51
|
Zhao X, Hendriks I, Le Gras S, Ye T, Ramos-Alonso L, Nguéa P A, Lien G, Ghasemi F, Klungland A, Jost B, Enserink J, Nielsen M, Chymkowitch P. OUP accepted manuscript. Nucleic Acids Res 2022; 50:1351-1369. [PMID: 35100417 PMCID: PMC8860575 DOI: 10.1093/nar/gkac027] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/05/2022] [Accepted: 01/10/2022] [Indexed: 11/20/2022] Open
Abstract
Tight control of gene expression networks required for adipose tissue formation and plasticity is essential for adaptation to energy needs and environmental cues. However, the mechanisms that orchestrate the global and dramatic transcriptional changes leading to adipocyte differentiation remain to be fully unraveled. We investigated the regulation of nascent transcription by the sumoylation pathway during adipocyte differentiation using SLAMseq and ChIPseq. We discovered that the sumoylation pathway has a dual function in differentiation; it supports the initial downregulation of pre-adipocyte-specific genes, while it promotes the establishment of the mature adipocyte transcriptional program. By characterizing endogenous sumoylome dynamics in differentiating adipocytes by mass spectrometry, we found that sumoylation of specific transcription factors like PPARγ/RXR and their co-factors are associated with the transcription of adipogenic genes. Finally, using RXR as a model, we found that sumoylation may regulate adipogenic transcription by supporting the chromatin occurrence of transcription factors. Our data demonstrate that the sumoylation pathway supports the rewiring of transcriptional networks required for formation of functional adipocytes. This study also provides the scientists in the field of cellular differentiation and development with an in-depth resource of the dynamics of the SUMO-chromatin landscape, SUMO-regulated transcription and endogenous sumoylation sites during adipocyte differentiation.
Collapse
Affiliation(s)
- Xu Zhao
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway
- Department of Microbiology, Oslo University Hospital, 0372 Oslo, Norway
| | | | | | - Tao Ye
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, Inserm U964, Université de Strasbourg, Illkirch, France
| | - Lucía Ramos-Alonso
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway
- Department of Microbiology, Oslo University Hospital, 0372 Oslo, Norway
| | - Aurélie Nguéa P
- Department of Microbiology, Oslo University Hospital, 0372 Oslo, Norway
| | - Guro Flor Lien
- Department of Microbiology, Oslo University Hospital, 0372 Oslo, Norway
| | - Fatemeh Ghasemi
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway
| | - Arne Klungland
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway
- Department of Microbiology, Oslo University Hospital, 0372 Oslo, Norway
| | - Bernard Jost
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, Inserm U964, Université de Strasbourg, Illkirch, France
| | - Jorrit M Enserink
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research,Oslo University Hospital, 0372 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318 Oslo, Norway
| | - Michael L Nielsen
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research (NNF-CPR), Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | | |
Collapse
|
52
|
Jang SY, Kim J, Park JT, Liu CY, Korn BS, Kikkawa DO, Lee EJ, Yoon JS. Therapeutic Potential of Targeting Periostin in the Treatment of Graves' Orbitopathy. Front Endocrinol (Lausanne) 2022; 13:900791. [PMID: 35707463 PMCID: PMC9189304 DOI: 10.3389/fendo.2022.900791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Periostin is a matricellular protein that is ubiquitously expressed in normal human tissues and is involved in pathologic mechanism of chronic inflammatory and fibrotic disease. In this study we investigate periostin in the pathogenesis of Graves' orbitopathy (GO) using human orbital adipose tissue obtained from surgery and primary cultured orbital fibroblasts in vitro. POSTN (gene encoding periostin) expression in Graves' orbital tissues and healthy control tissues was studied, and the role of periostin in GO pathologic mechanism was examined through small-interfering RNA (siRNA)-mediated silencing. POSTN gene expression was significantly higher in Graves' orbital tissues than healthy control tissues in real-time PCR results, and immunohistochemical staining revealed higher expression of periostin in Graves' orbital tissues than normal tissues. Silencing periostin using siRNA transfection significantly attenuated TGF-β-induced profibrotic protein production and phosphorylated p38 and SMAD protein production. Knockdown of periostin inhibited interleukin-1 β -induced proinflammatory cytokines production as well as phosphorylation of NF-κB and Ak signaling protein. Adipocyte differentiation was also suppressed in periostin-targeting siRNA transfected GO cells. We hypothesize that periostin contributes to the pathogenic process of inflammation, fibrosis and adipogenesis of GO. Our study provides in vitro evidence that periostin may be a novel potential therapeutic target for the treatment of GO.
Collapse
Affiliation(s)
- Sun Young Jang
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, South Korea
| | - Jinjoo Kim
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Jung Tak Park
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, South Korea
| | - Catherine Y. Liu
- Division of Oculofacial Plastic and Reconstructive Surgery, University of California, San Diego, La Jolla, CA, United States
| | - Bobby S. Korn
- Division of Oculofacial Plastic and Reconstructive Surgery, University of California, San Diego, La Jolla, CA, United States
| | - Don O. Kikkawa
- Division of Oculofacial Plastic and Reconstructive Surgery, University of California, San Diego, La Jolla, CA, United States
| | - Eun Jig Lee
- Division of Endocrinology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Jin Sook Yoon
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea
- *Correspondence: Jin Sook Yoon,
| |
Collapse
|
53
|
Impact of oral lipid and glucose tolerance tests on the postprandial concentrations of angiopoietin-like proteins (Angptl) 3 and 4. Eur J Nutr 2021; 61:1919-1929. [PMID: 34951653 PMCID: PMC9106616 DOI: 10.1007/s00394-021-02748-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/16/2021] [Indexed: 11/22/2022]
Abstract
Background The postprandial regulation of angiopoietin-like proteins (Angptls) and their expression in adipocytes is poorly characterized. Objective Circulating Angptl3 and 4 were analyzed in healthy individuals undergoing either an oral lipid tolerance test (OLTT; n = 98) or an oral glucose tolerance test (OGTT; n = 99). Venous blood was drawn after 0, 2, 4, and 6 h during OLTT and after 0, 1, and 2 h during OGTT. Anthropometric and laboratory parameters were assessed and concentrations of Angptls were quantified by enzyme-linked immunosorbent assay. Angptl gene expression in 3T3-L1 adipocytes and in murine adipose tissues and cellular fractions was analyzed by quantitative real-time PCR. Results Angptl3 concentrations significantly decreased while Angptl4 levels continuously increased during OLTT. Both proteins remained unaffected during OGTT. Angptl3 and Angptl4 were expressed in murine subcutaneous and visceral AT with higher mRNA levels in mature adipocytes when compared to the stroma-vascular cell fraction. Both proteins were strongly induced during 3T3-L1 adipocyte differentiation and they were unresponsive to glucose in mature fat cells. Adipocyte Angptl3 (but not Angptl4) mRNA expression was inhibited by the polyunsaturated fatty acids arachidonic acid and docosahexaenoic acid, whereas nine types of dietary fatty acids remained without any effect. Conclusions There is evidence of short-time regulation of Angptl3/4 levels upon metabolic stress. Angptl4 expression is high and Angptl3 expression is low in AT and restricted mainly to mature adipocytes without any differences concerning fat compartments. Whereas dietary fatty acids and glucose are without any effect, omega-3/-6-polyunsaturated fatty acids inhibited Anptl3 expression in adipocytes.
Collapse
|
54
|
Schmidt V, Horváth C, Dong H, Blüher M, Qvist P, Wolfrum C, Willnow TE. SORLA is required for insulin-induced expansion of the adipocyte precursor pool in visceral fat. J Cell Biol 2021; 220:e202006058. [PMID: 34779857 PMCID: PMC8598079 DOI: 10.1083/jcb.202006058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 06/19/2021] [Accepted: 09/08/2021] [Indexed: 01/24/2023] Open
Abstract
Visceral adipose tissue shows remarkable plasticity, constantly replacing mature adipocytes from an inherent pool of adipocyte precursors. The number of precursors is set in the juvenile organism and remains constant in adult life. Which signals drive precursor pool expansion in juveniles and why they operate in visceral but not in subcutaneous white adipose tissue (WAT) are unclear. Using mouse models, we identified the insulin-sensitizing receptor SORLA as a molecular factor explaining the distinct proliferative capacity of visceral WAT. High levels of SORLA activity in precursors of juvenile visceral WAT prime these cells for nutritional stimuli provided through insulin, promoting mitotic expansion of the visceral precursor cell pool in overfed juvenile mice. SORLA activity is low in subcutaneous precursors, blunting their response to insulin and preventing diet-induced proliferation of this cell type. Our findings provide a molecular explanation for the unique proliferative properties of juvenile visceral WAT, and for the genetic association of SORLA with visceral obesity in humans.
Collapse
Affiliation(s)
- Vanessa Schmidt
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Carla Horváth
- Laboratory of Translational Nutrition Biology, ETH Zurich, Schwerzenbach, Switzerland
| | - Hua Dong
- Laboratory of Translational Nutrition Biology, ETH Zurich, Schwerzenbach, Switzerland
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Per Qvist
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Centre for Genomics and Personalized Medicine, Aarhus University, Aarhus, Denmark
| | - Christian Wolfrum
- Laboratory of Translational Nutrition Biology, ETH Zurich, Schwerzenbach, Switzerland
| | - Thomas E. Willnow
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
55
|
Al-Sayegh M, Ali H, Jamal MH, ElGindi M, Chanyong T, Al-Awadi K, Abu-Farha M. Mouse Embryonic Fibroblast Adipogenic Potential: A Comprehensive Transcriptome Analysis. Adipocyte 2021; 10:1-20. [PMID: 33345692 PMCID: PMC7757854 DOI: 10.1080/21623945.2020.1859789] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Our understanding of adipose tissue has progressed from an inert tissue for energy storage to be one of the largest endocrine organs regulating metabolic homoeostasis through its ability to synthesize and release various adipokines that regulate a myriad of pathways. The field of adipose tissue biology is growing due to this association with various chronic metabolic diseases. An important process in the regulation of adipose tissue biology is adipogenesis, which is the formation of new adipocytes. Investigating adipogenesis in vitro is currently a focus for identifying factors that might be utilized in clinically. A powerful tool for such work is high-throughput sequencing which can rapidly identify changes at gene expression level. Various cell models exist for studying adipogenesis and has been used in high-throughput studies, yet little is known about transcriptome profile that underlies adipogenesis in mouse embryonic fibroblasts. This study utilizes RNA-sequencing and computational analysis with DESeq2, gene ontology, protein–protein networks, and robust rank analysis to understand adipogenesis in mouse embryonic fibroblasts in-depth. Our analyses confirmed the requirement of mitotic clonal expansion prior to adipogenesis in this cell model and highlight the role of Cebpa and Cebpb in regulating adipogenesis through interactions of large numbers of genes.
Collapse
Affiliation(s)
- Mohamed Al-Sayegh
- New York University Abu Dhabi, Division of Biology, Abu Dhabi, United Arab Emirates
| | - Hamad Ali
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Health Sciences Center (HSC), Kuwait University, Kuwait City, State of Kuwait
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute (DDI), Kuwait City, State of Kuwait
| | - Mohammad H Jamal
- Department of Surgery, Faculty of Medicine, Health Sciences Center (HSC), Kuwait University, Kuwait City, State of Kuwait
| | - Mei ElGindi
- New York University Abu Dhabi, Division of Biology, Abu Dhabi, United Arab Emirates
| | - Tina Chanyong
- New York University Abu Dhabi, Division of Biology, Abu Dhabi, United Arab Emirates
| | - Khulood Al-Awadi
- New York University Abu Dhabi, Design Studio, Abu Dhabi, United Arab Emirates
| | - Mohamed Abu-Farha
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute (DDI), Kuwait City, State of Kuwait
| |
Collapse
|
56
|
Suchý T, Kaczmarek I, Maricic T, Zieschang C, Schöneberg T, Thor D, Liebscher I. Evaluating the feasibility of Cas9 overexpression in 3T3-L1 cells for generation of genetic knock-out adipocyte cell lines. Adipocyte 2021; 10:631-645. [PMID: 34915813 PMCID: PMC8735834 DOI: 10.1080/21623945.2021.1990480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Cell lines recapitulating physiological processes can represent alternatives to animal or human studies. The 3T3-L1 cell line is used to mimic adipocyte function and differentiation. Since transfection of 3T3-L1 cells is difficult, we used a modified 3T3-L1 cell line overexpressing Cas9 for a straightforward generation of gene knock-outs. As an example, we intended to generate 3T3-L1 cell lines deficient for adhesion G protein-coupled receptors Gpr64/Adgr2 and Gpr126/Adgr6 using the CRISPR/Cas approach. Surprisingly, all the generated knock-out as well as scramble control cell lines were unresponsive to isoprenaline in respect to adiponectin secretion and lipolysis in contrast to the wild type 3T3-L1 cells. We, therefore, analysed the properties of these stable Cas9-overexpressing 3T3-L1 cells. We demonstrate that this commercially available cell line exhibits dysfunction in cAMP signalling pathways as well as reduced insulin sensitivity independent of gRNA transfection. We tried transient transfection of plasmids harbouring Cas9 as well as direct introduction of the Cas9 protein as alternate approaches to the stable expression of this enzyme. We find that transfection of the Cas9 protein is not only feasible but also does not impair adipogenesis and, therefore, represents a preferable alternative to achieve genetic knock-out.
Collapse
Affiliation(s)
- Tomás Suchý
- Department of Molecular Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Isabell Kaczmarek
- Department of Molecular Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Tomislav Maricic
- Department of Evolutionary Genetics, Max-Planck-Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Christian Zieschang
- Department of Molecular Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Torsten Schöneberg
- Department of Molecular Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Doreen Thor
- Department of Molecular Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Ines Liebscher
- Department of Molecular Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| |
Collapse
|
57
|
Ahmed M, Lai TH, Kim DR. A Small Fraction of Progenitors Differentiate Into Mature Adipocytes by Escaping the Constraints on the Cell Structure. Front Cell Dev Biol 2021; 9:753042. [PMID: 34708046 PMCID: PMC8542793 DOI: 10.3389/fcell.2021.753042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 09/10/2021] [Indexed: 11/13/2022] Open
Abstract
Differentiating 3T3-L1 pre-adipocytes are a mixture of non-identical culture cells. It is vital to identify the cell types that respond to the induction stimulus to understand the pre-adipocyte potential and the mature adipocyte behavior. To test this hypothesis, we deconvoluted the gene expression profiles of the cell culture of MDI-induced 3T3-L1 cells. Then we estimated the fractions of the sub-populations and their changes in time. We characterized the sub-populations based on their specific expression profiles. Initial cell cultures comprised three distinct phenotypes. A small fraction of the starting cells responded to the induction and developed into mature adipocytes. Unresponsive cells were probably under structural constraints or were committed to differentiating into alternative phenotypes. Using the same population gene markers, similar proportions were found in induced human primary adipocyte cell cultures. The three sub-populations had diverse responses to treatment with various drugs and compounds. Only the response of the maturating sub-population resembled that estimated from the profiles of the mixture. We then showed that even at a low division rate, a small fraction of cells could increase its share in a dynamic two-populations model. Finally, we used a cell cycle expression index to validate that model. To sum, pre-adipocytes are a mixture of different cells of which a limited fraction become mature adipocytes.
Collapse
Affiliation(s)
- Mahmoud Ahmed
- Department of Biochemistry and Convergence Medical Sciences, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, South Korea
| | - Trang Huyen Lai
- Department of Biochemistry and Convergence Medical Sciences, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, South Korea
| | - Deok Ryong Kim
- Department of Biochemistry and Convergence Medical Sciences, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, South Korea
| |
Collapse
|
58
|
Meteorin-Like Protein (Metrnl) in Obesity, during Weight Loss and in Adipocyte Differentiation. J Clin Med 2021; 10:jcm10194338. [PMID: 34640356 PMCID: PMC8509786 DOI: 10.3390/jcm10194338] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/08/2021] [Accepted: 09/13/2021] [Indexed: 12/05/2022] Open
Abstract
Meteorin-like protein (Metrnl) is an adipo-myokine with pleiotropic effects in adipose tissue (AT). Its systemic regulation in obesity and under weight loss is unclear. Circulating Metrnl concentrations were analyzed by ELISA in severely obese patients undergoing bariatric surgery (BS) or low calorie diet (LCD). Metrnl mRNA expression was analyzed in human and murine tissues and cell lines by quantitative real-time PCR. About 312 morbidly obese individuals underwent BS (n = 181; BMI 53.4 + 6.8 kg/m2) or LCD (n = 131; BMI 43.5 + 6.7 kg/m2). Serum samples were obtained at baseline and 3, 6, and 12 months after intervention. AT specimen from subcutaneous and visceral adipose tissue were resected during BS. Serum Metrnl levels were lower in type 2 diabetic patients and negatively correlated with HbA1c. In BS and LCD patients, Metrnl concentrations significantly increased after 3 months and returned to baseline levels after 12 months. There was no gender-specific effect. Metrnl mRNA expression did not differ between visceral and subcutaneous AT in n = 130 patients. In contrast, Metrnl gene expression in mice was highest in intra-abdominal AT followed by subcutaneous, peri-renal, and brown AT. In the murine 3T3-L1 cell line, Metrnl expression was high in pre-adipocytes and mature adipocytes with a transient downregulation during adipocyte differentiation. Metrnl expression remained unaffected upon treatment with glucose, insulin, fatty acids, bile acids, and incretins. Polyunsaturated omega-3 and omega-6 fatty acids downregulated Metrnl expression. Systemic Metrnl is transiently upregulated during massive weight loss and gene expression in adipocytes is differentially regulated.
Collapse
|
59
|
Heydt Q, Xintaropoulou C, Clear A, Austin M, Pislariu I, Miraki-Moud F, Cutillas P, Korfi K, Calaminici M, Cawthorn W, Suchacki K, Nagano A, Gribben JG, Smith M, Cavenagh JD, Oakervee H, Castleton A, Taussig D, Peck B, Wilczynska A, McNaughton L, Bonnet D, Mardakheh F, Patel B. Adipocytes disrupt the translational programme of acute lymphoblastic leukaemia to favour tumour survival and persistence. Nat Commun 2021; 12:5507. [PMID: 34535653 PMCID: PMC8448863 DOI: 10.1038/s41467-021-25540-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 08/17/2021] [Indexed: 11/09/2022] Open
Abstract
The specific niche adaptations that facilitate primary disease and Acute Lymphoblastic Leukaemia (ALL) survival after induction chemotherapy remain unclear. Here, we show that Bone Marrow (BM) adipocytes dynamically evolve during ALL pathogenesis and therapy, transitioning from cellular depletion in the primary leukaemia niche to a fully reconstituted state upon remission induction. Functionally, adipocyte niches elicit a fate switch in ALL cells towards slow-proliferation and cellular quiescence, highlighting the critical contribution of the adipocyte dynamic to disease establishment and chemotherapy resistance. Mechanistically, adipocyte niche interaction targets posttranscriptional networks and suppresses protein biosynthesis in ALL cells. Treatment with general control nonderepressible 2 inhibitor (GCN2ib) alleviates adipocyte-mediated translational repression and rescues ALL cell quiescence thereby significantly reducing the cytoprotective effect of adipocytes against chemotherapy and other extrinsic stressors. These data establish how adipocyte driven restrictions of the ALL proteome benefit ALL tumours, preventing their elimination, and suggest ways to manipulate adipocyte-mediated ALL resistance.
Collapse
Affiliation(s)
- Q Heydt
- Centre for Haemato-Oncology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - C Xintaropoulou
- Centre for Haemato-Oncology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - A Clear
- Centre for Haemato-Oncology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - M Austin
- Centre for Haemato-Oncology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - I Pislariu
- Centre for Haemato-Oncology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - F Miraki-Moud
- Centre for Haemato-Oncology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - P Cutillas
- Centre for Haemato-Oncology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - K Korfi
- Centre for Haemato-Oncology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - M Calaminici
- Centre for Haemato-Oncology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - W Cawthorn
- BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, Scotland, UK
| | - K Suchacki
- BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, Scotland, UK
| | - A Nagano
- Centre for Molecular Oncology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - J G Gribben
- Centre for Haemato-Oncology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - M Smith
- Department of Haemato-Oncology, St Bartholomew's Hospital, West Smithfield, London, UK
| | - J D Cavenagh
- Department of Haemato-Oncology, St Bartholomew's Hospital, West Smithfield, London, UK
| | - H Oakervee
- Department of Haemato-Oncology, St Bartholomew's Hospital, West Smithfield, London, UK
| | - A Castleton
- Christie NHS Foundation Trust, Manchester, UK
| | - D Taussig
- Haemato-oncology Unit, The Royal Marsden Hospital, Sutton, UK
| | - B Peck
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - A Wilczynska
- CRUK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - L McNaughton
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, UK
| | - D Bonnet
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, UK
| | - F Mardakheh
- Centre for Molecular Oncology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - B Patel
- Centre for Haemato-Oncology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK.
| |
Collapse
|
60
|
Reproducibility of adipogenic responses to metabolism disrupting chemicals in the 3T3-L1 pre-adipocyte model system: An interlaboratory study. Toxicology 2021; 461:152900. [PMID: 34411659 DOI: 10.1016/j.tox.2021.152900] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/02/2021] [Accepted: 08/13/2021] [Indexed: 12/17/2022]
Abstract
The 3T3-L1 murine pre-adipocyte line is an established cell culture model for screening Metabolism Disrupting Chemicals (MDCs). Despite a need to accurately identify MDCs for further evaluation, relatively little research has been performed to comprehensively evaluate reproducibility across laboratories, assess factors that might contribute to varying degrees of differentiation between laboratories (media additives, plastics, cell source, etc.), or to standardize protocols. As such, the goals of this study were to assess interlaboratory variability of efficacy and potency outcomes for triglyceride accumulation and pre-adipocyte proliferation using the mouse 3T3-L1 pre-adipocyte cell assay to test chemicals. Ten laboratories from five different countries participated. Each laboratory evaluated one reference chemical (rosiglitazone) and three blinded test chemicals (tributyltin chloride, pyraclostrobin, and bisphenol A) using: 1) their Laboratory-specific 3T3-L1 Cells (LC) and their Laboratory-specific differentiation Protocol (LP), 2) Shared 3T3-L1 Cells (SC) with LP, 3) LC with a Shared differentiation Protocol (SP), and 4) SC with SP. Blinded test chemical responses were analyzed by the coordinating laboratory. The magnitude and range of bioactivities reported varied considerably across laboratories and test conditions, though the presence or absence of activity for each tested chemical was more consistent. Triglyceride accumulation activity determinations for rosiglitazone ranged from 90 to 100% across test conditions, but 30-70 % for pre-adipocyte proliferation; this was 40-80 % for triglyceride accumulation induced by pyraclostrobin, 80-100 % for tributyltin, and 80-100 % for bisphenol A. Consistency was much lower for pre-adipocyte proliferation, with 30-70 % active determinations for pyraclostrobin, 30-50 % for tributyltin, and 20-40 % for bisphenol A. Greater consistency was observed for the SC/SP assessment. As such, working to develop a standardized adipogenic differentiation protocol represents the best strategy for improving consistency of adipogenic responses using the 3T3-L1 model to reproducibly identify MDCs and increase confidence in reported outcomes.
Collapse
|
61
|
Karrasch T, Höpfinger A, Schäffler A, Schmid A. The adipokine C1q/TNF-related protein-3 (CTRP-3) inhibits Toll-like receptor (TLR)-induced expression of Cathelicidin antimicrobial peptide (CAMP) in adipocytes. Cytokine 2021; 148:155663. [PMID: 34388476 DOI: 10.1016/j.cyto.2021.155663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND AIM CAMP (Cathelicidin antimicrobial peptide) expression in adipocytes is regulated by Toll-like receptor (TLR) agonists. Secreted adipokines such as CTRP-3 have been suggested to participate in innate immune signaling in adipose tissue (AT). This study investigates whether TLR-induced CAMP expression in adipocytes is antagonized by CTRP-3. METHODS 3T3-L1 adipocytes were co-stimulated with TLR agonists (LPS, MALP-2, Pam3CSK4, pI:C) and recombinant CTRP-3. In a SIRS model, C57BL/6 wild-type mice were intraperitoneally (ip) injected with recombinant CTRP-3 prior to LPS. CAMP expression was analyzed by real-time PCR in AT of wild-type mice and in AT and primary adipocytes from transgenic mice lacking adipocyte CTRP-3 expression. Comparative transcriptome analysis by RNA seq. was applied in CTRP-3 KO adipocytes. RESULTS In vitro, CTRP-3 antagonized TLR4- and TLR1/2-induced CAMP expression in adipocytes whereas TLR3- and TLR2/6-mediated induction of CAMP was not affected. in vivo, application of exogenous CTRP-3 dose-dependently antagonized LPS-induced CAMP expression in intra-abdominal AT. CAMP expression in total AT and in primary adipocytes of subcutaneous and intra-abdominal AT did not differ between wild-type mice and transgenic mice lacking adipocyte CTRP-3 expression. CONCLUSIONS The study suggests a hypothetical role of CAMP in host defense not only against Gram-positive bacteria sensed by TLR1/2 and TLR2/6 but also against Gram-negative bacteria sensed by TLR4 and potentially against viruses sensed by TLR3. The machinery of TLR-mediated pro-inflammatory activation of the CAMP gene in adipocytes seems to be partly modulated by secreted adipokines belonging to the growing family of C1q/TNF-related proteins such as CTRP-3.
Collapse
Affiliation(s)
- Thomas Karrasch
- Department of Internal Medicine III, University of Giessen, Germany
| | | | | | - Andreas Schmid
- Department of Internal Medicine III, University of Giessen, Germany.
| |
Collapse
|
62
|
Kim S, Reed E, Monti S, Schlezinger JJ. A Data-Driven Transcriptional Taxonomy of Adipogenic Chemicals to Identify White and Brite Adipogens. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:77006. [PMID: 34323617 PMCID: PMC8320370 DOI: 10.1289/ehp6886] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
BACKGROUND Chemicals in disparate structural classes activate specific subsets of the transcriptional programs of peroxisome proliferator-activated receptor-γ (PPARγ) to generate adipocytes with distinct phenotypes. OBJECTIVES Our objectives were to a) establish a novel classification method to predict PPARγ ligands and modifying chemicals; and b) create a taxonomy to group chemicals on the basis of their effects on PPARγ's transcriptome and downstream metabolic functions. We tested the hypothesis that environmental adipogens highly ranked by the taxonomy, but segregated from therapeutic PPARγ ligands, would induce white but not brite adipogenesis. METHODS 3T3-L1 cells were differentiated in the presence of 76 chemicals (negative controls, nuclear receptor ligands known to influence adipocyte biology, potential environmental PPARγ ligands). Differentiation was assessed by measuring lipid accumulation. mRNA expression was determined by RNA-sequencing (RNA-Seq) and validated by reverse transcription-quantitative polymerase chain reaction. A novel classification model was developed using an amended random forest procedure. A subset of environmental contaminants identified as strong PPARγ agonists were analyzed by their effects on lipid handling, mitochondrial biogenesis, and cellular respiration in 3T3-L1 cells and human preadipocytes. RESULTS We used lipid accumulation and RNA-Seq data to develop a classification system that a) identified PPARγ agonists; and b) sorted chemicals into likely white or brite adipogens. Expression of Cidec was the most efficacious indicator of strong PPARγ activation. 3T3-L1 cells treated with two known environmental PPARγ ligands, tetrabromobisphenol A and triphenyl phosphate, which sorted distinctly from therapeutic ligands, had higher expression of white adipocyte genes but no difference in Pgc1a and Ucp1 expression, and higher fatty acid uptake but not mitochondrial biogenesis. Moreover, cells treated with two chemicals identified as highly ranked PPARγ agonists, tonalide and quinoxyfen, induced white adipogenesis without the concomitant health-promoting characteristics of brite adipocytes in mouse and human preadipocytes. DISCUSSION A novel classification procedure accurately identified environmental chemicals as PPARγ ligands distinct from known PPARγ-activating therapeutics. CONCLUSION The computational and experimental framework has general applicability to the classification of as-yet uncharacterized chemicals. https://doi.org/10.1289/EHP6886.
Collapse
Affiliation(s)
- Stephanie Kim
- Boston University Superfund Research Program, Boston University, Massachusetts, USA
- Department of Environmental Health, Boston University School of Public Health, Massachusetts, USA
| | - Eric Reed
- Boston University Superfund Research Program, Boston University, Massachusetts, USA
- Section of Computational Biomedicine, Boston University School of Medicine, Massachusetts, USA
- Boston University Bioinformatics Program, Boston University, Massachusetts, USA
| | - Stefano Monti
- Boston University Superfund Research Program, Boston University, Massachusetts, USA
- Section of Computational Biomedicine, Boston University School of Medicine, Massachusetts, USA
- Boston University Bioinformatics Program, Boston University, Massachusetts, USA
| | - Jennifer J. Schlezinger
- Boston University Superfund Research Program, Boston University, Massachusetts, USA
- Department of Environmental Health, Boston University School of Public Health, Massachusetts, USA
| |
Collapse
|
63
|
Schmid A, Schäffler A, Karrasch T. CTRP-3 Regulates NOD1-mediated Inflammation and NOD1 Expression in Adipocytes and Adipose Tissue. Inflammation 2021; 44:2260-2269. [PMID: 34165676 PMCID: PMC8616866 DOI: 10.1007/s10753-021-01497-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 11/29/2022]
Abstract
The anti-inflammatory adipokine CTRP-3 might affect innate immune reactions such as NOD1. The impact of CTRP-3 on NOD1-mediated inflammation in adipocytes and monocytic cells as well as on NOD1 expression was investigated. Murine 3T3-L1 pre-adipocytes and adipocytes as well as human THP-1 monocyte-like cells were co-stimulated with the synthetic NOD1 agonist Tri-DAP and recombinant CTRP-3. Gonadal adipose tissue and primary adipocytes were obtained from a murine model carrying a knockout (KO) of CTRP-3 in adipocytes but not in stroma-vascular cells. Wildtype mice with lipopolysaccharide (LPS)-induced elevated NOD1 expression were treated with CTRP-3. Secreted inflammatory cytokines in cell supernatants were measured by ELISA and mRNA levels were quantified by RT-PCR. Pro-inflammatory chemokine and cytokine secretion (MCP-1, RANTES, TNFα) was induced by NOD1 activation in adipocytes and monocyte-like cells, and MCP-1 and RANTES release was effectively inhibited by pre-incubation of cells with CTRP-3. CTRP-3 also antagonized LPS-triggered induction of NOD1 gene expression in murine adipose tissue, whereas adipocyte CTRP-3 deficiency upregulated NOD1 expression in adipose tissue. CTRP-3 is an effective antagonist of peptidoglycan-induced, NOD1-mediated inflammation and of LPS-induced NOD1 expression. Since basal NOD1 expression is increased by adipocyte CTRP-3 deficiency, there have to be also inflammation-independent mechanisms of NOD1 expression regulation by CTRP-3.
Collapse
Affiliation(s)
- Andreas Schmid
- Department of Internal Medicine III, University Hospital of Giessen, Giessen, Germany.
| | - Andreas Schäffler
- Department of Internal Medicine III, University Hospital of Giessen, Giessen, Germany
| | - Thomas Karrasch
- Department of Internal Medicine III, University Hospital of Giessen, Giessen, Germany
| |
Collapse
|
64
|
Derici GE, Özdaş S, Canatar İ, Koç M. Antidiabetic activities of Bolanthus spergulifolius (Caryophyllaceae) extracts on insulin-resistant 3T3-L1 adipocytes. PLoS One 2021; 16:e0252707. [PMID: 34133443 PMCID: PMC8208533 DOI: 10.1371/journal.pone.0252707] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 05/20/2021] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus (DM) is a metabolic disorder with chronic hyperglycemia featured by metabolic outcomes owing to insufficient insulin secretion and/or insulin effect defect. It is critical to investigate new therapeutic approaches for T2DM and alternative, natural agents that target molecules in potential signal pathways. Medicinal plants are significant resources in the research of alternative new drug active ingredients. Bolanthus spergulifolius (B. spergulifolius) is one of the genera of the family Caryophyllaceae. In this study, it was explored the potential anti-diabetic effects in vitro of B. spergulifolius extracts on 3T3-L1 adipocytes. The total phenolic contents (TPC) of methanolic (MeOH), ethyl acettate (EA) and aqueous extracts of B. spergulifolius were evaluated via Folin-Ciocateau. B. spergulifolius extracts showing highly TPC (Aqueous< MeOH< EA) and their different concentrations were carried out on preadipocytes differentiated in to mature 3T3-L1 adipocytes to investigate their half-maximal (50%) inhibitory concentration (IC50) value by using Thiazolyl blue tetrazolium bromide (MTT) assay. The IC50 of MeOH, EA and Aqueous extracts were observed as 305.7 ± 5.583 μg/mL, 567.4 ± 3.008 μg/mL, and 418.3 ± 4.390 μg/mL and used for further experiments. A live/dead assay further confirmed the cytotoxic effects of MeOH, EA and Aqueous extracts (respectively, 69.75 ± 1.70%, 61.75 ± 1.70%, 70 ± 4.24%, and for all p< 0.05). Also, effects of extracts on lipid accumulation in mature 3T3-L1 adipocytes were evaluated by Oil-Red O staining assay. The extracts effectively decreased lipid-accumulation compared to untreated adipocytes (for all p< 0.05). Moreover, effect of extracts on apoptosis regulated by the Bax and Bcl-2 was investigated by quantitative reverse transcription polymerase chain reaction (qRT-PCR). The extracts significantly induced apoptosis by up-regulating pro-apoptotic Bax expression but down-regulated anti-apoptotic Bcl-2 gene expression compared to untreated adipocytes (for all p< 0.05). The Glut-4 expression linked with insulin resistance was determined by qRT-PCR, Western-blot analysis, and immunofluorescence staining. In parallel, the expression of Glut-4 in adipocytes treated with extracts was significantly higher compared to untreated adipocytes (for all p< 0.05). Extracts significantly suppressed cell migration after 30 h of wounding in a scratch-assay (for all p< 0.05). Cell morphology and diameter were further evaluated by phase-contrast microscopy, scanning electron microscopy, Immunofluorescence with F-Actin and Giemsa staining. The adipocytes treated with extracts partially lost spherical morphology and showed smaller cell-diameter compared to untreated adipocytes (for all p< 0.05). In conclusion, these results suggest that extracts of B. spergulifolius cause to an induce apoptosis, decrease lipid-accumulation, wound healing, up-regulating Glut-4 level and might contribute to reducing of insulin-resistance in DM.
Collapse
Affiliation(s)
- Gizem Ece Derici
- Faculty of Engineering Sciences, Department of Bioengineering, Adana Alpaslan Türkeş Science and Technology University, Adana, Turkey
| | - Sibel Özdaş
- Faculty of Engineering Sciences, Department of Bioengineering, Adana Alpaslan Türkeş Science and Technology University, Adana, Turkey
- * E-mail:
| | - İpek Canatar
- Faculty of Engineering Sciences, Department of Bioengineering, Adana Alpaslan Türkeş Science and Technology University, Adana, Turkey
| | - Murat Koç
- Graduate School of Public Health, Department of Traditional, Complementary and Integrative Medicine, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| |
Collapse
|
65
|
Pinto C, Ibáñez MR, Loyola G, León L, Salvatore Y, González C, Barraza V, Castañeda F, Aldunate R, Contreras-Porcia L, Fuenzalida K, Bronfman FC. Characterization of an Agarophyton chilense Oleoresin Containing PPARγ Natural Ligands with Insulin-Sensitizing Effects in a C57Bl/6J Mouse Model of Diet-Induced Obesity and Antioxidant Activity in Caenorhabditis elegans. Nutrients 2021; 13:1828. [PMID: 34071972 PMCID: PMC8227508 DOI: 10.3390/nu13061828] [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: 03/31/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 12/27/2022] Open
Abstract
The biomedical potential of the edible red seaweed Agarophyton chilense (formerly Gracilaria chilensis) has not been explored. Red seaweeds are enriched in polyunsaturated fatty acids and eicosanoids, which are known natural ligands of the PPARγ nuclear receptor. PPARγ is the molecular target of thiazolidinediones (TZDs), drugs used as insulin sensitizers to treat type 2 diabetes mellitus. Medical use of TZDs is limited due to undesired side effects, a problem that has triggered the search for selective PPARγ modulators (SPPARMs) without the TZD side effects. We produced Agarophyton chilense oleoresin (Gracilex®), which induces PPARγ activation without inducing adipocyte differentiation, similar to SPPARMs. In a diet-induced obesity model of male mice, we showed that treatment with Gracilex® improves insulin sensitivity by normalizing altered glucose and insulin parameters. Gracilex® is enriched in palmitic acid, arachidonic acid, oleic acid, and lipophilic antioxidants such as tocopherols and β-carotene. Accordingly, Gracilex® possesses antioxidant activity in vitro and increased antioxidant capacity in vivo in Caenorhabditis elegans. These findings support the idea that Gracilex® represents a good source of natural PPARγ ligands and antioxidants with the potential to mitigate metabolic disorders. Thus, its nutraceutical value in humans warrants further investigation.
Collapse
Affiliation(s)
- Claudio Pinto
- Postgraduate Department, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia 5110566, Chile;
- Center for Aging and Regeneration (CARE), Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
| | - María Raquel Ibáñez
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (M.R.I.); (G.L.); (L.L.); (Y.S.); (C.G.); (V.B.)
- Institute of Biomedical Sciences (ICB), Faculty of Medicine, Universidad Andres Bello, Santiago 8320000, Chile
| | - Gloria Loyola
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (M.R.I.); (G.L.); (L.L.); (Y.S.); (C.G.); (V.B.)
- Institute of Biomedical Sciences (ICB), Faculty of Medicine, Universidad Andres Bello, Santiago 8320000, Chile
| | - Luisa León
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (M.R.I.); (G.L.); (L.L.); (Y.S.); (C.G.); (V.B.)
| | - Yasmin Salvatore
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (M.R.I.); (G.L.); (L.L.); (Y.S.); (C.G.); (V.B.)
| | - Carla González
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (M.R.I.); (G.L.); (L.L.); (Y.S.); (C.G.); (V.B.)
| | - Víctor Barraza
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (M.R.I.); (G.L.); (L.L.); (Y.S.); (C.G.); (V.B.)
| | - Francisco Castañeda
- Department of Ecology and Biodiversity, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8320000, Chile; (F.C.); (L.C.-P.)
- Quintay Marine Research Center (CIMARQ), Faculty of Life Sciences, Universidad Andres Bello, Valparaiso, Quintay 2480000, Chile
- Center of Applied Ecology and Sustainability (CAPES), Santiago 8331150, Chile
- Instituto Milenio en Socio-Ecología Costera (SECOS), Santiago 8370251, Chile
| | - Rebeca Aldunate
- Faculty of Sciences, School of Biotechnology, Universidad Santo Tomas, Santiago 8320000, Chile;
| | - Loretto Contreras-Porcia
- Department of Ecology and Biodiversity, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8320000, Chile; (F.C.); (L.C.-P.)
- Quintay Marine Research Center (CIMARQ), Faculty of Life Sciences, Universidad Andres Bello, Valparaiso, Quintay 2480000, Chile
- Center of Applied Ecology and Sustainability (CAPES), Santiago 8331150, Chile
- Instituto Milenio en Socio-Ecología Costera (SECOS), Santiago 8370251, Chile
| | - Karen Fuenzalida
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (M.R.I.); (G.L.); (L.L.); (Y.S.); (C.G.); (V.B.)
- Institute of Biomedical Sciences (ICB), Faculty of Medicine, Universidad Andres Bello, Santiago 8320000, Chile
| | - Francisca C. Bronfman
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (M.R.I.); (G.L.); (L.L.); (Y.S.); (C.G.); (V.B.)
- Institute of Biomedical Sciences (ICB), Faculty of Medicine, Universidad Andres Bello, Santiago 8320000, Chile
| |
Collapse
|
66
|
Role of the Endocannabinoid System in the Adipose Tissue with Focus on Energy Metabolism. Cells 2021; 10:cells10061279. [PMID: 34064024 PMCID: PMC8224009 DOI: 10.3390/cells10061279] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/12/2021] [Accepted: 05/15/2021] [Indexed: 12/15/2022] Open
Abstract
The endocannabinoid system is involved in a wide range of processes including the control of energy acquisition and expenditure. Endocannabinoids and their receptors are present in the central nervous system but also in peripheral tissues, notably the adipose tissues. The endocannabinoid system interacts with two main hormones regulating appetite, namely leptin and ghrelin. The inhibitory effect of the cannabinoid receptor 1 (CB1) antagonist rimonabant on fat mass suggested that the endocannabinoid system can also have a peripheral action in addition to its effect on appetite reduction. Thus, several investigations have focused on the peripheral role of the endocannabinoid system in the regulation of metabolism. The white adipose tissue stores energy as triglycerides while the brown adipose tissue helps to dissipate energy as heat. The endocannabinoid system regulates several functions of the adipose tissues to favor energy accumulation. In this review we will describe the presence of the endocannabinoid system in the adipose tissue. We will survey the role of the endocannabinoid system in the regulation of white and brown adipose tissue metabolism and how the eCB system participates in obesity and metabolic diseases.
Collapse
|
67
|
Lee KW, An YJ, Lee J, Lee JH, Yim HS. α-Poly-L-lysine functions as an adipogenic inducer in 3T3-L1 preadipocytes. Amino Acids 2021; 53:587-596. [PMID: 33743070 PMCID: PMC8107076 DOI: 10.1007/s00726-020-02932-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023]
Abstract
α-Poly-l-lysine (PLL) has been used for various purposes such as cell attachment, immunization, and molecular delivery, and is known to be cytotoxic to several cell lines. Here, we studied the effect of PLL on the adipogenesis of 3T3-L1 cells and investigated the underlying mechanism. Differentiation media containing PLL with a molecular weight (MW) greater than 4 kDa enhanced lipid droplet formation and increased adipogenic marker levels, indicating an increase in adipocyte differentiation. PLL with a molecular weight between 30 and 70 kDa was more effective than PLL of other sizes in 3T3-L1 cell differentiation. Moreover, PLL induced 3T3-L1 adipogenesis in insulin-free adipocyte differentiation medium. Incubation with insulin and PLL exhibited greater adipogenesis than insulin treatment only even at a high concentration. PLL stimulated insulin signaling and augmented the signaling pathway when it was added with insulin. While PLL did not activate the glucocorticoid receptor, which is phosphorylated by dexamethasone (DEX), it showed a positive effect on the cAMP signal pathway when preadipocytes were treated with PLL and 3-isobutyl-1-methylxanthine (IBMX). Consistent with these results, incubation with PLL and DEX without IBMX induced adipocyte differentiation. We also observed that the mitotic clonal expansion phase was the critical stage in adipogenesis for inducing the effects of PLL. These results suggest that PLL functions as an adipogenic inducer in 3T3-L1 preadipocytes and PLL has a direct effect on insulin signaling, one of the main regulatory pathways.
Collapse
Affiliation(s)
- Kyeong Won Lee
- Marine Biotechnology Research Center, Korea Institute of Ocean Science and Technology, 385 Haeyang-ro, Busan, 49111, Republic of Korea
| | - Young Jun An
- Marine Biotechnology Research Center, Korea Institute of Ocean Science and Technology, 385 Haeyang-ro, Busan, 49111, Republic of Korea
| | - Janet Lee
- Marine Biotechnology Research Center, Korea Institute of Ocean Science and Technology, 385 Haeyang-ro, Busan, 49111, Republic of Korea
| | - Jung-Hyun Lee
- Marine Biotechnology Research Center, Korea Institute of Ocean Science and Technology, 385 Haeyang-ro, Busan, 49111, Republic of Korea. .,Department of Ocean Science, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea.
| | - Hyung-Soon Yim
- Marine Biotechnology Research Center, Korea Institute of Ocean Science and Technology, 385 Haeyang-ro, Busan, 49111, Republic of Korea.
| |
Collapse
|
68
|
Hochberg A, Patz M, Karrasch T, Schäffler A, Schmid A. Serum Levels and Adipose Tissue Gene Expression of Cathelicidin Antimicrobial Peptide (CAMP) in Obesity and During Weight Loss. Horm Metab Res 2021; 53:169-177. [PMID: 33434932 PMCID: PMC7924992 DOI: 10.1055/a-1323-3050] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
CAMP (Cathelicidin antimicrobial peptide) is synthesized and secreted by adipocytes and involved in adipose tissue (AT) innate immune response and host defense of subcutaneous AT against Gram positive bacteria. Data on the regulation of CAMP in obesity and during weight loss are scarce and reference values do not exist. Serum CAMP levels (ELISA) and AT gene expression levels (quantitative real time PCR) were investigated in two large and longitudinal (12 months) cohorts of severely obese patients undergoing either a low calorie diet (LCD; n=79) or bariatric surgery (BS; n=156). The impact of metabolic factors on CAMP expression in vitro was investigated in differentiated 3T3-L1 adipocytes. CAMP serum levels significantly increased after BS but not during LCD. Females had lower CAMP serum levels and lower gene expression levels in subcutaneous AT. CAMP was positively correlated to unfavorable metabolic factors/adipokines and negatively to favorable factors/adipokines. CAMP gene expression was higher in subcutaneous than in visceral AT but serum CAMP levels were not correlated to levels of AT gene expression. While certain bile acids upregulated CAMP expression in vitro, high glucose/insulin as well as GLP-1 had an inhibitory effect. There exist gender-specific and AT compartment-specific effects on the regulation of CAMP gene expression. Weight loss induced by BS (but not by LCD) upregulated CAMP serum levels suggesting the involvement of weight loss-independent mechanisms in CAMP regulation such as bile acids, incretins and metabolic factors. CAMP might represent an adipokine at the interface between metabolism and innate immune response.
Collapse
Affiliation(s)
- Alexandra Hochberg
- Department of Internal Medicine III, University of Giessen, Giessen,
Germany
| | - Marissa Patz
- Department of Internal Medicine III, University of Giessen, Giessen,
Germany
| | - Thomas Karrasch
- Department of Internal Medicine III, University of Giessen, Giessen,
Germany
| | - Andreas Schäffler
- Department of Internal Medicine III, University of Giessen, Giessen,
Germany
| | - Andreas Schmid
- Department of Internal Medicine III, University of Giessen, Giessen,
Germany
- Correspondence Andreas Schmid PhD. Department of Internal Medicine IIIGiessen University HospitalKlinikstraße 3335392 GießenGermany+49 641 99 30641+49 641 99 30649
| |
Collapse
|
69
|
Izuchi R, Katsuki T. Pomolic acid in persimmon peel suppresses the increase in glycerol-3 phosphate dehydrogenase activity in 3T3-L1 adipocytes. Biosci Biotechnol Biochem 2021; 85:691-696. [PMID: 33624785 DOI: 10.1093/bbb/zbaa079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/06/2020] [Indexed: 11/13/2022]
Abstract
Persimmon peels, though usually discarded, are useful sources of nutraceuticals. In this study, persimmon peel-derived pomolic acid was found to suppress the increase in the activity of glycerol-3 phosphate dehydrogenase, a neutral fat synthesis-related enzyme, in 3T3-L1 adipocytes, whereas oleanolic and ursolic acids did not exert this effect. Therefore, persimmon peel may be an effective functional food material.
Collapse
Affiliation(s)
- Ryoichi Izuchi
- Toyo Institute of Food Technology, 23-2 4-chome, Minami-Hanayashiki, Kawanishi-shi, Hyogo, Japan
| | - Takahiro Katsuki
- Toyo Institute of Food Technology, 23-2 4-chome, Minami-Hanayashiki, Kawanishi-shi, Hyogo, Japan
| |
Collapse
|
70
|
Multilevel Regulation of Protein Kinase CδI Alternative Splicing by Lithium Chloride. Mol Cell Biol 2021; 41:e0033820. [PMID: 33288642 PMCID: PMC8088272 DOI: 10.1128/mcb.00338-20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Lithium chloride (LiCl) is commonly used in treatment of mood disorders; however, its usage leads to weight gain, which promotes metabolic disorders. Protein kinase C delta (PKCδ), a serine/threonine kinase, is alternatively spliced to PKCδI and PKCδII in 3T3-L1 cells. We previously demonstrated that PKCδI is the predominantly expressed isoform in 3T3-L1 preadipocytes. Here, we demonstrate that LiCl treatment decreases PKCδI levels, increases formation of lipid droplets, and increases oxidative stress. Hence, we investigated the molecular mechanisms underlying the regulation of PKCδI alternative splicing by LiCl. We previously demonstrated that the splice factor SFRS10 is essential for PKCδI splicing. Our results demonstrate that glycogen synthase kinase 3 beta (GSK3β) phosphorylates SFRS10, and SFRS10 is in a complex with long noncoding RNA NEAT1 to promote PKCδI splicing. Using PKCδ splicing minigene and RNA immunoprecipitation assays, our results demonstrate that upon LiCl treatment, NEAT1 levels are reduced, GSK3β activity is inhibited, and SFRS10 phosphorylation is decreased, which leads to decreased expression of PKCδI. Integration of the GSK3β signaling pathway with the ribonucleoprotein complex of long noncoding RNA (lncRNA) NEAT1 and SFRS10 enables fine-tuning of PKCδI expression during adipogenesis. Knowledge of the molecular pathways impacted by LiCl provides an understanding of the ascent of obesity as a comorbidity in disease management.
Collapse
|
71
|
Schmid A, Roderfeld M, Gehl J, Roeb E, Nist A, Chung HR, Stiewe T, Karrasch T, Schäffler A. C1q/TNF-Related Protein 3 (CTRP-3) Deficiency of Adipocytes Affects White Adipose Tissue Mass but Not Systemic CTRP-3 Concentrations. Int J Mol Sci 2021; 22:ijms22041670. [PMID: 33562308 PMCID: PMC7915696 DOI: 10.3390/ijms22041670] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 12/20/2022] Open
Abstract
CTRP-3 (C1q/TNF-related protein-3) is an adipokine with endocrine and immunological function. The impact of adipocyte CTRP-3 production on systemic CTRP-3 concentrations and on adipocyte biology is unknown. A murine model of adipocyte CTRP-3 knockout (KO) was established (via the Cre/loxP system). Serum adipokine levels were quantified by ELISA and adipose tissue (AT) gene expression by real-time PCR. Preadipocytes were isolated from AT and differentiated into adipocytes. Comparative transcriptome analysis was applied in adipocytes and liver tissue. Body weight and AT mass were reduced in CTRP-3 KO mice together with decreased serum leptin. In primary cells from visceral AT of KO mice, expression of adiponectin, progranulin, and resistin was induced, while peroxisome proliferator activated receptor γ (PPARγ) was decreased. M1/M2 macrophage polarization markers were shifted to a more anti-inflammatory phenotype. CTRP-3 expression in AT did not contribute to serum concentrations. AT and liver morphology remained unaffected by CTRP-3 KO. Myelin transcription factor 1-like (Myt1l) was identified as a highly upregulated gene. In conclusion, adipocyte CTRP-3 has a role in adipogenesis and AT weight gain whereas adipocyte differentiation is not impaired by CTRP-3 deficiency. Since no effects on circulating CTRP-3 levels were observed, the impact of adipocyte CTRP-3 KO is limited to adipose tissue. Modified AT gene expression indicates a rather anti-inflammatory phenotype.
Collapse
Affiliation(s)
- Andreas Schmid
- Department of Internal Medicine III, University of Giessen, 35390 Giessen, Germany; (J.G.); (T.K.); (A.S.)
- Correspondence: ; Tel.: +49-641-99-30641
| | - Martin Roderfeld
- Department of Gastroenterology, University of Giessen, 35390 Giessen, Germany; (M.R.); (E.R.)
| | - Jonas Gehl
- Department of Internal Medicine III, University of Giessen, 35390 Giessen, Germany; (J.G.); (T.K.); (A.S.)
| | - Elke Roeb
- Department of Gastroenterology, University of Giessen, 35390 Giessen, Germany; (M.R.); (E.R.)
| | - Andrea Nist
- Institute of Molecular Oncology and Genomics Core Facility, University of Marburg, 35037 Marburg, Germany; (A.N.); (T.S.)
| | - Ho-Ryun Chung
- Institute of Medical Bioinformatics and Biostatistics, University of Marburg, 35037 Marburg, Germany;
| | - Thorsten Stiewe
- Institute of Molecular Oncology and Genomics Core Facility, University of Marburg, 35037 Marburg, Germany; (A.N.); (T.S.)
| | - Thomas Karrasch
- Department of Internal Medicine III, University of Giessen, 35390 Giessen, Germany; (J.G.); (T.K.); (A.S.)
| | - Andreas Schäffler
- Department of Internal Medicine III, University of Giessen, 35390 Giessen, Germany; (J.G.); (T.K.); (A.S.)
| |
Collapse
|
72
|
Xu Q, Fan Y, Loor JJ, Liang Y, Sun X, Jia H, Zhao C, Xu C. All-trans retinoic acid controls differentiation, proliferation, and lipolysis in isolated subcutaneous adipocytes from peripartal Holstein cows. J Dairy Sci 2021; 104:4999-5008. [PMID: 33551168 DOI: 10.3168/jds.2020-19408] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/05/2020] [Indexed: 11/19/2022]
Abstract
Preadipocyte proliferation and differentiation are critical for normal adipose tissue development, including achieving a mature phenotype, characterized by its ability to accumulate triacylglycerol and release fatty acids. In nonruminants, it is well known that all-trans retinoic acid (ATRA), the most-active form of vitamin A, helps regulate proliferation, differentiation, and apoptosis in several types of cells including adipocytes. The purpose of this study was to evaluate the effect of ATRA on proliferation, apoptosis, differentiation, and lipolysis of primary bovine adipocytes isolated from subcutaneous adipose tissue of 5 healthy Holstein cows at 17 (±4 standard deviations) d postpartum. Cells were stimulated with increasing concentrations of ATRA (0.2, 2, and 20 nM) at the preconfluent (2 d) and postconfluent (8 d) preadipocyte stage or at the mature adipocyte stage (2 d). All concentrations of ATRA inhibited preconfluent preadipocyte proliferation with decreased proportion of S-phase cells and reduced protein abundance of cyclins (CCND1, CCND2, CCND3, CCNE1) and cyclin-dependent kinases (CDK2, CDK4, CDK6). Compared with vehicle, ATRA treatment induced apoptosis in preconfluent preadipocytes. Additionally, ATRA (0.2, 2, and 20 nM) supplementation also inhibited differentiation of postconfluent preadipocytes through downregulation of protein abundance of PPARγ and C/EBPα. After induction of differentiation, basal lipolysis in mature adipocytes increased upon treatment with all concentrations of ATRA. However, data on phosphorylated hormone-sensitive lipase or PLIN1 indicated that ATRA had no effect on epinephrine-stimulated lipolysis in mature adipocytes. Overall, these results demonstrate that ATRA might inhibit lipid accumulation by suppressing preadipocyte proliferation and differentiation, subsequently leading to apoptosis in postconfluent preadipocytes and promoting basal lipolysis in mature adipocytes. Overall, these in vitro responses provide some insights into the potential for nutritional management to modulate adipose tissue lipolysis, particularly in overconditioned cows during the dry period, which are more susceptible to suffer metabolic disorders due to excessive fat mobilization postpartum.
Collapse
Affiliation(s)
- Qiushi Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yunhui Fan
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Juan J Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Yusheng Liang
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Xudong Sun
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Hongdou Jia
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Chenxu Zhao
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Chuang Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China.
| |
Collapse
|
73
|
Höpfinger A, Karrasch T, Schäffler A, Schmid A. Regulation of CAMP (cathelicidin antimicrobial peptide) expression in adipocytes by TLR 2 and 4. Innate Immun 2021; 27:184-191. [PMID: 33509002 PMCID: PMC7882808 DOI: 10.1177/1753425920988167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Recent data argue for a pro-inflammatory role of CAMP (cathelicidin antimicrobial peptide) in adipocytes and adipose tissue (AT) and for regulatory circuits involving TLRs. In order to investigate regulatory effects of TLR2 and TLR4, 3T3-L1 adipocytes were stimulated with TLR2 agonistic lipopeptide MALP-2 and with TLR4 agonist LPS in presence or absence of signal transduction inhibitors. CAMP gene expression was analysed by quantitative real-time PCR in adipocytes and in murine AT compartments and cellular subfractions. CAMP expression was higher in gonadal than in subcutaneous AT and there was a gender-specific effect with higher levels in males. Adipocytes had higher CAMP expression than the stroma-vascular cell (SVC) fraction. MALP-2 up-regulated CAMP expression significantly, mediated by STAT3 and PI3K and potentially (non-significant trend) by NF-κB and MAPK, but not by raf-activated MEK-1/-2. Moreover, LPS proved to act as a potent inducer of CAMP via NF-κB, PI3K and STAT3, whereas specific inhibition of MAPK and MEK-1/-2 had no effect. In conclusion, activation of TLR2 and TLR4 by classical ligands up-regulates adipocyte CAMP expression involving classical signal transduction elements. These might represent future drug targets for pharmacological modulation of CAMP expression in adipocytes, especially in the context of metabolic and infectious diseases.
Collapse
Affiliation(s)
| | - Thomas Karrasch
- Department of Internal Medicine III, University of Giessen, Germany
| | | | - Andreas Schmid
- Department of Internal Medicine III, University of Giessen, Germany
| |
Collapse
|
74
|
Dufau J, Shen JX, Couchet M, De Castro Barbosa T, Mejhert N, Massier L, Griseti E, Mouisel E, Amri EZ, Lauschke VM, Rydén M, Langin D. In vitro and ex vivo models of adipocytes. Am J Physiol Cell Physiol 2021; 320:C822-C841. [PMID: 33439778 DOI: 10.1152/ajpcell.00519.2020] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adipocytes are specialized cells with pleiotropic roles in physiology and pathology. Several types of fat cells with distinct metabolic properties coexist in various anatomically defined fat depots in mammals. White, beige, and brown adipocytes differ in their handling of lipids and thermogenic capacity, promoting differences in size and morphology. Moreover, adipocytes release lipids and proteins with paracrine and endocrine functions. The intrinsic properties of adipocytes pose specific challenges in culture. Mature adipocytes float in suspension culture due to high triacylglycerol content and are fragile. Moreover, a fully differentiated state, notably acquirement of the unilocular lipid droplet of white adipocyte, has so far not been reached in two-dimensional culture. Cultures of mouse and human-differentiated preadipocyte cell lines and primary cells have been established to mimic white, beige, and brown adipocytes. Here, we survey various models of differentiated preadipocyte cells and primary mature adipocyte survival describing main characteristics, culture conditions, advantages, and limitations. An important development is the advent of three-dimensional culture, notably of adipose spheroids that recapitulate in vivo adipocyte function and morphology in fat depots. Challenges for the future include isolation and culture of adipose-derived stem cells from different anatomic location in animal models and humans differing in sex, age, fat mass, and pathophysiological conditions. Further understanding of fat cell physiology and dysfunction will be achieved through genetic manipulation, notably CRISPR-mediated gene editing. Capturing adipocyte heterogeneity at the single-cell level within a single fat depot will be key to understanding diversities in cardiometabolic parameters among lean and obese individuals.
Collapse
Affiliation(s)
- Jérémy Dufau
- Inserm, Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR1297, Toulouse, France.,Faculté de Médecine, I2MC, UMR1297, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Joanne X Shen
- Karolinska Institutet, Department of Physiology and Pharmacology, Stockholm, Sweden
| | - Morgane Couchet
- Karolinska Institutet, Department of Medicine (H7), Stockholm, Sweden
| | | | - Niklas Mejhert
- Karolinska Institutet, Department of Medicine (H7), Stockholm, Sweden
| | - Lucas Massier
- Karolinska Institutet, Department of Medicine (H7), Stockholm, Sweden
| | - Elena Griseti
- Inserm, Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR1297, Toulouse, France.,Faculté de Médecine, I2MC, UMR1297, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Etienne Mouisel
- Inserm, Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR1297, Toulouse, France.,Faculté de Médecine, I2MC, UMR1297, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | | | - Volker M Lauschke
- Karolinska Institutet, Department of Physiology and Pharmacology, Stockholm, Sweden
| | - Mikael Rydén
- Karolinska Institutet, Department of Medicine (H7), Stockholm, Sweden
| | - Dominique Langin
- Inserm, Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR1297, Toulouse, France.,Faculté de Médecine, I2MC, UMR1297, Université de Toulouse, Université Paul Sabatier, Toulouse, France.,Toulouse University Hospitals, Department of Biochemistry, Toulouse, France
| |
Collapse
|
75
|
Pant R, Firmal P, Shah VK, Alam A, Chattopadhyay S. Epigenetic Regulation of Adipogenesis in Development of Metabolic Syndrome. Front Cell Dev Biol 2021; 8:619888. [PMID: 33511131 PMCID: PMC7835429 DOI: 10.3389/fcell.2020.619888] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Obesity is one of the biggest public health concerns identified by an increase in adipose tissue mass as a result of adipocyte hypertrophy and hyperplasia. Pertaining to the importance of adipose tissue in various biological processes, any alteration in its function results in impaired metabolic health. In this review, we discuss how adipose tissue maintains the metabolic health through secretion of various adipokines and inflammatory mediators and how its dysfunction leads to the development of severe metabolic disorders and influences cancer progression. Impairment in the adipocyte function occurs due to individuals' genetics and/or environmental factor(s) that largely affect the epigenetic profile leading to altered gene expression and onset of obesity in adults. Moreover, several crucial aspects of adipose biology, including the regulation of different transcription factors, are controlled by epigenetic events. Therefore, understanding the intricacies of adipogenesis is crucial for recognizing its relevance in underlying disease conditions and identifying the therapeutic interventions for obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Richa Pant
- National Centre for Cell Science, SP Pune University Campus, Pune, India
| | - Priyanka Firmal
- National Centre for Cell Science, SP Pune University Campus, Pune, India
| | - Vibhuti Kumar Shah
- National Centre for Cell Science, SP Pune University Campus, Pune, India
| | - Aftab Alam
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Samit Chattopadhyay
- National Centre for Cell Science, SP Pune University Campus, Pune, India.,Department of Biological Sciences, BITS Pilani, Goa, India
| |
Collapse
|
76
|
Transcriptional analyses of the effects of Catharanthus roseus L. medicinal plant extracts on some markers related to obesity and inflammation in 3T3-L1 mouse cell lines. Biologia (Bratisl) 2021. [DOI: 10.2478/s11756-020-00567-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
77
|
Lee JS, Chae MK, Kikkawa DO, Lee EJ, Yoon JS. Glycogen Synthase Kinase-3β Mediates Proinflammatory Cytokine Secretion and Adipogenesis in Orbital Fibroblasts from Patients with Graves' Orbitopathy. Invest Ophthalmol Vis Sci 2021; 61:51. [PMID: 32735324 PMCID: PMC7426624 DOI: 10.1167/iovs.61.8.51] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Purpose We sought to determine the role of glycogen synthase kinase-3β (GSK-3β) in the pathogenesis of Graves’ orbitopathy(GO). Methods Expression of the GSK-3β gene in whole orbital tissue explants was compared between GO and non-GO donors using quantitative real-time PCR (RT-PCR). The expression of proinflammatory molecules in the presence of the GSK-3β inhibitor CHIR 99021 was analyzed using RT-PCR, western blot, and ELISA. Adipogenic differentiation was identified using Oil Red O staining, and the levels of peroxisome proliferator activator gamma (PPARγ) and CCAAT-enhancer-binding proteins (C/EBPs) α and β were determined by western blot. Results The expression of GSK-3β was significantly higher in GO tissues than in control tissues. The addition of CHIR 99021 led to a decrease in the active form of the kinase in which the Y216 residue is phosphorylated. When GO and non-GO fibroblasts were stimulated with IL-1β or TNF-α, IL-6, IL-8, intercellular adhesion molecule-1 (ICAM-1), cyclooxygenase-1 (COX-1), and monocyte chemoattractant protein 1 (MCP-1) showed increased production, which was blunted when CHIR 99021 was added. The activation of Akt, PI3K, nuclear factor (NF)-κB, Erk, Jnk, and p38 kinase by IL-1β and TNF-α was diminished with CHIR 99021 in GO cells. A decrease in lipid droplets and expression of PPARγ and c/EBPα and -β was noted in fibroblasts treated with CHIR 99021 during adipocyte differentiation. The inhibition of Wnt and β-catenin in adipogenesis was reversed by CHIR 99021. Conclusions GSK-3β plays a significant role in GO pathogenesis. The inhibition of the kinase attenuated the proinflammatory cytokines production and fibroblast differentiation into adipocytes. GSK-3β may be a potential target for anti-inflammatory and anti-adipogenic treatment of GO.
Collapse
|
78
|
Ahmed M, Min DS, Kim DR. Curated gene expression dataset of differentiating 3T3-L1 adipocytes under pharmacological and genetic perturbations. Adipocyte 2020; 9:600-608. [PMID: 33016192 PMCID: PMC7553567 DOI: 10.1080/21623945.2020.1829852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The 3T3-L1 cell line is used as an adipocyte differentiation model for the analysis of genes specifically expressed during the differentiation course. This cell model has several applications in obesity and insulin resistance research. We built a data resource to model gene expression of differentiating and mature adipocytes in response to several drugs and gene manipulations. We surveyed the literature survey for microarray datasets of differentiating 3T3-L1 cell line sampled at one or more time points under genetic or pharmacological perturbations. Data and metadata were obtained from the gene expression omnibus. The metadata were manually curated using unified language across the studies. Probe intensities were mapped and collapsed to genes using a reproducible pipeline. Samples were classified into none, genetically or pharmacologically modified. In addition to the clean datasets, two aggregated sets were further homogenized for illustration purposes. The curated datasets are available as an R/Bioconductor experimental data package curatedAdipoArray. The package documents the source code of the data collection, curation and processing. Finally, we used a subset of the data to effectively remove batch effects and reproduce biological observations. Database URL https://bioconductor.org/packages/curatedAdipoArray
Collapse
Affiliation(s)
- Mahmoud Ahmed
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
| | - Do Sik Min
- College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Deok Ryong Kim
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
| |
Collapse
|
79
|
Merrett JE, Bo T, Psaltis PJ, Proud CG. Identification of DNA response elements regulating expression of CCAAT/enhancer-binding protein (C/EBP) β and δ and MAP kinase-interacting kinases during early adipogenesis. Adipocyte 2020; 9:427-442. [PMID: 32787498 PMCID: PMC7469549 DOI: 10.1080/21623945.2020.1796361] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 07/07/2020] [Accepted: 07/09/2020] [Indexed: 12/15/2022] Open
Abstract
Given the high and increasing prevalence of obesity and associated disorders, such as type-2 diabetes, it is important to understand the mechanisms that regulate lipid storage and the differentiation of fat cells, a process termed adipogenesis. Using the well-established mouse 3T3-L1 in vitro model of adipogenesis, we refine how the induction of two key adipogenic transcription factors, CCAAT/enhancer-binding proteins (C/EBPs) β and δ are regulated during early adipogenesis. We identify, in the gene promoters of Cebpb and Cebpd, the DNA response elements responsible for binding transcription factors that are activated by cAMP or glucocorticoids. We also show that mitogen-activated protein kinase (MAPK)-interacting kinase 2 (MNK2; Mknk2), which plays a distinct role in diet-induced obesity, is induced during early adipogenesis and identify the functional DNA response elements responsible for regulating its expression. Mknk2 expression is maintained in differentiated 3T3-L1 adipocytes and is expressed at high levels across a range of mouse adipose tissue depots. Together, these new insights help to clarify the transcriptional programme of early adipogenesis and identify Mknk2 as one of potentially many genes up-regulated during adipogenesis.
Collapse
Affiliation(s)
- James E. Merrett
- Lifelong Health, South Australian Health and Medical Research Institute, Adelaide, Australia
- Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - Tao Bo
- Lifelong Health, South Australian Health and Medical Research Institute, Adelaide, Australia
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, China
| | - Peter J. Psaltis
- Lifelong Health, South Australian Health and Medical Research Institute, Adelaide, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | - Christopher G. Proud
- Lifelong Health, South Australian Health and Medical Research Institute, Adelaide, Australia
- Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| |
Collapse
|
80
|
Zysman M, Baptista BR, Essari LA, Taghizadeh S, Thibault de Ménonville C, Giffard C, Issa A, Franco-Montoya ML, Breau M, Souktani R, Aissat A, Caeymaex L, Lizé M, Van Nhieu JT, Jung C, Rottier R, Cruzeiro MD, Adnot S, Epaud R, Chabot F, Lanone S, Boczkowski J, Boyer L. Targeting p16 INK4a Promotes Lipofibroblasts and Alveolar Regeneration after Early-Life Injury. Am J Respir Crit Care Med 2020; 202:1088-1104. [PMID: 32628504 DOI: 10.1164/rccm.201908-1573oc] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Rationale: Promoting endogenous pulmonary regeneration is crucial after damage to restore normal lungs and prevent the onset of chronic adult lung diseases.Objectives: To investigate whether the cell-cycle inhibitor p16INK4a limits lung regeneration after newborn bronchopulmonary dysplasia (BPD), a condition characterized by the arrest of alveolar development, leading to adult sequelae.Methods: We exposed p16INK4a-/- and p16INK4a ATTAC (apoptosis through targeted activation of caspase 8) transgenic mice to postnatal hyperoxia, followed by pneumonectomy of the p16INK4a-/- mice. We measured p16INK4a in blood mononuclear cells of preterm newborns, 7- to 15-year-old survivors of BPD, and the lungs of patients with BPD.Measurements and Main Results: p16INK4a concentrations increased in lung fibroblasts after hyperoxia-induced BPD in mice and persisted into adulthood. p16INK4a deficiency did not protect against hyperoxic lesions in newborn pups but promoted restoration of the lung architecture by adulthood. Curative clearance of p16INK4a-positive cells once hyperoxic lung lesions were established restored normal lungs by adulthood. p16INK4a deficiency increased neutral lipid synthesis and promoted lipofibroblast and alveolar type 2 (AT2) cell development within the stem-cell niche. Besides, lipofibroblasts support self-renewal of AT2 cells into alveolospheres. Induction with a PPARγ (peroxisome proliferator-activated receptor γ) agonist after hyperoxia also increased lipofibroblast and AT2 cell numbers and restored alveolar architecture in hyperoxia-exposed mice. After pneumonectomy, p16INK4a deficiency again led to an increase in lipofibroblast and AT2 cell numbers in the contralateral lung. Finally, we observed p16INK4a mRNA overexpression in the blood and lungs of preterm newborns, which persisted in the blood of older survivors of BPD.Conclusions: These data demonstrate the potential of targeting p16INK4a and promoting lipofibroblast development to stimulate alveolar regeneration from childhood to adulthood.
Collapse
Affiliation(s)
- Maéva Zysman
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
| | - Bruno Ribeiro Baptista
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France.,Service de Pneumologie, Centre Hospitalier Universitaire, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Laure-Aléa Essari
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France.,Service de Pneumologie, Centre Hospitalier Universitaire, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Sara Taghizadeh
- Department of Pulmonary and Critical Care Medicine, Key Laboratory of Interventional Pulmonology of Zheijiang Province, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | | | | | - Amelle Issa
- Centre de Recherche Clinique, Centre de Ressource Biologique, Centre Hospitalier Intercommunal, Creteil, France
| | | | | | | | - Abdel Aissat
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
| | - Laurence Caeymaex
- Soins Intensifs Néonataux, Centre Hospitalier Intercommunal, Creteil, France
| | - Muriel Lizé
- Molecular and Experimental Pneumology Group, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Goettingen, Germany
| | - Jeanne Tran Van Nhieu
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France.,Service de Pathologie, Hôpital Henri Mondor, AP-HP, Hôpital Henri Mondor, Creteil, France
| | - Camille Jung
- Centre de Recherche Clinique, Centre de Ressource Biologique, Centre Hospitalier Intercommunal, Creteil, France
| | - Robert Rottier
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands.,Department of Cell Biology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Marcio Do Cruzeiro
- INSERM U1016, Institut Cochin, Paris, France.,UMR 8104, Centre National de la Recherche Scientifique, Paris, France.,Université Paris Descartes, Sorbonne, Paris, France
| | - Serge Adnot
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France.,Service de Physiologie, Hôpital Henri Mondor, AP-HP, Creteil, France; and
| | - Ralph Epaud
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France.,Service de Pédiatrie, Centre des Maladies Respiratoire Rares, Centre Hospitalier Intercommunal, Creteil, France
| | - François Chabot
- Service de Pneumologie, Centre Hospitalier Universitaire, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Sophie Lanone
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
| | | | - Laurent Boyer
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France.,Service de Physiologie, Hôpital Henri Mondor, AP-HP, Creteil, France; and
| |
Collapse
|
81
|
Cui H, Chang Y, Jiang X, Li M. Triphenyl phosphate exposure induces kidney structural damage and gut microbiota disorders in mice under different diets. ENVIRONMENT INTERNATIONAL 2020; 144:106054. [PMID: 32818822 DOI: 10.1016/j.envint.2020.106054] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 08/08/2020] [Accepted: 08/10/2020] [Indexed: 06/11/2023]
Abstract
Exposure of humans to organophosphate flame retardants (OPFRs) and the consequent health risk have increased owing to the latter's widespread application. Although triphenyl phosphate (TPP), an OPFR, is a potential chemical determinant of liver function damage, its effects on kidney function in mice under high fructose/fat (HFF) diet are still unclear. In this study, C57BL/6J mice were fed HFF to generate an obesity model and mice were exposed to low dose (0.01 mg/kg/day; TPP-L) and high dose (1 mg/kg/day; TPP-H) of TPP for 12 weeks. Results showed that TPP-L and TPP-H combined with HFF, as well as TPP-H alone, caused kidney structural damage and gut microbiota disorders in mice. Inflammatory response induced by nuclear factor kappa B (NF-κB p65)/nod-like receptor protein 3 (NLRP3) and caspase-3 promoted kidney structure damage, as well as accumulation of triglyceride and total cholesterol and the protein residues in urine. Although TPP-L did not cause obvious structural damage in the kidneys, 0.01 mg/kg TPP induced significant inflammation and gut microbiota disorders. These findings provide new insights regarding health risk assessment after chronic exposure to TPP and HFF alone, as well as a combination of TPP with HFF in mice.
Collapse
Affiliation(s)
- Haiyan Cui
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Yeqian Chang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Xiaofeng Jiang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Mei Li
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China.
| |
Collapse
|
82
|
Pomegranate flower extract bidirectionally regulates the proliferation, differentiation and apoptosis of 3T3-L1 cells through regulation of PPARγ expression mediated by PI3K-AKT signaling pathway. Biomed Pharmacother 2020; 131:110769. [DOI: 10.1016/j.biopha.2020.110769] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/31/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
|
83
|
Downregulation of CTRP-3 by Weight Loss In Vivo and by Bile Acids and Incretins in Adipocytes In Vitro. Int J Mol Sci 2020; 21:ijms21218168. [PMID: 33142914 PMCID: PMC7662344 DOI: 10.3390/ijms21218168] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 01/11/2023] Open
Abstract
The adipokine CTRP-3 (C1q/TNF-related protein-3) exerts anti-inflammatory and anti-diabetic effects. Its regulation in obesity and during weight loss is unknown. Serum and adipose tissue (AT) samples were obtained from patients (n = 179) undergoing bariatric surgery (BS). Moreover, patients (n = 131) participating in a low-calorie diet (LCD) program were studied. CTRP 3 levels were quantified by ELISA and mRNA expression was analyzed in AT and in 3T3-L1 adipocytes treated with bile acids and incretins. There was a persistent downregulation of CTRP-3 serum levels during weight loss. CTRP-3 expression was higher in subcutaneous than in visceral AT and serum levels of CTRP-3 were positively related to AT expression levels. A rapid decrease of circulating CTRP-3 was observed immediately upon BS, suggesting weight loss-independent regulatory mechanisms. Adipocytes CTRP-3 expression was inhibited by primary bile acid species and GLP 1. Adipocyte-specific CTRP-3 deficiency increased bile acid receptor expression. Circulating CTRP-3 levels are downregulated during weight loss, with a considerable decline occurring immediately upon BS. Mechanisms dependent and independent of weight loss cause the post-surgical decline of CTRP-3. The data strongly argue for regulatory interrelations of CTRP-3 with bile acids and incretin system.
Collapse
|
84
|
Lu HP, Lin CJ, Chen WC, Chang YJ, Lin SW, Wang HH, Chang CJ. TRIM28 Regulates Dlk1 Expression in Adipogenesis. Int J Mol Sci 2020; 21:ijms21197245. [PMID: 33008113 PMCID: PMC7582669 DOI: 10.3390/ijms21197245] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/20/2020] [Accepted: 09/27/2020] [Indexed: 12/23/2022] Open
Abstract
The tripartite motif-containing protein 28 (TRIM28) is a transcription corepressor, interacting with histone deacetylase and methyltransferase complexes. TRIM28 is a crucial regulator in development and differentiation. We would like to investigate its function and regulation in adipogenesis. Knockdown of Trim28 by transducing lentivirus-carrying shRNAs impairs the differentiation of 3T3-L1 preadipocytes, demonstrated by morphological observation and gene expression analysis. To understand the molecular mechanism of Trim28-mediated adipogenesis, the RNA-seq was performed to find out the possible Trim28-regulated genes. Dlk1 (delta-like homolog 1) was increased in Trim28 knockdown 3T3-L1 cells both untreated and induced to differentiation. Dlk1 is an imprinted gene and known as an inhibitor of adipogenesis. Further knockdown of Dlk1 in Trim28 knockdown 3T3-L1 would rescue cell differentiation. The epigenetic analysis showed that DNA methylation of Dlk1 promoter and differentially methylated regions (DMRs) was not altered significantly in Trim28 knockdown cells. However, compared to control cells, the histone methylation on the Dlk1 promoter was increased at H3K4 and decreased at H3K27 in Trim28 knockdown cells. Finally, we found Trim28 might be recruited by transcription factor E2f1 to regulate Dlk1 expression. The results imply Trim28-Dlk1 axis is critical for adipogenesis.
Collapse
Affiliation(s)
- Hsin-Pin Lu
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan; (H.-P.L.); (C.-J.L.); (W.-C.C.)
| | - Chieh-Ju Lin
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan; (H.-P.L.); (C.-J.L.); (W.-C.C.)
| | - Wen-Ching Chen
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan; (H.-P.L.); (C.-J.L.); (W.-C.C.)
| | - Yao-Jen Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; (Y.-J.C.); (S.-W.L.)
| | - Sheng-Wei Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; (Y.-J.C.); (S.-W.L.)
| | - Hsin-Hui Wang
- Department of Pediatrics, Division of Pediatric Immunology and Nephrology, Taipei Veterans General Hospital, Taipei 11217, Taiwan;
- Department of Pediatrics, Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan
- Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan
| | - Ching-Jin Chang
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan; (H.-P.L.); (C.-J.L.); (W.-C.C.)
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; (Y.-J.C.); (S.-W.L.)
- Correspondence:
| |
Collapse
|
85
|
Ohno S, Quek LE, Krycer JR, Yugi K, Hirayama A, Ikeda S, Shoji F, Suzuki K, Soga T, James DE, Kuroda S. Kinetic Trans-omic Analysis Reveals Key Regulatory Mechanisms for Insulin-Regulated Glucose Metabolism in Adipocytes. iScience 2020; 23:101479. [PMID: 32891058 PMCID: PMC7479629 DOI: 10.1016/j.isci.2020.101479] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/17/2020] [Accepted: 08/17/2020] [Indexed: 12/24/2022] Open
Abstract
Insulin regulates glucose metabolism through thousands of regulatory mechanisms; however, which regulatory mechanisms are keys to control glucose metabolism remains unknown. Here, we performed kinetic trans-omic analysis by integrating isotope-tracing glucose flux and phosphoproteomic data from insulin-stimulated adipocytes and built a kinetic mathematical model to identify key allosteric regulatory and phosphorylation events for enzymes. We identified nine reactions regulated by allosteric effectors and one by enzyme phosphorylation and determined the regulatory mechanisms for three of these reactions. Insulin stimulated glycolysis by promoting Glut4 activity by enhancing phosphorylation of AS160 at S595, stimulated fatty acid synthesis by promoting Acly activity through allosteric activation by glucose 6-phosphate or fructose 6-phosphate, and stimulated glutamate synthesis by alleviating allosteric inhibition of Gls by glutamate. Most of glycolytic reactions were regulated by amounts of substrates and products. Thus, phosphorylation or allosteric modulator-based regulation of only a few key enzymes was sufficient to change insulin-induced metabolism.
Collapse
Affiliation(s)
- Satoshi Ohno
- Molecular Genetics Research Laboratory, Graduate School of Science, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Lake-Ee Quek
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW 2006, Australia
| | - James R. Krycer
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Katsuyuki Yugi
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
- Laboratory for Integrated Cellular Systems, RIKEN Center for Integrative Medical Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
- PRESTO, Japan Science and Technology Agency, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
- AMED-CREST, AMED, 1-7-1 Otemachi, Chiyoda-Ku, Tokyo 100-0004, Japan
| | - Satsuki Ikeda
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
| | - Futaba Shoji
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
| | - Kumi Suzuki
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
- AMED-CREST, AMED, 1-7-1 Otemachi, Chiyoda-Ku, Tokyo 100-0004, Japan
| | - David E. James
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Shinya Kuroda
- Molecular Genetics Research Laboratory, Graduate School of Science, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
86
|
Jiang S, Wu X, Wang Y, Zou J, Zhao X. The potential DPP-4 inhibitors from Xiao-Ke-An improve the glucolipid metabolism via the activation of AKT/GSK-3β pathway. Eur J Pharmacol 2020; 882:173272. [PMID: 32535096 DOI: 10.1016/j.ejphar.2020.173272] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 11/29/2022]
Abstract
Dipeptidyl Peptidase-4 (DPP-4) is a specific enzyme hydrolyzing the incretin hormone glucagon like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) to reduce insulin secretion, meanwhile DPP-4 inhibitors play an important role in diabetic therapy. In present study, 14 potential inhibitors were screened with an inhibition over 50% on DPP-4 activity from Xiao-Ke-An formula (XKA) and 12 of them exhibited a dose-dependently inhibitory effect at concentrations of 5-50 μmol/l. We found 10 DPP-4 inhibitors restrained differentiation of 3T3-L1 pre-adipocytes as well as reducing the triglycerides and total cholesterol content in 3T3-L1 adipocytes. Furthermore, 7 DPP-4 inhibitors promoted the glucose consumption in insulin-resistance BNL CL.2 cells. Thereinto, ginsenoside Rk1 up-regulated the protein kinase B (AKT) and glycogen synthase kinase-3 (GSK-3β) phosphorylation expression, while kukoamine B and coptisine hydrochloride obviously increased the phosphorylation of AKT protein and columbamine, panaxadiol, ginsenoside Ro, timosaponin AI significantly promoted the phosphorylation of GSK-3β protein. It's our first effort to confirm those seven compounds could serve as DPP-4 inhibitors to attenuate DPP-4 activities, accompanied with the ability to adjust glucolipid metabolism. Moreover, activating the AKT/GSK-3β signaling pathway to ameliorate insulin resistant may be the anti-diabetic mechanism of XKA.
Collapse
Affiliation(s)
- Shumin Jiang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Xueli Wu
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Yi Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Jingtao Zou
- Tonghua Huaxia Pharmaceutical Company, JiLin, 134000, China.
| | - Xiaoping Zhao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
87
|
Oldoni F, Cheng H, Banfi S, Gusarova V, Cohen JC, Hobbs HH. ANGPTL8 has both endocrine and autocrine effects on substrate utilization. JCI Insight 2020; 5:138777. [PMID: 32730227 PMCID: PMC7526440 DOI: 10.1172/jci.insight.138777] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/22/2020] [Indexed: 12/17/2022] Open
Abstract
The angiopoietin-like protein ANGPTL8 (A8) is one of 3 ANGPTLs (A8, A3, A4) that coordinate changes in triglyceride (TG) delivery to tissues by inhibiting lipoprotein lipase (LPL), an enzyme that hydrolyzes TG. Previously we showed that A8, which is expressed in liver and adipose tissue, is required to redirect dietary TG from oxidative to storage tissues following food intake. Here we show that A8 from liver and adipose tissue have different roles in this process. Mice lacking hepatic A8 have no circulating A8, high intravascular LPL activity, low plasma TG levels, and evidence of decreased delivery of dietary lipids to adipose tissue. In contrast, mice lacking A8 in adipose tissue have higher postprandial TG levels and similar intravascular LPL activity and plasma A8 levels and higher levels of plasma TG. Expression of A8, together with A4, in cultured cells reduced A4 secretion and A4-mediated LPL inhibition. Thus, hepatic A8 (with A3) acts in an endocrine fashion to inhibit intravascular LPL in oxidative tissues, whereas A8 in adipose tissue enhances LPL activity by autocrine/paracrine inhibition of A4. These combined actions of A8 ensure that TG stores are rapidly replenished and sufficient energy is available until the next meal. Angiopoietin-like protein ANGPTL8 expressed in liver and adipose tissue partner with ANGPTL3 and ANGPTL4 respectively, and replenish adipose tissue triglyceride stores by distinct endocrine and autocrine/paracrine mechanisms.
Collapse
Affiliation(s)
- Federico Oldoni
- Departments of Molecular Genetics and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Haili Cheng
- Departments of Molecular Genetics and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Serena Banfi
- Departments of Molecular Genetics and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | | | - Helen H Hobbs
- Departments of Molecular Genetics and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Howard Hughes Medical Institute, University of Texas Southwestern (UTSW) Medical Center, Dallas, Texas, USA
| |
Collapse
|
88
|
Huang D, Camacho CV, Setlem R, Ryu KW, Parameswaran B, Gupta RK, Kraus WL. Functional Interplay between Histone H2B ADP-Ribosylation and Phosphorylation Controls Adipogenesis. Mol Cell 2020; 79:934-949.e14. [PMID: 32822587 DOI: 10.1016/j.molcel.2020.08.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 07/30/2020] [Accepted: 08/03/2020] [Indexed: 01/17/2023]
Abstract
Although ADP-ribosylation of histones by PARP-1 has been linked to genotoxic stress responses, its role in physiological processes and gene expression has remained elusive. We found that NAD+-dependent ADP-ribosylation of histone H2B-Glu35 by small nucleolar RNA (snoRNA)-activated PARP-1 inhibits AMP kinase-mediated phosphorylation of adjacent H2B-Ser36, which is required for the proadipogenic gene expression program. The activity of PARP-1 on H2B requires NMNAT-1, a nuclear NAD+ synthase, which directs PARP-1 catalytic activity to Glu and Asp residues. ADP-ribosylation of Glu35 and the subsequent reduction of H2B-Ser36 phosphorylation inhibits the differentiation of adipocyte precursors in cultured cells. Parp1 knockout in preadipocytes in a mouse lineage-tracing genetic model increases adipogenesis, leading to obesity. Collectively, our results demonstrate a functional interplay between H2B-Glu35 ADP-ribosylation and H2B-Ser36 phosphorylation that controls adipogenesis.
Collapse
Affiliation(s)
- Dan Huang
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cardiology, Clinical Center for Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, P.R. China
| | - Cristel V Camacho
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rohit Setlem
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Keun Woo Ryu
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Balaji Parameswaran
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rana K Gupta
- Department of Internal Medicine, Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - W Lee Kraus
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
89
|
Chen H, Yin C, Zhang X, Zhu Y. Preparation and characterisation of bifunctional surface-modified silicone catheter in lumen. J Glob Antimicrob Resist 2020; 23:46-54. [PMID: 32795517 DOI: 10.1016/j.jgar.2020.07.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/08/2020] [Accepted: 07/20/2020] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES The purpose of this study was to evaluate the coating of antimicrobial peptides (AMPs) and polyvinylpyrrolidone (PVP) to the surface of a silicone catheter to reduce bacterial growth and to increase hydrophilicity, respectively. METHODS Surface characterisation was performed on bare silicone, AMP-coated, PVP-coated and AMP + PVP-coated silicone catheters using attenuated total reflectance-infrared (ATR-IR) spectroscopy, X-ray photoelectron spectroscopy (XPS) and water contact angle. Antibacterial activity, antibacterial biofilm growth and XTT assay were performed on bare silicone, AMP-coated, PVP-coated and AMP + PVP-coated silicone catheters. Statistical analysis was performed by one-way ANOVA. RESULTS The water contact angle of the AMP + PVP-coated silicone catheter was 21.37 ± 2.17° compared with 107.23 ± 0.96°, 74.40 ± 1.76° and 20.77 ± 0.32° for bare silicone, AMP-coated and PVP-coated silicone catheters. Based on in vitro antimicrobial tests, the AMP + PVP-coated silicone catheter had 6.2, 2.2 and 2.5 greater antibacterial activity than that of the bare silicone catheter against Escherichia coli, Staphylococcus aureus and Pseudomonas aeruginosa, respectively. Moreover, bacterial biofilm growth on the surface of the AMP + PVP-coated silicone catheter was minimal as characterised by scanning electron microscopy. MTT assay showed that bare silicone, AMP-coated, AMP + PVP-coated and PVP-coated silicone catheters were non-cytotoxic to 3T3 and human colon cancer (Caco-2) cells. CONCLUSIONS This work demonstrates that AMP + PVP-coated silicone catheters have potential clinical application prospects with improved hydrophilicity, excellent biocompatibility, antibacterial activity and a certain antibacterial biofilm effect.
Collapse
Affiliation(s)
- Haiyan Chen
- College of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Caiyun Yin
- College of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Xin Zhang
- College of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Yishen Zhu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China.
| |
Collapse
|
90
|
MAPK-interacting kinase 2 (MNK2) regulates adipocyte metabolism independently of its catalytic activity. Biochem J 2020; 477:2735-2754. [DOI: 10.1042/bcj20200433] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 11/17/2022]
Abstract
The mitogen-activated protein kinase (MAPK)-interacting kinases (MNKs) are serine/threonine protein kinases that are activated by the ERK1/2 (extracellular regulated kinase) and p38α/β MAPK pathways. The MNKs have previously been implicated in metabolic disease and shown to mediate diet-induced obesity. In particular, knockout of MNK2 in mice protects from the weight gain induced by a high-fat diet. These and other data suggest that MNK2 regulates the expansion of adipose tissue (AT), a stable, long-term energy reserve that plays an important role in regulating whole-body energy homeostasis. Using the well-established mouse 3T3-L1 in vitro model of adipogenesis, the role of the MNKs in adipocyte differentiation and lipid storage was investigated. Inhibition of MNK activity using specific inhibitors failed to impair adipogenesis or lipid accumulation, suggesting that MNK activity is not required for adipocyte differentiation and does not regulate lipid storage. However, small-interfering RNA (siRNA) knock-down of MNK2 did reduce lipid accumulation and regulated the levels of two major lipogenic transcriptional regulators, ChREBP (carbohydrate response element-binding protein) and LPIN1 (Lipin-1). These factors are responsible for controlling the expression of genes for proteins involved in de novo lipogenesis and triglyceride synthesis. The knock-down of MNK2 also increased the expression of hormone-sensitive lipase which catalyses the breakdown of triglyceride. These findings identify MNK2 as a regulator of adipocyte metabolism, independently of its catalytic activity, and reveal some of the mechanisms by which MNK2 drives AT expansion. The development of an MNK2-targeted therapy may, therefore, be a useful intervention for reducing weight caused by excessive nutrient intake.
Collapse
|
91
|
Herranz-López M, Olivares-Vicente M, Rodríguez Gallego E, Encinar JA, Pérez-Sánchez A, Ruiz-Torres V, Joven J, Roche E, Micol V. Quercetin metabolites from Hibiscus sabdariffa contribute to alleviate glucolipotoxicity-induced metabolic stress in vitro. Food Chem Toxicol 2020; 144:111606. [PMID: 32738368 DOI: 10.1016/j.fct.2020.111606] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 12/17/2022]
Abstract
Polyphenols from Hibiscus sabdariffa (HS) alleviate obesity-related metabolic complications but the metabolites responsible for such effects are unknown. We aimed to elucidate which of the potential plasma metabolites from a polyphenol-enriched HS (PEHS) extract contributed for the reversion of glucolipotoxicity-induced metabolic stress using 3T3-L1 adipocyte and INS 832/13 pancreatic β-cell models under glucolipotoxic conditions. PEHS extract, quercetin (Q) and quercetin-3-O-glucuronide (Q3GA) showed stronger capacity to decrease glucolipotoxicity-induced ROS generation than ascorbic acid or chlorogenic acid. PEHS extract, Q and Q3GA decreased secretion of cytokines (leptin, TNF-α, IGF-1, IL-6, VEGF, IL-1α, IL-1β and CCL2) and reduced CCL2 expression at transcriptional level. In addition, PEHS extract, Q and Q3GA reduced triglyceride accumulation, which occurred through fatty acid synthase (FASN) downregulation, AMPK activation and mitochondrial mass and biogenesis restoration via PPARα upregulation. Electron microscopy confirmed that PEHS extract and Q3GA decreased mitochondrial remodeling and mitophagy. Virtual screening leads us to postulate that Q and Q3GA might act as agonists of these protein targets at specific sites. These data suggest that Q and Q3GA may be the main responsible compounds for the capacity of PEHS extract to revert glucolipotoxicity-induced metabolic stress through AMPK-mediated decrease in fat storage and increase in fatty acid oxidation, though other compounds of the extract may contribute to this capacity.
Collapse
Affiliation(s)
- María Herranz-López
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE) e Instituto de Biología Molecular y Celular (IBMC). Miguel Hernández University (UMH), Elche, 03202, Alicante, Spain.
| | - Mariló Olivares-Vicente
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE) e Instituto de Biología Molecular y Celular (IBMC). Miguel Hernández University (UMH), Elche, 03202, Alicante, Spain
| | - Esther Rodríguez Gallego
- Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43007, Tarragona, Spain
| | - Jose Antonio Encinar
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE) e Instituto de Biología Molecular y Celular (IBMC). Miguel Hernández University (UMH), Elche, 03202, Alicante, Spain
| | - Almudena Pérez-Sánchez
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE) e Instituto de Biología Molecular y Celular (IBMC). Miguel Hernández University (UMH), Elche, 03202, Alicante, Spain
| | - Verónica Ruiz-Torres
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE) e Instituto de Biología Molecular y Celular (IBMC). Miguel Hernández University (UMH), Elche, 03202, Alicante, Spain
| | - Jorge Joven
- Universitat Rovira i Virgili, Departament de Cirurgia i Medicina, Unitat de Recerca Biomèdica, 43201, Reus, Tarragona, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), 43204, Reus, Spain
| | - Enrique Roche
- Institute of Bioengineering and Department of Applied Biology-Nutrition, University Miguel Hernandez, Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain; CIBER, Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Instituto de Salud Carlos III (CB12/03/30038), 28220, Madrid, Spain
| | - Vicente Micol
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE) e Instituto de Biología Molecular y Celular (IBMC). Miguel Hernández University (UMH), Elche, 03202, Alicante, Spain; CIBER, Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Instituto de Salud Carlos III (CB12/03/30038), 28220, Madrid, Spain
| |
Collapse
|
92
|
Rivero-Pino F, Espejo-Carpio FJ, Guadix EM. Antidiabetic Food-Derived Peptides for Functional Feeding: Production, Functionality and In Vivo Evidences. Foods 2020; 9:E983. [PMID: 32718070 PMCID: PMC7466190 DOI: 10.3390/foods9080983] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/15/2020] [Accepted: 07/21/2020] [Indexed: 12/11/2022] Open
Abstract
Bioactive peptides released from the enzymatic hydrolysis of food proteins are currently a trending topic in the scientific community. Their potential as antidiabetic agents, by regulating the glycemic index, and thus to be employed in food formulation, is one of the most important functions of these peptides. In this review, we aimed to summarize the whole process that must be considered when talking about including these molecules as a bioactive ingredient. In this regard, at first, the production, purification and identification of bioactive peptides is summed up. The detailed metabolic pathways described included carbohydrate hydrolases (glucosidase and amylase) and dipeptidyl-peptidase IV inhibition, due to their importance in the food-derived peptides research field. Then, their characterization, concerning bioavailability in vitro and in situ, stability and functionality in food matrices, and ultimately, the in vivo evidence (from invertebrate animals to humans), was described. The future applicability that these molecules have due to their biological potential as functional ingredients makes them an important field of research, which could help the world population avoid suffering from several diseases, such as diabetes.
Collapse
Affiliation(s)
- Fernando Rivero-Pino
- Department of Chemical Engineering, University of Granada, 18071 Granada, Spain; (F.J.E.-C.); (E.M.G.)
| | | | | |
Collapse
|
93
|
Kim BR, Kim J, Lee JE, Lee EJ, Yoon JS. Therapeutic Effect of Guggulsterone in Primary Cultured Orbital Fibroblasts Obtained From Patients with Graves' Orbitopathy. Invest Ophthalmol Vis Sci 2020; 61:39. [PMID: 32196098 PMCID: PMC7401490 DOI: 10.1167/iovs.61.3.39] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Purpose Inflammation, hyaluronan production, and adipogenesis are the main pathological events leading to Graves' orbitopathy (GO). Guggulsterone (GS), a phytosterol found in the resin of the guggul plant, is a well-known treatment for several inflammatory disorders, such as arthritis, obesity, and hyperlipidemia. Here we investigated the effects of GS treatment on GO pathology. Methods Using primary cultures of orbital fibroblasts from GO patients and non-GO controls, we examined the effects of GS on hyaluronan production and the production of proinflammatory cytokines induced by interleukin (IL)-1β, using real-time reverse transcription-polymerase chain reaction analysis, western blots, and enzyme-linked immunosorbent assays. Further, adipogenic differentiation was evaluated by quantification of Oil Red O staining and assessment of protein levels of peroxisome proliferator activator gamma (PPARγ), CCAAT-enhancer-binding proteins (C/EBP) α and β, and sterol regulatory element-binding protein-1 (SREBP-1). Results Treatment with noncytotoxic concentrations of GS resulted in the dose-dependent inhibition of IL-1β-induced inflammatory cytokines, including IL-6, IL-8, MCP-1, and COX-2, at both mRNA and protein levels. The hyaluronan level was also significantly suppressed by GS. Moreover, GS significantly decreased the formation of lipid droplets and expression of PPARγ, C/EBP α/β, and SREBP-1 in a dose-dependent manner. GS pretreatment attenuated the phosphorylation of nuclear factor-kappa B induced by IL-1β. Conclusions Our data show significant inhibitory effects of GS on inflammation, production of hyaluronan, and adipogenesis in orbital fibroblasts. To our knowledge, this is the first in vitro preclinical evidence of the therapeutic effect of GS in GO.
Collapse
|
94
|
Cottingham CM, Patrick T, Richards MA, Blackburn KD. Tricyclic antipsychotics promote adipogenic gene expression to potentiate preadipocyte differentiation in vitro. Hum Cell 2020; 33:502-511. [PMID: 32447572 PMCID: PMC10805149 DOI: 10.1007/s13577-020-00372-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/02/2020] [Indexed: 11/30/2022]
Abstract
Antipsychotic-induced weight gain is a well-established but poorly understood clinical phenomenon. New mechanistic insights into how antipsychotics modulate adipose physiology are sorely needed, in hopes of either devising a therapeutic intervention to ameliorate weight gain or contributing to improved design of future agents. In this study, we have hypothesized that the weight gain-associated tricyclic antipsychotics clozapine and chlorpromazine directly impact adipose tissue by potentiating adipogenic differentiation of preadipocytes. Utilizing a well-established in vitro model system (3T3-L1 preadipocyte cell line), we demonstrate that, when applied specifically during induction of adipogenic differentiation, both clozapine and chlorpromazine significantly potentiate in vitro adipogenesis, observed as morphological changes and increased intracellular lipid accumulation. These persistent effects, observed at endpoints well after the end of antipsychotic exposure, are accompanied by increased transcript- and protein-level expression of the mature adipocyte marker perilipin-1, as indicated by RT-qPCR and Western blotting, but not by further upregulation of pro-adipogenic transcription factors versus positive controls. Our findings point to a possible physiological mechanism of antipsychotic-induced hyperplasia, with potentiated expression of mature adipocyte markers enhancing the differentiation and maturation of preadipocytes.
Collapse
Affiliation(s)
- Christopher M Cottingham
- Department of Biology, University of North Alabama, UNA, One Harrison Plaza, Box 5048, Florence, AL, 35632, USA.
| | - Taylor Patrick
- Department of Biology and Chemistry, Morehead State University, 103 Lappin Hall, Morehead, KY, 40351, USA
- University of Kentucky College of Medicine, UK Medical Center MN 150, Lexington, KY, 40536, USA
| | - Morgan A Richards
- Department of Biology and Chemistry, Morehead State University, 103 Lappin Hall, Morehead, KY, 40351, USA
- School of Rehabilitation and Health Sciences, Ohio State University, 453 West 10th Avenue, Columbus, OH, 43210, USA
| | - Kirkland D Blackburn
- Department of Biology and Chemistry, Morehead State University, 103 Lappin Hall, Morehead, KY, 40351, USA
- Kentucky College of Osteopathic Medicine, 147 Sycamore Street, Pikeville, KY, 41501, USA
| |
Collapse
|
95
|
Coleman RA. The "discovery" of lipid droplets: A brief history of organelles hidden in plain sight. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158762. [PMID: 32622088 DOI: 10.1016/j.bbalip.2020.158762] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 12/14/2022]
Abstract
Mammalian lipid droplets (LDs), first described as early as the 1880s, were virtually ignored for more than 100 years. Between 1991 and the early 2000s, however, a series of discoveries and conceptual breakthroughs led to a resurgent interest in obesity as a disease, in the metabolism of intracellular triacylglycerol (TAG), and in the physical locations of LDs as cellular structures with their associated proteins. Insights included the recognition that obesity underlies major chronic diseases, that appetite is hormonally controlled, that hepatic steatosis is not a benign finding, and that diabetes might fundamentally be a disorder of lipid metabolism. In this brief review, I describe the metamorphosis of LDs from overlooked globs of stored fat to dynamic organelles that control insulin resistance, mitochondrial oxidation, and viral replication.
Collapse
Affiliation(s)
- Rosalind A Coleman
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States of America.
| |
Collapse
|
96
|
Cárdenas-León CG, Montoya-Contreras A, Mäemets-Allas K, Jaks V, Salazar-Olivo LA. A human preadipocyte cell strain with multipotent differentiation capability as an in vitro model for adipogenesis. In Vitro Cell Dev Biol Anim 2020; 56:399-411. [PMID: 32535758 DOI: 10.1007/s11626-020-00468-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 05/14/2020] [Indexed: 12/15/2022]
Abstract
Murine 3T3 cell lines constitute a standard model system for in vitro study of mammalian adipogenesis although they do not faithfully reflect the biology of the human adipose cells. Several human adipose cell lines and strains have been used to recapitulate human adipogenesis in vitro, but to date there is no generally accepted in vitro model for human adipogenesis. We obtained a clonal strain of human subcutaneous adipose stromal cells, IPI-SA3-C4, and characterized its utility as an in vitro model for human subcutaneous adipogenesis. IPI-SA3-C4 cells showed a high proliferative potential for at least 30 serial passages, reached 70 cumulative population doublings and exhibited a population doubling time of 47 h and colony forming efficiency of 12% at the 57th cumulative population doublings. IPI-SA3-C4 cells remained diploid (46XY) even at the 56th cumulative population doublings and expressed the pluripotency markers POU5F1, NANOG, KLF4, and MYC even at 50th cumulative population doublings. Under specific culture conditions, IPI-SA3-C4 cells displayed cellular hallmarks and molecular markers of adipogenic, osteogenic, and chondrogenic lineages and showed adipogenic capacity even at the 66th cumulative population doublings. These characteristics show IPI-SA3-C4 cells as a promising potential model for human subcutaneous adipogenesis in vitro.
Collapse
Affiliation(s)
- Claudia G Cárdenas-León
- División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica, Camino a la Presa San José 2055, 78216, San Luis Potosí, SLP, Mexico
| | - Angélica Montoya-Contreras
- División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica, Camino a la Presa San José 2055, 78216, San Luis Potosí, SLP, Mexico
| | - Kristina Mäemets-Allas
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Viljar Jaks
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Luis A Salazar-Olivo
- División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica, Camino a la Presa San José 2055, 78216, San Luis Potosí, SLP, Mexico.
| |
Collapse
|
97
|
Human Adipose Derived Cells in Two- and Three-Dimensional Cultures: Functional Validation of an In Vitro Fat Construct. Stem Cells Int 2020; 2020:4242130. [PMID: 32587620 PMCID: PMC7303735 DOI: 10.1155/2020/4242130] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/20/2020] [Accepted: 03/13/2020] [Indexed: 02/06/2023] Open
Abstract
Obesity, defined as a body mass index of 30 kg/m2 or above, has increased considerably in incidence and frequency within the United States and globally. Associated comorbidities including cardiovascular disease, type 2 diabetes mellitus, metabolic syndrome, and nonalcoholic fatty liver disease have led to a focus on the mechanisms promoting the prevention and treatment of obesity. Commonly utilized in vitro models employ human or mouse preadipocyte cell lines in a 2-dimensional (2D) format. Due to the structural, biochemical, and biological limitations of these models, increased attention has been placed on "organ on a chip" technologies for a 3-dimensional (3D) culture. Herein, we describe a method employing cryopreserved primary human stromal vascular fraction (SVF) cells and a human blood product-derived biological scaffold to create a 3D adipose depot in vitro. The "fat-on-chip" 3D cultures have been validated relative to 2D cultures based on proliferation, flow cytometry, adipogenic differentiation, confocal microscopy/immunofluorescence, and functional assays (adipokine secretion, glucose uptake, and lipolysis). Thus, the in vitro culture system demonstrates the critical characteristics required for a humanized 3D white adipose tissue (WAT) model.
Collapse
|
98
|
Yu B, Pu Y, Liu J, Liao J, Chen K, Zhang J, Zhong W, Hu Y, Wang XQ, Liu B, Liu H, Tan W. Targeted delivery of emodin to adipocytes by aptamer-functionalized PEG-PLGA nanoparticles in vitro. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
99
|
Andrews FV, Kim SM, Edwards L, Schlezinger JJ. Identifying adipogenic chemicals: Disparate effects in 3T3-L1, OP9 and primary mesenchymal multipotent cell models. Toxicol In Vitro 2020; 67:104904. [PMID: 32473317 DOI: 10.1016/j.tiv.2020.104904] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/18/2020] [Accepted: 05/22/2020] [Indexed: 12/19/2022]
Abstract
3T3-L1 pre-adipocytes are used commonly to identify new adipogens, but this cell line has been shown to produce variable results. Here, potential adipogenic chemicals (identified in the ToxCast dataset using the Toxicological Priority Index) were tested for their ability to induce adipocyte differentiation in 3T3-L1 cells, OP9 cells and primary mouse bone marrow multipotent stromal cells (BM-MSC). Ten of the 36 potential adipogens stimulated lipid accumulation in at least one model (novel: fenthion, quinoxyfen, prallethrin, allethrin, pyrimethanil, tebuconzaole, 2,4,6-tris (tert-butyl)phenol; known: fentin, pioglitazone, 3,3',5,5'-tetrabromobisphenol A). Only prallethrin and pioglitazone enhanced lipid accumulation in all models. OP9 cells were significantly more sensitive to chemicals known to activate PPARγ through RXR than the other models. Coordinate effects on adipocyte and osteoblast differentiation were investigated further in BM-MSCs. Lipid accumulation was correlated with the ability to stimulate expression of the PPARγ target gene, Plin1. Induction of lipid accumulation also was associated with reduction in alkaline phosphatase activity. Allethrin, prallethrin, and quinoxyfen strongly suppressed osteogenic gene expression. BM-MSCs were useful in coordinately investigating pro-adipogenic and anti-osteogenic effects. Overall, the results show that additional models should be used in conjunction with 3T3-L1 cells to identify a broader spectrum of adipogens and their coordinate effects on osteogenesis.
Collapse
Affiliation(s)
- Faye V Andrews
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Stephanie M Kim
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Lariah Edwards
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Jennifer J Schlezinger
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA.
| |
Collapse
|
100
|
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: 1.8] [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
|