1
|
Abdillah AM, Lee JY, Lee YR, Yun JW. Modulatory roles of capsaicin on thermogenesis in C2C12 myoblasts and the skeletal muscle of mice. Chem Biol Interact 2025; 407:111380. [PMID: 39800145 DOI: 10.1016/j.cbi.2025.111380] [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: 07/23/2024] [Revised: 11/25/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Capsaicin, a polyphenol, is known to regulate energy expenditure and thermogenesis in adipocytes and muscles. However, its role in modulating uncoupling proteins (UCPs) and adenosine triphosphate (ATP)-dependent thermogenesis in muscles remains unclear. This study investigated the mechanisms underlying the role of capsaicin in modulating the UCP- and ATP-dependent thermogenesis in C2C12 myoblasts, as well as the gastrocnemius (GM) and soleus muscles (SM) of mice. We employed molecular dynamics (MD), quantitative real-time polymerase chain reactions (qRT-PCR), immunoblots, staining methods, and assay kits to investigate the role of capsaicin on thermogenesis and its modulatory roles on the transient receptor potential cation channel subfamily V member 1 (TRPV1) and α-/β-adrenergic receptors (ARs) using in vitro and in vivo models. Our findings demonstrate that capsaicin treatment in high-fat diet-induced obese mice reduces weight gain and elevates the expression of UCP- and ATP-dependent thermogenic effectors through ATP-consuming calcium and creatine futile cycles. In vitro and in vivo models capsaicin treatment elevated the expression of sarcoendoplasmic/endoplasmic reticulum calcium ATPases (SERCA-1 and -2), ryanodine receptors (RYR-1 and -2), uncoupling proteins (UCP-2 and -3), creatine kinase B (CKB), and creatine kinase mitochondrial 2 (CKMT2), through activation of TRPV1, α1-, β2-, and β3-AR as well as the suppressed expression of α2-AR. Furthermore, our results also indicate that capsaicin promotes myotube development and enhances lipid metabolism in C2C12 cells. We found that capsaicin increased intracellular Ca2+ levels and the expression of the voltage-dependent anion channel (VDAC) and mitochondrial calcium uniporter (MCU), suggesting that elevated mitochondrial Ca2+ levels boost the expression of oxidative phosphorylation protein complexes via the activation of the ATP-futile cycle. Mechanistic studies in C2C12 cells revealed that TRPV1 is likely dispensable for capsaicin-induced thermogenesis, and TRPV1 and α1-AR may synergistically induce thermogenesis. Collectively, our findings have uncovered a novel mechanism of UCP- and ATP-dependent thermogenesis and its associated pathways in both cellular and animal models which is crucial for designing therapeutic strategies to address obesity and associated metabolic diseases.
Collapse
Affiliation(s)
- Alfin Mohammad Abdillah
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea
| | - Jae Young Lee
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea
| | - Young Rok Lee
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea
| | - Jong Won Yun
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea.
| |
Collapse
|
2
|
Luo J, Molbay M, Chen Y, Horvath I, Kadletz K, Kick B, Zhao S, Al-Maskari R, Singh I, Ali M, Bhatia HS, Minde DP, Negwer M, Hoeher L, Calandra GM, Groschup B, Su J, Kimna C, Rong Z, Galensowske N, Todorov MI, Jeridi D, Ohn TL, Roth S, Simats A, Singh V, Khalin I, Pan C, Arús BA, Bruns OT, Zeidler R, Liesz A, Protzer U, Plesnila N, Ussar S, Hellal F, Paetzold J, Elsner M, Dietz H, Erturk A. Nanocarrier imaging at single-cell resolution across entire mouse bodies with deep learning. Nat Biotechnol 2025:10.1038/s41587-024-02528-1. [PMID: 39809933 DOI: 10.1038/s41587-024-02528-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025]
Abstract
Efficient and accurate nanocarrier development for targeted drug delivery is hindered by a lack of methods to analyze its cell-level biodistribution across whole organisms. Here we present Single Cell Precision Nanocarrier Identification (SCP-Nano), an integrated experimental and deep learning pipeline to comprehensively quantify the targeting of nanocarriers throughout the whole mouse body at single-cell resolution. SCP-Nano reveals the tissue distribution patterns of lipid nanoparticles (LNPs) after different injection routes at doses as low as 0.0005 mg kg-1-far below the detection limits of conventional whole body imaging techniques. We demonstrate that intramuscularly injected LNPs carrying SARS-CoV-2 spike mRNA reach heart tissue, leading to proteome changes, suggesting immune activation and blood vessel damage. SCP-Nano generalizes to various types of nanocarriers, including liposomes, polyplexes, DNA origami and adeno-associated viruses (AAVs), revealing that an AAV2 variant transduces adipocytes throughout the body. SCP-Nano enables comprehensive three-dimensional mapping of nanocarrier distribution throughout mouse bodies with high sensitivity and should accelerate the development of precise and safe nanocarrier-based therapeutics.
Collapse
Affiliation(s)
- Jie Luo
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Deep Piction, Munich, Germany
| | - Muge Molbay
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
- Munich Medical Research School (MMRS), Munich, Germany
- Pharmaceutical Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Ying Chen
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Munich Medical Research School (MMRS), Munich, Germany
- Faculty of Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Izabela Horvath
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Deep Piction, Munich, Germany
- TUM School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | - Karoline Kadletz
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Deep Piction, Munich, Germany
| | - Benjamin Kick
- Department of Biosciences, School of Natural Sciences, Technical University of Munich, Garching, Germany
- Munich Institute of Biomedical Engineering, Technical University of Munich, Garching, Germany
| | - Shan Zhao
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
- Munich Medical Research School (MMRS), Munich, Germany
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- ETH Zurich, Institute for Molecular Health Sciences, Zurich, Switzerland
| | - Rami Al-Maskari
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Deep Piction, Munich, Germany
- TUM School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | - Inderjeet Singh
- Research Unit Adipocytes & Metabolism (ADM), Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Medicine, Technische Universität München, Munich, Germany
| | - Mayar Ali
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
- Graduate School of Neuroscience (GSN), Munich, Germany
- Institute of Computational Biology, Helmholtz Munich, Neuherberg, Germany
| | - Harsharan Singh Bhatia
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Deep Piction, Munich, Germany
| | - David-Paul Minde
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
| | - Moritz Negwer
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
- Deep Piction, Munich, Germany
| | - Luciano Hoeher
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
| | - Gian Marco Calandra
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Bernhard Groschup
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Jinpeng Su
- Institute of Virology, Technical University of Munich / Helmholtz Munich, Munich, Germany
- German Center for Infection Research (DZIF), Munich partner site, Munich, Germany
| | - Ceren Kimna
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Deep Piction, Munich, Germany
| | - Zhouyi Rong
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Munich Medical Research School (MMRS), Munich, Germany
| | - Nikolas Galensowske
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
| | - Mihail Ivilinov Todorov
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
- Deep Piction, Munich, Germany
| | - Denise Jeridi
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
- Deep Piction, Munich, Germany
| | - Tzu-Lun Ohn
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
| | - Stefan Roth
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
| | - Alba Simats
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
| | - Vikramjeet Singh
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Igor Khalin
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @Caen-Normandie (BB@C), Caen, France
| | - Chenchen Pan
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
| | - Bernardo A Arús
- Department of Functional Imaging in Surgical Oncology, National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Oliver T Bruns
- Department of Functional Imaging in Surgical Oncology, National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Reinhard Zeidler
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Structural Biology, Munich, Germany
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich / Helmholtz Munich, Munich, Germany
- German Center for Infection Research (DZIF), Munich partner site, Munich, Germany
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Siegfried Ussar
- Research Unit Adipocytes & Metabolism (ADM), Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Farida Hellal
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Johannes Paetzold
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
- Deep Piction, Munich, Germany
- Department of Computing, Imperial College London, London, UK
| | - Markus Elsner
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany
- Deep Piction, Munich, Germany
| | - Hendrik Dietz
- Department of Biosciences, School of Natural Sciences, Technical University of Munich, Garching, Germany.
- Munich Institute of Biomedical Engineering, Technical University of Munich, Garching, Germany.
| | - Ali Erturk
- Institute for Intelligent Biotechnologies (iBIO), Helmholtz Center Munich, Neuherberg, Germany.
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
- Deep Piction, Munich, Germany.
- School of Medicine, Koç University, İstanbul, Turkey.
| |
Collapse
|
3
|
Zhang Z, Cui Y, Zhang X, Hu X, Li S, Li T. Gut microbiota combined with serum metabolites to reveal the effect of Morchella esculenta polysaccharides on lipid metabolism disordered in high-fat diet mice. Int J Biol Macromol 2024; 281:136380. [PMID: 39389515 DOI: 10.1016/j.ijbiomac.2024.136380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/25/2024] [Accepted: 10/05/2024] [Indexed: 10/12/2024]
Abstract
The ameliorating effects and mechanisms of Morchella esculenta polysaccharides (MEP-1) on lipid metabolism were investigated in high-fat diet (HFD) mice. The results showed that MEP-1 intervention significantly reduced serum TC, TG, LDL-C, and inflammatory factors (TNF-α, IL-1β and IL-6) in HFD mice in a dose-dependent manner, and high-dose (400 mg/kg/d) exhibited the most significant reductive effects. In addition, MEP-1 significantly recovered the gut microbiota disorders caused by HFD, especially decreasing the ratio of Firmicutes and Bacteroidetes (F/B) and increasing the dominant bacterial of Muribaculaceae_genus, Bacteroides, Alistipes and Enterococcus. Moreover, MEP-1 promoted the production of SCFAs and increased the expression levels of Occludin, Claudin and Muc2, also regulated lipid metabolism disorder and inflammation by inhibiting TLR4/MyD88/NF-κB via the gut-liver axis. In addition, serum metabolomic analysis revealed that l-phenylalanine, l-arginine and acetylcholine were significantly upregulated with MEP-1 intervention, and were negatively correlated with blood lipid level, in which l-arginine could activate NO/PPARα/CPT1A pathway to ameliorate lipid metabolism disorders. Such results demonstrated that gut microbiota, amino acid metabolic and insulin secretion pathways might be the important factors that mediated the regulation of MEP-1 in lipid metabolism. The results also provided new evidence and strategies for the application of MEP-1 as functional foods.
Collapse
Affiliation(s)
- Zuoyi Zhang
- College of Food Science, Shenyang Agricultural University, Shenyang 110161, China
| | - Yanmin Cui
- College of Food Science, Shenyang Agricultural University, Shenyang 110161, China
| | - Xiushan Zhang
- College of Food Science, Shenyang Agricultural University, Shenyang 110161, China
| | - Xiaopei Hu
- College of Food Science, Shenyang Agricultural University, Shenyang 110161, China.
| | - Suhong Li
- College of Food Science, Shenyang Agricultural University, Shenyang 110161, China
| | - Tuoping Li
- College of Food Science, Shenyang Agricultural University, Shenyang 110161, China.
| |
Collapse
|
4
|
Cazzola R, Della Porta M, Piuri G, Maier JA. Magnesium: A Defense Line to Mitigate Inflammation and Oxidative Stress in Adipose Tissue. Antioxidants (Basel) 2024; 13:893. [PMID: 39199139 PMCID: PMC11351329 DOI: 10.3390/antiox13080893] [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: 06/28/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 09/01/2024] Open
Abstract
Magnesium (Mg) is involved in essential cellular and physiological processes. Globally, inadequate consumption of Mg is widespread among populations, especially those who consume processed foods, and its homeostasis is impaired in obese individuals and type 2 diabetes patients. Since Mg deficiency triggers oxidative stress and chronic inflammation, common features of several frequent chronic non-communicable diseases, interest in this mineral is growing in clinical medicine as well as in biomedicine. To date, very little is known about the role of Mg deficiency in adipose tissue. In obesity, the increase in fat tissue leads to changes in the release of cytokines, causing low-grade inflammation and macrophage infiltration. Hypomagnesemia in obesity can potentiate the excessive production of reactive oxygen species, mitochondrial dysfunction, and decreased ATP production. Importantly, Mg plays a role in regulating intracellular calcium concentration and is involved in carbohydrate metabolism and insulin receptor activity. This narrative review aims to consolidate existing knowledge, identify research gaps, and raise awareness of the critical role of Mg in supporting adipose tissue metabolism and preventing oxidative stress.
Collapse
Affiliation(s)
| | | | | | - Jeanette A. Maier
- Department of Biomedical and Clinical Sciences, University of Milano, 20174 Milan, Italy; (R.C.); (M.D.P.); (G.P.)
| |
Collapse
|
5
|
Carrasco Del Amor A, Bautista RH, Ussar S, Cristobal S, Urbatzka R. Insights into the mechanism of action of the chlorophyll derivative 13- 2-hydroxypheophytine a on reducing neutral lipid reserves in zebrafish larvae and mice adipocytes. Eur J Pharmacol 2023; 960:176158. [PMID: 37898286 DOI: 10.1016/j.ejphar.2023.176158] [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: 07/06/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 10/30/2023]
Abstract
Obesity is a worldwide epidemic and natural products may hold promise in its treatment. The chlorophyll derivative 13-2-hydroxypheophytine (hpa) was isolated in a screen with zebrafish larvae to identify lipid reducing molecules from cyanobacteria. However, the mechanisms underlying the lipid-reducing effects of hpa in zebrafish larvae remain poorly understood. Thus, investigating the mechanism of action of hpa and validation in other model organisms such as mice represents important initial steps. In this study, we identified 14 protein targets of hpa in zebrafish larvae by thermal proteome profiling, and selected two targets (malate dehydrogenase and pyruvate kinase) involved in cellular metabolism for further validation by enzymatic measurements. Our findings revealed a dose-dependent inhibition of pyruvate kinase by hpa exposure using protein extracts of zebrafish larvae in vitro, and in exposure experiments from 3 to 5 days post fertilization in vivo. Analysis of untargeted metabolomics of zebrafish larvae detected 940 mass peaks (66 increased, 129 decreased) and revealed that hpa induced the formation of various phospholipid species (phosphoinositol, phosphoethanolamine, phosphatidic acid). Inter-species validation showed that brown adipocytes exposed to hpa significantly reduced the size of lipid droplets, increased maximal mitochondrial respiratory capacity, and the expression of PPARy during adipocyte differentiation. In line with our data, previous work described that reduced pyruvate kinase activity lowered hepatic lipid content via reduced pyruvate and citrate, and improved mitochondrial function via phospholipids. Thus, our data provide new insights into the molecular mechanism underlying the lipid reducing activities of hpa in zebrafish larvae, and species overlapping functions in reduction of lipids.
Collapse
Affiliation(s)
- Ana Carrasco Del Amor
- Department of Biomedical and Clinical Sciences, Cell Biology, Faculty of Medicine, Linköping University, SE-58185, Linköping, Sweden.
| | - Rene Hernandez Bautista
- RG Adipocyte and Metabolism, Institute for Diabetes and Obesity, Helmholtz Center Munich, 85764, Neuherberg, Germany.
| | - Siegfried Ussar
- RG Adipocyte and Metabolism, Institute for Diabetes and Obesity, Helmholtz Center Munich, 85764, Neuherberg, Germany.
| | - Susana Cristobal
- Department of Biomedical and Clinical Sciences, Cell Biology, Faculty of Medicine, Linköping University, SE-58185, Linköping, Sweden; Ikerbasque, Basque Foundation for Sciences, Department of Physiology, Faculty of Medicine, and Nursing, University of the Basque Country UPV/EHU, Spain.
| | - Ralph Urbatzka
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, Avenida General Norton de Matos, s/n, 4450-208, Matosinhos, Portugal.
| |
Collapse
|
6
|
Ribeiro T, Jónsdóttir K, Hernandez-Bautista R, Silva NG, Sánchez-Astráin B, Samadi A, Eiriksson FF, Thorsteinsdóttir M, Ussar S, Urbatzka R. Metabolite Profile Characterization of Cyanobacterial Strains with Bioactivity on Lipid Metabolism Using In Vivo and In Vitro Approaches. Mar Drugs 2023; 21:498. [PMID: 37755111 PMCID: PMC10533020 DOI: 10.3390/md21090498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/28/2023] Open
Abstract
Cyanobacteria have demonstrated their therapeutic potential for many human diseases. In this work, cyanobacterial extracts were screened for lipid reducing activity in zebrafish larvae and in fatty-acid-overloaded human hepatocytes, as well as for glucose uptake in human hepatocytes and ucp1 mRNA induction in murine brown adipocytes. A total of 39 cyanobacteria strains were grown and their biomass fractionated, resulting in 117 chemical fractions. Reduction of neutral lipids in zebrafish larvae was observed for 12 fractions and in the human hepatocyte steatosis cell model for five fractions. The induction of ucp1 expression in murine brown adipocytes was observed in six fractions, resulting in a total of 23 bioactive non-toxic fractions. All extracts were analyzed by untargeted UPLC-Q-TOF-MS mass spectrometry followed by multivariate statistical analysis to prioritize bioactive strains. The metabolite profiling led to the identification of two markers with lipid reducing activity in zebrafish larvae. Putative compound identification using mass spectrometry databases identified them as phosphatidic acid and aromatic polyketides derivatives-two compound classes, which were previously associated with effects on metabolic disorders. In summary, we have identified cyanobacterial strains with promising lipid reducing activity, whose bioactive compounds needs to be identified in the future.
Collapse
Affiliation(s)
- Tiago Ribeiro
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, Avenida General Norton de Matos, s/n, 4450-208 Matosinhos, Portugal; (T.R.); (N.G.S.); (B.S.-A.)
- Faculty of Sciences, University of Porto, Rua do Campo Alegre, 1021, 4169-007 Porto, Portugal
| | - Kristín Jónsdóttir
- Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, 107 Reykjavik, Iceland; (K.J.); (A.S.); (F.F.E.); (M.T.)
| | - Rene Hernandez-Bautista
- RG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Diabetes Center, Helmholtz Munich, 85764 Neuherberg, Germany; (R.H.-B.); (S.U.)
| | - Natália Gonçalves Silva
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, Avenida General Norton de Matos, s/n, 4450-208 Matosinhos, Portugal; (T.R.); (N.G.S.); (B.S.-A.)
- Faculty of Sciences, University of Porto, Rua do Campo Alegre, 1021, 4169-007 Porto, Portugal
| | - Begoña Sánchez-Astráin
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, Avenida General Norton de Matos, s/n, 4450-208 Matosinhos, Portugal; (T.R.); (N.G.S.); (B.S.-A.)
| | - Afshin Samadi
- Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, 107 Reykjavik, Iceland; (K.J.); (A.S.); (F.F.E.); (M.T.)
- Joint Laboratory of Applied Ecotoxicology, Korea Institute of Science and Technology Europe (KIST EU), Campus E7.1, 66123 Saarbrucken, Germany
| | - Finnur F. Eiriksson
- Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, 107 Reykjavik, Iceland; (K.J.); (A.S.); (F.F.E.); (M.T.)
- ArcticMass, Sturlugata 8, 102 Reykjavik, Iceland
| | - Margrét Thorsteinsdóttir
- Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, 107 Reykjavik, Iceland; (K.J.); (A.S.); (F.F.E.); (M.T.)
- ArcticMass, Sturlugata 8, 102 Reykjavik, Iceland
| | - Siegfried Ussar
- RG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Diabetes Center, Helmholtz Munich, 85764 Neuherberg, Germany; (R.H.-B.); (S.U.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Ralph Urbatzka
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, Avenida General Norton de Matos, s/n, 4450-208 Matosinhos, Portugal; (T.R.); (N.G.S.); (B.S.-A.)
| |
Collapse
|
7
|
Liu J, Lu W, Yan D, Guo J, Zhou L, Shi B, Su X. Mitochondrial respiratory complex I deficiency inhibits brown adipogenesis by limiting heme regulation of histone demethylation. Mitochondrion 2023; 72:22-32. [PMID: 37451354 DOI: 10.1016/j.mito.2023.07.004] [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: 11/14/2022] [Revised: 06/13/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Mitochondrial functions play a crucial role in determining the metabolic and thermogenic status of brown adipocytes. Increasing evidence reveals that the mitochondrial oxidative phosphorylation (OXPHOS) system plays an important role in brown adipogenesis, but the mechanistic insights are limited. Herein, we explored the potential metabolic mechanisms leading to OXPHOS regulation of brown adipogenesis in pharmacological and genetic models of mitochondrial respiratory complex I deficiency. OXPHOS deficiency inhibits brown adipogenesis through disruption of the brown adipogenic transcription circuit without affecting ATP levels. Neither blockage of calcium signaling nor antioxidant treatment can rescue the suppressed brown adipogenesis. Metabolomics analysis revealed a decrease in levels of tricarboxylic acid cycle intermediates and heme. Heme supplementation specifically enhances respiratory complex I activity without affecting complex II and partially reverses the inhibited brown adipogenesis by OXPHOS deficiency. Moreover, the regulation of brown adipogenesis by the OXPHOS-heme axis may be due to the suppressed histone methylation status by increasing histone demethylation. In summary, our findings identified a heme-sensing retrograde signaling pathway that connects mitochondrial OXPHOS to the regulation of brown adipocyte differentiation and metabolic functions.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Biochemistry and Molecular Biology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Wen Lu
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Dongyue Yan
- Department of Biochemistry and Molecular Biology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Junyuan Guo
- Department of Biochemistry and Molecular Biology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Li Zhou
- Department of Nutrition, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Bimin Shi
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xiong Su
- Department of Biochemistry and Molecular Biology, Suzhou Medical College of Soochow University, Suzhou 215123, China.
| |
Collapse
|
8
|
Zhong W, Chhibber A, Luo L, Mehrotra DV, Shen J. A fast and powerful linear mixed model approach for genotype-environment interaction tests in large-scale GWAS. Brief Bioinform 2023; 24:6955097. [PMID: 36545787 DOI: 10.1093/bib/bbac547] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/26/2022] [Accepted: 11/12/2022] [Indexed: 12/24/2022] Open
Abstract
Genotype-by-environment interaction (GEI or GxE) plays an important role in understanding complex human traits. However, it is usually challenging to detect GEI signals efficiently and accurately while adjusting for population stratification and sample relatedness in large-scale genome-wide association studies (GWAS). Here we propose a fast and powerful linear mixed model-based approach, fastGWA-GE, to test for GEI effect and G + GxE joint effect. Our extensive simulations show that fastGWA-GE outperforms other existing GEI test methods by controlling genomic inflation better, providing larger power and running hundreds to thousands of times faster. We performed a fastGWA-GE analysis of ~7.27 million variants on 452 249 individuals of European ancestry for 13 quantitative traits and five environment variables in the UK Biobank GWAS data and identified 96 significant signals (72 variants across 57 loci) with GEI test P-values < 1 × 10-9, including 27 novel GEI associations, which highlights the effectiveness of fastGWA-GE in GEI signal discovery in large-scale GWAS.
Collapse
Affiliation(s)
- Wujuan Zhong
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - Aparna Chhibber
- Translational Bioinformatics, Bristol Myers Squibb, Lawrenceville, NJ 08540, USA
| | - Lan Luo
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., North Wales, PA 19454, USA
| | - Devan V Mehrotra
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., North Wales, PA 19454, USA
| | - Judong Shen
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., Rahway, NJ 07065, USA
| |
Collapse
|
9
|
Talukdar FR, Escobar Marcillo DI, Laskar RS, Novoloaca A, Cuenin C, Sbraccia P, Nisticò L, Guglielmi V, Gheit T, Tommasino M, Dogliotti E, Fortini P, Herceg Z. Bariatric surgery-induced weight loss and associated genome-wide DNA-methylation alterations in obese individuals. Clin Epigenetics 2022; 14:176. [PMID: 36528638 PMCID: PMC9759858 DOI: 10.1186/s13148-022-01401-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Obesity is a multifactorial and chronic condition of growing universal concern. It has recently been reported that bariatric surgery is a more successful treatment for severe obesity than other noninvasive interventions, resulting in rapid significant weight loss and associated chronic disease remission. The identification of distinct epigenetic patterns in patients who are obese or have metabolic imbalances has suggested a potential role for epigenetic alterations in causal or mediating pathways in the development of obesity-related pathologies. Specific changes in the epigenome (DNA methylome), associated with metabolic disorders, can be detected in the blood. We investigated whether such epigenetic changes are reversible after weight loss using genome-wide DNA methylome analysis of blood samples from individuals with severe obesity (mean BMI ~ 45) undergoing bariatric surgery. RESULTS Our analysis revealed 41 significant (Bonferroni p < 0.05) and 1169 (false discovery rate p < 0.05) suggestive differentially methylated positions (DMPs) associated with weight loss due to bariatric surgery. Among the 41 significant DMPs, 5 CpGs were replicated in an independent cohort of BMI-discordant monozygotic twins (the heavier twin underwent diet-induced weight loss). The effect sizes of these 5 CpGs were consistent across discovery and replication sets (p < 0.05). We also identified 192 differentially methylated regions (DMRs) among which SMAD6 and PFKFB3 genes were the top hypermethylated and hypomethylated regions, respectively. Pathway enrichment analysis of the DMR-associated genes showed that functional pathways related to immune function and type 1 diabetes were significant. Weight loss due to bariatric surgery also significantly decelerated epigenetic age 12 months after the intervention (mean = - 4.29; p = 0.02). CONCLUSIONS We identified weight loss-associated DNA-methylation alterations targeting immune and inflammatory gene pathways in blood samples from bariatric-surgery patients. The top hits were replicated in samples from an independent cohort of BMI-discordant monozygotic twins following a hypocaloric diet. Energy restriction and bariatric surgery thus share CpGs that may represent early indicators of response to the metabolic effects of weight loss. The analysis of bariatric surgery-associated DMRs suggests that epigenetic regulation of genes involved in endothelial and adipose tissue function is key in the pathophysiology of obesity.
Collapse
Affiliation(s)
- Fazlur Rahman Talukdar
- Epigenomics and Mechanisms Branch, International Agency for Research On Cancer (IARC), 150 Cours Albert Thomas, Lyon, France
| | - David Israel Escobar Marcillo
- Section of Mechanisms, Biomarkers and Models, Dept Environment and Health, Istituto Superiore Di Sanità, Viale Regina Elena, No. 299, 00161 Rome, Italy
- Obesity Center-Internal Medicine Unit, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Ruhina Shirin Laskar
- Nutrition and Metabolism Branch, International Agency for Research On Cancer (IARC), 150 Cours Albert Thomas, Lyon, France
| | - Alexei Novoloaca
- Epigenomics and Mechanisms Branch, International Agency for Research On Cancer (IARC), 150 Cours Albert Thomas, Lyon, France
| | - Cyrille Cuenin
- Epigenomics and Mechanisms Branch, International Agency for Research On Cancer (IARC), 150 Cours Albert Thomas, Lyon, France
| | - Paolo Sbraccia
- Obesity Center-Internal Medicine Unit, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Lorenza Nisticò
- Centre for Behavioral Sciences and Mental Health, Istituto Superiore Di Sanità, Viale Regina Elena, No. 299, 00161 Rome, Italy
| | - Valeria Guglielmi
- Obesity Center-Internal Medicine Unit, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Tarik Gheit
- Early Detection, Prevention, and Infections Branch, International Agency for Research On Cancer (IARC), 150 Cours Albert Thomas, Lyon, France
| | | | - Eugenia Dogliotti
- Section of Mechanisms, Biomarkers and Models, Dept Environment and Health, Istituto Superiore Di Sanità, Viale Regina Elena, No. 299, 00161 Rome, Italy
| | - Paola Fortini
- Section of Mechanisms, Biomarkers and Models, Dept Environment and Health, Istituto Superiore Di Sanità, Viale Regina Elena, No. 299, 00161 Rome, Italy
| | - Zdenko Herceg
- Epigenomics and Mechanisms Branch, International Agency for Research On Cancer (IARC), 150 Cours Albert Thomas, Lyon, France
| |
Collapse
|
10
|
Kenmochi M, Kawarasaki S, Takizawa S, Okamura K, Goto T, Uchida K. Involvement of mechano-sensitive Piezo1 channel in the differentiation of brown adipocytes. J Physiol Sci 2022; 72:13. [PMID: 35725398 PMCID: PMC10717802 DOI: 10.1186/s12576-022-00837-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 05/22/2022] [Indexed: 11/10/2022]
Abstract
Brown adipocytes expend energy via heat production and are a potential target for the prevention of obesity and related metabolic disorders. Piezo1 is a Ca2+-permeable non-selective cation channel activated by mechanical stimuli. Piezo1 is reported to be involved in mechano-sensation in non-sensory tissues. However, the expression and roles of Piezo1 in brown adipocytes have not been well clarified. Here, we generated a brown adipocyte line derived from UCP1-mRFP1 transgenic mice and showed that Piezo1 is expressed in pre-adipocytes. Application of Yoda-1, a Piezo1 agonist, suppressed brown adipocyte differentiation, and this suppression was significantly attenuated by treatment with a Piezo1 antagonist and by Piezo1 knockdown. Furthermore, the suppression of brown adipocyte differentiation by Yoda-1 was abolished by co-treatment with a calcineurin inhibitor. Thus, these results suggest that activation of Piezo1 suppresses brown adipocyte differentiation via the calcineurin pathway.
Collapse
Affiliation(s)
- Manato Kenmochi
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Satoko Kawarasaki
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, 611-0011, Japan
| | - Satsuki Takizawa
- Laboratory of Functional Physiology, Department of Environmental and Life Sciences, School of Food and Nutritional Sciences, University of Shizuoka, Yada 52-1, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Kazuhiko Okamura
- Department of Morphological Biology, Fukuoka Dental College, Fukuoka, 814-0193, Japan
| | - Tsuyoshi Goto
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, 611-0011, Japan
| | - Kunitoshi Uchida
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan.
- Laboratory of Functional Physiology, Department of Environmental and Life Sciences, School of Food and Nutritional Sciences, University of Shizuoka, Yada 52-1, Suruga-ku, Shizuoka, 422-8526, Japan.
| |
Collapse
|
11
|
Wang J, Onogi Y, Krueger M, Oeckl J, Karlina R, Singh I, Hauck SM, Feederle R, Li Y, Ussar S. PAT2 regulates vATPase assembly and lysosomal acidification in brown adipocytes. Mol Metab 2022; 61:101508. [PMID: 35513259 PMCID: PMC9114668 DOI: 10.1016/j.molmet.2022.101508] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 04/26/2022] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE Brown adipocytes play a key role in maintaining body temperature as well as glucose and lipid homeostasis. However, brown adipocytes need to adapt their thermogenic activity and substrate utilization to changes in nutrient availability. Amongst the multiple factors influencing brown adipocyte activity, autophagy is an important regulatory element of thermogenic capacity and activity. Nevertheless, a specific sensing mechanism of extracellular amino acid availability linking autophagy to nutrient availability in brown adipocytes is unknown. METHODS To characterize the role of the amino acid transporter PAT2/SLC36A2 in brown adipocytes, loss or gain of function of PAT2 were studied with respect to differentiation, subcellular localization, lysosomal activity and autophagy. Activity of vATPase was evaluated by quenching of EGFP fused to LC3 or FITC-dextran loaded lysosomes in brown adipocytes upon amino acid starvation, whereas the effect of PAT2 on assembly of the vATPase was investigated by Native-PAGE. RESULTS We show that PAT2 translocates from the plasma membrane to the lysosome in response to amino acid withdrawal. Loss or overexpression of PAT2 impair lysosomal acidification and starvation induced S6K re-phosphorylation, as PAT2 facilitates the assembly of the lysosomal vATPase, by recruitment of the cytoplasmic V1 subunit to the lysosome. CONCLUSION PAT2 is an important sensor of extracellular amino acids and regulator of lysosomal acidification in brown adipocytes.
Collapse
Affiliation(s)
- Jiefu Wang
- RG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Yasuhiro Onogi
- RG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Martin Krueger
- Institute for Anatomy, University of Leipzig, 04103, Leipzig, Germany
| | - Josef Oeckl
- Chair for Molecular Nutritional Medicine TUM School for Life Sciences,Technical University Munich, Munich, Germany
| | - Ruth Karlina
- RG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Inderjeet Singh
- RG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Stefanie M Hauck
- German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany; Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Regina Feederle
- German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany; Monoclonal Antibody Core Facility, Institute for Diabetes & Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany
| | - Yongguo Li
- Chair for Molecular Nutritional Medicine TUM School for Life Sciences,Technical University Munich, Munich, Germany
| | - Siegfried Ussar
- RG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany; Department of Medicine, Technische Universität München, Munich, Germany.
| |
Collapse
|
12
|
Abstract
Obesity is a chronic and complex psychosomatic disease that is becoming increasingly prevalent worldwide. This study aimed to analyze whole methylation profiles to uncover the epigenetic mechanisms associated with obesity. DNA methylation profiles in blood samples from patients with obesity and normal controls were studied using the Illumina 850 K methylation microarray. The diagnostic value of the differentially methylated genes was determined using receiver operating characteristic (ROC) analysis. The expression of selected candidate genes was verified using reverse transcription quantitative polymerase chain reaction (RT-qPCR) and pyrosequencing. A total of 9,371 significantly differentially methylated sites (7,974 hypermethylated sites and 1,397 hypomethylated sites) were identified in 4,571 genes. A difference in the distribution of differentially methylated sites (hypermethylated and hypomethylated) in both gene structures and CpG islands was observed. A total of 114 key differentially methylated sites were identified in the CpG islands. ROC results indicated that Inhibin Subunit Beta B (INHBB), Homeobox A9 (HOXA9), Troponin T3 (TNNT3), Cyclic adenosine monophosphate (cAMP)-responsive element binding protein (CREB)-regulated transcription coactivator 1 (CRTC1) and Zinc finger and BTB domain-containing 7 B (ZBTB7B) could discriminate patients with obesity from normal controls. RT-qPCR results of CRTC1 and ZBTB7B were consistent with our methylation profile results. The pyrosequencing results showed that the methylation levels of CRTC1 CpG sites (CpG1 and CpG2-cg11660071) and INHBB CpG sites (CpG2) were significantly changed in patients with obesity compared with normal controls, which was consistent with our DNA methylation profile results. Our study provides new insights into the pathological mechanism of obesity.
Collapse
Affiliation(s)
- Chunhu Wang
- 17th Department of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Meng Wang
- 17th Department of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiguang Ma
- 17th Department of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
13
|
Kuan CY, Lin YY, Yang IH, Chen CY, Chi CY, Li CH, Chen ZY, Lin LZ, Yang CC, Lin FH. The Synthesis of Europium-Doped Calcium Carbonate by an Eco-Method as Free Radical Generator Under Low-Intensity Ultrasonic Irradiation for Body Sculpture. Front Bioeng Biotechnol 2021; 9:765630. [PMID: 34869278 PMCID: PMC8639516 DOI: 10.3389/fbioe.2021.765630] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/20/2021] [Indexed: 11/17/2022] Open
Abstract
Body sculpture is a common method to remove excessive fat. The diet and exercise are the first suggestion to keep body shape; however, those are difficult to keep adherence. Ultrasound has been developed for fat ablation; however, it could only serve as the side treatment along with liposuction. In the study, a sonosensitizer of europium-doped calcium carbonate (CaCO3: Eu) would be synthesized by an eco-method and combined with low-intensity ultrasound for lipolysis. The crystal structure of CaCO3: Eu was identified by x-ray diffractometer (XRD). The morphology of CaCO3: Eu was analyzed by scanning electron microscope (SEM). The chemical composition of CaCO3: Eu was evaluated by energy-dispersed spectrophotometer (EDS) and inductively coupled plasma mass spectrometer (ICP-MS). The electronic diffraction pattern was to further check crystal structure of the synthesized individual grain by transmission electron microscope (TEM). The particle size was determined by Zeta-sizer. Water-soluble tetrazolium salt (WST-1) were used to evaluate the cell viability. Chloromethyl-2′,7′-dichlorofluorescein diacetate (CM-H2DCFDA) and live/dead stain were used to evaluate feasibility in vitro. SD-rat was used to evaluate the safety and efficacy in vivo. The results showed that CaCO3: Eu had good biocompatibility and could produce reactive oxygen species (ROS) after treated with low-intensity ultrasound. After 4-weeks, the CaCO3: Eu exposed to ultrasound irradiation on SD rats could significantly decrease body weight, waistline, and subcutaneous adipose tissue. We believe that ROS from sonoluminescence, CO2-bomb and locally increasing Ca2+ level would be three major mechanisms to remove away adipo-tissue and inhibit adipogenesis. We could say that the combination of the CaCO3: Eu and low-intensity ultrasound would be a non-invasive treatment for the body sculpture.
Collapse
Affiliation(s)
- Che-Yung Kuan
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan.,Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County, Taiwan
| | - Yu-Ying Lin
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County, Taiwan.,Ph.D. Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung, Taiwan
| | - I-Hsuan Yang
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan.,Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County, Taiwan
| | - Ching-Yun Chen
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Chih-Ying Chi
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County, Taiwan.,Ph.D. Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung, Taiwan.,Biomaterials Translational Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Chi-Han Li
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County, Taiwan.,Ph.D. Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Zhi-Yu Chen
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan.,Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County, Taiwan
| | - Li-Ze Lin
- Department of Materials Science and Engineering, National United University, Miaoli County, Taiwan
| | - Chun-Chen Yang
- Department of Materials Science and Engineering, National Taiwan University, Taipei, Taiwan
| | - Feng-Huei Lin
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan.,Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County, Taiwan
| |
Collapse
|
14
|
Lee D, Kwak HJ, Kim BH, Kim SH, Kim DW, Kang KS. Combined Anti-Adipogenic Effects of Hispidulin and p-Synephrine on 3T3-L1 Adipocytes. Biomolecules 2021; 11:biom11121764. [PMID: 34944408 PMCID: PMC8698582 DOI: 10.3390/biom11121764] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 12/14/2022] Open
Abstract
Hispidulin is abundant in Arrabidaea chica, Crossostephium chinense, and Grindelia argentina, among others. p-Synephrine is the main phytochemical constituent of Citrus aurantium. It has been used in combination with various other phytochemicals to determine synergistic effects in studies involving human participants. However, there have been no reports comparing the anti-adipogenic effects of the combination of hispidulin and p-synephrine. The current study explores the anti-adipogenic effects of hispidulin alone and in combination with p-synephrine in a murine preadipocyte cell line, 3T3-L1. Co-treatment resulted in a greater inhibition of the formation of red-labeled lipid droplets than the hispidulin or p-synephrine-alone treatments. Co-treatment with hispidulin and p-synephrine also significantly inhibited adipogenic marker proteins, including Akt, mitogen-activated protein kinases, peroxisome proliferator-activated receptor gamma, CCAAT/enhancer-binding protein alpha, glucocorticoid receptor, and CCAAT/enhancer-binding protein β. Although further studies are required to assess the effects of each drug on pharmacokinetic parameters, a combination treatment with hispidulin and p-synephrine may be a potential alternative strategy for developing novel anti-obesity drugs.
Collapse
Affiliation(s)
- Dahae Lee
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea; (D.L.); (S.H.K.)
| | - Hee Jae Kwak
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, Korea;
| | | | - Seung Hyun Kim
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea; (D.L.); (S.H.K.)
| | - Dong-Wook Kim
- Department of Pharmaceutical Engineering, Cheongju University, Cheongju 28530, Korea
- Correspondence: (D.-W.K.); (K.S.K.); Tel.: +82-43-229-7984 (D.-W.K.); +82-31-750-5402 (K.S.K.)
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea; (D.L.); (S.H.K.)
- Correspondence: (D.-W.K.); (K.S.K.); Tel.: +82-43-229-7984 (D.-W.K.); +82-31-750-5402 (K.S.K.)
| |
Collapse
|
15
|
Chung CW, Kaminski Schierle GS. Intracellular Thermometry at the Micro-/Nanoscale and its Potential Application to Study Protein Aggregation Related to Neurodegenerative Diseases. Chembiochem 2021; 22:1546-1558. [PMID: 33326160 DOI: 10.1002/cbic.202000765] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/14/2020] [Indexed: 11/11/2022]
Abstract
Temperature is a fundamental physical parameter that influences biological processes in living cells. Hence, intracellular temperature mapping can be used to derive useful information reflective of thermodynamic properties and cellular behaviour. Herein, existing publications on different thermometry systems, focusing on those that employ fluorescence-based techniques, are reviewed. From developments based on fluorescent proteins and inorganic molecules to metal nanoclusters and fluorescent polymers, the general findings of intracellular measurements from different research groups are discussed. Furthermore, the contradiction of mitochondrial thermogenesis and nuclear-cytoplasmic temperature differences to current thermodynamic understanding are highlighted. Lastly, intracellular thermometry is proposed as a tool to quantify the energy flow and cost associated with amyloid-β42 (Aβ42) aggregation, a hallmark of Alzheimer's disease.
Collapse
Affiliation(s)
- Chyi Wei Chung
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Phillipa Fawcett Drive, Cambridge, CB3 0AS, UK
| | - Gabriele S Kaminski Schierle
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Phillipa Fawcett Drive, Cambridge, CB3 0AS, UK
| |
Collapse
|
16
|
Muley C, Kotschi S, Bartelt A. Role of Ubiquilins for Brown Adipocyte Proteostasis and Thermogenesis. Front Endocrinol (Lausanne) 2021; 12:739021. [PMID: 34650520 PMCID: PMC8505974 DOI: 10.3389/fendo.2021.739021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/14/2021] [Indexed: 11/13/2022] Open
Abstract
The acclimatization of brown adipose tissue (BAT) to sustained cold exposure requires an adaptive increase in proteasomal protein quality control. Ubiquilins represent a recently identified family of shuttle proteins with versatile functions in protein degradation, such as facilitating substrate targeting and proteasomal degradation. However, whether ubiquilins participate in brown adipocyte function has not been investigated so far. Here, we determine the role of ubiquilins for proteostasis and non-shivering thermogenesis in brown adipocytes. We found that Ubqln1, 2 and 4 are highly expressed in BAT and their expression was induced by cold and proteasomal inhibition. Surprisingly, silencing of ubiquilin gene expression (one or multiple in combinations) did not lead to aggravated ER stress or inflammation. Moreover, ubiquitin level and proteasomal activity under basal conditions were not impacted by loss of ubiquilins. Also, non-shivering thermogenesis measured by norepinephrine-induced respiration remained intact after loss of ubiquilins. In conclusion, ubiquilin proteins are highly abundant in BAT and regulated by cold, but they are dispensable for brown adipocyte proteostasis and thermogenesis.
Collapse
Affiliation(s)
- Carolin Muley
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Stefan Kotschi
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Technische Universität München, Munich, Germany
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Molecular Metabolism & Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA, United States
- *Correspondence: Alexander Bartelt,
| |
Collapse
|
17
|
Abstract
As an important second messenger in adipocytes, calcium ions (Ca2+) are essential in regulating various intracellular signalling pathways that control critical cellular functions. Calcium channels show selective permeability to Ca2+ and facilitate Ca2+ entry into the cytoplasm, which are normally located in the plasmatic and intracellular membranes. The increase of cytosolic Ca2+ modulates a variety of signalling pathways and results in the transcription of target genes that contribute to adipogenesis, a key cellular event includes proliferation and differentiation of adipocyte. In the past decades, the involvement of some Ca2+-permeable ion channels, such as Ca2+ release-activated Ca2+ channels, transient receptor potential channels, voltage-gated calcium channels and others, in adipogenesis has been extensively explored. In the present review, we provided a summary of the expression and contributions of these Ca2+-permeable channels in mediating Ca2+ influxes that drive adipogenesis. Moreover, we discussed their potentials as future therapeutic targets.
Collapse
Affiliation(s)
- Mingzhu Zhai
- Huazhong University of Science and Technology Union Shenzhen Hospital and the 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
- Department of Orthopaedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Dazhi Yang
- Huazhong University of Science and Technology Union Shenzhen Hospital and the 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
- Department of Orthopaedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Weihong Yi
- Huazhong University of Science and Technology Union Shenzhen Hospital and the 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
- Department of Orthopaedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Wuping Sun
- Huazhong University of Science and Technology Union Shenzhen Hospital and the 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
18
|
Jiao Y, Liu L, Gu H, Liang X, Meng X, Gao J, Xu Y, Nuermaimaiti N, Guan Y. Ad36 promotes differentiation of hADSCs into brown adipocytes by up-regulating LncRNA ROR. Life Sci 2020; 265:118762. [PMID: 33189825 DOI: 10.1016/j.lfs.2020.118762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/28/2020] [Accepted: 11/10/2020] [Indexed: 01/04/2023]
Abstract
AIMS This study is to investigate the role of adenovirus type 36 (Ad36) in inducing differentiation of human adipose-derived stem cells (hADSCs) into brown adipocytes. MAIN METHODS The hADSCs were induced to differentiate into adipocytes by a cocktail method and Ad36, respectively. They were collected on the 2nd, 4th, 6th, and 8th day, respectively. LncRNA ROR was silenced by siRNA. RT-qPCR and Western-blot were used to detect the mRNA and protein levels. Transmission electron microscopy was used to observe the mitochondria. KEY FINDINGS The mRNA and protein expression levels of LncRNA ROR, Cidea, Dio2, Fgf21, Ucp1, Prdm16, Cox5b, Atp5o, Atp6, and Nd2 in the Ad36 induction group were significantly higher than those in the cocktail induction group. The expression levels of Leptin mRNA and protein in the Ad36 induction group were significantly lower than those in the cocktail induction group. After siRNA knockdown of LncRNA ROR, mRNA and protein expression levels of Cidea, Dio2, Fgf21, Ucp1, Prdm16, Cox5b, Atp5o, Atp6 and Nd2 were significantly lower than the control group during the induction of hADSC differentiation into adipocytes by Ad36. Additionally, mitochondria in the Ad36 induction group was increased compared to that in the cocktail induction group. SIGNIFICANCE Ad36 may promote the differentiation of hADSCs into brown adipocytes by up-regulating LncRNA ROR.
Collapse
Affiliation(s)
- Yi Jiao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830011, Xinjiang, China
| | - Ling Liu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830011, Xinjiang, China
| | - Hao Gu
- Department of Liver and Laparoscopic Surgery, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Xiaodi Liang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830011, Xinjiang, China
| | - Xuanyu Meng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830011, Xinjiang, China
| | - Jiale Gao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830011, Xinjiang, China
| | - Youzongsheng Xu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830011, Xinjiang, China
| | - Nuerbiye Nuermaimaiti
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830011, Xinjiang, China
| | - Yaqun Guan
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830011, Xinjiang, China.
| |
Collapse
|
19
|
Friend MA, Bhanugopan MS, McGrath SR, Edwards JH, Hancock S, Loudon K, Miller D, McGilchrist P, Refshauge G, Robertson SM, Thompson AN, Masters DG. Do calcium and magnesium deficiencies in reproducing ewes contribute to high lamb mortality? ANIMAL PRODUCTION SCIENCE 2020. [DOI: 10.1071/an17588] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
High lamb mortality continues to be a significant economic and welfare problem within the Australian sheep industry, with 20–30% of lambs born in commercial flocks dying mostly within 3 days of birth. Clinical hypocalcaemia and hypomagnesaemia cause ewe mortality, and, subsequently, either fetal or lamb death, but it is not known whether subclinical deficiencies of calcium (Ca) and magnesium (Mg) compromise lamb survival. This review considers the potential mechanisms through which Ca and Mg deficiencies may influence lamb survival, and factors influencing the risk of deficiency. Pastures grazed by lambing ewes may be marginal in calcium (Ca; <4 g/kg DM) and magnesium (Mg; <0.9 g/kg DM) but also have a high dietary cation–anion difference (>12 meq/100 g DM) and high concentrations of potassium (K; >30 g/kg DM) and nitrogen. In young cereal crops, sodium concentrations are also often low (<0.9 g/kg DM). This combination of minerals and other nutrients creates an imbalance in supply and increases susceptibility to acute Ca (hypocalcaemia) and Mg (hypomagnesaemia) deficiency. Calcium is required for smooth muscle function and has a direct role in uterine contraction, so may influence the duration of parturition. Low Ca and Mg intake both influence insulin release and sensitivity, low Mg results in poor glycaemic control and insulin resistance by impairing both insulin secretion and its action on peripheral tissues, also potentially altering the duration of parturition as well as risk of metabolic disease. Magnesium is also a neuroprotectant that slows the neuronal damage during hypoxia and has been linked with thermogenesis in offspring and increased immunoglobulins in colostrum. These functions indicate potential importance in improving the ease of parturition and improved ability of the newborn lamb to thermoregulate and survive after birth. Subclinical Ca and Mg deficiencies commonly occur in 20% of lambing ewes grazing temperate pastures, so further studies are warranted to investigate whether correction of these deficiencies can improve lamb survival.
Collapse
|
20
|
Kanojia D, Dakle P, Mayakonda A, Parameswaran R, Puhaindran ME, Min VLK, Madan V, Koeffler P. Identification of somatic alterations in lipoma using whole exome sequencing. Sci Rep 2019; 9:14370. [PMID: 31591430 PMCID: PMC6779901 DOI: 10.1038/s41598-019-50805-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/19/2019] [Indexed: 01/09/2023] Open
Abstract
Lipomas are benign fatty tumors with a high prevalence rate, mostly found in adults but have a good prognosis. Until now, reason for lipoma occurrence not been identified. We performed whole exome sequencing to define the mutational spectrum in ten lipoma patients along with their matching control samples. We presented genomic insight into the development of lipomas, the most common benign tumor of soft tissue. Our analysis identified 412 somatic variants including missense mutations, splice site variants, frameshift indels, and stop gain/lost. Copy number variation analysis highlighted minor aberrations in patients. Kinase genes and transcriptions factors were among the validated mutated genes critical for cell proliferation and survival. Pathway analysis revealed enrichment of calcium, Wnt and phospholipase D signaling in patients. In conclusion, whole exome sequencing in lipomas identified mutations in genes with a possible role in development and progression of lipomas.
Collapse
Affiliation(s)
- Deepika Kanojia
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
| | - Pushkar Dakle
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Anand Mayakonda
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rajeev Parameswaran
- Division of Surgical Oncology, National University Cancer Institute, Singapore, Singapore
| | - Mark E Puhaindran
- Department of Hand and Reconstructive Microsurgery, National University Hospital, Singapore, Singapore
| | | | - Vikas Madan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Phillip Koeffler
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Division of Hematology/Oncology, Cedars-Sinai Medical Center, University of California, School of Medicine, Los Angeles, California, USA
- National University Cancer Institute, National University Hospital, Singapore, Singapore
| |
Collapse
|
21
|
Schaar A, Sun Y, Sukumaran P, Rosenberger TA, Krout D, Roemmich JN, Brinbaumer L, Claycombe-Larson K, Singh BB. Ca 2+ entry via TRPC1 is essential for cellular differentiation and modulates secretion via the SNARE complex. J Cell Sci 2019; 132:jcs.231878. [PMID: 31182642 PMCID: PMC6633397 DOI: 10.1242/jcs.231878] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/27/2019] [Indexed: 12/17/2022] Open
Abstract
Properties of adipocytes, including differentiation and adipokine secretion, are crucial factors in obesity-associated metabolic syndrome. Here, we provide evidence that Ca2+ influx in primary adipocytes, especially upon Ca2+ store depletion, plays an important role in adipocyte differentiation, functionality and subsequently metabolic regulation. The endogenous Ca2+ entry channel in both subcutaneous and visceral adipocytes was found to be dependent on TRPC1–STIM1, and blocking Ca2+ entry with SKF96365 or using TRPC1−/− knockdown adipocytes inhibited adipocyte differentiation. Additionally, TRPC1−/− mice have decreased organ weight, but increased adipose deposition and reduced serum adiponectin and leptin concentrations, without affecting total adipokine expression. Mechanistically, TRPC1-mediated Ca2+ entry regulated SNARE complex formation, and agonist-mediated secretion of adipokine-loaded vesicles was inhibited in TRPC1−/− adipose. These results suggest an unequivocal role of TRPC1 in adipocyte differentiation and adiponectin secretion, and that loss of TRPC1 disturbs metabolic homeostasis. This article has an associated First Person interview with the first author of the paper. Summary: TRPC1 modulates Ca2+ entry, which is essential in adipocyte differentiation and adiponectin secretion, through facilitating SNARE complex formation, thereby maintaining metabolic homeostasis.
Collapse
Affiliation(s)
- Anne Schaar
- Department of Biomedical Science, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - Yuyang Sun
- Department of Biomedical Science, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - Pramod Sukumaran
- Department of Biomedical Science, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - Thad A Rosenberger
- Department of Biomedical Science, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - Danielle Krout
- US Department of Agriculture-Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA
| | - James N Roemmich
- US Department of Agriculture-Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA
| | - Lutz Brinbaumer
- Neurobiology Laboratory, NIHES, NIH, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA.,Institute of Biomedical Research, (BIOMED) Catholic University of Argentina, Av. Alicia Moreau de Justo 1300, Edificio San Jose Piso 3, Buenos Aires C1107AAZ, Argentina
| | - Kate Claycombe-Larson
- US Department of Agriculture-Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA
| | - Brij B Singh
- Department of Biomedical Science, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| |
Collapse
|
22
|
Zhang F, Su H, Song M, Zheng J, Liu F, Yuan C, Fu Q, Chen S, Zhu X, Wang L, Gao P, Shu G, Jiang Q, Wang S. Calcium Supplementation Alleviates High-Fat Diet-Induced Estrous Cycle Irregularity and Subfertility Associated with Concomitantly Enhanced Thermogenesis of Brown Adipose Tissue and Browning of White Adipose Tissue. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:7073-7081. [PMID: 31240927 DOI: 10.1021/acs.jafc.9b02663] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Obesity has been demonstrated as a disruptor of female fertility. Our previous study showed the antiobesity effects of calcium on HFD-fed male mice. However, the role of calcium in alleviating reproductive dysfunction of HFD-fed female mice remains unclear. Here, we found that HFD led to estrus cycle irregularity (longer cycle duration and shorter estrus period) and subfertility (longer conception time, lower fertility index, and less implantations) in mice. However, the HFD-induced reproductive abnormality was alleviated by calcium supplementation. Additionally, calcium supplementation enhanced activation/thermogenesis of BAT and browning of WAT in HFD-fed mice. Consequently, the abnormality of energy metabolism and glucose homeostasis induced by HFD were improved by calcium supplementation, with elevated metabolic rates and core temperature. In conclusion, these data showed that calcium supplementation alleviated HFD-induced estrous cycle irregularity and subfertility associated with concomitantly enhanced BAT thermogenesis and WAT browning, suggesting the potential application of calcium in improving obesity-related reproductive disorders.
Collapse
MESH Headings
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/physiopathology
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/physiopathology
- Animals
- Calcium/administration & dosage
- Diet, High-Fat/adverse effects
- Dietary Supplements/analysis
- Energy Metabolism/drug effects
- Estrous Cycle/drug effects
- Female
- Genital Diseases, Female/drug therapy
- Genital Diseases, Female/etiology
- Genital Diseases, Female/metabolism
- Genital Diseases, Female/physiopathology
- Humans
- Infertility/drug therapy
- Infertility/etiology
- Infertility/metabolism
- Infertility/physiopathology
- Male
- Mice
- Mice, Inbred C57BL
- Obesity/complications
- Thermogenesis/drug effects
Collapse
|
23
|
Zhang F, Ye J, Zhu X, Wang L, Gao P, Shu G, Jiang Q, Wang S. Anti-Obesity Effects of Dietary Calcium: The Evidence and Possible Mechanisms. Int J Mol Sci 2019; 20:E3072. [PMID: 31234600 PMCID: PMC6627166 DOI: 10.3390/ijms20123072] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/18/2019] [Accepted: 06/20/2019] [Indexed: 02/07/2023] Open
Abstract
Obesity is a serious health challenge worldwide and is associated with various comorbidities, including dyslipidemia, type 2 diabetes, and cardiovascular disease. Developing effective strategies to prevent obesity is therefore of paramount importance. One potential strategy to reduce obesity is to consume calcium, which has been implicated to be involved in reducing body weight/fat. In this review, we compile the evidence for the anti-obesity roles of calcium in cells, animals, and humans. In addition, we summarize the possible anti-obesity mechanisms of calcium, including regulation of (a) adipogenesis, (b) fat metabolism, (c) adipocyte (precursor) proliferation and apoptosis, (d) thermogenesis, (e) fat absorption and excretion, and (f) gut microbiota. Although the exact anti-obesity roles of calcium in different subjects and how calcium induces the proposed anti-obesity mechanisms need to be further investigated, the current evidence demonstrates the anti-obesity effects of calcium and suggests the potential application of dietary calcium for prevention of obesity.
Collapse
Affiliation(s)
- Fenglin Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- National Engineering Research Center for Breeding Swine Industry and ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou 510642, China.
| | - Jingjing Ye
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- National Engineering Research Center for Breeding Swine Industry and ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou 510642, China.
| | - Xiaotong Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- National Engineering Research Center for Breeding Swine Industry and ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou 510642, China.
| | - Lina Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- National Engineering Research Center for Breeding Swine Industry and ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou 510642, China.
| | - Ping Gao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- National Engineering Research Center for Breeding Swine Industry and ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou 510642, China.
| | - Gang Shu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- National Engineering Research Center for Breeding Swine Industry and ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou 510642, China.
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- National Engineering Research Center for Breeding Swine Industry and ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou 510642, China.
| | - Songbo Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- National Engineering Research Center for Breeding Swine Industry and ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
24
|
Calcium Signaling Pathways: Key Pathways in the Regulation of Obesity. Int J Mol Sci 2019; 20:ijms20112768. [PMID: 31195699 PMCID: PMC6600289 DOI: 10.3390/ijms20112768] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 05/29/2019] [Accepted: 05/31/2019] [Indexed: 02/07/2023] Open
Abstract
Nowadays, high epidemic obesity-triggered hypertension and diabetes seriously damage social public health. There is now a general consensus that the body's fat content exceeding a certain threshold can lead to obesity. Calcium ion is one of the most abundant ions in the human body. A large number of studies have shown that calcium signaling could play a major role in increasing energy consumption by enhancing the metabolism and the differentiation of adipocytes and reducing food intake through regulating neuronal excitability, thereby effectively decreasing the occurrence of obesity. In this paper, we review multiple calcium signaling pathways, including the IP3 (inositol 1,4,5-trisphosphate)-Ca2+ (calcium ion) pathway, the p38-MAPK (mitogen-activated protein kinase) pathway, and the calmodulin binding pathway, which are involved in biological clock, intestinal microbial activity, and nerve excitability to regulate food intake, metabolism, and differentiation of adipocytes in mammals, resulting in the improvement of obesity.
Collapse
|
25
|
Identification of Cyanobacterial Strains with Potential for the Treatment of Obesity-Related Co-Morbidities by Bioactivity, Toxicity Evaluation and Metabolite Profiling. Mar Drugs 2019; 17:md17050280. [PMID: 31083362 PMCID: PMC6562398 DOI: 10.3390/md17050280] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 05/03/2019] [Accepted: 05/07/2019] [Indexed: 12/13/2022] Open
Abstract
Obesity is a complex disease resulting in several metabolic co-morbidities and is increasing at epidemic rates. The marine environment is an interesting resource of novel compounds and in particular cyanobacteria are well known for their capacity to produce novel secondary metabolites. In this work, we explored the potential of cyanobacteria for the production of compounds with relevant activities towards metabolic diseases using a blend of target-based, phenotypic and zebrafish assays as whole small animal models. A total of 46 cyanobacterial strains were grown and biomass fractionated, yielding in total 263 fractions. Bioactivities related to metabolic function were tested in different in vitro and in vivo models. Studying adipogenic and thermogenic gene expression in brown adipocytes, lipid metabolism and glucose uptake in hepatocytes, as well as lipid metabolism in zebrafish larvae, we identified 66 (25%) active fractions. This together with metabolite profiling and the evaluation of toxicity allowed the identification of 18 (7%) fractions with promising bioactivity towards different aspects of metabolic disease. Among those, we identified several known compounds, such as eryloside T, leptosin F, pheophorbide A, phaeophytin A, chlorophyll A, present as minor peaks. Those compounds were previously not described to have bioactivities in metabolic regulation, and both known or unknown compounds could be responsible for such effects. In summary, we find that cyanobacteria hold a huge repertoire of molecules with specific bioactivities towards metabolic diseases, which needs to be explored in the future.
Collapse
|
26
|
Al-Anazi A, Parhar R, Saleh S, Al-Hijailan R, Inglis A, Al-Jufan M, Bazzi M, Hashmi S, Conca W, Collison K, Al-Mohanna F. Intracellular calcium and NF- kB regulate hypoxia-induced leptin, VEGF, IL-6 and adiponectin secretion in human adipocytes. Life Sci 2018; 212:275-284. [PMID: 30308181 DOI: 10.1016/j.lfs.2018.10.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/19/2018] [Accepted: 10/06/2018] [Indexed: 11/26/2022]
Abstract
AIMS Hypoxia-induced adipokine release has been attributed mainly to HIF-1α. Here we investigate the role of intracellular calcium and NF-kB in the hypoxia-dependent release of leptin, VEGF, IL-6 and the hypoxia-induced inhibition of adiponectin release in human adipocytes. MAIN METHODS We used intracellular calcium imaging to compare calcium status in preadipocytes and in adipocytes. We subjected both cell types to hypoxic conditions and measured the release of adipokines induced by hypoxia in the presence and absence of HIF-1α inhibitor YC-1, NF-κB inhibitor SN50 and intracellular calcium chelator BAPTA-AM. KEY FINDINGS We demonstrate reduced intracellular calcium oscillations and increased oxidative stress as the cells transitioned from preadipocytes to adipocytes. We show that differentiation of preadipocytes to adipocytes is associated with distinct morphological changes in the mitochondria. We also show that hypoxia-induced secretion of leptin, VEGF, IL-6 and hypoxia-induced inhibition of adiponectin secretion are independent of HIF-1α expression. The hypoxia-induced leptin, VEGF and IL-6 release are [Ca++]i dependent whereas adiponectin is NF-kB dependent. SIGNIFICANCE Our work suggests a major role for [Ca++]i in preadipocyte differentiation to adipocytes and that changes in mitochondrial morphology in the adipocytes might underlie the reduced calcium oscillations observed in the adipocytes. It also demonstrates that multiple signaling pathways are associated with the hypoxia-induced adipokine secretion.
Collapse
Affiliation(s)
- Azizah Al-Anazi
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Ranjit Parhar
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Soad Saleh
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Reem Al-Hijailan
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Angela Inglis
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Mansour Al-Jufan
- Heart Centre, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Mohammed Bazzi
- Department of Biochemistry, College of Science, King Saud University, Riyadh 12372, Saudi Arabia
| | - Sarwar Hashmi
- Developmental Biology, Center for Vector Biology, Rutgers University, New Brunswick, NJ 08901, United States
| | - Walter Conca
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| | - Kate Collison
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Futwan Al-Mohanna
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia.
| |
Collapse
|
27
|
Bishnoi M, Khare P, Brown L, Panchal SK. Transient receptor potential (TRP) channels: a metabolic TR(i)P to obesity prevention and therapy. Obes Rev 2018; 19:1269-1292. [PMID: 29797770 DOI: 10.1111/obr.12703] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/26/2018] [Accepted: 04/11/2018] [Indexed: 12/13/2022]
Abstract
Cellular transport of ions, especially by ion channels, regulates physiological function. The transient receptor potential (TRP) channels, with 30 identified so far, are cation channels with high calcium permeability. These ion channels are present in metabolically active tissues including adipose tissue, liver, gastrointestinal tract, brain (hypothalamus), pancreas and skeletal muscle, which suggests a potential role in metabolic disorders including obesity. TRP channels have potentially important roles in adipogenesis, obesity development and its prevention and therapy because of their physiological properties including calcium permeability, thermosensation and taste perception, involvement in cell metabolic signalling and hormone release. This wide range of actions means that organ-specific actions are unlikely, thus increasing the possibility of adverse effects. Delineation of responses to TRP channels has been limited by the poor selectivity of available agonists and antagonists. Food constituents that can modulate TRP channels are of interest in controlling metabolic status. TRP vanilloid 1 channels modulated by capsaicin have been the most studied, suggesting that this may be the first target for effective pharmacological modulation in obesity. This review shows that most of the TRP channels are potential targets to reduce metabolic disorders through a range of mechanisms.
Collapse
Affiliation(s)
- M Bishnoi
- Department of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute, S.A.S. Nagar (Mohali), Punjab, India.,Functional Foods Research Group, Institute for Agriculture and the Environment, University of Southern Queensland, Toowoomba, QLD, Australia
| | - P Khare
- Department of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute, S.A.S. Nagar (Mohali), Punjab, India
| | - L Brown
- Functional Foods Research Group, Institute for Agriculture and the Environment, University of Southern Queensland, Toowoomba, QLD, Australia.,School of Health and Wellbeing, University of Southern Queensland, Toowoomba, QLD, Australia
| | - S K Panchal
- Functional Foods Research Group, Institute for Agriculture and the Environment, University of Southern Queensland, Toowoomba, QLD, Australia
| |
Collapse
|
28
|
Schoettl T, Fischer IP, Ussar S. Heterogeneity of adipose tissue in development and metabolic function. ACTA ACUST UNITED AC 2018. [PMID: 29514879 DOI: 10.1242/jeb.162958] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adipose tissue is a central metabolic organ. Unlike other organs, adipose tissue is compartmentalized into individual depots and distributed throughout the body. These different adipose depots show major functional differences and risk associations for developing metabolic syndrome. Recent advances in lineage tracing demonstrate that individual adipose depots are composed of adipocytes that are derived from distinct precursor populations, giving rise to different populations of energy-storing white adipocytes. Moreover, distinct lineages of energy-dissipating brown and beige adipocytes exist in discrete depots or within white adipose tissue depots. In this Review, we discuss developmental and functional heterogeneity, as well as sexual dimorphism, between and within individual adipose tissue depots. We highlight current data relating to the differences between subcutaneous and visceral white adipose tissue in the development of metabolic dysfunction, with special emphasis on adipose tissue expansion and remodeling of the extracellular matrix. Moreover, we provide a detailed overview of adipose tissue development as well as the consensus and controversies relating to adult adipocyte precursor populations.
Collapse
Affiliation(s)
- Theresa Schoettl
- JRG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, 85748 Garching, Germany.,German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Ingrid P Fischer
- JRG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, 85748 Garching, Germany.,German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany.,Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 80333 Munich, Germany
| | - Siegfried Ussar
- JRG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, 85748 Garching, Germany .,German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| |
Collapse
|