1
|
Takahashi T, Takase Y, Shiraishi A, Matsubara S, Watanabe T, Kirimoto S, Yamagaki T, Osawa M. Weight Gain With Advancing Age Is Controlled by the Muscarinic Acetylcholine Receptor M4 in Male Mice. Endocrinology 2025; 166:bqaf064. [PMID: 40179260 PMCID: PMC12012353 DOI: 10.1210/endocr/bqaf064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/04/2025] [Accepted: 04/01/2025] [Indexed: 04/05/2025]
Abstract
Obesity is characterized by the excessive accumulation of adipose tissue, and it is a serious global health issue. Understanding the pathology of obesity is crucial for developing effective interventions. In this study, we investigated the role of muscarinic acetylcholine receptor M4 (mAChR-M4) in the regulation of obesity in Chrm4-knockout (M4-KO) mice. Male M4-KO mice showed higher weight gain and accumulation of white adipose tissue (WAT) with advancing age than the wild-type mice. The M4-KO mice also showed increased leptin expression at both the transcription and the translation levels. RNA sequencing and quantitative reverse transcription polymerase chain reaction analyses of subcutaneous adipose tissues revealed that the expression of WAT marker genes was significantly enhanced in the M4-KO mice. In contrast, the expression levels of brown adipose tissue/beige adipose tissue markers were strongly decreased in the M4-KO mice. To identify the Chrm4-expressing cell types, we generated Chrm4-mScarlet reporter mice and examined the localization of the mScarlet fluorescent signals in subcutaneous tissues. Fluorescent signals were prominently detected in WAT and mesenchymal stem cells. Additionally, we also found that choline acetyltransferase was expressed in macrophages, suggesting their involvement in acetylcholine (ACh) secretion. Corroborating this notion, we were able to quantitatively measure the ACh in subcutaneous tissues by liquid chromatography tandem mass spectrometry. Collectively, our findings suggest that endogenous ACh released from macrophages maintains the homeostasis of adipose cell growth and differentiation via mAChR-M4 in male mice. This study provides new insights into the molecular mechanisms underlying obesity and potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Toshio Takahashi
- Suntory Foundation for Life Sciences, Bioorganic Research Institute, Kyoto 619-0284, Japan
| | - Yuta Takase
- Suntory Foundation for Life Sciences, Bioorganic Research Institute, Kyoto 619-0284, Japan
| | - Akira Shiraishi
- Suntory Foundation for Life Sciences, Bioorganic Research Institute, Kyoto 619-0284, Japan
| | - Shin Matsubara
- Suntory Foundation for Life Sciences, Bioorganic Research Institute, Kyoto 619-0284, Japan
| | - Takehiro Watanabe
- Suntory Foundation for Life Sciences, Bioorganic Research Institute, Kyoto 619-0284, Japan
| | - Shinji Kirimoto
- Animal Science Business Unit, KAC Co., Ltd., Kyoto 604-8423, Japan
| | - Tohru Yamagaki
- Suntory Foundation for Life Sciences, Bioorganic Research Institute, Kyoto 619-0284, Japan
| | - Masatake Osawa
- Department of Regenerative Medicine and Applied Biomedical Sciences, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan
- Center for Highly Advanced Integration of Nano and Life Sciences, Gifu University, Gifu 501-1194, Japan
| |
Collapse
|
2
|
Wing A, Jeffery E, Church CD, Goodell J, Saavedra-Peña RDM, Saha M, Holtrup B, Voisin M, Alavi NS, Floody M, Wang Z, Zapadka TE, Garabedian MJ, Varshney R, Rudolph MC, Rodeheffer MS. Dietary oleic acid drives obesogenic adipogenesis via modulation of LXRα signaling. Cell Rep 2025; 44:115527. [PMID: 40208790 DOI: 10.1016/j.celrep.2025.115527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 02/17/2025] [Accepted: 03/15/2025] [Indexed: 04/12/2025] Open
Abstract
Dietary fat composition has changed substantially during the obesity epidemic. As adipocyte hyperplasia is a major mechanism of adipose expansion, we aim to ascertain how dietary fats affect adipogenesis during obesity. We employ an unbiased dietary screen to identify oleic acid (OA) as the only dietary fatty acid that induces obesogenic hyperplasia at physiologic levels and show that plasma monounsaturated fatty acids (MUFAs), which are mostly OA, are associated with human obesity. OA stimulates adipogenesis in mouse and human adipocyte precursor cells (APCs) by increasing AKT2 signaling, a hallmark of obesogenic hyperplasia, and reducing LXR activity. High OA consumption decreases LXRα Ser196 phosphorylation in APCs, while blocking LXRα phosphorylation results in APC hyperproliferation. As OA is increasingly being incorporated into dietary fats due to purported health benefits, our finding that OA is a unique physiologic regulator of adipose biology underscores the importance of understanding how high OA consumption affects metabolic health.
Collapse
Affiliation(s)
- Allison Wing
- Department of Molecular, Cell, and Developmental Biology, Yale University, 219 Prospect St., New Haven, CT 06520, USA
| | - Elise Jeffery
- Department of Cell Biology, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06520, USA
| | - Christopher D Church
- Department of Comparative Medicine, Yale University School of Medicine, 310 Cedar St., New Haven, CT 06520, USA
| | - Jennifer Goodell
- Department of Comparative Medicine, Yale University School of Medicine, 310 Cedar St., New Haven, CT 06520, USA
| | - Rocío Del M Saavedra-Peña
- Department of Molecular, Cell, and Developmental Biology, Yale University, 219 Prospect St., New Haven, CT 06520, USA
| | - Moumita Saha
- Department of Comparative Medicine, Yale University School of Medicine, 310 Cedar St., New Haven, CT 06520, USA
| | - Brandon Holtrup
- Department of Molecular, Cell, and Developmental Biology, Yale University, 219 Prospect St., New Haven, CT 06520, USA
| | - Maud Voisin
- Department of Microbiology, NYU School of Medicine, New York, NY 10016, USA
| | - N Sima Alavi
- Department of Comparative Medicine, Yale University School of Medicine, 310 Cedar St., New Haven, CT 06520, USA
| | - Mariana Floody
- Department of Comparative Medicine, Yale University School of Medicine, 310 Cedar St., New Haven, CT 06520, USA
| | - Zenan Wang
- Department of Molecular, Cell, and Developmental Biology, Yale University, 219 Prospect St., New Haven, CT 06520, USA
| | - Thomas E Zapadka
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Michael J Garabedian
- Department of Microbiology, NYU School of Medicine, New York, NY 10016, USA; Department of Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Rohan Varshney
- Department of Biochemistry and Physiology and Harold Hamm Diabetes Center, Oklahoma University Health Sciences, Oklahoma City, OK 73104, USA
| | - Michael C Rudolph
- Department of Biochemistry and Physiology and Harold Hamm Diabetes Center, Oklahoma University Health Sciences, Oklahoma City, OK 73104, USA.
| | - Matthew S Rodeheffer
- Department of Molecular, Cell, and Developmental Biology, Yale University, 219 Prospect St., New Haven, CT 06520, USA; Department of Cell Biology, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06520, USA; Yale Stem Cell Center, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06520, USA; Yale Center of Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
3
|
Dumesic PA, Wilensky SE, Bose S, Van Vranken JG, Gygi SP, Spiegelman BM. RBM43 controls PGC1α translation and a PGC1α-STING signaling axis. Cell Metab 2025; 37:742-757.e8. [PMID: 39965564 PMCID: PMC11885043 DOI: 10.1016/j.cmet.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 10/17/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025]
Abstract
Obesity is associated with systemic inflammation that impairs mitochondrial function. This disruption curtails oxidative metabolism, limiting adipocyte lipid metabolism and thermogenesis, a metabolically beneficial program that dissipates chemical energy as heat. Here, we show that PGC1α, a key governor of mitochondrial biogenesis, is negatively regulated at the level of its mRNA translation by the RNA-binding protein RBM43. RBM43 is induced by inflammatory cytokines and suppresses mitochondrial biogenesis in a PGC1α-dependent manner. In mice, adipocyte-selective Rbm43 disruption elevates PGC1α translation and oxidative metabolism. In obesity, Rbm43 loss improves glucose tolerance, reduces adipose inflammation, and suppresses activation of the innate immune sensor cGAS-STING in adipocytes. We further identify a role for PGC1α in safeguarding against cytoplasmic accumulation of mitochondrial DNA, a cGAS ligand. The action of RBM43 defines a translational regulatory axis by which inflammatory signals dictate cellular energy metabolism and contribute to metabolic disease pathogenesis.
Collapse
Affiliation(s)
- Phillip A Dumesic
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah E Wilensky
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Symanthika Bose
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
4
|
Johnston EK, Dassau T, Muraskin NA, Abbott RD. Unilocular adipocyte and lipid tracer for immunofluorescent images. Sci Rep 2025; 15:4643. [PMID: 39920142 PMCID: PMC11805913 DOI: 10.1038/s41598-024-80613-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 11/20/2024] [Indexed: 02/09/2025] Open
Abstract
Adipose tissue is a highly dynamic endocrine organ that serves as the body's primary energy reservoir through the storage and mobilization of lipids. Adipocyte cellular size has been recognized as an indicator of cellular status; hypertrophic adipocytes are more prone to insulin resistance and the secretion of pro-inflammatory cytokines. Thus, the size and number of lipids is important to consider both in the clinic with a biopsy and when developing disease models and regenerative tissue constructs. Tools available to analyze adipocyte size are finely tuned for hematoxylin-eosin images and tend to be challenged by confocal derived z-stack images which contain intensity gradients. Therefore, ImageJ manual analysis is the commonly utilized tool to measure these images. With there being heterogeneity in different researcher's analytical approach when conducted manually, the MATLAB script, PixCell, was developed to reduce the subjectivity and time involved in adipocyte size analysis. Given its stepwise thresholding and masking steps, PixCell retains on average a >80% accuracy when tested on excised human adipose tissue, adipocyte-laden collagen gels, and lipoaspirate seeded silk scaffolds. PixCell is able to consistently detect and measure lipids within regions of varying pixel intensities. This makes PixCell an appealing tool for use in both the clinical and pre-clinical setting, while greatly enhancing and streamlining the user experience of analyzing lipid sizes.
Collapse
Affiliation(s)
- Elizabeth K Johnston
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Tal Dassau
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Nickia A Muraskin
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Rosalyn D Abbott
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
5
|
Das P, Mukherjee T, Adhikary K, Mohanty S. Progressive Approaches in Adipose Radio Imaging: Cancer Utilization and Necessity for Advancements. Infect Disord Drug Targets 2025; 25:e220524230197. [PMID: 38778619 DOI: 10.2174/0118715265314722240508092646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/14/2024] [Accepted: 04/27/2024] [Indexed: 05/25/2024]
Affiliation(s)
- Priyamjeet Das
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India
| | - Tuhin Mukherjee
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India
| | - Krishnendu Adhikary
- Department of Interdisciplinary Science, Centurion University of Technology and Management, Odisha, 761211, India
| | - Satyajit Mohanty
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India
| |
Collapse
|
6
|
Di Rocco G, Trivisonno A, Trivisonno G, Toietta G. Dissecting human adipose tissue heterogeneity using single-cell omics technologies. Stem Cell Res Ther 2024; 15:322. [PMID: 39334440 PMCID: PMC11437900 DOI: 10.1186/s13287-024-03931-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Single-cell omics technologies that profile genes (genomic and epigenomic) and determine the abundance of mRNA (transcriptomic), protein (proteomic and secretomic), lipids (lipidomic), and extracellular matrix (matrisomic) support the dissection of adipose tissue heterogeneity at unprecedented resolution in a temporally and spatially defined manner. In particular, cell omics technologies may provide innovative biomarkers for the identification of rare specific progenitor cell subpopulations, assess transcriptional and proteomic changes affecting cell proliferation and immunomodulatory potential, and accurately define the lineage hierarchy and differentiation status of progenitor cells. Unraveling adipose tissue complexity may also provide for the precise assessment of a dysfunctional state, which has been associated with cancer, as cancer-associated adipocytes play an important role in shaping the tumor microenvironment supporting tumor progression and metastasis, obesity, metabolic syndrome, and type 2 diabetes mellitus. The information collected by single-cell omics has relevant implications for regenerative medicine because adipose tissue is an accessible source of multipotent cells; alternative cell-free approaches, including the use of adipose tissue stromal cell-conditioned medium, extracellular vesicles, or decellularized extracellular matrix, are clinically valid options. Subcutaneous white adipose tissue, which is generally harvested via liposuction, is highly heterogeneous because of intrinsic biological variability and extrinsic inconsistencies in the harvesting and processing procedures. The current limited understanding of adipose tissue heterogeneity impinges on the definition of quality standards appropriate for clinical translation, which requires consistency and uniformity of the administered product. We review the methods used for dissecting adipose tissue heterogeneity and provide an overview of advances in omics technology that may contribute to the exploration of heterogeneity and dynamics of adipose tissue at the single-cell level.
Collapse
Affiliation(s)
- Giuliana Di Rocco
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Angelo Trivisonno
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
| | | | - Gabriele Toietta
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Via E. Chianesi, 53, 00144, Rome, Italy.
| |
Collapse
|
7
|
Duarte Afonso Serdan T, Cervantes H, Frank B, Tian Q, Choi CHJ, Hoffmann A, Cohen P, Blüher M, Schwartz GJ, Shamsi F. Slit3 Fragments Orchestrate Neurovascular Expansion and Thermogenesis in Brown Adipose Tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.613949. [PMID: 39386533 PMCID: PMC11463466 DOI: 10.1101/2024.09.24.613949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Brown adipose tissue (BAT) represents an evolutionary innovation enabling placental mammals to regulate body temperature through adaptive thermogenesis. Brown adipocytes are surrounded by a dense network of blood vessels and sympathetic nerves that support their development and thermogenic function. Cold exposure stimulates BAT thermogenesis through the coordinated induction of brown adipogenesis, angiogenesis, and sympathetic innervation. However, how these distinct processes are coordinated remains unclear. Here, we identify Slit guidance ligand 3 (Slit3) as a new niche factor that mediates the crosstalk among adipocyte progenitors, endothelial cells, and sympathetic nerves. We show that adipocyte progenitors secrete Slit3 which regulates both angiogenesis and sympathetic innervation in BAT and is essential for BAT thermogenesis in vivo. Proteolytic cleavage of Slit3 generates secreted Slit3-N and Slit3-C fragments, which activate distinct receptors to stimulate angiogenesis and sympathetic innervation, respectively. Moreover, we introduce bone morphogenetic protein-1 (Bmp1) as the first Slit protease identified in vertebrates. In summary, this study underscores the essential role of Slit3-mediated neurovascular network expansion in enabling cold-induced BAT adaptation. The co-regulation of neurovascular expansion by Slit3 fragments provides a bifurcated yet harmonized approach to ensure a synchronized response of BAT to environmental challenges. This study presents the first evidence that adipocyte progenitors regulate tissue innervation, revealing a previously unrecognized dimension of cellular interaction within adipose tissue.
Collapse
Affiliation(s)
| | - Heidi Cervantes
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA
| | - Benjamin Frank
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA
| | - Qiyu Tian
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA
| | - Chan Hee J Choi
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Anne Hoffmann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, 04103, Germany
| | - Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, 04103, Germany
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig 04103, Germany
| | - Gary J Schwartz
- Departments of Medicine and Neuroscience, Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Farnaz Shamsi
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA
- Departments of Cell Biology and Medicine, Grossman School of Medicine, New York University, New York, NY, USA
| |
Collapse
|
8
|
Zhu Q, Zhang P, Liu D, Tang L, Yu J, Zhang C, Jiang G. Glucosinolate extract from radish ( Raphanus sativus L.) seed attenuates high-fat diet-induced obesity: insights into gut microbiota and fecal metabolites. Front Nutr 2024; 11:1442535. [PMID: 39176030 PMCID: PMC11340518 DOI: 10.3389/fnut.2024.1442535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 07/22/2024] [Indexed: 08/24/2024] Open
Abstract
Background Radish seed is a functional food with many beneficial health effects. Glucosinolates are characteristic components in radish seed that can be transformed into bioactive isothiocyanates by gut microbiota. Objective The present study aims to assess anti-obesity efficacy of radish seed glucosinolates (RSGs) and explored the underlying mechanisms with a focus on gut microbiota and fecal metabolome. Methods High-fat diet-induced obese mice were supplemented with different doses of RSGs extract for 8 weeks. Changes in body weight, serum lipid, alanine aminotransferase (ALT), and aspartate aminotransferase (AST) levels; and pathological changes in the liver and adipose tissue were examined. Fecal metabolome and 16S rRNA gene sequencing were used to analyze alterations in fecal metabolite abundance and the gut microbiota, respectively. Results and conclusion Results showed that RSG extract prevented weight gain and decreased serum lipid, ALT, AST levels and lipid deposition in liver and epididymal adipocytes in obese mice. Treatment with RSG extract also increased gut microbiota diversity and altered the dominant bacteria genera in the gut microbiota, decreasing the abundance of Faecalibaculum and increasing the abundance of Allobaculum, Romboutsia, Turicibacter, and Akkermansia. Fecal metabolome results identified 570 differentially abundant metabolites, of which glucosinolate degradation products, such as sulforaphene and 7-methylsulfinylheptyl isothiocyanate, were significantly upregulated after RSG extract intervention. Furthermore, enrichment analysis of metabolic pathways showed that the anti-obesity effects of RSG extract may be mediated by alterations in bile secretion, fat digestion and absorption, and biosynthesis of plant secondary metabolites. Overall, RSG extract can inhibit the development of obesity, and the obesity-alleviating effects of RSG are related to alternative regulation of the gut microbiota and glucosinolate metabolites.
Collapse
Affiliation(s)
- Quanfeng Zhu
- Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Peng Zhang
- Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Daqun Liu
- Food Science Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Leilei Tang
- Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Jiawen Yu
- Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Chengcheng Zhang
- Food Science Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Guojun Jiang
- Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
9
|
Augustyniak K, Lesniak M, Latka H, Golan MP, Kubiak JZ, Zdanowski R, Malek K. Adipose-derived mesenchymal stem cells' adipogenesis chemistry analyzed by FTIR and Raman metrics. J Lipid Res 2024; 65:100573. [PMID: 38844049 PMCID: PMC11260339 DOI: 10.1016/j.jlr.2024.100573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/08/2024] [Accepted: 05/28/2024] [Indexed: 07/01/2024] Open
Abstract
The full understanding of molecular mechanisms of cell differentiation requires a holistic view. Here we combine label-free FTIR and Raman hyperspectral imaging with data mining to detect the molecular cell composition enabling noninvasive monitoring of cell differentiation and identifying biochemical heterogeneity. Mouse adipose-derived mesenchymal stem cells (AD-MSCs) undergoing adipogenesis were followed by Raman and FT-IR imaging, Oil Red, and immunofluorescence. A workflow of the data analysis (IRRSmetrics4stem) was designed to identify spectral predictors of adipogenesis and test machine-learning (ML) methods (hierarchical clustering, PCA, PLSR) for the control of the AD-MSCs differentiation degree. IRRSmetrics4stem provided insights into the chemism of adipogenesis. With single-cell tracking, we established IRRS metrics for lipids, proteins, and DNA variations during AD-MSCs differentiation. The over 90% predictive efficiency of the selected ML methods proved the high sensitivity of the IRRS metrics. Importantly, the IRRS metrics unequivocally recognize a switch from proliferation to differentiation. This study introduced a new bioassay identifying molecular markers indicating molecular transformations and delivering rapid and machine learning-based monitoring of adipogenesis that can be relevant to other differentiation processes. Thus, we introduce a novel, rapid, machine learning-based bioassay to identify molecular markers of adipogenesis. It can be relevant to identification of differentiation-related molecular processes in other cell types, and beyond the cell differentiation including progression of different cellular pathophysiologies reconstituted in vitro.
Collapse
Affiliation(s)
- Karolina Augustyniak
- Department of Chemical Physics, Faculty of Chemistry, Jagiellonian University in Krakow, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Krakow, Poland
| | - Monika Lesniak
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine - National Research Institute, Warszawa, Poland
| | - Hubert Latka
- Department of Chemical Physics, Faculty of Chemistry, Jagiellonian University in Krakow, Krakow, Poland
| | - Maciej P Golan
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine - National Research Institute, Warszawa, Poland; Institute of Psychology, The Maria Grzegorzewska University, Warsaw, Poland
| | - Jacek Z Kubiak
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine - National Research Institute, Warszawa, Poland; Dynamics and Mechanics of Epithelia Group, Institute of Genetics and Development of Rennes (IGDR), Faculty of Medicine, University of Rennes, CNRS, UMR 6290, Rennes, France.
| | - Robert Zdanowski
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine - National Research Institute, Warszawa, Poland.
| | - Kamilla Malek
- Department of Chemical Physics, Faculty of Chemistry, Jagiellonian University in Krakow, Krakow, Poland.
| |
Collapse
|
10
|
Pei J, Zou D, Li L, Kang L, Sun M, Li X, Chen Q, Chen D, Qu B, Gao X, Lin Z. Senp7 deficiency impairs lipid droplets maturation in white adipose tissues via Plin4 deSUMOylation. J Biol Chem 2024; 300:107319. [PMID: 38677512 PMCID: PMC11134554 DOI: 10.1016/j.jbc.2024.107319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/29/2024] Open
Abstract
Lipid metabolism is important for the maintenance of physiological homeostasis. Several members of the small ubiquitin-like modifier (SUMO)-specific protease (SENP) family have been reported as the regulators of lipid homeostasis. However, the function of Senp7 in lipid metabolism remains unclear. In this study, we generated both conventional and adipocyte-specific Senp7 KO mice to characterize the role of Senp7 in lipid metabolism homeostasis. Both Senp7-deficient mice displayed reduced white adipose tissue mass and decreased size of adipocytes. By analyzing the lipid droplet morphology, we demonstrated that the lipid droplet size was significantly smaller in Senp7-deficient adipocytes. Mechanistically, Senp7 could deSUMOylate the perilipin family protein Plin4 to promote the lipid droplet localization of Plin4. Our results reveal an important role of Senp7 in the maturation of lipid droplets via Plin4 deSUMOylation.
Collapse
Affiliation(s)
- Jingwen Pei
- State Key Laboratory of Pharmaceutical Biotechnology, Ministry of Education Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Dayuan Zou
- State Key Laboratory of Pharmaceutical Biotechnology, Ministry of Education Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Lu Li
- State Key Laboratory of Pharmaceutical Biotechnology, Ministry of Education Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Lulu Kang
- State Key Laboratory of Pharmaceutical Biotechnology, Ministry of Education Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Minli Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Ministry of Education Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Xu Li
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Qianyue Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Ministry of Education Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Danning Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Ministry of Education Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Bin Qu
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Xiang Gao
- State Key Laboratory of Pharmaceutical Biotechnology, Ministry of Education Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China.
| | - Zhaoyu Lin
- State Key Laboratory of Pharmaceutical Biotechnology, Ministry of Education Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China.
| |
Collapse
|
11
|
Gulnaz A, Lew LC, Park YH, Sabir JSM, Albiheyri R, Rather IA, Hor YY. Efficacy of Probiotic Strains Lactobacillus sakei Probio65 and Lactobacillus plantarum Probio-093 in Management of Obesity: An In Vitro and In Vivo Analysis. Pharmaceuticals (Basel) 2024; 17:676. [PMID: 38931347 PMCID: PMC11206994 DOI: 10.3390/ph17060676] [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: 02/05/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
The prevalence of obesity, characterized by an excessive accumulation of adipose tissue and adipocyte hypertrophy, presents a major public health challenge. This study investigates the therapeutic potential of two probiotic strains, Lactobacillus sakei Probio65 and Lactobacillus plantarum Probio-093, in the context of obesity. Utilizing 3T3-L1 cell-derived human adipocytes, we assessed Probio65's and Probio-093's capacity to mitigate triglyceride accumulation and influence adipocytokine production in vitro. Subsequently, an in vivo trial with male C57BL/6J mice examined the effects of both probiotic strains on adipose tissue characteristics, body weight, fat mass, and obesity-related gene expression. This study employed both live and ethanol-extracted bacterial cells. The results demonstrated significant reductions in the triglyceride deposition, body weight, and adipose tissue mass in the treated groups (p < 0.05). Furthermore, both strains modulated adipokine profiles by downregulating proinflammatory markers such as PAI-1, leptin, TNF-α, STAMP2, F4/80, resistin, and MCP-1, and upregulating the insulin-sensitive transporter GLUT4 and the anti-inflammatory adiponectin (p < 0.05). Our findings suggest that Lactobacillus sakei Probio65 and Lactobacillus plantarum Probio-093 are promising agents for microbiome-targeted anti-obesity therapies, offering the effective mitigation of obesity and improvement in adipocyte function in a murine model.
Collapse
Affiliation(s)
- Aneela Gulnaz
- Department of Biotechnology, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Gyeongbuk, Republic of Korea
| | - Lee-Ching Lew
- Probionic Corp., Jeonbuk Institute for Food-Bioindustry, 111-18, Wonjangdong-gil, Deokjin-gu, Jeonju-si 38541, Jeollabuk-do, Republic of Korea
| | - Yong-Ha Park
- Department of Biotechnology, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Gyeongbuk, Republic of Korea
- Probionic Corp., Jeonbuk Institute for Food-Bioindustry, 111-18, Wonjangdong-gil, Deokjin-gu, Jeonju-si 38541, Jeollabuk-do, Republic of Korea
| | - Jamal S. M. Sabir
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Raed Albiheyri
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Irfan A. Rather
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Yan-Yan Hor
- Department of Biotechnology, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Gyeongbuk, Republic of Korea
- Probionic Corp., Jeonbuk Institute for Food-Bioindustry, 111-18, Wonjangdong-gil, Deokjin-gu, Jeonju-si 38541, Jeollabuk-do, Republic of Korea
| |
Collapse
|
12
|
Chinnapaka S, Malekzadeh H, Tirmizi Z, Ejaz A. Caloric restriction mitigates age-associated senescence characteristics in subcutaneous adipose tissue-derived stem cells. Aging (Albany NY) 2024; 16:7535-7552. [PMID: 38728252 PMCID: PMC11131987 DOI: 10.18632/aging.205812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/27/2024] [Indexed: 05/12/2024]
Abstract
Adipose tissue regulates metabolic balance, but aging disrupts it, shifting fat from insulin-sensitive subcutaneous to insulin-resistant visceral depots, impacting overall metabolic health. Adipose-derived stem cells (ASCs) are crucial for tissue regeneration, but aging diminishes their stemness and regeneration potential. Our findings reveal that aging is associated with a decrease in subcutaneous adipose tissue mass and an increase in the visceral fat depots mass. Aging is associated with increase in adipose tissue fibrosis but no significant change in adipocyte size was observed with age. Long term caloric restriction failed to prevent fibrotic changes but resulted in significant decrease in adipocytes size. Aged subcutaneous ASCs displayed an increased production of ROS. Using mitochondrial membrane activity as an indicator of stem cell quiescence and senescence, we observed a significant decrease in quiescence ASCs with age exclusively in subcutaneous adipose depot. In addition, aged subcutaneous adipose tissue accumulated more senescent ASCs having defective autophagy activity. However, long-term caloric restriction leads to a reduction in mitochondrial activity in ASCs. Furthermore, caloric restriction prevents the accumulation of senescent cells and helps retain autophagy activity in aging ASCs. These results suggest that caloric restriction and caloric restriction mimetics hold promise as a potential strategy to rejuvenate the stemness of aged ASCs. Further investigations, including in vivo evaluations using controlled interventions in animals and human studies, will be necessary to validate these findings and establish the clinical potential of this well-established approach for enhancing the stemness of aged stem cells.
Collapse
Affiliation(s)
- Somaiah Chinnapaka
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Hamid Malekzadeh
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Zayaan Tirmizi
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Asim Ejaz
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
13
|
Waterstraat MG, Wang Z, Kogiso M, Caballero-Juarez R, Chen MH. Dissection, Histological Processing, and Gene Expression Analysis of Murine Supraclavicular Brown Adipose Tissue. J Vis Exp 2024:10.3791/66475. [PMID: 38619263 PMCID: PMC11614390 DOI: 10.3791/66475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024] Open
Abstract
Brown adipose tissue (BAT)-mediated thermogenesis plays an important role in the regulation of metabolism, and its morphology and function can be greatly impacted by environmental stimuli in mice and humans. Currently, murine interscapular BAT (iBAT), which is located between two scapulae in the upper dorsal flank of mice, is the main BAT depot used by research laboratories to study BAT function. Recently, a few previously unknown BAT depots were identified in mice, including one analogous to human supraclavicular brown adipose tissue. Unlike iBAT, murine supraclavicular brown adipose tissue (scBAT) is situated in the intermediate layer of the neck and thus cannot be accessed as readily. To facilitate the study of newly identified mouse scBAT, presented herein is a protocol detailing the steps to dissect intact scBAT from postnatal and adult mice. Due to scBAT's small size relative to other adipose depots, procedures have been modified and optimized specifically for processing scBAT. Among these modifications is the use of a dissecting microscope during tissue collection to increase the precision and homogenization of frozen scBAT samples to raise the efficiency of subsequent qPCR analysis. With these optimizations, the identification of, morphological appearance of, and molecular characterization of the scBAT can be determined in mice.
Collapse
Affiliation(s)
- Mark G Waterstraat
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine; Department of BioSciences, Rice University
| | - ZiYi Wang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine
| | - Mari Kogiso
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine
| | - Rommel Caballero-Juarez
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine; Department of BioSciences, Rice University
| | - Miao-Hsueh Chen
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine;
| |
Collapse
|
14
|
Klein Hazebroek M, Baars A, Mischke M, Oosting A, van Schothorst EM, Schipper L. Early-Life Exposure to Dietary Large Phospholipid-Coated Lipid Droplets Improves Markers of Metabolic and Immune Function in Adipose Tissue Later in Life in a Mouse Model. Mol Nutr Food Res 2024; 68:e2300470. [PMID: 37985953 DOI: 10.1002/mnfr.202300470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/11/2023] [Indexed: 11/22/2023]
Abstract
SCOPE Human milk (HM) is considered optimal nutrition for infants, beneficially programming adult health outcomes including reduced obesity risk. Early life exposure to infant formula with lipid droplets closely resembling the structural properties of HM lipid globules (Nuturis) attenuated white adipose tissue (WAT) accumulation in mice upon adult western-style diet (WSD) feeding. Here, the study aims to elucidate underlying mechanisms. METHODS AND RESULTS Mice are raised on control or Nuturis diets between postnatal days 16-42 followed by either standard diet or WSD for 16 weeks. While the adult body composition of mice on a standard diet is not significantly affected, Nuturis reduced adiposity in mice on WSD. Morphologically, mean adipocyte size is reduced in Nuturis-raised mice, independent of adult diet exposure, and WAT macrophage content is reduced, albeit not significantly. Transcriptomics of epididymal WAT indicate potential beneficial effects on energy metabolism and macrophage function by Nuturis. CONCLUSION Reduced adult adiposity by early life exposure to Nuturis appears to be associated with smaller adipocytes and alterations in WAT immune and energy metabolism. These results suggest that early modulation of WAT structure and/or function may contribute to the protective programming effects of the early-life Nuturis diet on later-life adiposity.
Collapse
Affiliation(s)
- Marlou Klein Hazebroek
- Danone Nutricia Research, Utrecht, 3584 CT, The Netherlands
- Human and Animal Physiology, Wageningen University, Wageningen, 6708 WD, The Netherlands
| | | | - Mona Mischke
- Danone Nutricia Research, Utrecht, 3584 CT, The Netherlands
| | | | - Evert M van Schothorst
- Human and Animal Physiology, Wageningen University, Wageningen, 6708 WD, The Netherlands
| | | |
Collapse
|
15
|
Ware TD, Butcher D, Edmondson E, Stern ST. Evaluation of Nanomedicine Tissue Distribution and Stability by Alexa Fluor 488 and PEG Immunohistochemistry. Methods Mol Biol 2024; 2789:313-322. [PMID: 38507013 DOI: 10.1007/978-1-0716-3786-9_28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
A primary issue with nanomedicine biological evaluation is determination of nanoparticle carrier tissue distribution and stability. Here we present a method to evaluate nanomedicine distribution in tissues that is applicable to most nanomedicine constructs. This method utilizes immunohistochemical (IHC) analysis of an Alexa Fluor 488-tag and/or polyethylene glycol (PEG), a very common nanomedicine component, for tissue localization. Using specific Alexa Fluor 488- and/or PEG antibody-based IHC staining procedures allows evaluation of high-resolution nanoparticle tissue distribution, nanoparticle tissue stability, and also allows correlation of distribution with morphological changes. This protocol outlines the methods to follow to ensure proper tissue collection and optimized immunohistochemical staining of Alexa Fluor 488-tag and PEG in tissues.
Collapse
Affiliation(s)
- Teagan D Ware
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Donna Butcher
- Molecular Histopathology Laboratory, Laboratory of Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Elijah Edmondson
- Molecular Histopathology Laboratory, Laboratory of Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Stephan T Stern
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
| |
Collapse
|
16
|
Park YJ, Kim HY, Shin S, Lee J, Heo I, Cha YY, An HJ. Anti-obesity effect of Lythri herba water extracts in vitro and in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116789. [PMID: 37328083 DOI: 10.1016/j.jep.2023.116789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/07/2023] [Accepted: 06/13/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lythrum salicaria L., also called purple loosestrife, has traditionally been used as a medicinal plant to treat internal dysfunction, such as gastrointestinal disorders or hemorrhages. It contains numerous phytochemical compounds, including orientin, and has been reported to have anti-diarrheal, anti-inflammatory, antioxidant, and antimicrobial properties. AIM OF THE STUDY The effects of Lythrum salicaria L. on obesity have not been explored. Therefore, we investigated the anti-obesity effects of Lythri Herba, the aerial part of this plant, in vitro and in vivo. MATERIALS AND METHODS Using distilled water, Lythri Herba water extracts (LHWE) were prepared by extracting Lythri Herba at 100°Ϲ. The contents of orientin in LHWE were identified using High Performance Liquid Chromatography (HPLC) analysis. To evaluate the anti-obesity effect of LHWE, 3T3-L1 adipocytes and a high-fat diet (HFD)-fed mice were used. Oil-red O staining was performed to examine the anti-adipogenic effects of LHWE in vitro. The histological changes in epididymal white adipose tissue (epiWAT) by LHWE were examined using hematoxylin and eosin staining. Serum leptin levels were measured by enzyme-linked immunosorbent assay. Specific quantification kits measured total cholesterol and triglyceride levels in the serum. The relative fold induction of protein and mRNA was determined using western blot and Quantitative real-time Polymerase Chain Reaction analysis, respectively. RESULTS HPLC analysis demonstrated the presence of orientin in LHWE. LHWE treatment markedly reduced lipid accumulation in differentiated 3T3-L1 adipocytes. LHWE administration also conferred resistance to HFD-induced weight gain in mice and reduced epiWAT mass. Mechanistically, LHWE significantly decreased lipogenesis by downregulating lipoprotein lipase (LPL), glucose-6-phosphate dehydrogenase, ATP-citrate lyase, fatty acid synthase, stearoyl-CoA desaturase 1, sterol regulatory element binding transcription factor 1, and carbohydrate response element binding protein expression and increased the expression of genes involved in fatty acid oxidation (FAO), peroxisome proliferator-activated receptor α and carnitine palmitoyltransferase 1 in 3T3-L1 adipocytes and epiWAT. Furthermore, LHWE significantly up-regulated the phosphorylation of AMP-activated protein kinase in 3T3-L1 adipocytes and epiWAT. CONCLUSION LHWE decreases white adipogenesis in vitro and HFD-induced weight gain in vivo, which is associated with reduced lipogenesis and enhanced FAO.
Collapse
Affiliation(s)
- Yea-Jin Park
- Department of Rehabilitative Medicine of Korean Medicine and Neuropsychiatry, College of Korean Medicine, Sangji University, Wonju, Gangwon-do, 26339, Republic of Korea; Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Hee-Young Kim
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Integrated Drug Development and Natural Products, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Su Shin
- Research Institute, BIO PORT KOREA INC., 36, Ballyongsandan 1-ro, Jangan-eup, Gijang-gun, Busan, 46034, Republic of Korea.
| | - JungHyun Lee
- Research Institute, BIO PORT KOREA INC., 36, Ballyongsandan 1-ro, Jangan-eup, Gijang-gun, Busan, 46034, Republic of Korea.
| | - In Heo
- School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea; Department of Rehabilitation Medicine of Korean Medicine, Pusan National University Korean Medicine Hospital, Yangsan, Gyeongsangnam-do, Republic of Korea.
| | - Yun-Yeop Cha
- Department of Rehabilitative Medicine of Korean Medicine and Neuropsychiatry, College of Korean Medicine, Sangji University, Wonju, Gangwon-do, 26339, Republic of Korea.
| | - Hyo-Jin An
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Integrated Drug Development and Natural Products, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
17
|
Li Y, Dong D, Qu Y, Li J, Chen S, Zhao H, Zhang Q, Jiao Y, Fan L, Sun D. A Multidrug Delivery Microrobot for the Synergistic Treatment of Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301889. [PMID: 37423966 DOI: 10.1002/smll.202301889] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 06/21/2023] [Indexed: 07/11/2023]
Abstract
Multidrug combination therapy provides an effective strategy for malignant tumor treatment. This paper presents the development of a biodegradable microrobot for on-demand multidrug delivery. By combining magnetic targeting transportation with tumor therapy, it is hypothesized that loading multiple drugs on different regions of a single magnetic microrobot can enhance a synergistic effect for cancer treatment. The synergistic effect of using two drugs together is greater than that of using each drug separately. Here, a 3D-printed microrobot inspired by the fish structure with three hydrogel components: skeleton, head, and body structures is demonstrated. Made of iron oxide (Fe3 O4 ) nanoparticles embedded in poly(ethylene glycol) diacrylate (PEGDA), the skeleton can respond to magnetic fields for microrobot actuation and drug-targeted delivery. The drug storage structures, head, and body, made by biodegradable gelatin methacryloyl (GelMA) exhibit enzyme-responsive cargo release. The multidrug delivery microrobots carrying acetylsalicylic acid (ASA) and doxorubicin (DOX) in drug storage structures, respectively, exhibit the excellent synergistic effects of ASA and DOX by accelerating HeLa cell apoptosis and inhibiting HeLa cell metastasis. In vivo studies indicate that the microrobots improve the efficiency of tumor inhibition and induce a response to anti-angiogenesis. The versatile multidrug delivery microrobot conceptualized here provides a way for developing effective combination therapy for cancer.
Collapse
Affiliation(s)
- Yanfang Li
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Dingran Dong
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Yun Qu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Junyang Li
- Center for Robotics and Automation, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518000, China
- Department of Electronic Engineering, Ocean University of China, Qingdao, 266000, China
| | - Shuxun Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Han Zhao
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Qi Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Yang Jiao
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Lei Fan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
- Center for Robotics and Automation, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518000, China
| | - Dong Sun
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
- Center for Robotics and Automation, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518000, China
| |
Collapse
|
18
|
Singh P, Ali SA. Mature white adipocyte plasticity during mammary gland remodelling and cancer. CELL INSIGHT 2023; 2:100123. [PMID: 37771567 PMCID: PMC10522874 DOI: 10.1016/j.cellin.2023.100123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 09/30/2023]
Abstract
Mammary gland growth and differentiation predominantly rely on stromal-epithelial cellular communication. Specifically, mammary adipocytes play a crucial role in ductal morphogenesis, as well as in the proliferation and differentiation of mammary epithelial cells. The process of lactation entails a reduction in the levels of white adipose tissue associated with the MG, allowing for the expansion of milk-producing epithelial cells. Subsequently, during involution and the regression of the milk-producing unit, adipocyte layers resurface, occupying the vacated space. This dynamic phenomenon underscores the remarkable plasticity and expansion of adipose tissue. Traditionally considered terminally differentiated, adipocytes have recently been found to exhibit plasticity in certain contexts. Unraveling the significance of this cell type within the MG could pave the way for novel approaches to reduce the risk of breast cancer and enhance lactation performance. Moreover, a comprehensive understanding of adipocyte trans- and de-differentiation processes holds promise for the development of innovative therapeutic interventions targeting cancer, fibrosis, obesity, type 2 diabetes, and other related diseases. Additionally, adipocytes may find utility in the realm of regenerative medicine. This review article provides a comprehensive examination of recent advancements in our understanding of MG remodelling, with a specific focus on the tissue-specific functions of adipocytes and their role in the development of cancer. By synthesizing current knowledge in this field, it aims to consolidate our understanding of adipocyte biology within the context of mammary gland biology, thereby fostering further research and discovery in this vital area.
Collapse
Affiliation(s)
- Parul Singh
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, 132001, India
- Division of Radiation Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, 132001, India
- Division Proteomics of Stem Cells and Cancer, German Cancer Research Center, 69120, Heidelberg, Germany
| |
Collapse
|
19
|
Chinnapaka S, Malekzadeh H, Tirmizi Z, Arellano JA, Ejaz A. Nicotinamide Riboside Improves Stemness of Human Adipose-Derived Stem Cells and Inhibits Terminal Adipocyte Differentiation. Pharmaceuticals (Basel) 2023; 16:1134. [PMID: 37631051 PMCID: PMC10458272 DOI: 10.3390/ph16081134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Adipose tissue plays a crucial role in maintaining metabolic homeostasis by serving as a storage site for excess fat and protecting other organs from the detrimental effects of lipotoxicity. However, the aging process is accompanied by a redistribution of fat, characterized by a decrease in insulin-sensitive subcutaneous adipose depot and an increase in insulin-resistant visceral adipose depot. This age-related alteration in adipose tissue distribution has implications for metabolic health. Adipose-derived stem cells (ASCs) play a vital role in the regeneration of adipose tissue. However, aging negatively impacts the stemness and regenerative potential of ASCs. The accumulation of oxidative stress and mitochondrial dysfunction-associated cellular damage contributes to the decline in stemness observed in aged ASCs. Nicotinamide adenine dinucleotide (NAD+) is a crucial metabolite that is involved in maintaining cellular homeostasis and stemness. The dysregulation of NAD+ levels with age has been associated with metabolic disorders and the loss of stemness. In this study, we aimed to investigate the effects of nicotinamide riboside (NR), a precursor of NAD+, on the stemness of human ASCs in cell culture. Our findings reveal that adipogenesis is accompanied by an increase in mitochondrial activity and the production of reactive oxygen species (ROS). However, treatment with NR leads to a reduction in mitochondrial activity and ROS production in ASCs. Furthermore, NR administration improves the stemness-related genes expression in ASCs and mitigates their propensity for adipocyte differentiation. These results suggest that NR treatment holds promise as a potential strategy to rejuvenate the stemness of aged ASCs. Further investigations, including in vivo evaluations using animal models and human studies, will be necessary to validate these findings and establish the clinical potential of this well-established drug for enhancing the stemness of aged stem cells.
Collapse
Affiliation(s)
| | | | | | | | - Asim Ejaz
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
20
|
Saavedra-Peña RDM, Taylor N, Flannery C, Rodeheffer MS. Estradiol cycling drives female obesogenic adipocyte hyperplasia. Cell Rep 2023; 42:112390. [PMID: 37053070 PMCID: PMC10567995 DOI: 10.1016/j.celrep.2023.112390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 12/21/2022] [Accepted: 03/29/2023] [Indexed: 04/14/2023] Open
Abstract
White adipose tissue (WAT) distribution is sex dependent. Adipocyte hyperplasia contributes to WAT distribution in mice driven by cues in the tissue microenvironment, with females displaying hyperplasia in subcutaneous and visceral WAT, while males and ovariectomized females have visceral WAT (VWAT)-specific hyperplasia. However, the mechanism underlying sex-specific hyperplasia remains elusive. Here, transcriptome analysis in female mice shows that high-fat diet (HFD) induces estrogen signaling in adipocyte precursor cells (APCs). Analysis of APCs throughout the estrous cycle demonstrates increased proliferation only when proestrus (high estrogen) coincides with the onset of HFD feeding. We further show that estrogen receptor α (ERα) is required for this proliferation and that estradiol treatment at the onset of HFD feeding is sufficient to drive it. This estrous influence on APC proliferation leads to increased obesity driven by adipocyte hyperplasia. These data indicate that estrogen drives ERα-dependent obesogenic adipocyte hyperplasia in females, exacerbating obesity and contributing to the differential fat distribution between the sexes.
Collapse
Affiliation(s)
- Rocío Del M Saavedra-Peña
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Natalia Taylor
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Clare Flannery
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University, New Haven, CT 06520, USA; Section of Endocrinology and Metabolism, Yale University, New Haven, CT 06520, USA
| | - Matthew S Rodeheffer
- Department of Comparative Medicine, Yale University, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA; Yale Center for Molecular and Systems Metabolism, Yale University, New Haven, CT 06520, USA; Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
21
|
Vohnoutka RB, Kuppa A, Hegde Y, Chen Y, Pant A, Tohme ME, (Karen) Choi EY, McCarty SM, Bagchi DP, Du X, Chen Y, Chen VL, Mori H, Bielak LF, Maguire LH, Handelman SK, Sexton JZ, Saunders TL, Halligan BD, Speliotes EK. Knockout of murine Lyplal1 confers sex-specific protection against diet-induced obesity. J Mol Endocrinol 2023; 70:e220131. [PMID: 36748836 PMCID: PMC10947332 DOI: 10.1530/jme-22-0131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/06/2023] [Indexed: 02/08/2023]
Abstract
Human genome-wide association studies found single-nucleotide polymorphisms (SNPs) near LYPLAL1 (Lysophospholipase-like protein 1) that have sex-specific effects on fat distribution and metabolic traits. To determine whether altering LYPLAL1 affects obesity and metabolic disease, we created and characterized a mouse knockout (KO) of Lyplal1. We fed the experimental group of mice a high-fat, high-sucrose (HFHS) diet for 23 weeks, and the controls were fed regular chow diet. Here, we show that CRISPR-Cas9 whole-body Lyplal1 KO mice fed an HFHS diet showed sex-specific differences in weight gain and fat accumulation as compared to chow diet. Female, not male, KO mice weighed less than WT mice, had reduced body fat percentage, had white fat mass, and had adipocyte diameter not accounted for by changes in the metabolic rate. Female, but not male, KO mice had increased serum triglycerides, decreased aspartate, and decreased alanine aminotransferase. Lyplal1 KO mice of both sexes have reduced liver triglycerides and steatosis. These diet-specific effects resemble the effects of SNPs near LYPLAL1 in humans, suggesting that LYPLAL1 has an evolutionary conserved sex-specific effect on adiposity. This murine model can be used to study this novel gene-by-sex-by-diet interaction to elucidate the metabolic effects of LYPLAL1 on human obesity.
Collapse
Affiliation(s)
- Rishel B. Vohnoutka
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, Ann Arbor, MI - 48109
- Abcam, Waltham, MA – 02453
| | - Annapurna Kuppa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, Ann Arbor, MI - 48109
| | - Yash Hegde
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, Ann Arbor, MI - 48109
- College of Human Medicine, Michigan State University, East Lansing, MI – 48824
| | - Yue Chen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, Ann Arbor, MI - 48109
| | - Asmita Pant
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, Ann Arbor, MI - 48109
| | - Maurice E. Tohme
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, Ann Arbor, MI - 48109
| | | | - Sean M. McCarty
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI - 48109
| | - Devika P. Bagchi
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI – 48019
| | - Xiaomeng Du
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, Ann Arbor, MI - 48109
| | - Yanhua Chen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, Ann Arbor, MI - 48109
| | - Vincent L. Chen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, Ann Arbor, MI - 48109
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI - 48109
| | - Hiroyuki Mori
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI – 48019
| | - Lawrence F. Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI – 48109
| | - Lillias H. Maguire
- Hospital of the University of Pennsylvania, Philadelphia, PA – 19104
- Corporal Michael Crescenz VAMC, Philadelphia PA – 19104-4551
| | - Samuel K. Handelman
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, Ann Arbor, MI - 48109
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI - 48109
| | - Jonathan Z. Sexton
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI - 48109
| | - Thomas L. Saunders
- University of Michigan Transgenic Animal Model Core, Biomedical Research Core Facilities, Ann Arbor, MI - 48109
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI – 48109
| | - Brian D. Halligan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, Ann Arbor, MI - 48109
| | - Elizabeth K. Speliotes
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, Ann Arbor, MI - 48109
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI - 48109
| |
Collapse
|
22
|
Kotzé-Hörstmann LM, Bedada DT, Johnson R, Mabasa L, Sadie-Van Gijsen H. The effects of a green Rooibos ( Aspalathus linearis) extract on metabolic parameters and adipose tissue biology in rats fed different obesogenic diets. Food Funct 2022; 13:12648-12663. [PMID: 36441182 DOI: 10.1039/d2fo02440c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Current pharmaceutical treatments addressing obesity are plagued by high costs, low efficacy and adverse side effects. Natural extracts are popular alternatives, but evidence for their anti-obesity properties is scant. We assessed the efficacy of a green (minimally-oxidized) Rooibos (Aspalathus linearis) extract (GRT) to ameliorate the effects of obesogenic feeding in rats, by examining body weight, metabolic measures, adipose tissue cellularity and tissue-resident adipose stem cells (ASCs). Furthermore, we performed statistical correlations to explore the relationships and interactions between metabolic and adipose tissue measures. Using an in vivo/ex vivo study design, male Wistar rats were maintained for 17 weeks on one of 3 diets: CON (laboratory chow), OB1 (high-sugar, medium fat) or OB2 (high-fat, high-cholesterol) (n = 24 each). From weeks 11-17, half of the animals in each group received oral GRT supplementation (60 mg per kg body weight daily). Blood and tissue samples were collected, and ASCs from each animal were cultured. Diets OB1 and OB2 induced divergent metabolic profiles compared to CON, but metabolic measures within dietary groups were mostly unaffected by GRT supplementation. Notably, diets OB1 and OB2 uncoupled the positive association between visceral adiposity and insulin resistance, while GRT uncoupled the positive association between elevated serum cholesterol and liver damage. Obesogenic feeding and GRT supplementation induced adipocyte enlargement in vivo, but lipid accumulation in cultured ASCs did not differ between dietary groups. Larger adipocyte size in subcutaneous fat was associated with favourable glucose metabolism measures in all GRT groups. In conclusion, GRT affected the associations between systemic, adipose tissue-level and cellular measures against the background of obesogenic diet-induced metabolic dysregulation.
Collapse
Affiliation(s)
- L M Kotzé-Hörstmann
- Centre for Cardio-metabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, PO Box 241, Cape Town 8000, South Africa. .,Institute for Sport and Exercise Medicine (ISEM), Department of Sport Science, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, PO Box 241, Cape Town 8000, South Africa
| | - D T Bedada
- Division of Epidemiology and Biostatistics, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, PO Box 241, Cape Town 8000, South Africa
| | - R Johnson
- Centre for Cardio-metabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, PO Box 241, Cape Town 8000, South Africa. .,Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), PO Box 19070, Parow 7505, South Africa
| | - L Mabasa
- Centre for Cardio-metabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, PO Box 241, Cape Town 8000, South Africa. .,Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), PO Box 19070, Parow 7505, South Africa
| | - H Sadie-Van Gijsen
- Centre for Cardio-metabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, PO Box 241, Cape Town 8000, South Africa.
| |
Collapse
|
23
|
Chen M, Zhang F, Chen B, Lau C, Xu K, Tong T, Huo C, Han Q, Su T, Kwan HY. Omics approach to reveal the effects of obesity on the protein profiles of the exosomes derived from different adipose depots. Cell Mol Life Sci 2022; 79:570. [PMID: 36306016 PMCID: PMC11803032 DOI: 10.1007/s00018-022-04597-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/24/2022] [Accepted: 10/07/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Obesity affects the cargo packaging of the adipocyte-derived exosomes. Furthermore, adipocytes in different adipose tissues have different genetic makeup, the cargo contents of the exosomes derived from different adipose tissues under obesity conditions should be different, and hence their impacts on the pathophysiological conditions. METHODS AND RESULTS iTRAQ-based quantitative proteomics show that obesity has more prominent effects on the protein profiles of the exosomes derived from subcutaneous adipose tissue (SAT-Exos) in the high fat diet-induced obesity (DIO) mice than those derived from epididymal adipose tissue (EAT-Exos) and visceral adipose tissue (VAT-Exos). The differentially expressed proteins (DEPs) in SAT-Exos and VAT-Exos are mainly involved in metabolism. Subsequent untargeted metabolomic and lipidomics analyses reveal that injection of these SAT-Exos into the B6/J-Rab27a-Cas9-KO mice significantly affects the mouse metabolism such as fatty acid metabolism. Some of the DEPs in SAT-Exos are correlated with fatty acid metabolism including ADP-ribosylation factor and mitogen-activated protein kinase kinase kinase-3. Pathway analysis also shows that SAT-Exos affect adipocyte lipolysis and glycerophospholipid metabolism, which is in parallel with the enhanced plasma levels of fatty acids, diglycerides, monoglycerides and the changes in glycerophospholipid levels in DIO mice. CONCLUSION Our data provide scientific evidence to suggest SAT-Exos contribute to the changes in plasma lipid profiles under obesity conditions.
Collapse
Affiliation(s)
- Minting Chen
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Fan Zhang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Baisen Chen
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Condon Lau
- Department of Physics, City University of Hong Kong, Hong Kong, China
| | - Keyang Xu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Tiejun Tong
- Department of Mathematics, Hong Kong Baptist University, Hong Kong, China
| | - Chuying Huo
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Quanbin Han
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Tao Su
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Hiu Yee Kwan
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, China.
| |
Collapse
|
24
|
Sieckmann K, Winnerling N, Huebecker M, Leyendecker P, Ribeiro D, Gnad T, Pfeifer A, Wachten D, Hansen JN. AdipoQ - a simple, open-source software to quantify adipocyte morphology and function in tissues and in vitro. Mol Biol Cell 2022; 33:br22. [PMID: 35947507 DOI: 10.1091/mbc.e21-11-0592] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The different adipose tissues can be distinguished according to their function. For example, white adipose tissue (WAT) stores energy in form of lipids, whereas brown adipose tissue (BAT) dissipates energy in the form of heat. These functional differences are represented in the respective adipocyte morphology: whereas white adipocytes contain large, unilocular lipid droplets, brown adipocytes contain smaller, multilocular lipid droplets. However, an automated, image-analysis pipeline to comprehensively analyze adipocytes in vitro in cell culture as well as ex vivo in tissue sections is missing. We here present AdipoQ, an open-source software implemented as ImageJ plugins that allows to analyze adipocytes in tissue sections and in vitro after histological and/or immunofluorescent labelling. AdipoQ is compatible with different imaging modalities and staining methods, allows batch processing of large datasets and simple post-hoc analysis, provides a broad band of parameters, and allows combining multiple fluorescent read-outs. Thereby, AdipoQ is of immediate use not only for basic research but also for clinical diagnosis.
Collapse
Affiliation(s)
- Katharina Sieckmann
- Institute of Innate Immunity, Biophysical Imaging, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Nora Winnerling
- Institute of Innate Immunity, Biophysical Imaging, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Mylene Huebecker
- Institute of Innate Immunity, Biophysical Imaging, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Philipp Leyendecker
- Institute of Innate Immunity, Biophysical Imaging, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Dalila Ribeiro
- Institute of Innate Immunity, Biophysical Imaging, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Thorsten Gnad
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Dagmar Wachten
- Institute of Innate Immunity, Biophysical Imaging, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Jan N Hansen
- Institute of Innate Immunity, Biophysical Imaging, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| |
Collapse
|
25
|
Herrera-García A, Pérez-Mendoza M, Arellanes-Licea EDC, Gasca-Martínez D, Carmona-Castro A, Díaz-Muñoz M, Miranda-Anaya M. Obesity in male volcano mice Neotomodon alstoni affects the daily rhythm of metabolism and thermoregulation. Front Nutr 2022; 9:963804. [PMID: 35990356 PMCID: PMC9386375 DOI: 10.3389/fnut.2022.963804] [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: 06/07/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
The mouse N. alstoni spontaneously develops the condition of obesity in captivity when fed regular chow. We aim to study the differences in metabolic performance and thermoregulation between adult lean and obese male mice. The experimental approach included indirect calorimetry using metabolic cages for VO2 intake and VCO2 production. In contrast, the body temperature was measured and analyzed using intraperitoneal data loggers. It was correlated with the relative presence of UCP1 protein and its gene expression from interscapular adipose tissue (iBAT). We also explored in this tissue the relative presence of Tyrosine Hydroxylase (TH) protein, the rate-limiting enzyme for catecholamine biosynthesis present in iBAT. Results indicate that obese mice show a daily rhythm persists in estimated parameters but with differences in amplitude and profile. Obese mice presented lower body temperature, and a low caloric expenditure, together with lower VO2 intake and VCO2 than lean mice. Also, obese mice present a reduced thermoregulatory response after a cold pulse. Results are correlated with a low relative presence of TH and UCP1 protein. However, qPCR analysis of Ucp1 presents an increase in gene expression in iBAT. Histology showed a reduced amount of brown adipocytes in BAT. The aforementioned indicates that the daily rhythm in aerobic metabolism, thermoregulation, and body temperature control have reduced amplitude in obese mice Neotomodon alstoni.
Collapse
Affiliation(s)
- Andrea Herrera-García
- Unidad Multidisciplinaria de Docencia e Investigación, Facultad de Ciencias, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, Mexico
- Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Juriquilla, Querétaro, Mexico
| | - Moisés Pérez-Mendoza
- Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Juriquilla, Querétaro, Mexico
| | - Elvira del Carmen Arellanes-Licea
- Unidad Multidisciplinaria de Docencia e Investigación, Facultad de Ciencias, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, Mexico
| | - Deisy Gasca-Martínez
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, Mexico
| | - Agustín Carmona-Castro
- Unidad Multidisciplinaria de Docencia e Investigación, Facultad de Ciencias, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, Mexico
| | - Mauricio Díaz-Muñoz
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, Mexico
| | - Manuel Miranda-Anaya
- Unidad Multidisciplinaria de Docencia e Investigación, Facultad de Ciencias, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, Mexico
| |
Collapse
|
26
|
Mekbib T, Suen TC, Rollins-Hairston A, Smith K, Armstrong A, Gray C, Owino S, Baba K, Baggs JE, Ehlen JC, Tosini G, DeBruyne JP. "The ubiquitin ligase SIAH2 is a female-specific regulator of circadian rhythms and metabolism". PLoS Genet 2022; 18:e1010305. [PMID: 35789210 PMCID: PMC9286287 DOI: 10.1371/journal.pgen.1010305] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/15/2022] [Accepted: 06/22/2022] [Indexed: 01/05/2023] Open
Abstract
Circadian clocks enable organisms to predict and align their behaviors and physiologies to constant daily day-night environmental cycle. Because the ubiquitin ligase Siah2 has been identified as a potential regulator of circadian clock function in cultured cells, we have used SIAH2-deficient mice to examine its function in vivo. Our experiments demonstrate a striking and unexpected sexually dimorphic effect of SIAH2-deficiency on the regulation of rhythmically expressed genes in the liver. The absence of SIAH2 in females, but not in males, altered the expression of core circadian clock genes and drastically remodeled the rhythmic transcriptome in the liver by increasing the number of day-time expressed genes, and flipping the rhythmic expression from nighttime expressed genes to the daytime. These effects are not readily explained by effects on known sexually dimorphic pathways in females. Moreover, loss of SIAH2 in females, not males, preferentially altered the expression of transcription factors and genes involved in regulating lipid and lipoprotein metabolism. Consequently, SIAH2-deficient females, but not males, displayed disrupted daily lipid and lipoprotein patterns, increased adiposity and impaired metabolic homeostasis. Overall, these data suggest that SIAH2 may be a key component of a female-specific circadian transcriptional output circuit that directs the circadian timing of gene expression to regulate physiological rhythms, at least in the liver. In turn, our findings imply that sex-specific transcriptional mechanisms may closely interact with the circadian clock to tailor overt rhythms for sex-specific needs.
Collapse
Affiliation(s)
- Tsedey Mekbib
- Neuroscience Institute, Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Ting-Chung Suen
- Neuroscience Institute, Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Aisha Rollins-Hairston
- Neuroscience Institute, Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Kiandra Smith
- Neuroscience Institute, Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Ariel Armstrong
- Neuroscience Institute, Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Cloe Gray
- Neuroscience Institute, Department of Neurobiology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Sharon Owino
- Neuroscience Institute, Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Kenkichi Baba
- Neuroscience Institute, Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Julie E. Baggs
- Neuroscience Institute, Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - J. Christopher Ehlen
- Neuroscience Institute, Department of Neurobiology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Gianluca Tosini
- Neuroscience Institute, Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Jason P. DeBruyne
- Neuroscience Institute, Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| |
Collapse
|
27
|
Effects of starch-rich or fat-rich diets on metabolism, adiposity, and glycemia in immune-biased, C57BL/6 and BALB/c mice. J Nutr Biochem 2022; 108:109086. [PMID: 35691592 DOI: 10.1016/j.jnutbio.2022.109086] [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: 08/14/2021] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 11/21/2022]
Abstract
Diet is an essential factor to maintain health by regulating host metabolism and immunity. Host immunity acts as a critical regulator of metabolic changes. By using differentially immune-biased mice C57BL/6 and BALB/c, we demonstrated the metabolic consequence of consuming diets rich in non-resistant starch (starch-rich), unsaturated fat (sunflower oil-rich), and saturated fat (coconut oil-rich) for shorter (four weeks) or longer (eight weeks) duration. Time kinetics of various diets on two differentially immune-biased mice revealed that starch-rich and unsaturated fat-rich diets reduced insulin resistance (IR) and visceral adiposity in BALB/c mice. In contrast, a saturated fat-rich diet enhanced both parameters. In C57BL/6 mice, a fat-rich diet enhanced IR with time while visceral adiposity remained unchanged. Eight weeks' consumption of a saturated fat-rich diet led to the highest visceral adiposity in C57BL/6 mice, while the same diet resulted in the maximum IR in BALB/c mice. The current report presented a detailed metabolomic analysis of treatments with various diets using a) uni- and b) multi-variate analyses. We also calculated the differential index for each treatment for each mouse strain using a vector analysis of the multivariate linear discriminant data. The outcome of the vector analysis of metabolite profiles identified metabolites that affected lipid and glucose metabolism to establish the inter-strain physiological differences.
Collapse
|
28
|
Bel JS, Tai TC, Khaper N, Lees SJ. Chronic glucocorticoid exposure causes brown adipose tissue whitening, alters whole-body glucose metabolism and increases tissue uncoupling protein-1. Physiol Rep 2022; 10:e15292. [PMID: 35510321 PMCID: PMC9069169 DOI: 10.14814/phy2.15292] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/05/2022] [Accepted: 04/11/2022] [Indexed: 04/14/2023] Open
Abstract
Adipose tissue (AT) has been found to exist in two predominant forms, white and brown. White adipose tissue (WAT) is the body's conventional storage organ, and brown adipose tissue (BAT) is responsible for non-shivering thermogenesis which allows mammals to produce heat and regulate body temperature. Studies examining BAT and its role in whole-body metabolism have found that active BAT utilizes glucose and circulating fatty acids and is associated with improved metabolic outcomes. While the beiging of WAT is a growing area of interest, the possibility of the BAT depot to "whiten" and store more triglycerides also has metabolic and health implications. Currently, there are limited studies that examine the effects of chronic stress and its ability to induce a white-like phenotype in the BAT depot. This research examined how chronic exposure to the murine stress hormone, corticosterone, for 4 weeks can affect the whitening process of BAT in C57BL/6 male mice. Separate treatments with mirabegron, a known β3-adrenergic receptor agonist, were used to directly compare the effects of corticosterone with a beiging phenotype. Corticosterone-treated mice had significantly higher body weight (p ≤ 0.05) and BAT mass (p ≤ 0.05), increased adipocyte area (p ≤ 0.05), were insulin resistant (p ≤ 0.05), and significantly elevated expressions of uncoupling protein 1 (UCP-1) in BAT (p ≤ 0.05) while mitochondrial content remained unchanged. This whitened phenotype has not been previously associated with increased uncoupling proteins under chronic stress and may represent a compensatory mechanism being initiated under these conditions. These findings have implications for the study of BAT in response to chronic glucocorticoid exposure potentially leading to BAT dysfunction and negative impacts on whole-body glucose metabolism.
Collapse
Affiliation(s)
- Jocelyn S. Bel
- Biotechnology ProgramLakehead UniversityThunder BayOntarioCanada
| | - T. C. Tai
- Northern Ontario School of MedicineThunder BayOntarioCanada
- BiologyLaurentian UniversitySudburyOntarioCanada
- Chemistry and BiochemistryLaurentian UniversitySudburyOntarioCanada
- Biomolecular Sciences ProgramLaurentian UniversitySudburyOntarioCanada
| | - Neelam Khaper
- Northern Ontario School of MedicineThunder BayOntarioCanada
- Biomolecular Sciences ProgramLaurentian UniversitySudburyOntarioCanada
- BiologyLakehead UniversityThunder BayOntarioCanada
| | - Simon J. Lees
- Northern Ontario School of MedicineThunder BayOntarioCanada
- BiologyLakehead UniversityThunder BayOntarioCanada
| |
Collapse
|
29
|
Wang Q, Liu Y, Xu Y, Jin Y, Wu J, Ren Z. Comparative transcriptome and Lipidome analyses suggest a lipid droplet-specific response to heat exposure of brown adipose tissue in normal and obese mice. Life Sci 2022; 299:120540. [PMID: 35398332 DOI: 10.1016/j.lfs.2022.120540] [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: 03/06/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 10/18/2022]
Abstract
AIMS In mammals, heat stress (HS) from high-temperature environments has multiple adverse effects on the well-being of the organism. Brown adipose tissue (BAT) is a thermogenesis tissue that protects against obesity, and as an endocrine organ that regulates the systemic metabolism, but it is unclear how heat stress affects BAT in normal and obese subjects. Understanding the transcriptomic profiles and lipidomics of BAT upon heat exposure provides insights into the adaptive changes associated with this process. MATERIALS AND METHODS We constructed heat treatment (40 °C, 4 h) models for normal and obese mice, observed the effect of heat treatment on interscapular BAT (iBAT) and performed an assay for iBAT with RNA-seq and lipidomics to compare transcriptional programs and lipid dynamics. KEY FINDINGS In normal mice, heat treatment caused an iBAT damage by decreasing the expression of genes involved in thermogenesis, adipogenesis and lipid metabolism. Furthermore, HS disturbed the acyl-chain composition of triacylglycerols (TAGs) and glycerophospholipids (PEs, PCs and CLs), accelerated the production of cholesterol esters, and caused the formation of giant lipid droplets rich in cholesterol esters in iBAT. Unexpectedly, in obese mice, heat treatment had a smaller effect on iBAT by improving the composition of the saturated glycerolipids, PEs and PCs and increasing the proportion of oxidized lipid in lipid droplets. SIGNIFICANCE Our findings proved lipid droplets participated in the regulation of lipid components of iBAT in normal and obese mice after heat treatment, which provided a new view for the understanding of the adaptation of iBAT to high-temperature environments.
Collapse
Affiliation(s)
- Qiankun Wang
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, College of Animal Science, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Yue Liu
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, College of Animal Science, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Yue Xu
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, College of Animal Science, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Yi Jin
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, College of Animal Science, Huazhong Agricultural University, Wuhan 430070, Hubei, China; Hubei Hongshan Laboratory, Hubei Province, PR China
| | - Jian Wu
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, College of Animal Science, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Zhuqing Ren
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, College of Animal Science, Huazhong Agricultural University, Wuhan 430070, Hubei, China; Hubei Hongshan Laboratory, Hubei Province, PR China.
| |
Collapse
|
30
|
Yang Q, Hinkle J, Reed JN, Aherrahrou R, Xu Z, Harris TE, Stephenson EJ, Musunuru K, Keller SR, Civelek M. Adipocyte-Specific Modulation of KLF14 Expression in Mice Leads to Sex-Dependent Impacts on Adiposity and Lipid Metabolism. Diabetes 2022; 71:677-693. [PMID: 35081256 PMCID: PMC8965685 DOI: 10.2337/db21-0674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022]
Abstract
Genome-wide association studies identified single nucleotide polymorphisms on chromosome 7 upstream of KLF14 to be associated with metabolic syndrome traits and increased risk for type 2 diabetes (T2D). The associations were more significant in women than in men. The risk allele carriers expressed lower levels of the transcription factor KLF14 in adipose tissues than nonrisk allele carriers. To investigate how adipocyte KLF14 regulates metabolic traits in a sex-dependent manner, we characterized high-fat diet-fed male and female mice with adipocyte-specific Klf14 deletion or overexpression. Klf14 deletion resulted in increased fat mass in female mice and decreased fat mass in male mice. Female Klf14-deficient mice had overall smaller adipocytes in subcutaneous fat depots but larger adipocytes in parametrial depots, indicating a shift in lipid storage from subcutaneous to visceral fat depots. They had reduced metabolic rates and increased respiratory exchange ratios consistent with increased use of carbohydrates as an energy source. Fasting- and isoproterenol-induced adipocyte lipolysis was defective in female Klf14-deficient mice, and concomitantly, adipocyte triglycerides lipase mRNA levels were downregulated. Female Klf14-deficient mice cleared blood triglyceride and nonesterified fatty acid less efficiently than wild-type. Finally, adipocyte-specific overexpression of Klf14 resulted in lower total body fat in female but not male mice. Taken together, consistent with human studies, adipocyte KLF14 deficiency in female but not in male mice causes increased adiposity and redistribution of lipid storage from subcutaneous to visceral adipose tissues. Increasing KLF14 abundance in adipocytes of females with obesity and T2D may provide a novel treatment option to alleviate metabolic abnormalities.
Collapse
Affiliation(s)
- Qianyi Yang
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA
- Corresponding authors: Qianyi Yang, , and Mete Civelek,
| | - Jameson Hinkle
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA
| | - Jordan N. Reed
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA
| | - Redouane Aherrahrou
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA
| | - Zhiwen Xu
- Department of Chemistry, College of Arts and Sciences, University of Virginia, Charlottesville, VA
| | - Thurl E. Harris
- Department of Pharmacology, School of Medicine, University of Virginia, Charlottesville, VA
| | - Erin J. Stephenson
- Department of Anatomy, College of Graduate Studies & Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL
| | - Kiran Musunuru
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Division of Cardiovascular Medicine, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Susanna R. Keller
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Virginia, Charlottesville, VA
| | - Mete Civelek
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA
- Corresponding authors: Qianyi Yang, , and Mete Civelek,
| |
Collapse
|
31
|
Perdikari A, Cacciottolo T, Henning E, Mendes de Oliveira E, Keogh JM, Farooqi IS. Visualization of sympathetic neural innervation in human white adipose tissue. Open Biol 2022; 12:210345. [PMID: 35291877 PMCID: PMC8924755 DOI: 10.1098/rsob.210345] [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] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/08/2022] [Indexed: 01/09/2023] Open
Abstract
Obesity, defined as an excess of adipose tissue that adversely affects health, is a major cause of morbidity and mortality. However, to date, understanding the structure and function of human adipose tissue has been limited by the inability to visualize cellular components due to the innate structure of adipocytes, which are characterized by large lipid droplets. Combining the iDISCO and the CUBIC protocols for whole tissue staining and optical clearing, we developed a protocol to enable immunostaining and clearing of human subcutaneous white adipose tissue (WAT) obtained from individuals with severe obesity. We were able to perform immunolabelling of sympathetic nerve terminals in whole WAT and subsequent optical clearing by eliminating lipids to render the opaque tissue completely transparent. We then used light sheet confocal microscopy to visualize sympathetic innervation of human WAT from obese individuals in a three-dimensional manner. We demonstrate the visualization of sympathetic nerve terminals in human WAT. This protocol can be modified to visualize other structures such as blood vessels involved in the development, maintenance and function of human adipose tissue in health and disease.
Collapse
Affiliation(s)
- Aliki Perdikari
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Tessa Cacciottolo
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Elana Henning
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Edson Mendes de Oliveira
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Julia M. Keogh
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - I. Sadaf Farooqi
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
32
|
Maqdasy S, Lecoutre S, Renzi G, Frendo-Cumbo S, Rizo-Roca D, Moritz T, Juvany M, Hodek O, Gao H, Couchet M, Witting M, Kerr A, Bergo MO, Choudhury RP, Aouadi M, Zierath JR, Krook A, Mejhert N, Rydén M. Impaired phosphocreatine metabolism in white adipocytes promotes inflammation. Nat Metab 2022; 4:190-202. [PMID: 35165448 PMCID: PMC8885409 DOI: 10.1038/s42255-022-00525-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 01/05/2022] [Indexed: 02/07/2023]
Abstract
The mechanisms promoting disturbed white adipocyte function in obesity remain largely unclear. Herein, we integrate white adipose tissue (WAT) metabolomic and transcriptomic data from clinical cohorts and find that the WAT phosphocreatine/creatine ratio is increased and creatine kinase-B expression and activity is decreased in the obese state. In human in vitro and murine in vivo models, we demonstrate that decreased phosphocreatine metabolism in white adipocytes alters adenosine monophosphate-activated protein kinase activity via effects on adenosine triphosphate/adenosine diphosphate levels, independently of WAT beigeing. This disturbance promotes a pro-inflammatory profile characterized, in part, by increased chemokine (C-C motif) ligand 2 (CCL2) production. These data suggest that the phosphocreatine/creatine system links cellular energy shuttling with pro-inflammatory responses in human and murine white adipocytes. Our findings provide unexpected perspectives on the mechanisms driving WAT inflammation in obesity and may present avenues to target adipocyte dysfunction.
Collapse
Grants
- SM was supported by the Université Clermont Auvergne, Société Francophone du Diabète and Fondation Bettencourt Schueller.
- S.F.C. is supported by a Novo Nordisk postdoctoral fellowship run in partnership with Karolinska Institutet.
- the NovoNordisk Foundation (NNF20OC0061149), CIMED, Swedish Research Council.
- Knut och Alice Wallenbergs Stiftelse (Knut and Alice Wallenberg Foundation)
- Margareta af Uggla’s foundation, the Swedish Research Council, ERC-SyG SPHERES (856404 to M.R.), the NovoNordisk Foundation (including the Tripartite Immuno-metabolism Consortium Grant Number NNF15CC0018486, the MSAM consortium NNF15SA0018346 and the MeRIAD consortium Grant number 0064142), Knut and Alice Wallenbergs Foundation, CIMED, the Swedish Diabetes Foundation, the Stockholm County Council and the Strategic Research Program in Diabetes at Karolinska Institutet.
Collapse
Affiliation(s)
- Salwan Maqdasy
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
- CHU Clermont-Ferrand, Service d'endocrinologie, diabétologie et maladies métaboliques, Clermont-Ferrand, France
- Laboratoire GReD, Université Clermont Auvergne, Faculté de Médecine, Clermont Ferrand, France
| | - Simon Lecoutre
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Gianluca Renzi
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Scott Frendo-Cumbo
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - David Rizo-Roca
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Moritz
- Swedish Metabolomics Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
- The NovoNordisk Foundation Centre for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marta Juvany
- Swedish Metabolomics Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Ondrej Hodek
- Swedish Metabolomics Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Hui Gao
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Morgane Couchet
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Michael Witting
- Metabolomics and proteomics core (MPC), Helmholtz Zentrum München, Neuherberg, Germany
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Analytical Food Chemistry, TUM School of Life Sciences, Freising, Germany
| | - Alastair Kerr
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Martin O Bergo
- Department of Biosciences and Nutrition, Karolinska Comprehensive Cancer Center, Karolinska Institutet, Huddinge, Sweden
| | | | - Myriam Aouadi
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Juleen R Zierath
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Anna Krook
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Niklas Mejhert
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden.
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden.
| |
Collapse
|
33
|
Hinz A, Szczęch M, Szczepanowicz K, Bzowska M. Fluorophore Localization Determines the Results of Biodistribution of Core-Shell Nanocarriers. Int J Nanomedicine 2022; 17:577-588. [PMID: 35173431 PMCID: PMC8840834 DOI: 10.2147/ijn.s343266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/16/2021] [Indexed: 11/23/2022] Open
Abstract
Introduction Methods Results Conclusion
Collapse
Affiliation(s)
- Alicja Hinz
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Marta Szczęch
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, Kraków, Poland
| | - Krzysztof Szczepanowicz
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, Kraków, Poland
- Krzysztof Szczepanowicz, Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, Niezapominajek 8 Street, Kraków, 30-239, Poland, Tel/Fax +48 12 639 51 21, Email
| | - Monika Bzowska
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
- Correspondence: Monika Bzowska, Department of Cell Biochemistry, Faculty of Biochemistry, Jagiellonian University in Kraków, Biophysics and Biotechnology, 7 Gronostajowa Street, Kraków, 30-387, Poland, Tel/Fax +48 12 664 63 88, Email
| |
Collapse
|
34
|
Lynes MD, Carlone DL, Townsend KL, Breault DT, Tseng YH. Telomerase Reverse Transcriptase Expression Marks a Population of Rare Adipose Tissue Stem Cells. Stem Cells 2022; 40:102-111. [PMID: 35511869 PMCID: PMC9199842 DOI: 10.1093/stmcls/sxab005] [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: 04/08/2021] [Accepted: 09/17/2021] [Indexed: 11/12/2022]
Abstract
In adult tissues such as adipose tissue, post-mitotic cells like adipocytes can be replaced by differentiation of a population of tissue-resident stem cells. Expression of mouse telomerase reverse transcriptase (mTert) is a hallmark of stem cell populations, and previous efforts to identify tissue-resident adult stem cells by measuring mTert expression have increased our understanding of stem cell biology significantly. Here, we used a doxycycline-inducible mouse model to perform longitudinal, live-animal lineage-tracing of mTert-expressing cells for more than 1 year. We identified a rare (<2%) population of stem cells in different fat depots that express putative preadipocyte markers. The adipose-derived mTert-positive cells are capable of self-renewal and possess adipogenic potential. Finally, we demonstrate that high-fat diet (HFD) can initiate differentiation of these cells in vivo. These data identify a population of adipose stem cells that contribute to the depot-specific response to HFD.
Collapse
Affiliation(s)
- Matthew D Lynes
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, USA,Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA,Matthew D. Lynes, PhD, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME 04074, USA. Tel: 207-396-8100;
| | - Diana L Carlone
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA,Division of Endocrinology, Boston Children’s Hospital, Boston, MA, USA,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Kristy L Townsend
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - David T Breault
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA,Division of Endocrinology, Boston Children’s Hospital, Boston, MA, USA,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA,Corresponding author: Yu-Hua Tseng, PhD, Joslin Diabetes Center, One Joslin Place, Boston, MA 02215, USA. Tel: 617-309-1967;
| |
Collapse
|
35
|
Almeida DL, Moreira VM, Cardoso LE, Junior MDF, Pavanelo A, Ribeiro TA, da Silva Franco CC, Tófolo LP, Peres MNC, Ribeiro MVG, Ferreira ARO, Gomes RM, Miranda RA, Trevenzoli IH, Armitage JA, Palma-Rigo K, de Freitas Mathias PC. Lean in one way, in obesity another: effects of moderate exercise in brown adipose tissue of early overfed male Wistar rats. Int J Obes (Lond) 2022; 46:137-143. [PMID: 34552207 DOI: 10.1038/s41366-021-00969-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 09/01/2021] [Accepted: 09/13/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Early postnatal overfeeding (PO) induces long-term overweight and reduces brown adipose tissue (BAT) thermogenesis. Exercise has been suggested as a possible intervention to increase BAT function. In this study, we investigated chronical effects of moderate-intensity exercise in BAT function in postnatal overfed male Wistar rats METHODS: Litters' delivery was on postnatal-day 0 - PN0. At PN2, litters were adjusted to nine (normal litter - NL) or three pups (small litter - SL) per dam. Animals were weaned on PN21 and in PN30 randomly divided into sedentary (NL-Sed and SL-Sed) or exercised (NL-Exe and SL-Exe), N of 14 litters per group. Exercise protocol started (PN30) with an effort test; training sessions were performed three times weekly at 60% of the VO2max achieved in effort test, until PN80. On PN81, a temperature transponder was implanted beneath the interscapular BAT, whose temperature was assessed in periods of lights-on and -off from PN87 to PN90. Sympathetic nerve activation of BAT was registered at PN90. Animals were euthanized at PN91 and tissues collected RESULTS: PO impaired BAT thermogenesis in lights-on (pPO < 0.0001) and -off (pPO < 0.01). Exercise increased BAT temperature in lights-on (pExe < 0.0001). In NL-Exe, increased BAT activity was associated with higher sympathetic activity (pExe < 0.05), β3-AR (pExe < 0.001), and UCP1 (pExe < 0.001) content. In SL-Exe, increasing BAT thermogenesis is driven by a combination of tissue morphology remodeling (pExe < 0.0001) with greater effect in increasing UCP1 (pExe < 0.001) and increased β3-AR (pExe < 0.001) content. CONCLUSION Moderate exercise chronically increased BAT thermogenesis in both, NL and SL groups. In NL-Exe by increasing Sympathetic activity, and in SL-Exe by a combination of increased β3-AR and UCP1 content with morphologic remodeling of BAT. Chronically increasing BAT thermogenesis in obese subjects may lead to higher overall energy expenditure, favoring the reduction of obesity and related comorbidities.
Collapse
Affiliation(s)
- Douglas Lopes Almeida
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil.
- Department of Physiology, State University of Londrina, Londrina, Paraná, Brazil.
| | - Veridiana Mota Moreira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
- Department of Physiology, State University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Lucas Eduardo Cardoso
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | | | - Audrei Pavanelo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Tatiane Aparecida Ribeiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Claudinéia Conationi da Silva Franco
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Laize Perón Tófolo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Maria Natália Chimirri Peres
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Maiara Vanusa Guedes Ribeiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Anna Rebeka Oliveira Ferreira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Rodrigo Mello Gomes
- Physiological Sciences Department, Federal University of Goiás, Av Esperança, Goiânia/GO, Brazil
| | - Rosiane Aparecida Miranda
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, 550 Av, Pedro Calmon, Rio de Janeiro, Brazil
| | - Isis Hara Trevenzoli
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, 550 Av, Pedro Calmon, Rio de Janeiro, Brazil
| | - James Andrew Armitage
- Deakin University, School of Medicine, Optometry, 75 Pigdons Rd, Waurn Ponds, VIC, Australia
| | - Kesia Palma-Rigo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Paulo Cesar de Freitas Mathias
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| |
Collapse
|
36
|
Kislev N, Izgilov R, Adler R, Benayahu D. Exploring the Cell Stemness and the Complexity of the Adipose Tissue Niche. Biomolecules 2021; 11:biom11121906. [PMID: 34944549 PMCID: PMC8699211 DOI: 10.3390/biom11121906] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/05/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue is a complex organ composed of different cellular populations, including mesenchymal stem and progenitor cells, adipocytes, and immune cells such as macrophages and lymphocytes. These cellular populations alter dynamically during aging or as a response to pathophysiology such as obesity. Changes in the various inflammatory cells are associated with metabolic complications and the development of insulin resistance, indicating that immune cells crosstalk with the adipocytes. Therefore, a study of the cell populations in the adipose tissue and the extracellular matrix maintaining the tissue niche is important for the knowledge on the regulatory state of the organ. We used a combination of methods to study various parameters to identify the composition of the resident cells in the adipose tissue and evaluate their profile. We analyzed the tissue structure and cells based on histology, immune fluorescence staining, and flow cytometry of cells present in the tissue in vivo and these markers’ expression in vitro. Any shift in cells’ composition influences self-renewal of the mesenchymal progenitors, and other cells affect the functionality of adipogenesis.
Collapse
|
37
|
Metformin Improves Stemness of Human Adipose-Derived Stem Cells by Downmodulation of Mechanistic Target of Rapamycin (mTOR) and Extracellular Signal-Regulated Kinase (ERK) Signaling. Biomedicines 2021; 9:biomedicines9121782. [PMID: 34944598 PMCID: PMC8698459 DOI: 10.3390/biomedicines9121782] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/03/2021] [Accepted: 10/25/2021] [Indexed: 12/05/2022] Open
Abstract
Adipose tissue plays an important role in regulating metabolic homeostasis by storing excess fat and protecting other organs from lipotoxicity. Aging is associated with central fat redistribution, culminating in a decrease in insulin-sensitive subcutaneous and an increase in insulin-resistant visceral adipose depots. Adipose-derived stem cells (ASCs) play an important role in the regeneration of adipose tissue. Aged ASCs show decreased stemness and regenerative potential due to the accumulation of oxidative stress and mitochondrial dysfunction-related cell damage. Metformin is a well-established anti-diabetic drug that has shown anti-aging effects in different organisms and animal models. In this study, we analyzed the effect of metformin treatment on the stemness of human ASCs in cell culture and whole adipose tissue culture models. Our results demonstrate that metformin improves the stemness of ASCs, reducing their rate of proliferation and adipocyte differentiation. Investigating the possible underlying mechanism, we observed a decrease in the mTOR and ERK activity in metformin-treated ASCs. In addition, we observed an increase in autophagy activity upon metformin treatment. We conclude that metformin treatment improves ASCs stemness by reducing mTOR and ERK signaling and enhancing autophagy. Future in vivo evaluations in animal models and humans will pave the way for the clinical adaptation of this well-established drug for reviving the stemness of aged stem cells.
Collapse
|
38
|
Ackermann J, Arndt L, Kirstein M, Hobusch C, Brinker G, Klöting N, Braune J, Gericke M. Myeloid Cell-Specific IL-4 Receptor Knockout Partially Protects from Adipose Tissue Inflammation. THE JOURNAL OF IMMUNOLOGY 2021; 207:3081-3089. [PMID: 34789558 DOI: 10.4049/jimmunol.2100699] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/18/2021] [Indexed: 11/19/2022]
Abstract
IL-4 receptor signaling is supposed to play a major role in anti-inflammatory polarization and proliferation of adipose tissue macrophages. In this study, we examined the metabolic and inflammatory phenotype of C57BL/6J mice (IIl4ra) with LysM-dependent knockout (IIl4ra Δmyel) of the IL-4 receptor α-chain (IL-4Rα), the mandatory signaling component of IL-4 and IL-13, on chow and high-fat diet. Lean IIl4ra Δmyel mice showed decreased insulin sensitivity, no divergent adipose tissue macrophage polarization, but an increased percentage of CD8+ T cells in visceral adipose tissue. After 20 wk of a high-fat diet, IIl4ra Δmyel mice exhibited higher glucose tolerance, no changes in the lymphocyte compartment and fewer M1 macrophages in visceral adipose tissue. In vivo adipose tissue macrophage proliferation measured by BrdU incorporation was unaffected by Il4ra knockout. Interestingly, we show that IL-4Rα signaling directly augmented Itgax (Cd11c) gene expression in bone marrow-derived macrophages and increased the amount of CD11c+ macrophages in adipose tissue explants. Myeloid cell-specific knockout of Il4ra deteriorated insulin sensitivity in lean mice but improved parameters of glucose homeostasis and partially protected from adipose tissue inflammation in obese mice. Hence, IL-4Rα signaling probably plays a minor role in maintaining the macrophage M2 population and proliferation rates in vivo. Moreover, our data indicate that IL-4 signaling plays a proinflammatory role in adipose tissue inflammation by directly upregulating CD11c on adipose tissue macrophages.
Collapse
Affiliation(s)
- Jan Ackermann
- Institute of Anatomy and Cell Biology, Martin-Luther-University Halle-Wittenberg, Halle, Germany.,Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Lilli Arndt
- Institute of Anatomy and Cell Biology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Michaela Kirstein
- Institute of Anatomy and Cell Biology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | | | - Georg Brinker
- Institute of Anatomy and Cell Biology, Martin-Luther-University Halle-Wittenberg, Halle, Germany.,Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research of the Helmholtz Zentrum München at the University of Leipzig; and.,Medical Department III, Leipzig University, Leipzig, Germany
| | - Julia Braune
- Institute of Anatomy and Cell Biology, Martin-Luther-University Halle-Wittenberg, Halle, Germany.,Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Martin Gericke
- Institute of Anatomy and Cell Biology, Martin-Luther-University Halle-Wittenberg, Halle, Germany; .,Institute of Anatomy, Leipzig University, Leipzig, Germany
| |
Collapse
|
39
|
Gilarska A, Hinz A, Bzowska M, Dyduch G, Kamiński K, Nowakowska M, Lewandowska-Łańcucka J. Addressing the Osteoporosis Problem-Multifunctional Injectable Hybrid Materials for Controlling Local Bone Tissue Remodeling. ACS APPLIED MATERIALS & INTERFACES 2021; 13:49762-49779. [PMID: 34643364 PMCID: PMC8554765 DOI: 10.1021/acsami.1c17472] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/01/2021] [Indexed: 05/08/2023]
Abstract
Novel multifunctional biomimetic injectable hybrid systems were synthesized. The physicochemical as well as biological in vitro and in vivo tests demonstrated that they are promising candidates for bone tissue regeneration. The hybrids are composed of a biopolymeric collagen/chitosan/hyaluronic acid matrix and amine group-functionalized silica particles decorated with apatite to which the alendronate molecules were coordinated. The components of these systems were integrated and stabilized by cross-linking with genipin, a compound of natural origin. They can be precisely injected into the diseased tissue in the form of a viscous sol or a partially cross-linked hydrogel, where they can serve as scaffolds for locally controlled bone tissue regeneration/remodeling by supporting the osteoblast formation/proliferation and maintaining the optimal osteoclast level. These materials lack systemic toxicity. They can be particularly useful for the repair of small osteoporotic bone defects.
Collapse
Affiliation(s)
- Adriana Gilarska
- Faculty
of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
- Faculty
of Physics and Applied Computer Science, AGH University of Science and Technology, Mickiewicza 30, 30-059 Kraków, Poland
| | - Alicja Hinz
- Department
of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Monika Bzowska
- Department
of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Grzegorz Dyduch
- Department
of Pathomorphology, Jagiellonian University
Medical College, 30-387 Kraków, Poland
| | - Kamil Kamiński
- Faculty
of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| | - Maria Nowakowska
- Faculty
of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| | | |
Collapse
|
40
|
Mesothelial cells are not a source of adipocytes in mice. Cell Rep 2021; 36:109388. [PMID: 34260927 PMCID: PMC8317472 DOI: 10.1016/j.celrep.2021.109388] [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: 03/26/2021] [Revised: 06/06/2021] [Accepted: 06/21/2021] [Indexed: 02/08/2023] Open
Abstract
Visceral adipose tissue (VAT) depots are associated with the adverse metabolic consequences of obesity, such as insulin resistance. The developmental origin of VAT depots and the identity and regulation of adipocyte progenitor cells have been active areas of investigation. In recent years, a paradigm of mesothelial cells as a source of VAT adipocyte progenitor cells has emerged based on lineage tracing studies using the Wilms’ tumor gene, Wt1, as a marker for cells of mesothelial origin. Here, we show that Wt1 expression in adipose tissue is not limited to the mesothelium but is also expressed by a distinct preadipocyte population in mice and humans. We identify keratin 19 (Krt19) as a highly specific marker for the adult mouse mesothelium and demonstrate that Krt19-expressing mesothelial cells do not differentiate into visceral adipocytes. These results contradict the assertion that the VAT mesothelium can serve as a source of adipocytes. Mesothelial differentiation into adipocytes has been proposed based on fate mapping experiments using Wt1 as a marker. Westcott et al. show that Wt1 is expressed in stromal preadipocytes in addition to mesothelium and that fate mapping using a specific mesothelial marker, Krt19, does not support adipocyte differentiation.
Collapse
|
41
|
Lumaquin D, Johns E, Montal E, Weiss JM, Ola D, Abuhashem A, White RM. An in vivo reporter for tracking lipid droplet dynamics in transparent zebrafish. eLife 2021; 10:e64744. [PMID: 34114952 PMCID: PMC8195600 DOI: 10.7554/elife.64744] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 05/14/2021] [Indexed: 01/22/2023] Open
Abstract
Lipid droplets are lipid storage organelles found in nearly all cell types from adipocytes to cancer cells. Although increasingly implicated in disease, current methods to study lipid droplets in vertebrate models rely on static imaging or the use of fluorescent dyes, limiting investigation of their rapid in vivo dynamics. To address this, we created a lipid droplet transgenic reporter in whole animals and cell culture by fusing tdTOMATO to Perilipin-2 (PLIN2), a lipid droplet structural protein. Expression of this transgene in transparent casper zebrafish enabled in vivo imaging of adipose depots responsive to nutrient deprivation and high-fat diet. Simultaneously, we performed a large-scale in vitro chemical screen of 1280 compounds and identified several novel regulators of lipolysis in adipocytes. Using our Tg(-3.5ubb:plin2-tdTomato) zebrafish line, we validated several of these novel regulators and revealed an unexpected role for nitric oxide in modulating adipocyte lipid droplets. Similarly, we expressed the PLIN2-tdTOMATO transgene in melanoma cells and found that the nitric oxide pathway also regulated lipid droplets in cancer. This model offers a tractable imaging platform to study lipid droplets across cell types and disease contexts using chemical, dietary, or genetic perturbations.
Collapse
Affiliation(s)
- Dianne Lumaquin
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD ProgramNew YorkUnited States
| | - Eleanor Johns
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Gerstner Sloan Kettering Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Emily Montal
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Joshua M Weiss
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD ProgramNew YorkUnited States
| | - David Ola
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Abderhman Abuhashem
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD ProgramNew YorkUnited States
- Developmental Biology Program, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Richard M White
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| |
Collapse
|
42
|
Gene Expression Analysis of Environmental Temperature and High-Fat Diet-Induced Changes in Mouse Supraclavicular Brown Adipose Tissue. Cells 2021; 10:cells10061370. [PMID: 34199472 PMCID: PMC8226907 DOI: 10.3390/cells10061370] [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] [Received: 02/23/2021] [Revised: 05/26/2021] [Accepted: 05/29/2021] [Indexed: 12/16/2022] Open
Abstract
Obesity, a dysregulation of adipose tissue, is a major health risk factor associated with many diseases. Brown adipose tissue (BAT)-mediated thermogenesis can potentially regulate energy expenditure, making it an attractive therapeutic target to combat obesity. Here, we characterize the effects of cold exposure, thermoneutrality, and high-fat diet (HFD) feeding on mouse supraclavicular BAT (scBAT) morphology and BAT-associated gene expression compared to other adipose depots, including the interscapular BAT (iBAT). scBAT was as sensitive to cold induced thermogenesis as iBAT and showed reduced thermogenic effect under thermoneutrality. While both scBAT and iBAT are sensitive to cold, the expression of genes involved in nutrient processing is different. The scBAT also showed less depot weight gain and more single-lipid adipocytes, while the expression of BAT thermogenic genes, such as Ucp1, remained similar or increased more under our HFD feeding regime at ambient and thermoneutral temperatures than iBAT. Together, these findings show that, in addition to its anatomical resemblance to human scBAT, mouse scBAT possesses thermogenic features distinct from those of other adipose depots. Lastly, this study also characterizes a previously unknown mouse deep neck BAT (dnBAT) depot that exhibits similar thermogenic characteristics as scBAT under cold exposure and thermoneutrality.
Collapse
|
43
|
Taherkhani S, Suzuki K, Ruhee RT. A Brief Overview of Oxidative Stress in Adipose Tissue with a Therapeutic Approach to Taking Antioxidant Supplements. Antioxidants (Basel) 2021; 10:594. [PMID: 33924341 PMCID: PMC8069597 DOI: 10.3390/antiox10040594] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/05/2021] [Accepted: 04/08/2021] [Indexed: 12/20/2022] Open
Abstract
One of the leading causes of obesity associated with oxidative stress (OS) is excessive consumption of nutrients, especially fast-foods, and a sedentary lifestyle, characterized by the ample accumulation of lipid in adipose tissue (AT). When the body needs energy, the lipid is broken down into glycerol (G) and free fatty acids (FFA) during the lipolysis process and transferred to various tissues in the body. Materials secreted from AT, especially adipocytokines (interleukin (IL)-1β, IL-6, and tumor necrosis factor-α (TNF-α)) and reactive oxygen species (ROS), are impressive in causing inflammation and OS of AT. There are several ways to improve obesity, but researchers have highly regarded the use of antioxidant supplements due to their neutralizing properties in removing ROS. In this review, we have examined the AT response to OS to antioxidant supplements focusing on animal studies. The results are inconsistent due to differences in the study duration and diversity in animals (strain, age, and sex). Therefore, there is a need for different studies, especially in humans.
Collapse
Affiliation(s)
- Shima Taherkhani
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Guilan, Rasht 4199843653, Iran
| | - Katsuhiko Suzuki
- Faculty of Sport Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa 359-1192, Japan
| | - Ruheea Taskin Ruhee
- Gradute School of Sport Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa 359-1192, Japan
| |
Collapse
|
44
|
Ortner VK, Sahu A, Cordova M, Kose K, Aleissa S, Alessi-Fox C, Haedersdal M, Rajadhyaksha M, Rossi AM. Exploring the utility of Deep Red Anthraquinone 5 for digital staining of ex vivo confocal micrographs of optically sectioned skin. JOURNAL OF BIOPHOTONICS 2021; 14:e202000207. [PMID: 33314673 PMCID: PMC8274380 DOI: 10.1002/jbio.202000207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 05/11/2023]
Abstract
We investigated the utility of the fluorescent dye Deep Red Anthraquinone 5 (DRAQ5) for digital staining of optically sectioned skin in comparison to acridine orange (AO). Eight fresh-frozen thawed Mohs discard tissue specimens were stained with AO and DRAQ5, and imaged using an ex vivo confocal microscope at three wavelengths (488 nm and 638 nm for fluorescence, 785 nm for reflectance). Images were overlaid (AO + Reflectance, DRAQ5 + Reflectance), digitally stained, and evaluated by three investigators for perceived image quality (PIQ) and histopathological feature identification. In addition to nuclear staining, AO seemed to stain dermal fibers in a subset of cases in digitally stained images, while DRAQ5 staining was more specific to nuclei. Blinded evaluation showed substantial agreement, favoring DRAQ5 for PIQ (82%, Cl 75%-90%, Gwet's AC 0.74) and for visualization of histopathological features in (81%, Cl 73%-89%, Gwet's AC 0.67), supporting its use in digital staining of multimodal confocal micrographs of skin.
Collapse
Affiliation(s)
- Vinzent Kevin Ortner
- Department of Dermatology, Copenhagen University Hospital, Bispebjerg and Frederiskberg, Denmark
| | - Aditi Sahu
- Dermatology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Miguel Cordova
- Dermatology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kivanc Kose
- Dermatology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Saud Aleissa
- Dermatology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Merete Haedersdal
- Department of Dermatology, Copenhagen University Hospital, Bispebjerg and Frederiskberg, Denmark
| | - Milind Rajadhyaksha
- Dermatology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anthony Mario Rossi
- Dermatology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
45
|
Wilson RC, Lutterschmidt DI. Energy Metrics of Red-Sided Garter Snakes ( Thamnophis sirtalis parietalis) Vary with Sex but Not Life-History Stage. Physiol Biochem Zool 2021; 93:347-357. [PMID: 32730193 DOI: 10.1086/709994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Because reproduction is energetically expensive, an organism's energy stores are likely involved in mediating transitions between reproductive and self-maintenance activities. We investigated whether body condition index, adipocyte follicle size, and liver glycogen differ with the life-history transition from reproduction to migration and foraging in red-sided garter snakes (Thamnophis sirtalis parietalis). Females primarily investing in mating behavior located at the den had a significantly higher body condition index than females migrating to summer feeding grounds. The body condition index of male snakes did not differ between snakes located at the den and those migrating to summer feeding grounds. Neither adipocyte follicle area nor liver glycogen stores differed significantly between snakes performing mating activities at the den and those migrating to summer feeding grounds. We did find a sexual dimorphism in that female red-sided garter snakes had significantly larger adipocyte follicles and higher liver glycogen compared with males. Our findings support the across-species phenomenon of females and males displaying a sexual dimorphism in stored energy substrates. Conversely, we did not find evidence to suggest that red-sided garter snakes primarily utilize fatty acids to fuel the initiation of migration, a finding that is not consistent with other long-distance migrators, such as birds. Because we did not find evidence to suggest that stored energy metrics influence the decision to migrate, a physiological mechanism that induces migration in red-sided garter snakes remains elusive.
Collapse
|
46
|
Sebo ZL, Rodeheffer MS. Testosterone metabolites differentially regulate obesogenesis and fat distribution. Mol Metab 2020; 44:101141. [PMID: 33307216 PMCID: PMC7772371 DOI: 10.1016/j.molmet.2020.101141] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/20/2020] [Accepted: 12/03/2020] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVE Low testosterone in men (hypogonadism) is associated with obesity and type II diabetes. Testosterone replacement therapy has been shown to reverse these effects. However, the mechanisms by which testosterone regulates total fat mass, fat distribution, and metabolic health are unclear. In this study, we clarify the impact of hypogonadism on these parameters, as well as parse the role of testosterone from its downstream metabolites, dihydrotestosterone (DHT), and estradiol, in the regulation of depot-specific adipose tissue mass. METHODS To achieve this objective, we utilized mouse models of male hypogonadism coupled with hormone replacement therapy, magnetic resonance imaging (MRI), glucose tolerance tests, flow cytometry, and immunohistochemical techniques. RESULTS We observed that castrated mice develop increased fat mass, reduced muscle mass, and impaired glucose metabolism compared with gonadally intact males. Interestingly, obesity is further accelerated in castrated mice fed a high-fat diet, suggesting hypogonadism increases susceptibility to obesogenesis when dietary consumption of fat is elevated. By performing hormone replacement therapy in castrated mice, we show that testosterone impedes visceral and subcutaneous fat mass expansion. Testosterone-derived estradiol selectively blocks visceral fat growth, and DHT selectively blocks the growth of subcutaneous fat. These effects are mediated by depot-specific alterations in adipocyte size. We also show that high-fat diet-induced adipogenesis is elevated in castrated mice and that this can be rescued by androgen treatment. Obesogenic adipogenesis is also elevated in mice where androgen receptor activity is inhibited. CONCLUSIONS These data indicate that hypogonadism impairs glucose metabolism and increases obesogenic fat mass expansion through adipocyte hypertrophy and adipogenesis. In addition, our findings highlight distinct roles for testosterone, DHT, and estradiol in the regulation of total fat mass and fat distribution and reveal that androgen signaling blocks obesogenic adipogenesis in vivo.
Collapse
Affiliation(s)
- Zachary L Sebo
- Yale University, Department of Molecular, Cellular and Developmental Biology, USA
| | - Matthew S Rodeheffer
- Yale University, Department of Molecular, Cellular and Developmental Biology, USA; Department of Comparative Medicine, Yale University, USA; Department of Physiology, Yale University, USA; Yale Stem Cell Center, USA; Yale Program in Integrative Cell Signaling and Neurobiology of Metabolism, USA.
| |
Collapse
|
47
|
Fernández-Vega A, Chicano-Gálvez E, Prentice BM, Anderson D, Priego-Capote F, López-Bascón MA, Calderón-Santiago M, Avendaño MS, Guzmán-Ruiz R, Tena-Sempere M, Fernández JA, Caprioli RM, Malagón MM. Optimization of a MALDI-Imaging protocol for studying adipose tissue-associated disorders. Talanta 2020; 219:121184. [PMID: 32887102 DOI: 10.1016/j.talanta.2020.121184] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/13/2020] [Accepted: 05/15/2020] [Indexed: 12/13/2022]
Abstract
Matrix-assisted laser desorption ionization (MALDI) imaging mass spectrometry (IMS) is increasingly recognized for its potential in the discovery of novel biomarkers directly from tissue sections. However, there are no MALDI IMS studies as yet on the adipose tissue, a lipid-enriched tissue that plays a pivotal role in the development of obesity-associated disorders. Herein, we aimed at developing an optimized method for analyzing adipose tissue lipid composition under both physiological and pathological conditions by MALDI IMS. Our studies showed an exacerbated lipid delocalization from adipose tissue sections when conventional strategies were applied. However, our optimized method using conductive-tape sampling and 2,5-dihydroxybenzoic acid (DHB) as a matrix, preserved the anatomical organization and minimized lipid diffusion from sample sections. This method enabled the identification of a total of 625 down-regulated and 328 up-regulated m/z values in the adipose tissue from a rat model of extreme obesity as compared to lean animals. Combination of MALDI IMS and liquid chromatography (LC)-MS/MS data identified 44 differentially expressed lipid species between lean and obese animals, including phospholipids and sphingomyelins. Among the lipids identified, SM(d18:0_18:2), PE(P-16:0_20:0), and PC(O-16:0_16:1) showed a differential spatial distribution in the adipose tissue of lean vs. obese animals. In sum, our method provides a valuable new tool for research on adipose tissue that may pave the way for the identification of novel biomarkers of obesity and metabolic disease.
Collapse
Affiliation(s)
- A Fernández-Vega
- Dept. Cell Biology, Physiology, and Immunology, IMIBIC/University of Cordoba (UCO)/Reina Sofia University Hospital (HURS), Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), ISCIII, Spain
| | | | - B M Prentice
- Department of Chemistry, University of Florida, Gainesville, FL, USA
| | - D Anderson
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, 37232, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN, 37232, USA
| | - F Priego-Capote
- Department of Analytical Chemistry, IMIBIC/UCO/HURS, Cordoba, Spain
| | - M A López-Bascón
- Department of Analytical Chemistry, IMIBIC/UCO/HURS, Cordoba, Spain
| | | | - M S Avendaño
- Dept. Cell Biology, Physiology, and Immunology, IMIBIC/University of Cordoba (UCO)/Reina Sofia University Hospital (HURS), Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), ISCIII, Spain
| | - R Guzmán-Ruiz
- Dept. Cell Biology, Physiology, and Immunology, IMIBIC/University of Cordoba (UCO)/Reina Sofia University Hospital (HURS), Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), ISCIII, Spain
| | - M Tena-Sempere
- Dept. Cell Biology, Physiology, and Immunology, IMIBIC/University of Cordoba (UCO)/Reina Sofia University Hospital (HURS), Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), ISCIII, Spain
| | - J A Fernández
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain
| | - R M Caprioli
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, 37232, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN, 37232, USA; Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain; Department of Chemistry, Vanderbilt University, Nashville, TN, 37232, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37232, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Department of Medicine, Vanderbilt University, Nashville, TN, 37232, USA
| | - M M Malagón
- Dept. Cell Biology, Physiology, and Immunology, IMIBIC/University of Cordoba (UCO)/Reina Sofia University Hospital (HURS), Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), ISCIII, Spain.
| |
Collapse
|
48
|
Wang L, Wang S, Shi Y, Li R, Günther S, Ong YT, Potente M, Yuan Z, Liu E, Offermanns S. YAP and TAZ protect against white adipocyte cell death during obesity. Nat Commun 2020; 11:5455. [PMID: 33116140 PMCID: PMC7595161 DOI: 10.1038/s41467-020-19229-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
The expansion of the white adipose tissue (WAT) in obesity goes along with increased mechanical, metabolic and inflammatory stress. How adipocytes resist this stress is still poorly understood. Both in human and mouse adipocytes, the transcriptional co-activators YAP/TAZ and YAP/TAZ target genes become activated during obesity. When fed a high-fat diet (HFD), mice lacking YAP/TAZ in white adipocytes develop severe lipodystrophy with adipocyte cell death. The pro-apoptotic factor BIM, which is downregulated in adipocytes of obese mice and humans, is strongly upregulated in YAP/TAZ-deficient adipocytes under HFD, and suppression of BIM expression reduces adipocyte apoptosis. In differentiated adipocytes, TNFα and IL-1β promote YAP/TAZ nuclear translocation via activation of RhoA-mediated actomyosin contractility and increase YAP/TAZ-mediated transcriptional regulation by activation of c-Jun N-terminal kinase (JNK) and AP-1. Our data indicate that the YAP/TAZ signaling pathway may be a target to control adipocyte cell death and compensatory adipogenesis during obesity.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/deficiency
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Adipocytes, White/metabolism
- Adipocytes, White/pathology
- Adipogenesis
- Animals
- Bcl-2-Like Protein 11/metabolism
- Cell Cycle Proteins/deficiency
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cell Death
- Cells, Cultured
- Diet, High-Fat
- Disease Models, Animal
- Gene Expression Regulation
- Humans
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Obesity/genetics
- Obesity/metabolism
- Obesity/pathology
- Trans-Activators/deficiency
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription Factors/metabolism
- Transcriptional Coactivator with PDZ-Binding Motif Proteins
- YAP-Signaling Proteins
Collapse
Affiliation(s)
- Lei Wang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany.
| | - ShengPeng Wang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany.
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Yanta District, Xi'an, China.
| | - Yue Shi
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Yanta District, Xi'an, China
| | - Rui Li
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Stefan Günther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Yu Ting Ong
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Michael Potente
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Zuyi Yuan
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Enqi Liu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center Xi'an Jiaotong University, Xi'an, China
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany.
- Center for Molecular Medicine, Medical Faculty, Goethe University, Frankfurt am Main, 60590, Germany.
| |
Collapse
|
49
|
Dean JM, He A, Tan M, Wang J, Lu D, Razani B, Lodhi IJ. MED19 Regulates Adipogenesis and Maintenance of White Adipose Tissue Mass by Mediating PPARγ-Dependent Gene Expression. Cell Rep 2020; 33:108228. [PMID: 33027649 PMCID: PMC7561447 DOI: 10.1016/j.celrep.2020.108228] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 08/11/2020] [Accepted: 09/14/2020] [Indexed: 12/13/2022] Open
Abstract
The Mediator complex relays regulatory signals from gene-specific transcription factors to the basal transcriptional machinery. However, the role of individual Mediator subunits in different tissues remains unclear. Here, we demonstrate that MED19 is essential for adipogenesis and maintenance of white adipose tissue (WAT) by mediating peroxisome proliferator-activated receptor gamma (PPARγ) transcriptional activity. MED19 knockdown blocks white adipogenesis, but not brown adipogenesis or C2C12 myoblast differentiation. Adipose-specific MED19 knockout (KO) in mice results in a striking loss of WAT, whitening of brown fat, hepatic steatosis, and insulin resistance. Inducible adipose-specific MED19 KO in adult animals also results in lipodystrophy, demonstrating its requirement for WAT maintenance. Global gene expression analysis reveals induction of genes involved in apoptosis and inflammation and impaired expression of adipose-specific genes, resulting from decreased PPARγ residency on adipocyte gene promoters and reduced association of PPARγ with RNA polymerase II. These results identify MED19 as a crucial facilitator of PPARγ-mediated gene expression in adipose tissue.
Collapse
Affiliation(s)
- John M Dean
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anyuan He
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Min Tan
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jun Wang
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dongliang Lu
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Babak Razani
- Cardiology Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Veterans Affairs St. Louis Healthcare System, John Cochran Division, St. Louis, MO 63106, USA
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
50
|
Glastonbury CA, Pulit SL, Honecker J, Censin JC, Laber S, Yaghootkar H, Rahmioglu N, Pastel E, Kos K, Pitt A, Hudson M, Nellåker C, Beer NL, Hauner H, Becker CM, Zondervan KT, Frayling TM, Claussnitzer M, Lindgren CM. Machine Learning based histology phenotyping to investigate the epidemiologic and genetic basis of adipocyte morphology and cardiometabolic traits. PLoS Comput Biol 2020; 16:e1008044. [PMID: 32797044 PMCID: PMC7449405 DOI: 10.1371/journal.pcbi.1008044] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 08/26/2020] [Accepted: 06/11/2020] [Indexed: 12/29/2022] Open
Abstract
Genetic studies have recently highlighted the importance of fat distribution, as well as overall adiposity, in the pathogenesis of obesity-associated diseases. Using a large study (n = 1,288) from 4 independent cohorts, we aimed to investigate the relationship between mean adipocyte area and obesity-related traits, and identify genetic factors associated with adipocyte cell size. To perform the first large-scale study of automatic adipocyte phenotyping using both histological and genetic data, we developed a deep learning-based method, the Adipocyte U-Net, to rapidly derive mean adipocyte area estimates from histology images. We validate our method using three state-of-the-art approaches; CellProfiler, Adiposoft and floating adipocytes fractions, all run blindly on two external cohorts. We observe high concordance between our method and the state-of-the-art approaches (Adipocyte U-net vs. CellProfiler: R2visceral = 0.94, P < 2.2 × 10-16, R2subcutaneous = 0.91, P < 2.2 × 10-16), and faster run times (10,000 images: 6mins vs 3.5hrs). We applied the Adipocyte U-Net to 4 cohorts with histology, genetic, and phenotypic data (total N = 820). After meta-analysis, we found that mean adipocyte area positively correlated with body mass index (BMI) (Psubq = 8.13 × 10-69, βsubq = 0.45; Pvisc = 2.5 × 10-55, βvisc = 0.49; average R2 across cohorts = 0.49) and that adipocytes in subcutaneous depots are larger than their visceral counterparts (Pmeta = 9.8 × 10-7). Lastly, we performed the largest GWAS and subsequent meta-analysis of mean adipocyte area and intra-individual adipocyte variation (N = 820). Despite having twice the number of samples than any similar study, we found no genome-wide significant associations, suggesting that larger sample sizes and a homogenous collection of adipose tissue are likely needed to identify robust genetic associations.
Collapse
Affiliation(s)
- Craig A. Glastonbury
- Big Data Institute, University of Oxford, Oxford, United Kingdom
- BenevolentAI, London, United Kingdom
| | - Sara L. Pulit
- Big Data Institute, University of Oxford, Oxford, United Kingdom
| | - Julius Honecker
- Else Kröner-Fresenius-Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Jenny C. Censin
- Big Data Institute, University of Oxford, Oxford, United Kingdom
- Wellcome Centre for Human Genetics (WCHG), Oxford, United Kingdom
| | - Samantha Laber
- Big Data Institute, University of Oxford, Oxford, United Kingdom
- Broad Institute of MIT and Harvard, Cambridge Massachusetts, United States of America
| | - Hanieh Yaghootkar
- Genetics of Complex Traits, University of Exeter Medical School, Royal Devon & Exeter Hospital, Exeter, United Kingdom
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, United Kingdom
| | - Nilufer Rahmioglu
- Wellcome Centre for Human Genetics (WCHG), Oxford, United Kingdom
- Endometriosis CaRe Centre Oxford, Nuffield Department of Women’s and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Emilie Pastel
- Genetics of Complex Traits, University of Exeter Medical School, Royal Devon & Exeter Hospital, Exeter, United Kingdom
| | - Katerina Kos
- Genetics of Complex Traits, University of Exeter Medical School, Royal Devon & Exeter Hospital, Exeter, United Kingdom
| | - Andrew Pitt
- NIHR Exeter Clinical Research Facility, University of Exeter Medical School, University of Exeter and Royal Devon and Exeter NHS Foundation Trust Exeter, United Kingdom
| | - Michelle Hudson
- NIHR Exeter Clinical Research Facility, University of Exeter Medical School, University of Exeter and Royal Devon and Exeter NHS Foundation Trust Exeter, United Kingdom
| | - Christoffer Nellåker
- Big Data Institute, University of Oxford, Oxford, United Kingdom
- Endometriosis CaRe Centre Oxford, Nuffield Department of Women’s and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Nicola L. Beer
- Novo Nordisk Research Centre Oxford (NNRCO), Oxford, United Kingdom
| | - Hans Hauner
- Else Kröner-Fresenius-Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising, Germany
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, Munich
- German Center of Diabetes Research, Helmholtz Center Munich, Neuherberg, Germany
| | - Christian M. Becker
- Endometriosis CaRe Centre Oxford, Nuffield Department of Women’s and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Krina T. Zondervan
- Wellcome Centre for Human Genetics (WCHG), Oxford, United Kingdom
- Endometriosis CaRe Centre Oxford, Nuffield Department of Women’s and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Timothy M. Frayling
- Genetics of Complex Traits, University of Exeter Medical School, Royal Devon & Exeter Hospital, Exeter, United Kingdom
- NIHR Exeter Clinical Research Facility, University of Exeter Medical School, University of Exeter and Royal Devon and Exeter NHS Foundation Trust Exeter, United Kingdom
| | - Melina Claussnitzer
- Broad Institute of MIT and Harvard, Cambridge Massachusetts, United States of America
- University of Hohenheim, Stuttgart, Germany
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Cecilia M. Lindgren
- Big Data Institute, University of Oxford, Oxford, United Kingdom
- Wellcome Centre for Human Genetics (WCHG), Oxford, United Kingdom
- Broad Institute of MIT and Harvard, Cambridge Massachusetts, United States of America
| |
Collapse
|