survival during metastatic dissemination and outgrowth. Epithelial-mesenchymal transition (EMT) plays a major role in facilitating cell plasticity in cancer and allows cancer cells to escape chemotherapies and targeted therapies through dedifferentiation and signaling adaptation processes. In our recent study, we showed that breast cancer cells that have undergone EMT can be terminally differentiated into adipocytes using the PPARγ agonist rosiglitazone combined with the MEK inhibitor trametinib. The conversion of invasive cancer cells into adipocytes repressed primary tumor invasion and metastasis formation in mouse models of breast cancer. The transdifferentiated cancer cell-derived adipocytes were growth-arrested and lost their cellular plasticity. These results indicate the high potential of utilizing the increased cell plasticity inherent to invasive cancer cells for transdifferentiation therapy.
Collapse
Affiliation(s)
- Dana Ishay-Ronen
- Department of Biomedicine, University of Basel, Basel, Switzerland.
- Oncology Institute, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
- Affiliated with Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | |
Collapse
|
38
|
Caron A, Reynolds RP, Castorena CM, Michael NJ, Lee CE, Lee S, Berdeaux R, Scherer PE, Elmquist JK. Adipocyte Gs but not Gi signaling regulates whole-body glucose homeostasis. Mol Metab 2019; 27:11-21. [PMID: 31279640 PMCID: PMC6717754 DOI: 10.1016/j.molmet.2019.06.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/20/2019] [Accepted: 06/20/2019] [Indexed: 01/24/2023] Open
Abstract
Objective The sympathetic nervous system (SNS) is a key regulator of the metabolic and endocrine functions of adipose tissue. Increased SNS outflow promotes fat mobilization, stimulates non-shivering thermogenesis, promotes browning, and inhibits leptin production. Most of these effects are attributed to norepinephrine activation of the Gs-coupled beta adrenergic receptors located on the surface of the adipocytes. Evidence suggests that other adrenergic receptor subtypes, including the Gi-coupled alpha 2 adrenergic receptors might also mediate the SNS effects on adipose tissue. However, the impact of acute stimulation of adipocyte Gs and Gi has never been reported. Methods We harness the power of chemogenetics to develop unique mouse models allowing the specific and spatiotemporal stimulation of adipose tissue Gi and Gs signaling. We evaluated the impact of chemogenetic stimulation of these pathways on glucose homeostasis, lipolysis, leptin production, and gene expression. Results Stimulation of Gs signaling in adipocytes induced rapid and sustained hypoglycemia. These hypoglycemic effects were secondary to increased insulin release, likely consequent to increased lipolysis. Notably, we also observed differences in gene regulation and ex vivo lipolysis in different adipose depots. In contrast, acute stimulation of Gi signaling in adipose tissue did not affect glucose metabolism or lipolysis, but regulated leptin production. Conclusion Our data highlight the significance of adipose Gs signaling in regulating systemic glucose homeostasis. We also found previously unappreciated heterogeneity across adipose depots following acute stimulation. Together, these results highlight the complex interactions of GPCR signaling in adipose tissue and demonstrate the usefulness of chemogenetic technology to better understand adipocyte function. Chemogenetic stimulation of Gs signaling in adipose tissue potently induces hypoglycemia in mice. The magnitude by which adipose Gs stimulation reduces blood glucose is similar to the hypoglycemic effects of insulin. Chemogenetic stimulation of Gs signaling in adipose tissue ex vivo stimulates lipolysis. Chemogenetic stimulation of adipose Gi signaling does not affect glycemia or lipolysis, but increases leptin levels. Our data demonstrate the usefulness of chemogenetic technology to understand adipocytes functions.
Collapse
Affiliation(s)
- Alexandre Caron
- Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Ryan P Reynolds
- Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Carlos M Castorena
- Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Natalie J Michael
- Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Charlotte E Lee
- Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Syann Lee
- Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rebecca Berdeaux
- Department of Integrative Biology and Pharmacology, Center for Metabolic and Degenerative Diseases at the Brown Foundation, Institute of Molecular Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Graduate Program in Biochemistry and Cell Biology, MD Anderson Cancer Center-UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joel K Elmquist
- Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
39
|
Li K, Wu Y, Yang H, Hong P, Fang X, Hu Y. H19/miR-30a/C8orf4 axis modulates the adipogenic differentiation process in human adipose tissue-derived mesenchymal stem cells. J Cell Physiol 2019; 234:20925-20934. [PMID: 31026067 DOI: 10.1002/jcp.28697] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/03/2019] [Accepted: 04/05/2019] [Indexed: 12/11/2022]
Abstract
The adipogenic differentiation of adipose tissue-derived mesenchymal stem cells (ADSCs) is a critical issue in many obesity-related disorders. Cytidine-cytidine-adenosine-adenosine-thymidine (CCAAT) enhancer binding protein α (CEBP-α) and peroxisome proliferator-activated receptor-γ are two important lipogenic and adipogenic transcription factors and markers in adipogenic differentiation. Noncoding RNAs participate in adipogenic differentiation. The long noncoding RNA (lncRNA) H19 is related to multiple cellular differentiation, including adipogenic differentiation; however, its function and precise molecular mechanism in human ADSCs (hADSCs) adipogenic differentiation are unclear. microRNAs that were differentially expressed in adipogenic differentiation and could be targeted by H19 were screened and selected; the regulation and interaction between H19 and miR-30a were verified. The interaction between miR-30a and predicted downstream target C8orf4 was validated. The dynamic effects of H19 and miR-30a on C8orf4 messenger RNA (mRNA) expression and protein and adipogenic differentiation were evaluated. miR-30a negatively regulated H19 with each other through direct binding. As predicted by TargetScan and verified using luciferase reporter gene assays, miR-30a directly bound to the 3'-untranslated region of C8orf4 to inhibit its expression; H19 knockdown suppressed while miR-30a inhibition promoted the mRNA expression and the protein levels of C8orf4 and adipogenic differentiation; the effect of H19 knockdown could be partially reversed by miR-30a inhibition. The lncRNA H19 serves as a competing endogenous RNA (ceRNA) for miR-30a to augment miR-30a downstream target C8orf4, therefore modulating adipogenic differentiation in hADSCs. From the perspective of lncRNA-miRNA-mRNA regulation, we provided a novel regulatory mechanism of hADSCs adipogenic differentiation.
Collapse
Affiliation(s)
- Kun Li
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, China
| | - Yue Wu
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, China
| | - Hao Yang
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, China
| | - Pengyu Hong
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, China
| | - Xiaodan Fang
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, China
| | - Yanjia Hu
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, China
| |
Collapse
|
40
|
Eyileten C, Mirowska-Guzel D, Milanowski L, Zaremba M, Rosiak M, Cudna A, Kaplon-Cieslicka A, Opolski G, Filipiak KJ, Malek L, Postula M. Serum Brain-Derived Neurotrophic Factor is Related to Platelet Reactivity and Metformin Treatment in Adult Patients With Type 2 Diabetes Mellitus. Can J Diabetes 2019; 43:19-26. [DOI: 10.1016/j.jcjd.2018.01.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 01/31/2018] [Indexed: 01/23/2023]
|
41
|
Abstract
Adipose tissue remains a cryptic organ. The ubiquitous presence of adipocytes, the different fat pads in distinct anatomical locations, the many different types of fat, in each case with their distinct precursor populations, and the ability to interchange into other types of fat cells or even de-differentiate altogether, offers a staggering amount of complexity to the adipose tissue organ as a whole. Adipose tissue holds the key to improving our understanding of systemic metabolic homeostasis. As such, understanding adipose tissue physiology offers the basis for a mechanistic understanding of the pathophysiology of diabetes. This review presents some of the lesser known aspects of this fascinating tissue, which consistently still offers much opportunity for the discovery of novel targets for pharmacological intervention.
Collapse
Affiliation(s)
- Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390-8549, USA.
| |
Collapse
|
42
|
Ishay-Ronen D, Diepenbruck M, Kalathur RKR, Sugiyama N, Tiede S, Ivanek R, Bantug G, Morini MF, Wang J, Hess C, Christofori G. Gain Fat-Lose Metastasis: Converting Invasive Breast Cancer Cells into Adipocytes Inhibits Cancer Metastasis. Cancer Cell 2019; 35:17-32.e6. [PMID: 30645973 DOI: 10.1016/j.ccell.2018.12.002] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 08/21/2018] [Accepted: 12/05/2018] [Indexed: 01/06/2023]
Abstract
Cancer cell plasticity facilitates the development of therapy resistance and malignant progression. De-differentiation processes, such as an epithelial-mesenchymal transition (EMT), are known to enhance cellular plasticity. Here, we demonstrate that cancer cell plasticity can be exploited therapeutically by forcing the trans-differentiation of EMT-derived breast cancer cells into post-mitotic and functional adipocytes. Delineation of the molecular pathways underlying such trans-differentiation has motivated a combination therapy with MEK inhibitors and the anti-diabetic drug Rosiglitazone in various mouse models of murine and human breast cancer in vivo. This combination therapy provokes the conversion of invasive and disseminating cancer cells into post-mitotic adipocytes leading to the repression of primary tumor invasion and metastasis formation.
Collapse
Affiliation(s)
- Dana Ishay-Ronen
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland.
| | - Maren Diepenbruck
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | | | - Nami Sugiyama
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Stefanie Tiede
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Robert Ivanek
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Glenn Bantug
- University Hospital Basel, Department of Biomedicine, University of Basel, Switzerland
| | - Marco Francesco Morini
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Junrong Wang
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Christoph Hess
- University Hospital Basel, Department of Biomedicine, University of Basel, Switzerland
| | - Gerhard Christofori
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland.
| |
Collapse
|
43
|
Alshammari GM, Balakrishnan A. Pumpkin ( Cucurbita ficifolia Bouché) extract attenuate the adipogenesis in human mesenchymal stem cells by controlling adipogenic gene expression. Saudi J Biol Sci 2018; 26:744-751. [PMID: 31048999 PMCID: PMC6486525 DOI: 10.1016/j.sjbs.2018.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/19/2018] [Accepted: 10/01/2018] [Indexed: 01/06/2023] Open
Abstract
Prevention and management of obesity through dietary modification is one of the top way to trim down its consequences. Development of adipose tissue requires the differentiation of less specialized cells, such as human mesenchymal stem cells (hMSCs), into adipocytes. Since food constituents play a major role in the cell differentiation and proliferation, we sought to determine if various extracts of Cucurbita ficifolia (C. ficifolia), could affect the adipogenic differentiation of hMSCs. Flow cytometry analysis with quantitative and qualitative Nile red, and quantitative PCR methods were employed to evaluate the C. ficifolia effect on hMSCs adipogenesis. Results revealed that, chloroform extract exhibits significant adipogenic inhibition than that of hexane and methanol extracts. Chloroform extract treated cells display the down-regulation of ADIPOQ, FABP4, PPARGC1A, CEBPB & LPL and up-regulation of ACACB & CEBPA genes. Further, various phytoconstituents present in the chloroform extract of C. ficifolia were analyzed though LC-MS and GC-MS. Our results indicates that chloroform extract of C. ficifolia might be used as a food supplement to control obesity and its related consequences.
Collapse
Affiliation(s)
- Ghedeir M Alshammari
- Adipocytes and Metabolic Disorders Lab, Department of Food Science and Nutrition, College of Food and Agricultural Science, King Saud University, P.O. Box 2460, Riyadh 11451, Saudi Arabia
| | - Aristatile Balakrishnan
- Adipocytes and Metabolic Disorders Lab, Department of Food Science and Nutrition, College of Food and Agricultural Science, King Saud University, P.O. Box 2460, Riyadh 11451, Saudi Arabia
| |
Collapse
|
44
|
Sadie-Van Gijsen H. Adipocyte biology: It is time to upgrade to a new model. J Cell Physiol 2018; 234:2399-2425. [PMID: 30192004 DOI: 10.1002/jcp.27266] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 07/25/2018] [Indexed: 12/15/2022]
Abstract
Globally, the obesity pandemic is profoundly affecting quality of life and economic productivity, but efforts to address this, especially on a pharmacological level, have generally proven unsuccessful to date, serving as a stark demonstration that our understanding of adipocyte biology and pathophysiology is incomplete. To deliver better insight into adipocyte function and obesity, we need improved adipocyte models with a high degree of fidelity in representing the in vivo state and with a diverse range of experimental applications. Adipocyte cell lines, especially 3T3-L1 cells, have been used extensively over many years, but these are limited in terms of relevance and versatility. In this review, I propose that primary adipose-derived stromal/stem cells (ASCs) present a superior model with which to study adipocyte biology ex vivo. In particular, ASCs afford us the opportunity to study adipocytes from different, functionally distinct, adipose depots and to investigate, by means of in vivo/ex vivo studies, the effects of many different physiological and pathophysiological factors, such as age, body weight, hormonal status, diet and nutraceuticals, as well as disease and pharmacological treatments, on the biology of adipocytes and their precursors. This study will give an overview of the characteristics of ASCs and published studies utilizing ASCs, to highlight the areas where our knowledge is lacking. More comprehensive studies in primary ASCs will contribute to an improved understanding of adipose tissue, in healthy and dysfunctional states, which will enhance our efforts to more successfully manage and treat obesity.
Collapse
Affiliation(s)
- Hanél Sadie-Van Gijsen
- Division of Endocrinology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Parow, South Africa.,Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Parow, South Africa
| |
Collapse
|
45
|
Liu Y, Wang Y, He X, Zhang S, Wang K, Wu H, Chen L. LncRNA TINCR/miR-31-5p/C/EBP-α feedback loop modulates the adipogenic differentiation process in human adipose tissue-derived mesenchymal stem cells. Stem Cell Res 2018; 32:35-42. [PMID: 30172905 DOI: 10.1016/j.scr.2018.08.016] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/30/2018] [Accepted: 08/20/2018] [Indexed: 12/12/2022] Open
Abstract
The adipogenic differentiation of adipose tissue-derived mesenchymal stem cells (ADSCs) is a critical issue in many obesity-related disorders and it can be regulated by a crucial transcription factor, CCAAT enhancer binding protein α (C/EBP-α). Apart from, the involvement of non-coding RNAs in adipogenic differentiation has also been reported. As we know, Terminal differentiation-induced ncRNA (TINCR) is required in somatic tissue differentiation. Recently, we found that TINCR could modulate adipogenic differentiation in hADSCs. As predicted by JASPAR and further confirmed by luciferase reporter gene and ChIP assays, C/EBP-α could bind to the promoter region of lncRNA TINCR to activate its expression. Further, miR-31 was confirmed as a direct target of TINCR and could be negatively regulated by TINCR via competing endogenous RNA (ceRNA) mechanism; miR-31 inhibition enhanced the adipogenic differentiation in hADSCs. More importantly, we found that miR-31 directly bound to the 3'-UTR of C/EBP-α to inhibit its expression. Taken together, in hADSCs, lncRNA TINCR, miR-31 and C/EBP-α formed a feedback loop to modulate the adipogenic differentiation process. From the perspective of lncRNA-miRNA-mRNA regulation, we provided a novel regulatory mechanism of hADSCs adipogenic differentiation.
Collapse
Affiliation(s)
- Yutong Liu
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, PR China
| | - Yiqun Wang
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, PR China
| | - Xifan He
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, PR China
| | - Sheng Zhang
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, PR China
| | - Kai Wang
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, PR China
| | - Hanjiang Wu
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, PR China
| | - Lin Chen
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, PR China.
| |
Collapse
|
46
|
Plubell DL, Fenton AM, Wilmarth PA, Bergstrom P, Zhao Y, Minnier J, Heinecke JW, Yang X, Pamir N. GM-CSF driven myeloid cells in adipose tissue link weight gain and insulin resistance via formation of 2-aminoadipate. Sci Rep 2018; 8:11485. [PMID: 30065264 PMCID: PMC6068153 DOI: 10.1038/s41598-018-29250-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/03/2018] [Indexed: 02/07/2023] Open
Abstract
In a GM-CSF driven myeloid cell deficient mouse model (Csf2−/−) that has preserved insulin sensitivity despite increased adiposity, we used unbiased three-dimensional integration of proteome profiles, metabolic profiles, and gene regulatory networks to understand adipose tissue proteome-wide changes and their metabolic implications. Multi-dimensional liquid chromatography mass spectrometry and extended multiplex mass labeling was used to analyze proteomes of epididymal adipose tissues isolated from Csf2+/+ and Csf2−/− mice that were fed low fat, high fat, or high fat plus cholesterol diets for 8 weeks. The metabolic health (as measured by body weight, adiposity, plasma fasting glucose, insulin, triglycerides, phospholipids, total cholesterol levels, and glucose and insulin tolerance tests) deteriorated with diet for both genotypes, while mice lacking Csf2 were protected from insulin resistance. Regardless of diet, 30 mostly mitochondrial, branch chain amino acids (BCAA), and lysine metabolism proteins were altered between Csf2−/− and Csf2+/+ mice (FDR < 0.05). Lack of GM-CSF driven myeloid cells lead to reduced adipose tissue 2-oxoglutarate dehydrogenase complex (DHTKD1) levels and subsequent increase in plasma 2-aminoadipate (2-AA) levels, both of which are reported to correlate with insulin resistance. Tissue DHTKD1 levels were >4-fold upregulated and plasma 2-AA levels were >2 fold reduced in Csf2−/− mice (p < 0.05). GM-CSF driven myeloid cells link peripheral insulin sensitivity to adiposity via lysine metabolism involving DHTKD1/2-AA axis in a diet independent manner.
Collapse
Affiliation(s)
- Deanna L Plubell
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Alexandra M Fenton
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Phillip A Wilmarth
- Proteomics Shared Resource, Oregon Health & Science University, Portland, OR, USA
| | - Paige Bergstrom
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Yuqi Zhao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Jessica Minnier
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Jay W Heinecke
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Nathalie Pamir
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
47
|
Lau FH, Vogel K, Luckett JP, Hunt M, Meyer A, Rogers CL, Tessler O, Dupin CL, St Hilaire H, Islam KN, Frazier T, Gimble JM, Scahill S. Sandwiched White Adipose Tissue: A Microphysiological System of Primary Human Adipose Tissue. Tissue Eng Part C Methods 2018; 24:135-145. [PMID: 29141507 DOI: 10.1089/ten.tec.2017.0339] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
White adipose tissue (WAT) is a critical organ in both health and disease. However, physiologically faithful tissue culture models of primary human WAT remain limited, at best. In this study we describe a novel WAT culture system in which primary human WAT is sandwiched between tissue-engineered sheets of adipose-derived stromal cells. This construct, called "sandwiched white adipose tissue" (SWAT), can be defined as a microphysiological system (MPS) since it is a tissue-engineered, multicellular, three-dimensional organ construct produced using human cells. We validated SWAT against the National Institutes of Health MPS standards and found that SWAT is viable in culture for 8 weeks, retains physiologic responses to exogenous signaling, secretes adipokines, and engrafts into animal models. These attributes position SWAT as a powerful tool for the study of WAT physiology, pathophysiology, personalized medicine, and pharmaceutical development.
Collapse
Affiliation(s)
- Frank H Lau
- 1 LSUHSC SOM's Department of Surgery, Louisiana State University Health Sciences Center School of Medicine , New Orleans, Louisiana
| | - Kelly Vogel
- 1 LSUHSC SOM's Department of Surgery, Louisiana State University Health Sciences Center School of Medicine , New Orleans, Louisiana
| | - John P Luckett
- 1 LSUHSC SOM's Department of Surgery, Louisiana State University Health Sciences Center School of Medicine , New Orleans, Louisiana
| | - Maxwell Hunt
- 1 LSUHSC SOM's Department of Surgery, Louisiana State University Health Sciences Center School of Medicine , New Orleans, Louisiana
| | - Alicia Meyer
- 1 LSUHSC SOM's Department of Surgery, Louisiana State University Health Sciences Center School of Medicine , New Orleans, Louisiana
| | - Camille L Rogers
- 1 LSUHSC SOM's Department of Surgery, Louisiana State University Health Sciences Center School of Medicine , New Orleans, Louisiana
| | - Oren Tessler
- 1 LSUHSC SOM's Department of Surgery, Louisiana State University Health Sciences Center School of Medicine , New Orleans, Louisiana
| | - Charles L Dupin
- 1 LSUHSC SOM's Department of Surgery, Louisiana State University Health Sciences Center School of Medicine , New Orleans, Louisiana
| | - Hugo St Hilaire
- 1 LSUHSC SOM's Department of Surgery, Louisiana State University Health Sciences Center School of Medicine , New Orleans, Louisiana
| | - Kazi N Islam
- 1 LSUHSC SOM's Department of Surgery, Louisiana State University Health Sciences Center School of Medicine , New Orleans, Louisiana
| | - Trivia Frazier
- 2 New Orleans BioInnovation Center , LaCell LLC, New Orleans Louisiana
| | - Jeffrey M Gimble
- 3 Center for Stem Cell Research and Regenerative Medicine, Tulane University , New Orleans, Louisiana
| | - Steven Scahill
- 1 LSUHSC SOM's Department of Surgery, Louisiana State University Health Sciences Center School of Medicine , New Orleans, Louisiana
| |
Collapse
|
48
|
Abstract
The adult human adipose tissue is predominantly composed of white adipocytes. However, within certain depots, adipose tissue contains thermogenically active brown-like adipocytes, which have been evolutionarily conserved in mammals. This chapter will give a brief overview on the methods used to genetically target and trace both white and brown adipocytes using techniques such as bacterial artificial chromosome (BAC) cloning to create transgenic mouse models and the tools with which genetic recombination is mediated in vivo (e.g., Cre-loxP, CreERT, and Tet-On). The chapter furthermore critically discusses the strength and limitation of the various systems used to target mature white and brown adipocytes (ap2-Cre, Adipoq-Cre, and Ucp1-Cre). Based on these systems, it is evident that our knowledge of mature adipocyte categorization into brown, white, brite, or beige adipocytes is strongly influenced by the use of the various genetic mouse models described in this chapter. Our evaluation of different studies using the aforementioned systems focuses on key genes, which have been reported to maintain adipocyte's function (insulin receptor, Raptor, or Atgl).
Collapse
Affiliation(s)
- Christian Wolfrum
- Institute of Food, Nutrition, and Health, ETH Zurich, Zürich, Switzerland
| | | |
Collapse
|
49
|
Ilex latifolia Thunb protects mice from HFD-induced body weight gain. Sci Rep 2017; 7:14660. [PMID: 29116160 PMCID: PMC5676986 DOI: 10.1038/s41598-017-15292-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 10/25/2017] [Indexed: 12/16/2022] Open
Abstract
Kuding tea is implicated in alleviating metabolic disorders in traditional Chinese medicine. However, the role of Ilex latifolia Thunb (kuding tea), one of the large leaf kuding tea species, in the prevention of the development of obesity remains to be determined. We show here that 7-week-old male mice treated with an Ilex latifolia Thunb supplement for 14 weeks were resistant to HFD-induced body weight gain and hepatic steatosis, accompanied by improved insulin sensitivity. Ilex latifolia Thunb supplementation dramatically reduced the systemic and tissue inflammation levels of mice via reducing pro-inflammatory cytokine levels, increasing anti-inflammatory cytokine levels in the circulation and inhibiting p38 MAPK and p65 NF-κB signaling in adipose tissue. Together, these results indicate that Ilex latifolia Thunb protects mice from the development of obesity and is a potential compound pool for the development of novel anti-obesity drugs.
Collapse
|
50
|
Lee YM, Yoon Y, Yoon H, Park HM, Song S, Yeum KJ. Dietary Anthocyanins against Obesity and Inflammation. Nutrients 2017; 9:nu9101089. [PMID: 28974032 PMCID: PMC5691706 DOI: 10.3390/nu9101089] [Citation(s) in RCA: 219] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 09/25/2017] [Accepted: 09/25/2017] [Indexed: 12/26/2022] Open
Abstract
Chronic low-grade inflammation plays a pivotal role in the pathogenesis of obesity, due to its associated chronic diseases such as type II diabetes, cardiovascular diseases, pulmonary diseases and cancer. Thus, targeting inflammation is an attractive strategy to counter the burden of obesity-induced health problems. Recently, food-derived bioactive compounds have been spotlighted as a regulator against various chronic diseases due to their low toxicity, as opposed to drugs that induce severe side effects. Here we describe the beneficial effects of dietary anthocyanins on obesity-induced metabolic disorders and inflammation. Red cabbage microgreen, blueberry, blackcurrant, mulberry, cherry, black elderberry, black soybean, chokeberry and jaboticaba peel contain a variety of anthocyanins including cyanidins, delphinidins, malvidins, pelargonidins, peonidins and petunidins, and have been reported to alter both metabolic markers and inflammatory markers in cells, animals, and humans. This review discusses the interplay between inflammation and obesity, and their subsequent regulation via the use of dietary anthocyanins, suggesting an alternative dietary strategy to ameliorate obesity and obesity associated chronic diseases.
Collapse
Affiliation(s)
- Yoon-Mi Lee
- Division of Food Bioscience, College of Biomedical and Health Sciences, Konkuk University, Chungju-si 27478, Korea.
- Nanotechnology Research Center, Konkuk University, Chungju-si 27478, Korea.
| | - Young Yoon
- Division of Food Bioscience, College of Biomedical and Health Sciences, Konkuk University, Chungju-si 27478, Korea.
| | - Haelim Yoon
- Division of Food Bioscience, College of Biomedical and Health Sciences, Konkuk University, Chungju-si 27478, Korea.
| | - Hyun-Min Park
- Division of Food Bioscience, College of Biomedical and Health Sciences, Konkuk University, Chungju-si 27478, Korea.
| | - Sooji Song
- Division of Food Bioscience, College of Biomedical and Health Sciences, Konkuk University, Chungju-si 27478, Korea.
| | - Kyung-Jin Yeum
- Division of Food Bioscience, College of Biomedical and Health Sciences, Konkuk University, Chungju-si 27478, Korea.
- Institute of Biomedical and Health Science, Konkuk University, Chungju-si 27478, Korea.
| |
Collapse
|