1
|
Quarta S, Santarpino G, Carluccio MA, Calabriso N, Cardetta F, Siracusa L, Strano T, Palamà I, Leccese G, Visioli F, Massaro M. Cardiac fat adipocytes: An optimized protocol for isolation of ready-to-use mature adipocytes from human pericardial adipose tissue. J Mol Cell Cardiol 2024; 196:12-25. [PMID: 39214497 DOI: 10.1016/j.yjmcc.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
A better understanding of the pathophysiology of cardiac fat depots is crucial to describe their role in the development of cardiovascular diseases. To this end, we have developed a method to isolate mature fat cells from the pericardial adipose tissue (PAT), the most accessible cardiac fat depot during cardiac surgery. Using enzymatic isolation, we were able to successfully obtain mature fat cells together with the corresponding cells of the stromal vascular fraction (SVF). We subjected the PAT adipocytes to thorough morphological and molecular characterization, including detailed fatty acid profiling, and simultaneously investigated their reactivity to external stimuli. Our approach resulted in highly purified fat cells with sustained viability for up to 72 h after explantation. Remarkably, these adipocytes responded to multiple challenges, including pro-inflammatory and metabolic stimuli, indicating their potential to trigger a pro-inflammatory response and modulate endothelial cell behavior. Furthermore, we have created conditions to maintain whole PAT in culture and preserve their viability and reactivity to external stimuli. The efficiency of cell recovery combined with minimal dedifferentiation underscores the promise for future applications as a personalized tool for screening and assessing individual patient responses to drugs and supplements or nutraceuticals.
Collapse
Affiliation(s)
- Stefano Quarta
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy.
| | - Giuseppe Santarpino
- Department of Clinical and Experimental Medicine, Magna Graecia University of Catanzaro, Italy; Department of Cardiac Surgery, Città di Lecce Hospital, GVM Care&Research, Lecce, Italy; Department of Cardiac Surgery, Paracelsus Medical University, Nuremberg, Germany.
| | | | - Nadia Calabriso
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy.
| | - Francesco Cardetta
- Department of Cardiac Surgery, University "Campus Biomedico", Rome, Italy.
| | - Laura Siracusa
- Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Catania section, Via Paolo Gaifami 18, 95126 Catania, Italy.
| | - Tonia Strano
- Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Catania section, Via Paolo Gaifami 18, 95126 Catania, Italy.
| | - Ilaria Palamà
- Institute Nanotechnology Institute, CNR-NANOTEC, 73100 Lecce, Italy.
| | - Gabriella Leccese
- Institute Nanotechnology Institute, CNR-NANOTEC, 73100 Lecce, Italy.
| | | | - Marika Massaro
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy.
| |
Collapse
|
2
|
Dayarathne LA, Ko SC, Yim MJ, Lee JM, Kim JY, Oh GW, Kim CH, Kim KW, Lee DS, Jung WK, Je JY. Purple Butter Clam ( Saxidomus Purpurata) as a Potential Functional Food Source for Obesity Treatment. J Med Food 2024. [PMID: 39382491 DOI: 10.1089/jmf.2024.k.0169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024] Open
Abstract
Saxidomus purpurata extract (SPE) is a highly consumable seafood worldwide with known health-related benefits. However, there are no reports of its' anti-obesity effect. This study explores the potential of SPE for anti-obesity effects by modulating adipogenesis and lipolysis. SPE reduced intracellular lipid and triglyceride accumulation while increasing free glycerol release in adipocytes. SPE inhibited lipogenesis protein expressions and increased the phosphorylation of hormone-sensitive lipase and Adenosine monophosphate-activated protein kinase (AMPK) to promote lipolysis. In addition, SPE suppressed adipogenesis by downregulating protein expression of key adipogenic markers, peroxisome proliferator-activated receptor gamma (PPARγ), CCAAT/enhancer-binding protein α (C/EBPα), and sterol regulatory element-binding protein 1 (SREBP1) via Wnt/β-catenin signaling. SPE augmented the heme oxygenase-1 (HO-1) expression. Thus, pharmacological intervention with Zinc protoporphyrin (ZnPP-HO-1 antagonist) was employed to validate the HO-1 role. The presence of ZnPP increased the lipid accumulation and reduced the free glycerol release. At the molecular level, adipogenic transcription factors (PPARγ, C/EBPα, and SREBP1) expressions were restored in the presence of ZnPP. GC-MS analysis revealed that SPE was comprised of several fatty acids, contributing to its anti-obesity activity. SPE is an effective nutraceutical that can be used to reduce the progression of obesity. HO-1 expression during adipogenesis might be the mechanism of action for the anti-obesity effect of SPE.
Collapse
Affiliation(s)
- Lakshi A Dayarathne
- Department of Food and Nutrition, Pukyong National University, Busan, Republic of Korea
| | - Seok-Chun Ko
- National Marine Biodiversity of Korea (MABIK), Seochun, Korea
| | - Mi-Jin Yim
- National Marine Biodiversity of Korea (MABIK), Seochun, Korea
| | - Jeong Min Lee
- National Marine Biodiversity of Korea (MABIK), Seochun, Korea
| | - Ji-Yul Kim
- National Marine Biodiversity of Korea (MABIK), Seochun, Korea
| | - Gun-Woo Oh
- National Marine Biodiversity of Korea (MABIK), Seochun, Korea
| | - Chul Hwan Kim
- National Marine Biodiversity of Korea (MABIK), Seochun, Korea
| | - Kyung Woo Kim
- National Marine Biodiversity of Korea (MABIK), Seochun, Korea
| | - Dae-Sung Lee
- National Marine Biodiversity of Korea (MABIK), Seochun, Korea
| | - Won-Kyo Jung
- Major of Biomedical Engineering, Division of Smart Healthcare, Pukyong National University, Busan, Korea
| | - Jae-Young Je
- Major of Human Bioconvergence, Division of Smart Healthcare, Pukyong National University, Busan, Korea
| |
Collapse
|
3
|
Won YS, Bak SG, Chandimali N, Park EH, Lim HJ, Kwon HS, Park SI, Lee SJ. 7-MEGA™ inhibits adipogenesis in 3T3-L1 adipocytes and suppresses obesity in high-fat-diet-induced obese C57BL/6 mice. Lipids Health Dis 2024; 23:192. [PMID: 38909257 PMCID: PMC11193219 DOI: 10.1186/s12944-024-02175-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/04/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND Overweight, often known as obesity, is the abnormal and excessive accumulation of fat that exposes the health of a person at risk by increasing the likelihood that they may experience many chronic conditions. Consequently, obesity has become a global health threat, presenting serious health issues, and attracting a lot of attention in the healthcare profession and the scientific community. METHOD This study aims to explore the anti-adipogenic properties of 7-MEGA™ in an attempt to address obesity, using both in vitro and in vivo research. The effects of 7MEGA™ at three distinct concentrations were investigated in obese mice who were given a high-fat diet (HFD) and 3T3-L1 adipocytes. RESULTS 7MEGA™ decreased the total fat mass, overall body weight, and the perirenal and subcutaneous white adipose tissue (PWAT and SWAT) contents in HFD mice. Additionally, 7MEGA™ showed promise in improving the metabolic health of individuals with obesity and regulate the levels of insulin hormone, pro-inflammatory cytokines and adipokines. Furthermore, Peroxisome proliferator-activated receptors (PPAR) α and γ, Uncoupling Protein 1 (UCP-1), Sterol Regulatory Element-Binding Protein 1 (SREBP-1), Fatty Acid-Binding Protein 4 (FABP4), Fatty Acid Synthase (FAS), Acetyl-CoA Carboxylase (ACC), Stearoyl-CoA Desaturase-1 (SCD-1) and CCAAT/Enhancer-Binding Protein (C/EBPα) were among the adipogenic regulators that 7MEGA™ could regulate. CONCLUSION In summary, this study uncovered that 7MEGA™ demonstrates anti-adipogenic and anti-obesity effects, suggesting its potential in combating obesity.
Collapse
Affiliation(s)
- Yeong-Seon Won
- Division of Research Management, Department of Bioresource Industrialization, Honam National Institute of Biological Resource, Mokpo, 58762, Republic of Korea
| | - Seon-Gyeong Bak
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-Gil, Jeongeup, 56212, Republic of Korea
| | - Nisansala Chandimali
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-Gil, Jeongeup, 56212, Republic of Korea
- Department of Applied Biotechnology, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Eun Hyun Park
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-Gil, Jeongeup, 56212, Republic of Korea
- Department of Veterinary Pathology, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyung-Jin Lim
- Scripps Korea Antibody Institute, Chuncheon, 24341, Republic of Korea
| | - Hyuck Se Kwon
- R&D Team, Food & Supplement Health Claims, Vitech, Jeonju, 55365, Republic of Korea
| | - Sang-Ik Park
- Department of Veterinary Pathology, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Seung Jae Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-Gil, Jeongeup, 56212, Republic of Korea.
- Department of Applied Biotechnology, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
| |
Collapse
|
4
|
Aragón-Herrera A, Feijóo-Bandín S, Vázquez-Abuín X, Anido-Varela L, Moraña-Fernández S, Bravo SB, Tarazón E, Roselló-Lletí E, Portolés M, García-Seara J, Seijas J, Rodríguez-Penas D, Bani D, Gualillo O, González-Juanatey JR, Lago F. Human recombinant relaxin-2 (serelaxin) regulates the proteome, lipidome, lipid metabolism and inflammatory profile of rat visceral adipose tissue. Biochem Pharmacol 2024; 223:116157. [PMID: 38518995 DOI: 10.1016/j.bcp.2024.116157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/19/2024] [Accepted: 03/19/2024] [Indexed: 03/24/2024]
Abstract
Recombinant human relaxin-2 (serelaxin) has been widely proven as a novel drug with myriad effects at different cardiovascular levels, which support its potential therapeutic efficacy in several cardiovascular diseases (CVD). Considering these effects, together with the influence of relaxin-2 on adipocyte physiology and adipokine secretion, and the connection between visceral adipose tissue (VAT) dysfunction and the development of CVD, we could hypothesize that relaxin-2 may regulate VAT metabolism. Our objective was to evaluate the impact of a 2-week serelaxin treatment on the proteome and lipidome of VAT from Sprague-Dawley rats. We found that serelaxin increased 1 polyunsaturated fatty acid and 6 lysophosphatidylcholines and decreased 4 triglycerides in VAT employing ultra-high performance liquid chromatography-mass spectrometry (UHPLC-MS) based platforms, and that regulates 47 phosphoproteins using SWATH/MS analysis. Through RT-PCR, we found that serelaxin treatment also caused an effect on VAT lipolysis through an increase in the mRNA expression of hormone-sensitive lipase (HSL) and a decrease in the expression of adipose triglyceride lipase (ATGL), together with a reduction in the VAT expression of the fatty acid transporter cluster of differentiation 36 (Cd36). Serelaxin also caused an anti-inflammatory effect in VAT by the decrease in the mRNA expression of tumor necrosis factor α (TNFα), interleukin-1β (IL-1β), chemerin, and its receptor. In conclusion, our results highlight the regulatory role of serelaxin in the VAT proteome and lipidome, lipolytic function, and inflammatory profile, suggesting the implication of several mechanisms supporting the potential benefit of serelaxin for the prevention of obesity and metabolic disorders.
Collapse
Affiliation(s)
- Alana Aragón-Herrera
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain.
| | - Sandra Feijóo-Bandín
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Xocas Vázquez-Abuín
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Laura Anido-Varela
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Moraña-Fernández
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Cardiology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Susana B Bravo
- Proteomics Unit, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Estefanía Tarazón
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Valencia, Spain
| | - Esther Roselló-Lletí
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Valencia, Spain
| | - Manuel Portolés
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Valencia, Spain
| | - Javier García-Seara
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Arrhytmia Unit, Cardiology Department, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - José Seijas
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Cardiology Department Clinical Trial Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Diego Rodríguez-Penas
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Cardiology Department Clinical Trial Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Daniele Bani
- Research Unit of Histology & Embryology, Department of Experimental & Clinical Medicine, University of Florence, Florence, Italy
| | - Oreste Gualillo
- Laboratory of Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - José Ramón González-Juanatey
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain; Cardiology Department, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Francisca Lago
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
5
|
Zhang J, Zhao M, Yu H, Wang Q, Shen F, Cai H, Feng F, Tang J. Palmitoleic Acid Ameliorates Metabolic Disorders and Inflammation by Modulating Gut Microbiota and Serum Metabolites. Mol Nutr Food Res 2024; 68:e2300749. [PMID: 38511225 DOI: 10.1002/mnfr.202300749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/24/2024] [Indexed: 03/22/2024]
Abstract
SCOPE Palmitoleic acid (POA) is an omega-7 monounsaturated fatty acid that has been suggested to improve metabolic disorders. However, it remains unclear whether gut microbiota plays a role in the amelioration of metabolic disorders by POA. This study aims to investigate the regulation of POA on metabolism, as well as systemic inflammation in HFD-fed mice from the perspective of serum metabolome and gut microbiome. METHODS AND RESULTS Thirty-six C57BL/6 male mice are randomly assigned to either a normal chow diet containing 1.9% w/w lard or an HFD containing 20.68% w/w lard or 20.68% w/w sea buckthorn pulp oil for 16 weeks. The study finds that POA significantly attenuated hyperlipidemia, insulin resistance, and inflammation in HFD-fed mice. POA supplementation significantly alters the composition of serum metabolites, particularly lipid metabolites in the glycerophospholipid metabolism pathway. POA obviously increases the abundance of Bifidobacterium and decreases the abundance of Allobaculum. Importantly, the study finds that glycerophosphocholine mediates the effect of Bifidobacterium on LDL-C, sphingomyelin mediates the effect of Bifidobacterium on IL-6, and maslinic acid mediates the effect of Allobaculum on IL-6. CONCLUSION The results suggest that exogenous POA can improve metabolic disorders and inflammation in HFD-fed mice, potentially by modulating the serum metabolome and gut microbiome.
Collapse
Affiliation(s)
- Junhui Zhang
- School of Life Sciences, Westlake University, Hangzhou, 310012, China
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310012, China
| | - Minjie Zhao
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310012, China
| | - Huilin Yu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310012, China
| | - Qianqian Wang
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310012, China
| | - Fei Shen
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310012, China
| | - Haiying Cai
- School of Biological & Chemical Engineering, Zhejiang University of Science &Technology, Hangzhou, 310012, China
| | - Fengqin Feng
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310012, China
| | - Jun Tang
- School of Life Sciences, Westlake University, Hangzhou, 310012, China
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310012, China
- Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310012, China
| |
Collapse
|
6
|
Valdés-Hernández J, Folch JM, Crespo-Piazuelo D, Passols M, Sebastià C, Criado-Mesas L, Castelló A, Sánchez A, Ramayo-Caldas Y. Identification of candidate regulatory genes for intramuscular fatty acid composition in pigs by transcriptome analysis. Genet Sel Evol 2024; 56:12. [PMID: 38347496 PMCID: PMC10860264 DOI: 10.1186/s12711-024-00882-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 01/31/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Intramuscular fat (IMF) content and its fatty acid (FA) composition are typically controlled by several genes, each with a small effect. In the current study, to pinpoint candidate genes and putative regulators involved in FA composition, we performed a multivariate integrative analysis between intramuscular FA and transcriptome profiles of porcine longissimus dorsi (LD) muscle. We also carried out a combination of network, regulatory impact factor (RIF), in silico prediction of putative target genes, and functional analyses to better support the biological relevance of our findings. RESULTS For this purpose, we used LD RNA-Seq and intramuscular FA composition profiles of 129 Iberian × Duroc backcrossed pigs. We identified 378 correlated variables (13 FA and 365 genes), including six FA (C20:4n-6, C18:2n-6, C20:3n-6, C18:1n-9, C18:0, and C16:1n-7) that were among the most interconnected variables in the predicted network. The detected FA-correlated genes include genes involved in lipid and/or carbohydrate metabolism or in regulation of IMF deposition (e.g., ADIPOQ, CHUK, CYCS, CYP4B1, DLD, ELOVL6, FBP1, G0S2, GCLC, HMGCR, IDH3A, LEP, LGALS12, LPIN1, PLIN1, PNPLA8, PPP1R1B, SDR16C5, SFRP5, SOD3, SNW1, and TFRC), meat quality (GALNT15, GOT1, MDH1, NEU3, PDHA1, SDHD, and UNC93A), and transport (e.g., EXOC7 and SLC44A2). Functional analysis highlighted 54 over-represented gene ontology terms, including well-known biological processes and pathways that regulate lipid and carbohydrate metabolism. RIF analysis suggested a pivotal role for six transcription factors (CARHSP1, LBX1, MAFA, PAX7, SIX5, and TADA2A) as putative regulators of gene expression and intramuscular FA composition. Based on in silico prediction, we identified putative target genes for these six regulators. Among these, TADA2A and CARHSP1 had extreme RIF scores and present novel regulators in pigs. In addition, the expression of TADA2A correlated (either positively or negatively) with C20:4n-6, C18:2n-6, C20:3n-6, C18:1n-9, and that of CARHSP1 correlated (positively) with the C16:1n-7 lipokine. We also found that these two transcription factors share target genes that are involved in lipid metabolism (e.g., GOT1, PLIN1, and TFRC). CONCLUSIONS This integrative analysis of muscle transcriptome and intramuscular FA profile revealed valuable information about key candidate genes and potential regulators for FA and lipid metabolism in pigs, among which some transcription factors are proposed to control gene expression and modulate FA composition differences.
Collapse
Affiliation(s)
- Jesús Valdés-Hernández
- Plant and Animal Genomics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, Spain.
- Departament de Ciència Animal i dels Aliments, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| | - Josep M Folch
- Plant and Animal Genomics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, Spain
- Departament de Ciència Animal i dels Aliments, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Daniel Crespo-Piazuelo
- Departament de Genètica i Millora Animal, Institut de Recerca y Tecnologia Agraroalimentàries (IRTA), Caldes de Montbui, Spain
| | - Magí Passols
- Plant and Animal Genomics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, Spain
| | - Cristina Sebastià
- Plant and Animal Genomics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, Spain
- Departament de Ciència Animal i dels Aliments, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Lourdes Criado-Mesas
- Plant and Animal Genomics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, Spain
| | - Anna Castelló
- Plant and Animal Genomics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, Spain
- Departament de Ciència Animal i dels Aliments, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Armand Sánchez
- Plant and Animal Genomics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, Spain
- Departament de Ciència Animal i dels Aliments, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Yuliaxis Ramayo-Caldas
- Departament de Genètica i Millora Animal, Institut de Recerca y Tecnologia Agraroalimentàries (IRTA), Caldes de Montbui, Spain.
| |
Collapse
|
7
|
Feyera T, Lashkari S, Johannsen JC, Llauradó-Calero E, Zhe L, Theil PK, Jensen SK. Supplementation of palmitoleic acid improved piglet growth and reduced body temperature drop upon cold exposure. J Anim Sci 2023; 101:skad372. [PMID: 37935407 PMCID: PMC10656293 DOI: 10.1093/jas/skad372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/01/2023] [Indexed: 11/09/2023] Open
Abstract
Piglet survival is a major challenge in the first few days postpartum and interventions during this period may improve survival and growth. This study investigated the effects of palmitoleic acid (C16:1n-7; PA) supplementation on growth performance, body temperature, fatty acid (FA), and energy metabolism in milk-replacer-fed piglets. Forty-eight piglets were stratified by body weight and randomly assigned to one of four dietary treatments (0%, 1%, 2%, and 3% PA supplementation as a percent of milk replacer) and given the diet through an orogastric tube. They were fed dietary treatments every 2 h for 4 d in the first week postpartum and all were sacrificed at the end of the experiment. The piglets were weighed daily, and half in each dietary treatment group, the same piglets each day, were exposed daily to a lower temperature for 2 h. Plasma samples were collected immediately before sacrifice for analyses of FA and other plasma metabolites. The weight of organs and empty body weight were determined after sacrifice. Liver and semimembranosus muscle tissue samples were collected and analyzed for FA content. Contents of C16:1n-7 and C18:1n-7 in both plasma and liver (P < 0.001), and C16:1n-7 in semimembranosus muscle (P < 0.001) increased linearly as PA supplementation increased. Most plasma FA levels (except C16:1n-7, C16:1n-9, and C22:5n-3) were lower in piglets exposed to lower temperatures than those that were not. Plasma glucose, triglycerides, and lactate dehydrogenase levels increased linearly with PA supplementation (P < 0.001). Piglets' average daily gain, liver glycogen pool, liver weight, and gallbladder weight increased linearly (P < 0.05, P < 0.01, P < 0.05, and P < 0.001, respectively), but lung weight, liver nitrogen content, and body temperature drop decreased linearly (P < 0.01, P < 0.001, and P < 0.05, respectively) with PA supplementation. Piglets exposed to low temperature had greater liver nitrogen (P < 0.05) and lactate dehydrogenase (P < 0.001) contents but had lower liver weight (P < 0.01) and plasma lactate concentration (P < 0.05) than those that were not. In conclusion, this study demonstrated the importance of PA on the growth performance of the piglets by increasing their average daily gain and decreasing a drop in body temperature upon cold exposure, most likely due to a modified energy metabolism.
Collapse
Affiliation(s)
- Takele Feyera
- Department of Animal and Veterinary Sciences, Aarhus University AU-Viborg, DK-8830 Tjele, Denmark
| | - Saman Lashkari
- Department of Animal and Veterinary Sciences, Aarhus University AU-Viborg, DK-8830 Tjele, Denmark
| | - Jakob C Johannsen
- Department of Animal and Veterinary Sciences, Aarhus University AU-Viborg, DK-8830 Tjele, Denmark
| | - Eudald Llauradó-Calero
- Department of Animal and Veterinary Sciences, Aarhus University AU-Viborg, DK-8830 Tjele, Denmark
| | - Li Zhe
- Department of Animal and Veterinary Sciences, Aarhus University AU-Viborg, DK-8830 Tjele, Denmark
| | - Peter K Theil
- Department of Animal and Veterinary Sciences, Aarhus University AU-Viborg, DK-8830 Tjele, Denmark
| | - Søren K Jensen
- Department of Animal and Veterinary Sciences, Aarhus University AU-Viborg, DK-8830 Tjele, Denmark
| |
Collapse
|
8
|
Muthuraj PG, Krishnamoorthy C, Anderson-Berry A, Hanson C, Natarajan SK. Novel Therapeutic Nutrients Molecules That Protect against Zika Virus Infection with a Special Note on Palmitoleate. Nutrients 2022; 15:124. [PMID: 36615782 PMCID: PMC9823984 DOI: 10.3390/nu15010124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/11/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
Zika virus (ZIKV) is a Flavivirus from the Flaviviridae family and a positive-sense single strand RNA virus. ZIKV infection can cause a mild infection to the mother but can be vertically transmitted to the developing fetus, causing congenital anomalies. The prevalence of ZIKV infections was relatively insignificant with sporadic outbreaks in the Asian and African continents until 2006. However, recent epidemic in the Caribbean showed significant increased incidence of Congenital Zika Syndrome. ZIKV infection results in placental pathology which plays a crucial role in disease transmission from mother to fetus. Currently, there is no Food and Drug Administration (FDA) approved vaccine or therapeutic drug against ZIKV. This review article summarizes the recent advances on ZIKV transmission and diagnosis and reviews nutraceuticals which can protect against the ZIKV infection. Further, we have reviewed recent advances related to the novel therapeutic nutrient molecules that have been shown to possess activity against Zika virus infected cells. We also review the mechanism of ZIKV-induced endoplasmic reticulum and apoptosis and the protective role of palmitoleate (nutrient molecule) against ZIKV-induced ER stress and apoptosis in the placental trophoblasts.
Collapse
Affiliation(s)
- Philma Glora Muthuraj
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Chandan Krishnamoorthy
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Ann Anderson-Berry
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Corrine Hanson
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Medical Nutrition Education, College of Allied Health Profession, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sathish Kumar Natarajan
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Medical Nutrition Education, College of Allied Health Profession, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
9
|
Simão JJ, Cruz MM, Abdala FM, Bolsoni-Lopes A, Armelin-Correa L, Alonso-Vale MIC. Palmitoleic Acid Acts on Adipose-Derived Stromal Cells and Promotes Anti-Hypertrophic and Anti-Inflammatory Effects in Obese Mice. Pharmaceuticals (Basel) 2022; 15:1194. [PMID: 36297306 PMCID: PMC9609051 DOI: 10.3390/ph15101194] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 04/16/2024] Open
Abstract
Adipose tissue (AT) secretes adipokines, modulators of low-grade chronic inflammation in obesity. Molecules that induce the emergence of new and functional adipocytes in AT can alleviate or prevent inflammatory and metabolic disorders. The objective of this study was to investigate the role of palmitoleic acid (n7) in 3T3-L1 and primary pre-adipocyte differentiation and AT inflammation. C57BL/6j mice were submitted to a control or high-fat diet (HFD) for 8 weeks, and treated with n7 for 4 weeks. Mice consuming a HFD presented an increase in body weight, epididymal (Epi) fat mass, and Epi adipocytes size. N7 treatment attenuated the body weight gain and completely prevented the hypertrophy of Epi adipocytes, but not the increment in Epi mass induced by the HFD, suggesting a greater adipocytes hyperplasia in animals treated with n7. It was agreed that n7 increased 3T3-L1 proliferation and differentiation, as well as the expression of genes involved in adipogenesis, such as Cebpa, Pparg, aP2, Perilipin, and Scl2a4. Furthermore, n7 decreased the inflammatory cytokines Mcp1, Tnfa, Il6, Cxcl10, and Nos2 genes in Epi vascular stromal cells, but not in the whole AT. These findings show that n7 exerts anti-hypertrophic effects in adipocytes which influence the surrounding cells by attenuating the overexpression of pro-inflammatory cytokines triggered by a HFD.
Collapse
Affiliation(s)
- Jussara J. Simão
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-030, SP, Brazil
| | - Maysa M. Cruz
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-030, SP, Brazil
| | - Fernanda M. Abdala
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-030, SP, Brazil
| | - Andressa Bolsoni-Lopes
- Department of Nursing, Health Sciences Center, Federal University of Espirito Santo, Vitoria 29075-910, ES, Brazil
| | - Lucia Armelin-Correa
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-030, SP, Brazil
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-030, SP, Brazil
| | - Maria Isabel C. Alonso-Vale
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-030, SP, Brazil
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-030, SP, Brazil
| |
Collapse
|
10
|
Roles of Palmitoleic Acid and Its Positional Isomers, Hypogeic and Sapienic Acids, in Inflammation, Metabolic Diseases and Cancer. Cells 2022; 11:cells11142146. [PMID: 35883589 PMCID: PMC9319324 DOI: 10.3390/cells11142146] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/04/2022] [Accepted: 07/07/2022] [Indexed: 12/19/2022] Open
Abstract
In the last few years, the monounsaturated hexadecenoic fatty acids are being increasingly considered as biomarkers of health with key functions in physiology and pathophysiology. Palmitoleic acid (16:1n-7) and sapienic acid (16:1n-10) are synthesized from palmitic acid by the action of stearoyl-CoA desaturase-1 and fatty acid desaturase 2, respectively. A third positional isomer, hypogeic acid (16:1n-9) is produced from the partial β-oxidation of oleic acid. In this review, we discuss the current knowledge of the effects of palmitoleic acid and, where available, sapienic acid and hypogeic acid, on metabolic diseases such as diabetes, cardiovascular disease, and nonalcoholic fatty liver disease, and cancer. The results have shown diverse effects among studies in cell lines, animal models and humans. Palmitoleic acid was described as a lipokine able to regulate different metabolic processes such as an increase in insulin sensitivity in muscle, β cell proliferation, prevention of endoplasmic reticulum stress and lipogenic activity in white adipocytes. Numerous beneficial effects have been attributed to palmitoleic acid, both in mouse models and in cell lines. However, its role in humans is not fully understood, and is sometimes controversial. Regarding sapienic acid and hypogeic acid, studies on their biological effects are still scarce, but accumulating evidence suggests that they also play important roles in metabolic regulation. The multiplicity of effects reported for palmitoleic acid and the compartmentalized manner in which they often occur, may suggest the overlapping actions of multiple isomers being present at the same or neighboring locations.
Collapse
|
11
|
Søfteland L, Olsvik PA. In vitro toxicity of glyphosate in Atlantic salmon evaluated with a 3D hepatocyte-kidney co-culture model. Food Chem Toxicol 2022; 164:113012. [PMID: 35429611 DOI: 10.1016/j.fct.2022.113012] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 04/05/2022] [Accepted: 04/09/2022] [Indexed: 12/11/2022]
Abstract
A novel 3D Atlantic salmon co-culture model was developed using primary hepatocytes and kidney epithelial cells isolated from the same fish. Mono and co-cultures of primary hepatocytes and kidney epithelial cells were exposed for 48 h to glyphosate (5, 50 and 500 μM). For comparison, cells were also exposed to chlorpyrifos, benzo(a)pyrene and cadmium. Cell staining, cell viability assessments, RT-qPCR and global metabolomic profiling were used to examine the toxicological effects on liver and renal function and to compare responses in 3D and 2D cultures. The 3D hepatocyte cell culture was considered superior to the 2D culture due to the ATP binding cassette subfamily B member 1 (Abcb1) response and was thus used further in co-culture with kidney cells. Metabolomic analysis of co-cultured cells showed that glyphosate exposure (500 μM) altered lipid metabolism in both hepatocytes and kidney cells. Elevated levels of several types of PUFAs and long-chain fatty acids were observed in exposed hepatocytes, owing to increased uptake and phospholipid remodelling. Glyphosate suppressed the expression of estrogen receptor 1 (Esr1) and vitellogenin (Vtg) and altered histidine metabolism in exposed hepatocytes. Increased levels of cholesterol and downregulation of clusterin (Clu) suggest that glyphosate treatment affected membrane stability in Atlantic salmon kidney cells. This study demonstrates the usefulness of applying 3D co-culture models in risk assessment.
Collapse
Affiliation(s)
- L Søfteland
- Institute of Marine Research, Nordnesgaten 50, 5005, Bergen, Norway.
| | - P A Olsvik
- Institute of Marine Research, Nordnesgaten 50, 5005, Bergen, Norway; Nord University, Universitetsalléen 11, 8049, Bodø, Norway
| |
Collapse
|
12
|
Maternal Fructose Intake Causes Developmental Reprogramming of Hepatic Mitochondrial Catalytic Activity and Lipid Metabolism in Weanling and Young Adult Offspring. Int J Mol Sci 2022; 23:ijms23020999. [PMID: 35055185 PMCID: PMC8780605 DOI: 10.3390/ijms23020999] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/03/2022] [Accepted: 01/06/2022] [Indexed: 11/16/2022] Open
Abstract
Excess dietary fructose is a major public health concern, yet little is known about its influence on offspring development and later-life disease when consumed in excess during pregnancy. To determine whether increased maternal fructose intake could have long-term consequences on offspring health, we investigated the effects of 10% w/v fructose water intake during preconception and pregnancy in guinea pigs. Female Dunkin Hartley guinea pigs were fed a control diet (CD) or fructose diet (FD; providing 16% of total daily caloric intake) ad libitum 60 days prior to mating and throughout gestation. Dietary interventions ceased at day of delivery. Offspring were culled at day 21 (D21) (weaning) and at 4 months (4 M) (young adult). Fetal exposure to excess maternal fructose intake significantly increased male and female triglycerides at D21 and 4 M and circulating palmitoleic acid and total omega-7 through day 0 (D0) to 4 M. Proteomic and functional analysis of significantly differentially expressed proteins revealed that FD offspring (D21 and 4 M) had significantly increased mitochondrial metabolic activities of β-oxidation, electron transport chain (ETC) and oxidative phosphorylation and reactive oxygen species production compared to the CD offspring. Western blotting analysis of both FD offspring validated the increased protein abundances of mitochondrial ETC complex II and IV, SREBP-1c and FAS, whereas VDAC1 expression was higher at D21 but lower at 4 M. We provide evidence demonstrating offspring programmed hepatic mitochondrial metabolism and de novo lipogenesis following excess maternal fructose exposure. These underlying asymptomatic programmed pathways may lead to a predisposition to metabolic dysfunction later in life.
Collapse
|
13
|
Miklankova D, Markova I, Hüttl M, Stankova B, Malinska H. The Different Insulin-Sensitising and Anti-Inflammatory Effects of Palmitoleic Acid and Oleic Acid in a Prediabetes Model. J Diabetes Res 2022; 2022:4587907. [PMID: 36147256 PMCID: PMC9489414 DOI: 10.1155/2022/4587907] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/01/2022] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Monounsaturated fatty acids (MUFA) are understood to have therapeutic and preventive effects on chronic complications associated with type 2 diabetes mellitus (T2DM); however, there are differences between individual MUFAs. Although the effects of palmitoleic acid (POA) are still debated, POA can regulate glucose homeostasis, lipid metabolism, and cytokine production, thus improving metabolic disorders. In this study, we investigated and compared the metabolic effects of POA and oleic acid (OA) supplementation on glucose and lipid metabolism, insulin sensitivity, and inflammation in a prediabetic model, the hereditary hypertriglyceridemic rat (HHTg). HHTg rats exhibiting genetically determined hypertriglyceridemia, insulin resistance, and impaired glucose tolerance were fed a standard diet. POA and OA were each administered intragastrically at a dose of 100 mg/kg b.wt. for four weeks. RESULTS Supplementation with both MUFAs significantly elevated insulin and glucagon levels, but only POA decreased nonfasting glucose. POA-treated rats showed elevated circulating NEFA associated with increased lipolysis, lipoprotein lipase gene expression, and fatty acid reesterification in visceral adipose tissue (VAT). The mechanism of improved insulin sensitivity of peripheral tissues (measured as insulin-stimulated lipogenesis and glycogenesis) in POA-treated HHTg rats could contribute increased circulating adiponectin and omentin levels together with elevated FADS1 gene expression in VAT. POA-supplemented rats exhibited markedly decreased proinflammatory cytokine production by VAT, which can alleviate chronic inflammation. OA-supplemented rats exhibited decreased arachidonic acid (AA) profiles and decreased proinflammatory AA-derived metabolites (20-HETE) in membrane phospholipids of peripheral tissues. Slightly increased FADS1 gene expression after OA along with increased adiponectin production by VAT was reflected in slightly ameliorated adipose tissue insulin sensitivity (increased insulin-stimulated lipogenesis). CONCLUSIONS Our results show that POA served as a lipokine, ameliorating insulin sensitivity in peripheral tissue and markedly modulating the metabolic activity of VAT including cytokine secretion. OA had a beneficial effect on lipid metabolism and improved inflammation by modulating AA metabolism.
Collapse
Affiliation(s)
- Denisa Miklankova
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Irena Markova
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Martina Hüttl
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Barbora Stankova
- First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Hana Malinska
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
14
|
Betz IR, Qaiyumi SJ, Goeritzer M, Thiele A, Brix S, Beyhoff N, Grune J, Klopfleisch R, Greulich F, Uhlenhaut NH, Kintscher U, Foryst-Ludwig A. Cardioprotective Effects of Palmitoleic Acid (C16:1n7) in a Mouse Model of Catecholamine-Induced Cardiac Damage Are Mediated by PPAR Activation. Int J Mol Sci 2021; 22:ijms222312695. [PMID: 34884498 PMCID: PMC8657733 DOI: 10.3390/ijms222312695] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 01/25/2023] Open
Abstract
Palmitoleic acid (C16:1n7) has been identified as a regulator of physiological cardiac hypertrophy. In the present study, we aimed to investigate the molecular pathways involved in C16:1n7 responses in primary murine cardiomyocytes (PCM) and a mouse model of isoproterenol (ISO)-induced cardiac damage. PCMs were stimulated with C16:1n7 or a vehicle. Afterwards, RNA sequencing was performed using an Illumina HiSeq sequencer. Confirmatory analysis was performed in PCMs and HL-1 cardiomyocytes. For an in vivo study, 129 sv mice were orally treated with a vehicle or C16:1n7 for 22 days. After 5 days of pre-treatment, the mice were injected with ISO (25 mg/kg/d s. c.) for 4 consecutive days. Cardiac phenotyping was performed using echocardiography. In total, 129 genes were differentially expressed in PCMs stimulated with C16:1n7, including Angiopoietin-like factor 4 (Angptl4) and Pyruvate Dehydrogenase Kinase 4 (Pdk4). Both Angptl4 and Pdk4 are proxisome proliferator-activated receptor α/δ (PPARα/δ) target genes. Our in vivo results indicated cardioprotective and anti-fibrotic effects of C16:1n7 application in mice. This was associated with the C16:1n7-dependent regulation of the cardiac PPAR-specific signaling pathways. In conclusion, our experiments demonstrated that C16:1n7 might have protective effects on cardiac fibrosis and inflammation. Our study may help to develop future lipid-based therapies for catecholamine-induced cardiac damage.
Collapse
Affiliation(s)
- Iris Rosa Betz
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- Berlin Institute of Health, Emergency Department Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Sarah Julia Qaiyumi
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
| | - Madeleine Goeritzer
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Arne Thiele
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Sarah Brix
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Niklas Beyhoff
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Jana Grune
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Robert Klopfleisch
- Department of Veterinary Pathology, College of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany;
| | - Franziska Greulich
- German Center for Environmental Health GmbH, Institute for Diabetes and Cancer (IDC), 85764 Munich, Germany; (F.G.); (N.H.U.)
- Metabolic Programming, School of Life Sciences Weihenstephan, Technische Universitaet Muenchen (TUM), 85354 Freising, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, 13125 Berlin, Germany
| | - Nina Henriette Uhlenhaut
- German Center for Environmental Health GmbH, Institute for Diabetes and Cancer (IDC), 85764 Munich, Germany; (F.G.); (N.H.U.)
- Metabolic Programming, School of Life Sciences Weihenstephan, Technische Universitaet Muenchen (TUM), 85354 Freising, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, 13125 Berlin, Germany
| | - Ulrich Kintscher
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Anna Foryst-Ludwig
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Correspondence:
| |
Collapse
|
15
|
Kulagina TP, Popova SS, Aripovsky AV. Seasonal Changes in the Content of Fatty Acids in the Myocardium and m. longissimus dorsi of the Long-Tailed Ground Squirrel Urocitellus undulatus. Biophysics (Nagoya-shi) 2021. [DOI: 10.1134/s0006350921060087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
16
|
Đurašević S, Ružičić A, Lakić I, Tosti T, Đurović S, Glumac S, Pavlović S, Borković-Mitić S, Grigorov I, Stanković S, Jasnić N, Đorđević J, Todorović Z. The Effects of a Meldonium Pre-Treatment on the Course of the Faecal-Induced Sepsis in Rats. Int J Mol Sci 2021; 22:ijms22189698. [PMID: 34575863 PMCID: PMC8464894 DOI: 10.3390/ijms22189698] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/29/2021] [Accepted: 09/01/2021] [Indexed: 12/29/2022] Open
Abstract
Sepsis is a life-threatening condition caused by the dysregulated and overwhelming response to infection, accompanied by an exaggerated pro-inflammatory state and lipid metabolism disturbance leading to sequential organ failure. Meldonium is an anti-ischemic and anti-inflammatory agent which negatively interferes with lipid metabolism by shifting energy production from fatty acid oxidation to glycolysis, as a less oxygen-demanding pathway. Thus, we investigated the effects of a four-week meldonium pre-treatment on faecal-induced sepsis in Sprague-Dawley male rats. Surprisingly, under septic conditions, meldonium increased animal mortality rate compared with the meldonium non-treated group. However, analysis of the tissue oxidative status did not provide support for the detrimental effects of meldonium, nor did the analysis of the tissue inflammatory status showing anti-inflammatory, anti-apoptotic, and anti-necrotic effects of meldonium. After performing tissue lipidomic analysis, we concluded that the potential cause of the meldonium harmful effect is to be found in the overall decreased lipid metabolism. The present study underlines the importance of uninterrupted energy production in sepsis, closely drawing attention to the possible harmful effects of lipid-mobilization impairment caused by certain therapeutics. This could lead to the much-needed revision of the existing guidelines in the clinical treatment of sepsis while paving the way for discovering new therapeutic approaches.
Collapse
Affiliation(s)
- Siniša Đurašević
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (A.R.); (I.L.); (N.J.); (J.Đ.)
- Correspondence: ; Tel.: +381-63-367108
| | - Aleksandra Ružičić
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (A.R.); (I.L.); (N.J.); (J.Đ.)
| | - Iva Lakić
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (A.R.); (I.L.); (N.J.); (J.Đ.)
| | - Tomislav Tosti
- Faculty of Chemistry, University of Belgrade, 11000 Belgrade, Serbia;
| | - Saša Đurović
- Institute of General and Physical Chemistry, University of Belgrade, 11000 Belgrade, Serbia;
| | - Sofija Glumac
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (S.G.); (Z.T.)
| | - Slađan Pavlović
- Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (S.P.); (S.B.-M.); (I.G.)
| | - Slavica Borković-Mitić
- Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (S.P.); (S.B.-M.); (I.G.)
| | - Ilijana Grigorov
- Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (S.P.); (S.B.-M.); (I.G.)
| | - Sanja Stanković
- Centre for Medical Biochemistry, University Clinical Centre of Serbia, 11000 Belgrade, Serbia;
- Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Nebojša Jasnić
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (A.R.); (I.L.); (N.J.); (J.Đ.)
| | - Jelena Đorđević
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (A.R.); (I.L.); (N.J.); (J.Đ.)
| | - Zoran Todorović
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (S.G.); (Z.T.)
- University Medical Centre “Bežanijska kosa”, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
17
|
A New Zealand green-lipped mussel oil-enriched high-fat diet exhibits beneficial effects on body weight and metabolism in mice. Br J Nutr 2021; 125:972-982. [PMID: 32594917 DOI: 10.1017/s0007114520002342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
To induce diet-induced obesity (DIO) in rodents, diets high in saturated fat and/or carbohydrates are commonly used. In the laboratory, standardised diets evolved over time without paying particular attention to the effect of fat composition on metabolic alterations. In the present study, customised high-fat diets (HFD) enriched with a combination of lard and different concentrations of New Zealand green-lipped mussel (Perna canaliculus) oil or MSC Hoki (Macruronus novaezelandiae, blue grenadier) liver oil, important sources of n-3 PUFA, in comparison with a solely lard-based diet, were fed to lean and DIO male C57BL/6 mice and their effects on metabolic parameters were monitored. Intriguingly, an isoenergetic HFD containing 63 % of total fat in the form of mussel oil and only 28 % in the form of lard attenuated HFD-induced body weight gain after 1 and 4 weeks, respectively. Consistently, changing a lard-enriched HFD to the mussel oil diet reduced body weight markedly even after mice had been exposed to the former diet for 10 months. The weight-reducing effect of the diet was not caused by altered energy intake or expenditure, but was associated with reduced visceral fat mass. Collectively, these data suggest a novel weight-reducing potential of green-lipped mussel oil.
Collapse
|
18
|
Kato H, Ario T, Kishida T, Tadano M, Osawa S, Maeda Y, Takakura H, Izawa T. Homeobox A5 and C10 genes modulate adaptation of brown adipose tissue during exercise training in juvenile rats. Exp Physiol 2021; 106:463-474. [PMID: 33369800 DOI: 10.1113/ep089114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022]
Abstract
NEW FINDINGS What is the central question of this study? Exercise can stimulate brown adipose tissue (BAT) with subsequent increase in uncoupling protein 1 expression and mitochondrial biogenesis. In that case, do BAT-specific Hox genes modify BAT functioning and cause uncoupling protein expression changes due to exercise? What is the main finding and its importance? Exercise enhanced brown adipocyte markers, with significant upregulation of HoxA5 and downregulation of HoxC10 mRNA expression in rat BAT. HoxA5 and HoxC10 are thus likely to play distinct roles in exercise-induced changes in BAT markers during the early postnatal period. These findings provide new insight into the mechanisms underlying exercise-induced changes in BAT function. ABSTRACT Brown adipose tissue (BAT) recruitment is involved in increased energy expenditure associated with cold exposure and exercise training. We explored whether exercise training induced changes in expression levels of brown adipocyte-selective factors and Homeobox (Hox) genes during the post-weaning growth period of male Wistar rats. Relative to total body weight, BAT weights alone were lower in exercise-trained (EX) rats compared to sedentary control (SED) rats. mRNA expression of HoxA5 was higher and that of HoxC10 was lower in EX rats than in SED rats, accompanied by both higher citrate synthase activity and protein expression levels for uncoupling protein 1 (UCP1), peroxisome proliferator-activated receptor (PPAR) α, and PPARγ-coactivator (PGC)-1α. HoxA5 knockdown with siRNA reduced the expression of PR-domain containing 16 (Prdm16), cell death-inducing DNA fragmentation factor-α-like effector A (Cidea) gene, type 2 deiodinase mRNA, and PRDM16 protein. Comparatively, HoxC10 knockdown with siRNA enhanced mRNA expression of Prdm16, Pparα and Pgc1α and protein expression of UCP1, PPARα and PGC1α in brown adipocytes. The stimulation of brown adipocytes with isoproterenol, a β-adrenoceptor agonist, caused a phenomenon similar to the effect of exercise training on the genes tested: upregulation of HoxA5 mRNA, downregulation of HoxC10 mRNA, and increased protein expression for UCP1 and PGC1α. Collectively, HoxA5 and HoxC10 may have unique functions that contribute to modulating the expression of BAT-selective markers in BAT of juvenile rats during exercise training. The study findings regarding activation and recruitment of BAT during exercise training have implications for anti-obesity management.
Collapse
Affiliation(s)
- Hisashi Kato
- Faculty, Doshisha University, Kyotanabe, Kyoto, Japan.,Organization for Research Initiatives and Development, Doshisha University, Kyotanabe, Kyoto, Japan
| | - Takuto Ario
- Faculty, Doshisha University, Kyotanabe, Kyoto, Japan
| | | | - Manami Tadano
- Graduate School of Health and Sports Science, Doshisha University, Kyotanabe, Kyoto, Japan
| | - Seita Osawa
- Graduate School of Health and Sports Science, Doshisha University, Kyotanabe, Kyoto, Japan
| | - Yuki Maeda
- Graduate School of Health and Sports Science, Doshisha University, Kyotanabe, Kyoto, Japan
| | | | - Tetsuya Izawa
- Faculty, Doshisha University, Kyotanabe, Kyoto, Japan.,Organization for Research Initiatives and Development, Doshisha University, Kyotanabe, Kyoto, Japan
| |
Collapse
|
19
|
Huang WW, Hong BH, Bai KK, Tan R, Yang T, Sun JP, Yi RZ, Wu H. Cis- and Trans-Palmitoleic Acid Isomers Regulate Cholesterol Metabolism in Different Ways. Front Pharmacol 2020; 11:602115. [PMID: 33363473 PMCID: PMC7753117 DOI: 10.3389/fphar.2020.602115] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/23/2020] [Indexed: 11/13/2022] Open
Abstract
Hypercholesterolemia is a preventable risk factor for atherosclerosis and cardiovascular disease. However, the mechanisms whereby cis-palmitoleic acid (cPOA) and trans-palmitoleic acid (tPOA) promote cholesterol homeostasis and ameliorate hypercholesterolemia remain elusive. To investigate the effects of cPOA and tPOA on cholesterol metabolism and its mechanisms, we induced hypercholesterolemia in mice using a high-fat diet and then intragastrically administered cPOA or tPOA once daily for 4 weeks. tPOA administration reduced serum cholesterol, low-density lipoprotein, high-density lipoprotein, and hepatic free cholesterol and total bile acids (TBAs). Conversely, cPOA had no effect on these parameters except for TBAs. Histological examination of the liver, however, revealed that cPOA ameliorated hepatic steatosis more effectively than tPOA. tPOA significantly reduced the expression of 3-hydroxy-3-methyl glutaryl coenzyme reductase (HMGCR), LXRα, and intestinal Niemann-Pick C1-Like 1 (NPC1L1) and increased cholesterol 7-alpha hydroxylase (CYP7A1) in the liver, whereas cPOA reduced the expression of HMGCR and CYP7A1 in the liver and had no effect on intestinal NPC1L1. In summary, our results suggest that cPOA and tPOA reduce cholesterol synthesis by decreasing HMGCR levels. Furthermore, tPOA, but not cPOA, inhibited intestinal cholesterol absorption by downregulating NPC1L1. Both high-dose tPOA and cPOA may promote the conversion of cholesterol into bile acids by upregulating CYP7A1. tPOA and cPOA prevent hypercholesterolemia via distinct mechanisms.
Collapse
Affiliation(s)
- Wen-Wen Huang
- Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China.,Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bi-Hong Hong
- Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
| | - Kai-Kai Bai
- Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
| | - Ran Tan
- Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
| | - Ting Yang
- Fujian Provincial Key Laboratory on Hematology, Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fuzhou, China
| | - Ji-Peng Sun
- Zhejiang Marine Development Research Institute, Zhoushan, China
| | - Rui-Zao Yi
- Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
| | - Hao Wu
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
20
|
Xiang W, Cheng S, Zhou Y, Ma L. Effects of 1,25(OH) 2 D 3 on lipid droplet growth in adipocytes. Biofactors 2020; 46:943-954. [PMID: 31904171 DOI: 10.1002/biof.1610] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 12/22/2019] [Indexed: 02/06/2023]
Abstract
This study aimed to explore the effects of 1,25(OH)2 D3 on lipid droplet (LD) growth in 3T3-L1 adipocytes of hypertrophy model. Cocktail method was used to induce differentiation in 3T3-L1 cells. After 8 days, the cells were modeled by 100, 300, 600, and 900 μM palmitic acid (PA) for 24 hr. The best concentration of modeling was screened by MTT results and triglycerides (TG) content. The model cells were intervened by 1, 10, and 100 nM 1,25(OH)2 D3 for 24 hr. Then, the TG content of cells were detected and stained by oil red O. The diameter and quantity of LDs were analyzed. mRNA relative expression levels of genes related to LD (CIDE-a, Fsp27, PLIN-1), upstream response factor (PPAR-α, PPAR-γ, and VDR), and TG metabolism (long chain acyl-CoA synthetase 3, 1-acylglycerol-3-phosphate O-acyltransferase 1, adipose triglyceride lipase, diacylglycerol acyltransferase 1, diacylglycerol acyltransferase 2, glycerol-3-phosphate O-acyltransferase 3, glycerol-3-phosphate O-acyltransferase 4, hormone-sensitive lipase, mannosyl (alpha-1,3-)-glycoprotein beta-1,2-N-acetyl glucosaminyl transferase, phosphatidic acid phosphatase, and uncoupling protein-1) were detected by RT-qPCR. A total of 300 μM PA was selected as the optimum concentration. Compared with model group, 10 and 100 nM 1,25(OH)2 D3 decreased the average diameter, increased the quantity of LDs, upregulated PPAR-α and PLIN-1 mRNA expression levels, and downregulated CIDE-a and Fsp27 mRNA expression levels significantly (p < .05). However, 1 nM 1,25(OH)2 D3 did not alter LD morphology and TG content. mRNA expression levels of long chain acyl-CoA synthetase 3, 1-acylglycerol-3-phosphate O-acyltransferase 1, diacylglycerol acyltransferase 2, glycerol-3-phosphate O-acyltransferase 3, and glycerol-3-phosphate O-acyltransferase 4 in 10 and 100 nM groups were significantly lower than those in the model group (p < .05); mRNA expression levels of adipose triglyceride lipase, diacylglycerol acyltransferase 1, hormone-sensitive lipase, mannosyl (alpha-1,3-)-glycoprotein beta-1,2-N-acetyl glucosaminyl transferase, phosphatidic acid phosphatase, and uncoupling protein-1 were significantly increased in the 100 nM group (p < .05). The 10 and 100 nM 1,25(OH)2 D3 can inhibit LD fusion, promote LD decomposition, reduce LD volume, and inhibit lipogenesis through the PPAR-α signaling pathway.
Collapse
Affiliation(s)
- Wei Xiang
- Department of Nutrition and Diet, Changzhou Traditional Chinese Medicine Hospital, Changzhou, China
| | - Shi Cheng
- Department of Nutrition and Food Hygiene, School of Public Health, Xinjiang Medical University, Urumqi, China
| | - Yong Zhou
- Department of Medical Cell Biology and Genetics, College of Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Ling Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Southwest Medical University, Luzhou, China
| |
Collapse
|
21
|
Zhang Y, Zheng Y, Wang X, Qiu J, Liang C, Cheng G, Wang H, Zhao C, Yang W, Zan L, Li A. Bovine Stearoyl-CoA Desaturase 1 Promotes Adipogenesis by Activating the PPARγ Receptor. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:12058-12066. [PMID: 33052678 DOI: 10.1021/acs.jafc.0c05147] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Stearoyl-CoA desaturase 1 (SCD1) is a rate-limiting enzyme that mainly catalyzes the saturated fatty acids (SFAs) into the monounsaturated fatty acids (MUFAs). The expression level of SCD1 is positively correlated with the marbling score. However, the functional mechanism of SCD1 in adipogenesis is still unclear. In this study, we identified SCD1 as highly expressed in subcutaneous and visceral fat, peaking at 2 days after differentiation in bovine stromal vascular fraction (SVF) cells. When the SCD1 was overexpressed in bovine SVF cells, lipid droplets accumulation was increased from 142.46 ± 21.77 to 254.89 ± 11.75 μg/mg (P < 0.01). Further, the expression levels of FABP4, FASN, and ACCα were increased (P < 0.01), while the expression of PPARγ or C/EBPα was not changed at mRNA or protein level (P > 0.05). Dual-luciferase reporter assay showed that the activity of the PPARγ receptor was enhanced by 3.69 times (P < 0.01). Moreover, the contents of palmitoleate (C16:1) and oleate (C18:1) were significantly increased (P < 0.05). Furthermore, 100 μM exogenous oleate increased the lipid accumulation by 22.28 times (P < 0.01). These results suggest that oleate is probably a strong ligand of the PPARγ receptor to enhance adipogenesis.
Collapse
Affiliation(s)
- Yu Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Yan Zheng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Xiaoyu Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Ju Qiu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Chengcheng Liang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Gong Cheng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Hongbao Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Chunping Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Wucai Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Anning Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| |
Collapse
|
22
|
Huang X, Yi S, Hu J, Du Z, Wang Q, Ye Z, Cao Q, Su G, Yuan G, Zhou C, Wang Y, Kijlstra A, Yang P. Analysis of the role of palmitoleic acid in acute anterior uveitis. Int Immunopharmacol 2020; 84:106552. [PMID: 32422526 DOI: 10.1016/j.intimp.2020.106552] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/09/2020] [Accepted: 04/27/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE To study the role of palmitoleic acid (PA) in the pathogenesis of acute anterior uveitis (AAU). METHODS PA levels in feces from AAU patients were measured by gas chromatography coupled with a mass spectrometer (GC-MS) and compared with samples obtained from healthy individuals. Enzyme linked immunosorbent assay (ELISA) and flow cytometry (FCM) were used to assess the effect of PA on dendritic cells (DCs) and CD4+T cells obtained from mice, AAU patients and healthy individuals. C57BL/6 mice were fed with PA or vehicle and experimental autoimmune uveitis (EAU) was induced with a human retinal IRBP651-670 peptide. Disease severity of EAU was evaluated by clinical manifestation and histology. Differentiation of splenic Type 1 helper T cells (Th1) and Th17 cells was evaluated by FCM. Tandem mass tag (TMT)-based proteomics analysis was used to identify differentially expressed proteins following incubation of DCs with PA. RESULTS The fecal concentration of PA was increased in AAU patients as compared with healthy individuals. In vitro, PA promoted apoptosis of DCs and inhibited the secretion of TNF-α from mouse bone-marrow-derived dendritic cells (BMDCs) as well as in DCs from AAU patients and healthy individuals. It only decreased DCs surface marker expression and IL-12p70 secretion in BMDCs and healthy individuals DCs but not in AAU patient DCs. PA-treated BMDCs inhibited Th cell differentiation from mouse naïve CD4+T cells and IL-17 and IFN-γ secretion in co-culture supernatants. PA also inhibited the differentiation of Th cells and secretion of IFN-γ and IL-17 in CD4+T cells from mice, AAU patients and healthy individuals. In vivo, PA-treated EAU mice showed milder clinical and histopathological intraocular manifestations as compared with the control group. PA feeding inhibited differentiation of splenic Th17 cells, whereas Th1 cells were not affected. Up to 30 upregulated and 77 downregulated proteins were identified when comparing PA-treated DCs with controls. CONCLUSION An increased expression of fecal PA was observed in AAU patients. PA was shown to have immunoregulatory effects on DCs and CD4+T cells and attenuated disease severity in EAU mice.
Collapse
Affiliation(s)
- Xinyue Huang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, PR China
| | - Shenglan Yi
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, PR China
| | - Jianping Hu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, PR China
| | - Ziyu Du
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, PR China
| | - Qingfeng Wang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, PR China
| | - Zi Ye
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, PR China
| | - Qingfeng Cao
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, PR China
| | - Guannan Su
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, PR China
| | - Gangxiang Yuan
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, PR China
| | - Chunjiang Zhou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, PR China
| | - Yao Wang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, PR China
| | - Aize Kijlstra
- University Eye Clinic Maastricht, Maastricht, the Netherlands
| | - Peizeng Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, PR China.
| |
Collapse
|
23
|
Iwase M, Tokiwa S, Seno S, Mukai T, Yeh YS, Takahashi H, Nomura W, Jheng HF, Matsumura S, Kusudo T, Osato N, Matsuda H, Inoue K, Kawada T, Goto T. Glycerol kinase stimulates uncoupling protein 1 expression by regulating fatty acid metabolism in beige adipocytes. J Biol Chem 2020; 295:7033-7045. [PMID: 32273338 DOI: 10.1074/jbc.ra119.011658] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 04/02/2020] [Indexed: 11/06/2022] Open
Abstract
Browning of adipose tissue is induced by specific stimuli such as cold exposure and consists of up-regulation of thermogenesis in white adipose tissue. Recently, it has emerged as an attractive target for managing obesity in humans. Here, we performed a comprehensive analysis to identify genes associated with browning in murine adipose tissue. We focused on glycerol kinase (GYK) because its mRNA expression pattern is highly correlated with that of uncoupling protein 1 (UCP1), which regulates the thermogenic capacity of adipocytes. Cold exposure-induced Ucp1 up-regulation in inguinal white adipose tissue (iWAT) was partially abolished by Gyk knockdown (KD) in vivo Consistently, the Gyk KD inhibited Ucp1 expression induced by treatment with the β-adrenergic receptors (βAR) agonist isoproterenol (Iso) in vitro and resulted in impaired uncoupled respiration. Gyk KD also suppressed Iso- and adenylate cyclase activator-induced transcriptional activation and phosphorylation of the cAMP response element-binding protein (CREB). However, we did not observe these effects with a cAMP analog. Therefore Gyk KD related to Iso-induced cAMP products. In Iso-treated Gyk KD adipocytes, stearoyl-CoA desaturase 1 (SCD1) was up-regulated, and monounsaturated fatty acids such as palmitoleic acid (POA) accumulated. Moreover, a SCD1 inhibitor treatment recovered the Gyk KD-induced Ucp1 down-regulation and POA treatment down-regulated Iso-activated Ucp1 Our findings suggest that Gyk stimulates Ucp1 expression via a mechanism that partially depends on the βAR-cAMP-CREB pathway and Gyk-mediated regulation of fatty acid metabolism.
Collapse
Affiliation(s)
- Mari Iwase
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Soshi Tokiwa
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Shigeto Seno
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Suita 565-0871, Japan
| | - Takako Mukai
- Faculty of Human Sciences, Tezukayama Gakuin University, Sakai 590-0113, Japan
| | - Yu-Sheng Yeh
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Haruya Takahashi
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Wataru Nomura
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan.,Research Unit for Physiological Chemistry, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8317, Japan
| | - Huei-Fen Jheng
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Sigenobu Matsumura
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Tatsuya Kusudo
- Faculty of Human Sciences, Tezukayama Gakuin University, Sakai 590-0113, Japan
| | - Naoki Osato
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Suita 565-0871, Japan
| | - Hideo Matsuda
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Suita 565-0871, Japan
| | - Kazuo Inoue
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Teruo Kawada
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan.,Research Unit for Physiological Chemistry, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8317, Japan
| | - Tsuyoshi Goto
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan .,Research Unit for Physiological Chemistry, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8317, Japan
| |
Collapse
|
24
|
da Cunha de Sá RDC, Cruz MM, de Farias TM, da Silva VS, de Jesus Simão J, Telles MM, Alonso-Vale MIC. Fish oil reverses metabolic syndrome, adipocyte dysfunction, and altered adipokines secretion triggered by high-fat diet-induced obesity. Physiol Rep 2020; 8:e14380. [PMID: 32109344 PMCID: PMC7048378 DOI: 10.14814/phy2.14380] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 02/06/2023] Open
Abstract
The effect of fish oil (FO) treatment on high-fat (HF) diet-induced obesity and metabolic syndrome was addressed by analyzing dysfunctions in cells of different adipose depots. For this purpose, mice were initially induced to obesity for 8 weeks following a treatment with FO containing high concentration of EPA compared to DHA (5:1), for additional 8 weeks (by gavage, 3 times per week). Despite the higher fat intake, the HF group showed lower food intake but higher body weight, glucose intolerance and insulin resistance, significant dyslipidemia and increased liver, subcutaneous (inguinal-ING) and visceral (retroperitoneal-RP) adipose depots mass, accompanied by adipocyte hypertrophy and decreased cellularity in both adipose tissue depots. FO treatment reversed all these effects, as well as it improved the metabolic activities of isolated adipocytes, such as glucose uptake and lipolysis in both depots, and de novo synthesis of fatty acids in ING adipocytes. HF diet also significantly increased both the pro and anti-inflammatory cytokines expression by adipocytes, while HF + FO did not differ from control group. Collectively, these data show that the concomitant administration of FO with the HF diet is able to revert metabolic changes triggered by the diet-induced obesity, as well as to promote beneficial alterations in adipose cell activities. The main mechanism underlying all systemic effects involves direct and differential effects on ING and RP adipocytes.
Collapse
Affiliation(s)
- Roberta D. C. da Cunha de Sá
- Post‐graduate Program in Chemical BiologyInstitute of Environmental Sciences, Chemical and PharmaceuticalFederal University of Sao Paulo ‐UNIFESPDiademaSao PauloBrazil
| | - Maysa M. Cruz
- Post‐graduate Program in Chemical BiologyInstitute of Environmental Sciences, Chemical and PharmaceuticalFederal University of Sao Paulo ‐UNIFESPDiademaSao PauloBrazil
| | - Talita M. de Farias
- Post‐graduate Program in Chemical BiologyInstitute of Environmental Sciences, Chemical and PharmaceuticalFederal University of Sao Paulo ‐UNIFESPDiademaSao PauloBrazil
| | - Viviane S. da Silva
- Post‐graduate Program in Chemical BiologyInstitute of Environmental Sciences, Chemical and PharmaceuticalFederal University of Sao Paulo ‐UNIFESPDiademaSao PauloBrazil
| | - Jussara de Jesus Simão
- Post‐graduate Program in Chemical BiologyInstitute of Environmental Sciences, Chemical and PharmaceuticalFederal University of Sao Paulo ‐UNIFESPDiademaSao PauloBrazil
| | - Monica M. Telles
- Post‐graduate Program in Chemical BiologyInstitute of Environmental Sciences, Chemical and PharmaceuticalFederal University of Sao Paulo ‐UNIFESPDiademaSao PauloBrazil
- Department of Biological SciencesInstitute of Environmental Sciences, Chemical and PharmaceuticalFederal University of Sao Paulo ‐ UNIFESPDiademaSao PauloBrazil
| | - Maria Isabel C. Alonso-Vale
- Post‐graduate Program in Chemical BiologyInstitute of Environmental Sciences, Chemical and PharmaceuticalFederal University of Sao Paulo ‐UNIFESPDiademaSao PauloBrazil
- Department of Biological SciencesInstitute of Environmental Sciences, Chemical and PharmaceuticalFederal University of Sao Paulo ‐ UNIFESPDiademaSao PauloBrazil
| |
Collapse
|
25
|
Cruz MM, Simão JJ, de Sá RDCC, Farias TSM, da Silva VS, Abdala F, Antraco VJ, Armelin-Correa L, Alonso-Vale MIC. Palmitoleic Acid Decreases Non-alcoholic Hepatic Steatosis and Increases Lipogenesis and Fatty Acid Oxidation in Adipose Tissue From Obese Mice. Front Endocrinol (Lausanne) 2020; 11:537061. [PMID: 33117273 PMCID: PMC7561405 DOI: 10.3389/fendo.2020.537061] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 08/28/2020] [Indexed: 01/06/2023] Open
Abstract
We recently demonstrated that palmitoleic acid (C16:1n7), a monounsaturated fatty acid, increases the metabolic and oxidative capacity of 3T3-L1 adipocytes. Herein, the effect of 16:1n7 supplementation on metabolic parameters on white adipose tissue (WAT) and liver of obese mice induced by a high-fat diet (HFD) was addressed by analyzing metabolic (dys)function and altered genes expression in adipose tissue, as well as liver and serum biochemistry analysis. For this purpose, mice were induced to obesity for 8 weeks, and from the 5th week, they received 16:1n7 (300 mg/kg per day) or water for 30 days, by gavage. Subcutaneous inguinal (ING) and epididymal (EPI) WAT were removed for analysis of metabolic, (anti)inflammatory, adipogenic, and thermogenic genes expression by real-time reverse transcriptase-polymerase chain reaction. Additionally, metabolic activities of isolated adipocytes, such as glucose uptake, lipogenesis (triacylglycerol esterification), β-oxidation, and lipolysis in ING adipocytes, were also assessed. Despite the higher fat intake, the HFD group showed lower food intake but higher body weight, increased glucose, significant dyslipidemia, and increased liver and adipose depot mass, accompanied by liver steatosis. The 16:1n7 supplementation slowed down the body mass gain and prevented the increase of lipids in the liver. HFD+n7 animals presented increased fatty acid oxidation and lipogenesis compared to control, but no effect was observed on lipolysis and glucose uptake in ING isolated adipocytes. Besides, 16:1n7 increased the content of the mRNA encoding FABP4, but partially prevented the expression of genes encoding ATGL, HSL, perilipin, lipin, C/EBP-α, PPAR-γ, C/EBP-β, CPT1, NRF1, TFAM, PRDM16, and nitric oxide synthase 2 in ING depot from HFD group of animals. Finally, HFD increased Mcp1 and Tnfα expression, and 16:1n7 promoted a more marked increase in it. In summary, the data show that palmitoleic acid promotes metabolic changes and partially prevents the increase in gene expression on adipocytes triggered by obesity, suggesting that HFD+n7 animals do not require the same magnitude of metabolic adaptation to cope with energy demand from the HFD. In the long term, the effects of 16:1n7 may be more evident and beneficial for the function/dysfunction of WAT from an obese organism, with relevant repercussions in the systemic metabolic homeostasis.
Collapse
Affiliation(s)
- Maysa M. Cruz
- Post-graduate Program in Chemical Biology – Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo - UNIFESP, Diadema, Brazil
| | - Jussara J. Simão
- Post-graduate Program in Chemical Biology – Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo - UNIFESP, Diadema, Brazil
| | - Roberta D. C. C. de Sá
- Post-graduate Program in Chemical Biology – Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo - UNIFESP, Diadema, Brazil
| | - Talita S. M. Farias
- Post-graduate Program in Chemical Biology – Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo - UNIFESP, Diadema, Brazil
| | - Viviane S. da Silva
- Post-graduate Program in Chemical Biology – Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo - UNIFESP, Diadema, Brazil
| | - Fernanda Abdala
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo - UNIFESP, Diadema, Brazil
| | - Vitor J. Antraco
- Post-graduate Program in Chemical Biology – Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo - UNIFESP, Diadema, Brazil
| | - Lucia Armelin-Correa
- Post-graduate Program in Chemical Biology – Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo - UNIFESP, Diadema, Brazil
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo - UNIFESP, Diadema, Brazil
| | - Maria Isabel C. Alonso-Vale
- Post-graduate Program in Chemical Biology – Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo - UNIFESP, Diadema, Brazil
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo - UNIFESP, Diadema, Brazil
- *Correspondence: Maria Isabel C. Alonso-Vale
| |
Collapse
|
26
|
The Novel Perspectives of Adipokines on Brain Health. Int J Mol Sci 2019; 20:ijms20225638. [PMID: 31718027 PMCID: PMC6887733 DOI: 10.3390/ijms20225638] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022] Open
Abstract
First seen as a fat-storage tissue, the adipose tissue is considered as a critical player in the endocrine system. Precisely, adipose tissue can produce an array of bioactive factors, including cytokines, lipids, and extracellular vesicles, which target various systemic organ systems to regulate metabolism, homeostasis, and immune response. The global effects of adipokines on metabolic events are well defined, but their impacts on brain function and pathology remain poorly defined. Receptors of adipokines are widely expressed in the brain. Mounting evidence has shown that leptin and adiponectin can cross the blood–brain barrier, while evidence for newly identified adipokines is limited. Significantly, adipocyte secretion is liable to nutritional and metabolic states, where defective circuitry, impaired neuroplasticity, and elevated neuroinflammation are symptomatic. Essentially, neurotrophic and anti-inflammatory properties of adipokines underlie their neuroprotective roles in neurodegenerative diseases. Besides, adipocyte-secreted lipids in the bloodstream can act endocrine on the distant organs. In this article, we have reviewed five adipokines (leptin, adiponectin, chemerin, apelin, visfatin) and two lipokines (palmitoleic acid and lysophosphatidic acid) on their roles involving in eating behavior, neurotrophic and neuroprotective factors in the brain. Understanding and regulating these adipokines can lead to novel therapeutic strategies to counteract metabolic associated eating disorders and neurodegenerative diseases, thus promote brain health.
Collapse
|
27
|
Hernández-Saavedra D, Stanford KI. The Regulation of Lipokines by Environmental Factors. Nutrients 2019; 11:E2422. [PMID: 31614481 PMCID: PMC6835582 DOI: 10.3390/nu11102422] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/06/2019] [Accepted: 10/09/2019] [Indexed: 01/08/2023] Open
Abstract
Adipose tissue is a highly metabolically-active tissue that senses and secretes hormonal and lipid mediators that facilitate adaptations to metabolic tissues. In recent years, the role of lipokines, which are lipid species predominantly secreted from adipose tissue that act as hormonal regulators in many metabolic tissues, has been an important area of research for obesity and diabetes. Previous studies have identified that these secreted lipids, including palmitoleate, 12,13-diHOME, and fatty acid-hydroxy-fatty acids (FAHFA) species, are important regulators of metabolism. Moreover, environmental factors that directly affect the secretion of lipokines such as diet, exercise, and exposure to cold temperatures constitute attractive therapeutic strategies, but the mechanisms that regulate lipokine stimulation have not been thoroughly reviewed. In this study, we will discuss the chemical characteristics of lipokines that position them as attractive targets for chronic disease treatment and prevention and the emerging roles of lipokines as regulators of inter-tissue communication. We will define the target tissues of lipokines, and explore the ability of lipokines to prevent or delay the onset and development of chronic diseases. Comprehensive understanding of the lipokine synthesis and lipokine-driven regulation of metabolic outcomes is instrumental for developing novel preventative and therapeutic strategies that harness adipose tissue-derived lipokines.
Collapse
Affiliation(s)
- Diego Hernández-Saavedra
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
28
|
Funcke JB, Scherer PE. Beyond adiponectin and leptin: adipose tissue-derived mediators of inter-organ communication. J Lipid Res 2019; 60:1648-1684. [PMID: 31209153 PMCID: PMC6795086 DOI: 10.1194/jlr.r094060] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/17/2019] [Indexed: 01/10/2023] Open
Abstract
The breakthrough discoveries of leptin and adiponectin more than two decades ago led to a widespread recognition of adipose tissue as an endocrine organ. Many more adipose tissue-secreted signaling mediators (adipokines) have been identified since then, and much has been learned about how adipose tissue communicates with other organs of the body to maintain systemic homeostasis. Beyond proteins, additional factors, such as lipids, metabolites, noncoding RNAs, and extracellular vesicles (EVs), released by adipose tissue participate in this process. Here, we review the diverse signaling mediators and mechanisms adipose tissue utilizes to relay information to other organs. We discuss recently identified adipokines (proteins, lipids, and metabolites) and briefly outline the contributions of noncoding RNAs and EVs to the ever-increasing complexities of adipose tissue inter-organ communication. We conclude by reflecting on central aspects of adipokine biology, namely, the contribution of distinct adipose tissue depots and cell types to adipokine secretion, the phenomenon of adipokine resistance, and the capacity of adipose tissue to act both as a source and sink of signaling mediators.
Collapse
Affiliation(s)
- Jan-Bernd Funcke
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
29
|
Liu X, Zhao K, Yang X, Zhao Y. Gut Microbiota and Metabolome Response of Decaisnea insignis Seed Oil on Metabolism Disorder Induced by Excess Alcohol Consumption. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:10667-10677. [PMID: 31483636 DOI: 10.1021/acs.jafc.9b04792] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
This study investigated the modulatory effects of Decaisnea insignis seed oil (DISO), which was rich in palmitoleic acid (55.25%), palmitic acid (12.25%), and oleic acid (28.74%), on alcohol-induced metabolism disorder in mice. Fifty mice were orally administered with 38% alcohol (0.4 mL/day) and without or with DISO (3, 6, and 12 g/kg) for consecutive 12 weeks. DISO inhibited the alcohol-induced weight loss and liver function abnormality (p < 0.01) and shifted the profiles of cecal microbiome: elevating the abundance of Lactobacillus, Ruminoccoceae_UCG_004 (p < 0.05) and decreasing abundance of Parabacteroides (p < 0.05). This treatment also regulated metabolome response of amino acid and lipid metabolism in cecal content: upregulating 5-hydroxyindole-3-acetic acid (p < 0.05), 6-hydroxynicotinic acid, 5-methoxytryptamine, nicotinamide, and nicotinic acid (p < 0.1) and downregulating androsterone, tryptophan, and indole-3-acetamide (p < 0.05). DISO protected against alcoholic liver injury and gut microbiota dysbiosis by enriching the relative abundance of Lactobacillus, which was positively associated with the improvement of intestinal permeability and tryptophan metabolism.
Collapse
|
30
|
Farias TDSMD, Paixao RID, Cruz MM, de Sa RDCDC, Simão JDJ, Antraco VJ, Alonso-Vale MIC. Melatonin Supplementation Attenuates the Pro-Inflammatory Adipokines Expression in Visceral Fat from Obese Mice Induced by A High-Fat Diet. Cells 2019; 8:E1041. [PMID: 31489938 PMCID: PMC6770101 DOI: 10.3390/cells8091041] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 12/16/2022] Open
Abstract
Obesity is defined as a condition of abnormal or excessive fat accumulation in white adipose tissue that results from the exacerbated consumption of calories associated with low energy expenditure. Fat accumulation in both adipose tissue and other organs contributes to a systemic inflammation leading to the development of metabolic disorders such as type 2 diabetes, hypertension, and dyslipidemia. Melatonin is a potent antioxidant and improves inflammatory processes and energy metabolism. Using male mice fed a high-fat diet (HFD-59% fat from lard and soybean oil; 9:1) as an obesity model, we investigated the effects of melatonin supplementation on the prevention of obesity-associated complications through an analysis of plasma biochemical profile, body and fat depots mass, adipocytes size and inflammatory cytokines expression in epididymal (EPI) adipose depot. Melatonin prevented a gain of body weight and fat depot mass as well as adipocyte hypertrophy. Melatonin also reversed the increase of total cholesterol, triglycerides and LDL-cholesterol. In addition, this neurohormone was effective in completely decreasing the inflammatory cytokines leptin and resistin in plasma. In the EPI depot, melatonin reversed the increase of leptin, Il-6, Mcp-1 and Tnf-α triggered by obesity. These data allow us to infer that melatonin presents an anti-obesity effect since it acts to prevent the progression of pro-inflammatory markers in the epididymal adipose tissue together with a reduction in adiposity.
Collapse
Affiliation(s)
- Talita da Silva Mendes de Farias
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-130, Brazil.
| | - Regislane Ino da Paixao
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-130, Brazil.
| | - Maysa Mariana Cruz
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-130, Brazil.
| | | | - Jussara de Jesus Simão
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-130, Brazil.
| | - Vitor Jaco Antraco
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-130, Brazil.
| | - Maria Isabel Cardoso Alonso-Vale
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of Sao Paulo, Diadema 09913-130, Brazil.
| |
Collapse
|
31
|
Transforming Growth Factor β Acts as a Regulatory Molecule for Lipogenic Pathways among Hepatitis C Virus Genotype-Specific Infections. J Virol 2019; 93:JVI.00811-19. [PMID: 31243135 DOI: 10.1128/jvi.00811-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 06/21/2019] [Indexed: 12/14/2022] Open
Abstract
Hepatitis C virus (HCV) infection promotes metabolic disorders, and the severity of lipogenic disease depends upon the infecting virus genotype. Here, we have examined HCV genotype 1-, 2-, or 3-specific regulation of lipid metabolism, involving transforming growth factor β (TGF-β)-regulated phospho-Akt (p-Akt) and peroxisome proliferator-activated receptor alpha (PPARα) axes. Since HCV core protein is one of the key players in metabolic regulation, we also examined its contribution in lipid metabolic pathways. The expression of regulatory molecules, TGF-β1/2, phospho-Akt (Ser473), PPARα, sterol regulatory element-binding protein 1 (SREBP-1), fatty acid synthase (FASN), hormone-sensitive lipase (HSL), and acyl dehydrogenases was analyzed in virus-infected hepatocytes. Interestingly, HCV genotype 3a exhibited much higher activation of TGF-β and p-Akt, with a concurrent decrease in PPARα expression and fatty acid oxidation. A significant and similar decrease in HSL, unlike in HCV genotype 1a, was observed with both genotypes 2a and 3a. Similar observations were made from ectopic expression of the core genomic region from each genotype. The key role of TGF-β was further verified using specific small interfering RNA (siRNA). Together, our results highlight a significant difference in TGF-β-induced activity for the HCV genotype 2a- or 3a-induced lipogenic pathway, exhibiting higher triglyceride synthesis and a decreased lipolytic mechanism. These results may help in therapeutic modalities for early treatment of HCV genotype-associated lipid metabolic disorders.IMPORTANCE Hepatic steatosis is a frequent complication associated with chronic hepatitis C virus (HCV) infection and is a key prognostic indicator for progression to fibrosis and cirrhosis. Several mechanisms are proposed for the development of steatosis, especially with HCV genotype 3a. Our observations suggest that transforming growth factor β (TGF-β) and peroxisome proliferator-activated receptor alpha (PPARα)-associated mechanistic pathways in hepatocytes infected with HCV genotype 2a and 3a differ from those in cells infected with genotype 1a. The results suggest that a targeted therapeutic approach for enhanced PPARα and lipolysis may reduce HCV genotype-associated lipid metabolic disorder in liver disease.
Collapse
|
32
|
Oliveira TE, Castro É, Belchior T, Andrade ML, Chaves-Filho AB, Peixoto AS, Moreno MF, Ortiz-Silva M, Moreira RJ, Inague A, Yoshinaga MY, Miyamoto S, Moustaid-Moussa N, Festuccia WT. Fish Oil Protects Wild Type and Uncoupling Protein 1-Deficient Mice from Obesity and Glucose Intolerance by Increasing Energy Expenditure. Mol Nutr Food Res 2019; 63:e1800813. [PMID: 30632684 DOI: 10.1002/mnfr.201800813] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 12/21/2018] [Indexed: 12/22/2022]
Abstract
SCOPE The mechanisms and involvement of uncoupling protein 1 (UCP1) in the protection from obesity and insulin resistance induced by intake of a high-fat diet rich in omega-3 (n-3) fatty acids are investigated. METHODS AND RESULTS C57BL/6J mice are fed either a low-fat (control group) or one of two isocaloric high-fat diets containing either lard (HFD) or fish oil (HFN3) as fat source and evaluated for body weight, adiposity, energy expenditure, glucose homeostasis, and inguinal white and interscapular brown adipose tissue (iWAT and iBAT, respectively) gene expression, lipidome, and mitochondrial bioenergetics. HFN3 intake protected from obesity, glucose and insulin intolerances, and hyperinsulinemia. This is associated with increased energy expenditure, iWAT UCP1 expression, and incorporation of n-3 eicosapentaenoic and docosahexaenoic fatty acids in iWAT and iBAT triacylglycerol. Importantly, HFN3 is equally effective in reducing body weight gain, adiposity, and glucose intolerance and increasing energy expenditure in wild-type and UCP1-deficient mice without recruiting other thermogenic processes in iWAT and iBAT, such as mitochondrial uncoupling and SERCA-mediated calcium and creatine-driven substrate cyclings. CONCLUSION Intake of a high-fat diet rich in omega-3 fatty acids protects both wild-type and UCP1-deficient mice from obesity and insulin resistance by increasing energy expenditure through unknown mechanisms.
Collapse
Affiliation(s)
- Tiago E Oliveira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Érique Castro
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Thiago Belchior
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Maynara L Andrade
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Adriano B Chaves-Filho
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Albert S Peixoto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Mayara F Moreno
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Milene Ortiz-Silva
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Rafael J Moreira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Alex Inague
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo, 05508-000, Brazil
| | - Marcos Y Yoshinaga
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo, 05508-000, Brazil
| | - Sayuri Miyamoto
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo, 05508-000, Brazil
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences and Obesity Research Cluster, Texas Tech University, Lubbock, 79409, TX, USA
| | - William T Festuccia
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| |
Collapse
|
33
|
de Souza CO, Valenzuela CA, Baker EJ, Miles EA, Rosa Neto JC, Calder PC. Palmitoleic Acid has Stronger Anti-Inflammatory Potential in Human Endothelial Cells Compared to Oleic and Palmitic Acids. Mol Nutr Food Res 2018; 62:e1800322. [PMID: 30102465 DOI: 10.1002/mnfr.201800322] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/18/2018] [Indexed: 12/13/2022]
Abstract
SCOPE Fatty acids (FAs) may affect endothelial cell (EC) function, influencing atherogenesis and inflammatory processes. Palmitoleic acid (POA) has been described as an anti-inflammatory FA. However, its effects on ECs are underexplored. This study compares the effects of POA with those of palmitic acid (PA) and oleic acid (OA) on EC inflammatory responses. METHODS AND RESULTS EAHy926 cells (EC lineage) are exposed to PA, OA, or POA, and stimulated with tumor necrosis factor (TNF)-α. Associated with the FA's own incorporation, PA induces a twofold increase in arachidonic acid, while POA increases the amount of cis-vaccenic acid. PA, but not OA, enhances the production of IL-6 and IL-8 in response to TNF-α. In contrast, POA decreases production of monocyte chemotactic protein (MCP)-1, IL-6, and IL-8 compared to PA. TNF-α increases surface intercellular adhesion molecule-1 expression previously decreased by POA. TNF-α stimulation increases the expression of NFκB, cyclooxygenase (COX)-2, MCP-1, and IL-6 genes and reduces the expression of peroxisome proliferator-activated receptor (PPAR)-α gene. PA enhances the expression of MCP-1, IL-6, and COX-2 genes, while POA downregulates these genes, decreases expression of NFκB, and upregulates PPAR-α gene expression. CONCLUSION POA has anti-inflammatory effects on ECs stimulated with TNF-α and may counter endothelial dysfunction.
Collapse
Affiliation(s)
- Camila Oliveira de Souza
- Department of Cell and Developmental Biology, University of São Paulo, 1524, Lineu prestes av, São Paulo, Brazil
| | - Carina A Valenzuela
- Human Development and Health Academic Unit, Faculty of Medicine, Tremona Rd, S016 6HT, University of Southampton, Southampton, UK.,School of Nutrition, Faculty of Pharmacy, University of Valparaíso, 1093, Gran Bretaña av, Playa Ancha, Valparaíso, Chile
| | - Ella J Baker
- Human Development and Health Academic Unit, Faculty of Medicine, Tremona Rd, S016 6HT, University of Southampton, Southampton, UK
| | - Elizabeth A Miles
- Human Development and Health Academic Unit, Faculty of Medicine, Tremona Rd, S016 6HT, University of Southampton, Southampton, UK
| | - José C Rosa Neto
- Department of Cell and Developmental Biology, University of São Paulo, 1524, Lineu prestes av, São Paulo, Brazil
| | - Philip C Calder
- Human Development and Health Academic Unit, Faculty of Medicine, Tremona Rd, S016 6HT, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Tremona Rd, S016 6HT, Southampton, UK
| |
Collapse
|
34
|
Araki M, Nakagawa Y, Oishi A, Han SI, Wang Y, Kumagai K, Ohno H, Mizunoe Y, Iwasaki H, Sekiya M, Matsuzaka T, Shimano H. The Peroxisome Proliferator-Activated Receptor α (PPARα) Agonist Pemafibrate Protects against Diet-Induced Obesity in Mice. Int J Mol Sci 2018; 19:ijms19072148. [PMID: 30041488 PMCID: PMC6073532 DOI: 10.3390/ijms19072148] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/13/2018] [Accepted: 07/13/2018] [Indexed: 12/12/2022] Open
Abstract
Peroxisome proliferator-activated receptor α (PPARα) is a therapeutic target for hyperlipidemia. Pemafibrate (K-877) is a new selective PPARα modulator activating PPARα transcriptional activity. To determine the effects of pemafibrate on diet-induced obesity, wild-type mice were fed a high-fat diet (HFD) containing pemafibrate for 12 weeks. Like fenofibrate, pemafibrate significantly suppressed HFD-induced body weight gain; decreased plasma glucose, insulin and triglyceride (TG) levels; and increased plasma fibroblast growth factor 21 (FGF21). However, compared to the dose of fenofibrate, a relatively low dose of pemafibrate showed these effects. Pemafibrate activated PPARα transcriptional activity in the liver, increasing both hepatic expression and plasma levels of FGF21. Additionally, pemafibrate increased the expression of genes involved in thermogenesis and fatty acid oxidation, including Ucp1, Cidea and Cpt1b in inguinal adipose tissue (iWAT) and the mitochondrial marker Elovl3 in brown adipose tissue (BAT). Therefore, pemafibrate activates thermogenesis in iWAT and BAT by increasing plasma levels of FGF21. Additionally, pemafibrate induced the expression of Atgl and Hsl in epididymal white adipose tissue, leading to the activation of lipolysis. Taken together, pemafibrate suppresses diet-induced obesity in mice and improves their obesity-related metabolic abnormalities. We propose that pemafibrate may be useful for the suppression and improvement of obesity-induced metabolic abnormalities.
Collapse
Affiliation(s)
- Masaya Araki
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Yoshimi Nakagawa
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Asayo Oishi
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Song-Iee Han
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Yunong Wang
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Kae Kumagai
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Hiroshi Ohno
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Yuhei Mizunoe
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Hitoshi Iwasaki
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Motohiro Sekiya
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Takashi Matsuzaka
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Hitoshi Shimano
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan.
- Japan Agency for Medical Research and Development⁻Core Research for Evolutional Science and Technology (AMED-CREST), Chiyoda-ku, Tokyo 100-1004, Japan.
| |
Collapse
|
35
|
Regulation of Metabolic Disease-Associated Inflammation by Nutrient Sensors. Mediators Inflamm 2018; 2018:8261432. [PMID: 30116154 PMCID: PMC6079375 DOI: 10.1155/2018/8261432] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/21/2018] [Accepted: 06/14/2018] [Indexed: 12/15/2022] Open
Abstract
Visceral obesity is frequently associated with the development of type 2 diabetes (T2D), a highly prevalent chronic disease that features insulin resistance and pancreatic β-cell dysfunction as important hallmarks. Recent evidence indicates that the chronic, low-grade inflammation commonly associated with visceral obesity plays a major role connecting the excessive visceral fat deposition with the development of insulin resistance and pancreatic β-cell dysfunction. Herein, we review the mechanisms by which nutrients modulate obesity-associated inflammation.
Collapse
|
36
|
Cruz MM, Lopes AB, Crisma AR, de Sá RCC, Kuwabara WMT, Curi R, de Andrade PBM, Alonso-Vale MIC. Palmitoleic acid (16:1n7) increases oxygen consumption, fatty acid oxidation and ATP content in white adipocytes. Lipids Health Dis 2018; 17:55. [PMID: 29554895 PMCID: PMC5859716 DOI: 10.1186/s12944-018-0710-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 03/13/2018] [Indexed: 12/27/2022] Open
Abstract
Background We have recently demonstrated that palmitoleic acid (16:1n7) increases lipolysis, glucose uptake and glucose utilization for energy production in white adipose cells. In the present study, we tested the hypothesis that palmitoleic acid modulates bioenergetic activity in white adipocytes. Methods For this, 3 T3-L1 pre-adipocytes were differentiated into mature adipocytes in the presence (or absence) of palmitic (16:0) or palmitoleic (16:1n7) acid at 100 or 200 μM. The following parameters were evaluated: lipolysis, lipogenesis, fatty acid (FA) oxidation, ATP content, oxygen consumption, mitochondrial mass, citrate synthase activity and protein content of mitochondrial oxidative phosphorylation (OXPHOS) complexes. Results Treatment with 16:1n7 during 9 days raised basal and isoproterenol-stimulated lipolysis, FA incorporation into triacylglycerol (TAG), FA oxidation, oxygen consumption, protein expression of subunits representing OXPHOS complex II, III, and V and intracellular ATP content. These effects were not observed in adipocytes treated with 16:0. Conclusions Palmitoleic acid, by concerted action on lipolysis, FA esterification, mitochondrial FA oxidation, oxygen consumption and ATP content, does enhance white adipocyte energy expenditure and may act as local hormone.
Collapse
Affiliation(s)
- Maysa M Cruz
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo, 210, Sao Nicolau St, Diadema, 09913-030, Brazil
| | - Andressa B Lopes
- Department of Nursing , Health Sciences Center, Federal University of Espírito Santo, Vitória, Brazil
| | - Amanda R Crisma
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Roberta C C de Sá
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo, 210, Sao Nicolau St, Diadema, 09913-030, Brazil
| | - Wilson M T Kuwabara
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Rui Curi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Interdisciplinary Postgraduate Program in Health Sciences, Institute of Physical Activity Sciences and Sports, Cruzeiro do Sul University, São Paulo, Brazil
| | - Paula B M de Andrade
- Interdisciplinary Postgraduate Program in Health Sciences, Institute of Physical Activity Sciences and Sports, Cruzeiro do Sul University, São Paulo, Brazil
| | - Maria I C Alonso-Vale
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo, 210, Sao Nicolau St, Diadema, 09913-030, Brazil.
| |
Collapse
|
37
|
de Souza CO, Vannice GK, Rosa Neto JC, Calder PC. Is Palmitoleic Acid a Plausible Nonpharmacological Strategy to Prevent or Control Chronic Metabolic and Inflammatory Disorders? Mol Nutr Food Res 2017; 62. [PMID: 28980402 DOI: 10.1002/mnfr.201700504] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 09/11/2017] [Indexed: 12/16/2022]
Abstract
Although dietary fatty acids can modulate metabolic and immune responses, the effects of palmitoleic acid (16:1n-7) remain unclear. Since this monounsaturated fatty acid is described as a lipokine, studies with cell culture and rodent models have suggested it enhances whole body insulin sensitivity, stimulates insulin secretion by β cells, increases hepatic fatty acid oxidation, improves the blood lipid profile, and alters macrophage differentiation. However, human studies report elevated blood levels of palmitoleic acid in people with obesity and metabolic syndrome. These findings might be reflection of the level or activity of stearoyl-CoA desaturase-1, which synthesizes palmitoleate and is enhanced in liver and adipose tissue of obese patients. The aim of this review is to describe the immune-metabolic effects of palmitoleic acid observed in cell culture, animal models, and humans to answer the question of whether palmitoleic acid is a plausible nonpharmacological strategy to prevent, control, or ameliorate chronic metabolic and inflammatory disorders. Despite the beneficial effects observed in cell culture and in animal studies, there are insufficient human intervention studies to fully understand the physiological effects of palmitoleic acid. Therefore, more human-based research is needed to identify whether palmitoleic acid meets the promising therapeutic potential suggested by the preclinical research.
Collapse
Affiliation(s)
- Camila O de Souza
- Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| | | | - José C Rosa Neto
- Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| | - Philip C Calder
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University of Southampton, Southampton, UK
| |
Collapse
|
38
|
Bond LM, Ntambi JM. UCP1 deficiency increases adipose tissue monounsaturated fatty acid synthesis and trafficking to the liver. J Lipid Res 2017; 59:224-236. [PMID: 29203476 DOI: 10.1194/jlr.m078469] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 11/28/2017] [Indexed: 12/26/2022] Open
Abstract
Uncoupling protein-1 (UCP1) facilitates thermogenesis in brown and beige adipocytes and can promote energy expenditure by decreasing mitochondrial respiratory efficiency. Defects in UCP1 and brown adipose tissue thermogenesis subject animals to chronic cold stress and elicit compensatory responses to generate heat. How UCP1 regulates white adipose tissue (WAT) lipid biology and tissue crosstalk is not completely understood. Here, we probed the effect of UCP1 deficiency on FA metabolism in inguinal and epididymal WAT and investigated how these metabolic perturbations influence hepatic lipid homeostasis. We report that at standard housing temperature (21°C), loss of UCP1 induces inguinal WAT de novo lipogenesis through transcriptional activation of the lipogenic gene program and elevated GLUT4. Inguinal adipocyte hyperplasia and depot expansion accompany the increase in lipid synthesis. We also found that UCP1 deficiency elevates adipose stearoyl-CoA desaturase gene expression, and increased inguinal WAT lipolysis supports the transport of adipose-derived palmitoleate (16:1n7) to the liver and hepatic triglyceride accumulation. The observed WAT and liver phenotypes were resolved by housing animals at thermoneutral housing (30°C). These data illustrate depot-specific responses to impaired BAT thermogenesis and communication between WAT and liver in UCP1-/- mice.
Collapse
Affiliation(s)
- Laura M Bond
- Departments of Biochemistry University of Wisconsin-Madison, Madison, WI 53706
| | - James M Ntambi
- Departments of Biochemistry University of Wisconsin-Madison, Madison, WI 53706 .,Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
39
|
Souza CO, Teixeira AA, Biondo LA, Silveira LS, Calder PC, Rosa Neto JC. Palmitoleic acid reduces the inflammation in LPS-stimulated macrophages by inhibition of NFκB, independently of PPARs. Clin Exp Pharmacol Physiol 2017; 44:566-575. [PMID: 28135761 DOI: 10.1111/1440-1681.12736] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 01/06/2017] [Accepted: 01/20/2017] [Indexed: 12/27/2022]
Abstract
Palmitoleic acid (PM, 16:1n-7) has anti-inflammatory properties that could be linked to higher expression of PPARα, an inhibitor of NFκB. Macrophages play a major role in the pathogenesis of chronic inflammation, however, the effects of PM on macrophages are underexplored. Thus, we aimed to investigate the effects of PM in activated macrophages as well the role of PPARα. Primary macrophages were isolated from C57BL/6 wild type (WT) and PPARα knockout (KO) mice, cultured under standard conditions and exposed to lipopolysaccharides LPS (2.5 μg/ml) and PM 600 μmol/L conjugated with albumin for 24 hours. The stimulation with LPS increased the production of interleukin (IL)-6 and IL-1β while PM decreased the production of IL-6 in WT macrophages. In KO macrophages, LPS increased the production of tumour necrosis factor (TNF)-α and IL-6 and PM decreased the production of TNFα. The expression of inflammatory markers such NFκB and IL1β were increased by LPS and decreased by PM in both WT and KO macrophages. PM reduced the expression of MyD88 and caspase-1 in KO macrophages, and the expression of TLR4 and HIF-1α in both WT and KO macrophages, although LPS had no effect. CD86, an inflammatory macrophage marker, was reduced by PM independently of genotype. PM increased PPARγ and reduced PPARβ gene expression in macrophages of both genotypes, and increased ACOX-1 expression in KO macrophages. In conclusion, PM promotes anti-inflammatory effects in macrophages exposed to LPS through inhibition of inflammasome pathway, which was independent of PPARα, PPARϒ and AMPK, thus the molecular mechanisms of anti-inflammatory response caused by PM is still unclear.
Collapse
Affiliation(s)
- Camila O Souza
- Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| | - Alexandre As Teixeira
- Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| | - Luana A Biondo
- Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| | - Loreana S Silveira
- Exercise and Immunometabolism Research Group, Department of Physical Education, State University of São Paulo, Presidente Prudente, Brazil
| | - Philip C Calder
- Human Development and Health, University of Southampton, Southampton, UK
| | - José C Rosa Neto
- Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
40
|
Bolsoni-Lopes A, Alonso-Vale MIC. Lipolysis and lipases in white adipose tissue - An update. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2017; 59:335-42. [PMID: 26331321 DOI: 10.1590/2359-3997000000067] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 04/22/2015] [Indexed: 11/22/2022]
Abstract
Lipolysis is defined as the sequential hydrolysis of triacylglycerol (TAG) stored in cell lipid droplets. For many years, it was believed that hormone-sensitive lipase (HSL) and monoacylglycerol lipase (MGL) were the main enzymes catalyzing lipolysis in the white adipose tissue. Since the discovery of adipose triglyceride lipase (ATGL) in 2004, many studies were performed to investigate and characterize the actions of this lipase, as well as of other proteins and possible regulatory mechanisms involved, which reformulated the concept of lipolysis. Novel findings from these studies include the identification of lipolytic products as signaling molecules regulating important metabolic processes in many non-adipose tissues, unveiling a previously underestimated aspect of lipolysis. Thus, we present here an updated review of concepts and regulation of white adipocyte lipolysis with a special emphasis in its role in metabolism homeostasis and as a source of important signaling molecules.
Collapse
Affiliation(s)
- Andressa Bolsoni-Lopes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, BR
| | - Maria Isabel C Alonso-Vale
- Departamento de Ciências Biológicas, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema, SP, BR
| |
Collapse
|
41
|
Masi LN, Martins AR, Crisma AR, do Amaral CL, Davanso MR, Serdan TDA, da Cunha de Sá RDC, Cruz MM, Alonso-Vale MIC, Torres RP, Mancini-Filho J, Pereira JNB, da Silva Righetti MM, Liberti EA, Hirabara SM, Curi R. Combination of a high-fat diet with sweetened condensed milk exacerbates inflammation and insulin resistance induced by each separately in mice. Sci Rep 2017. [PMID: 28638152 PMCID: PMC5479812 DOI: 10.1038/s41598-017-04308-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Obesogenic diets increase body weight and cause insulin resistance (IR), however, the association of these changes with the main macronutrient in the diet remains to be elucidated. Male C57BL/6 mice were fed with: control (CD), CD and sweetened condensed milk (HS), high-fat (HF), and HF and condensed milk (HSHF). After 2 months, increased body weight, glucose intolerance, adipocyte size and cholesterol levels were observed. As compared with CD, HS ingested the same amount of calories whereas HF and HSHF ingested less. HS had increased plasma AST activity and liver type I collagen. HF caused mild liver steatosis and hepatocellular damage. HF and HSHF increased LDL-cholesterol, hepatocyte and adipocyte hypertrophy, TNF-α by macrophages and decreased lipogenesis and adiponectin in adipose tissue (AT). HSHF exacerbated these effects, increasing IR, lipolysis, mRNA expression of F4/80 and leptin in AT, Tlr-4 in soleus muscle and IL-6, IL-1β, VCAM-1, and ICAM-1 protein in AT. The three obesogenic diets induced obesity and metabolic dysfunction. HS was more proinflammatory than the HF and induced hepatic fibrosis. The HF was more detrimental in terms of insulin sensitivity, and it caused liver steatosis. The combination HSHF exacerbated the effects of each separately on insulin resistance and AT inflammatory state.
Collapse
Affiliation(s)
- Laureane Nunes Masi
- Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro of Sul University, Sao Paulo, Brazil.
| | - Amanda Roque Martins
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Amanda Rabello Crisma
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Cátia Lira do Amaral
- Campus of Exact Sciences and Technology, State University of Goias, Anapolis, Brazil
| | - Mariana Rodrigues Davanso
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | | | - Maysa Mariana Cruz
- Department of Biological Sciences, Institute of Biomedical Sciences, Federal University of Sao Paulo, Sao Paulo, Brazil
| | | | - Rosângela Pavan Torres
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Jorge Mancini-Filho
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | | | - Edson Aparecido Liberti
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Sandro Massao Hirabara
- Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro of Sul University, Sao Paulo, Brazil.,Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Rui Curi
- Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro of Sul University, Sao Paulo, Brazil.,Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
42
|
de Souza CO, Teixeira AA, Biondo LA, Lima Junior EA, Batatinha HA, Rosa Neto JC. Palmitoleic Acid Improves Metabolic Functions in Fatty Liver by PPARα-Dependent AMPK Activation. J Cell Physiol 2017; 232:2168-2177. [DOI: 10.1002/jcp.25715] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 11/30/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Camila O. de Souza
- Department of Cell and Developmental Biology; University of São Paulo; São Paulo Brazil
| | | | - Luana A. Biondo
- Department of Cell and Developmental Biology; University of São Paulo; São Paulo Brazil
| | - Edson A. Lima Junior
- Department of Cell and Developmental Biology; University of São Paulo; São Paulo Brazil
| | - Helena A.P. Batatinha
- Department of Cell and Developmental Biology; University of São Paulo; São Paulo Brazil
| | - Jose C. Rosa Neto
- Department of Cell and Developmental Biology; University of São Paulo; São Paulo Brazil
| |
Collapse
|
43
|
Identification of Palmitoleic Acid Controlled by mTOR Signaling as a Biomarker of Polymyositis. J Immunol Res 2017; 2017:3262384. [PMID: 28194428 PMCID: PMC5282408 DOI: 10.1155/2017/3262384] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 09/04/2016] [Accepted: 12/04/2016] [Indexed: 02/05/2023] Open
Abstract
Polymyositis (PM) is a chronic disease characterized by muscle pain, weakness, and increase in muscle-related enzymes, accompanied with inflammations in lymphocytes. However, it is not well understood how the molecular alternations in lymphocytes contribute to the development of polymyositis. The mechanistic target of rapamycin (mTOR) signaling is the central regulator of metabolism and inflammation in mammalian cells. Based on previous studies, we proposed that mTOR signaling may control inflammatory reactions via lipid metabolism. In this study, we aim to figure out the role of mTOR signaling in the development of polymyositis and identify novel biomarkers for the detection and therapy of polymyositis. After screening and validation, we found that palmitoleic acid, a monounsaturated fatty acid, is highly regulated by mTOR signaling. Inhibition of mTORC1 activity decreases palmitoleic acid level. Moreover, mTORC1 regulates the level of palmitoleic acid by controlling its de novo synthesis. Importantly, increased palmitoleic acid has been proven to be a marker of polymyositis. Our work identifies palmitoleic acid in peripheral blood mononuclear cells (PBMC) as a biomarker of polymyositis and offers new targets to the clinical therapy.
Collapse
|
44
|
CREB3L3 controls fatty acid oxidation and ketogenesis in synergy with PPARα. Sci Rep 2016; 6:39182. [PMID: 27982131 PMCID: PMC5159891 DOI: 10.1038/srep39182] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 11/18/2016] [Indexed: 12/13/2022] Open
Abstract
CREB3L3 is involved in fatty acid oxidation and ketogenesis in a mutual manner with PPARα. To evaluate relative contribution, a combination of knockout and transgenic mice was investigated. On a ketogenic-diet (KD) that highlights capability of hepatic ketogenesis, Creb3l3−/− mice exhibited reduction of expression of genes for fatty oxidation and ketogenesis comparable to Ppara−/− mice. Most of the genes were further suppressed in double knockout mice indicating independent contribution of hepatic CREB3L3. During fasting, dependency of ketogenesis on CREB3L3 is lesser extents than Ppara−/− mice suggesting importance of adipose PPARα for supply of FFA and hyperlipidemia in Creb3l3−/− mice. In conclusion CREB3L3 plays a crucial role in hepatic adaptation to energy starvation via two pathways: direct related gene regulation and an auto-loop activation of PPARα. Furthermore, as KD-fed Creb3l3−/− mice exhibited severe fatty liver, activating inflammation, CREB3L3 could be a therapeutic target for NAFLD.
Collapse
|
45
|
Passos MEP, Alves HHO, Momesso CM, Faria FG, Murata G, Cury-Boaventura MF, Hatanaka E, Massao-Hirabara S, Gorjão R. Differential effects of palmitoleic acid on human lymphocyte proliferation and function. Lipids Health Dis 2016; 15:217. [PMID: 27964715 PMCID: PMC5154135 DOI: 10.1186/s12944-016-0385-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/02/2016] [Indexed: 02/13/2023] Open
Abstract
BACKGROUND Palmitoleic acid (PA) is a n-7 monounsaturated fatty acid (MUFA) secreted by adipose tissue and related to decreased insulin resistance in peripheral tissues. Evidences have been shown that PA also decreased proinflammatory cytokine expression in cultured macrophages. Although studies have shown that other fatty acids (FAs) modulate several lymphocyte functions, the specific effect of PA on these cells is unknown. The aim of the present study was to evaluate the possible influence of PA on activation and differentiation of human lymphocytes in comparison to oleic acid (OA). METHODS Human lymphocytes were isolated from peripheral blood of health men and cultured in the presence of growing concentrations of PA or OA (5 to 200 μM), for 24 h. After that, cells were collected and cytotoxicity evaluated by flow cytometry. Then, we analyzed proliferative capacity in lymphocytes treated with non toxic concentrations of PA and OA (25 and 50 μM, respectively), in the presence or absence of concanavalin A (ConA). The Th1/Th2/Th17 cytokine production was determined by the Cytometric Bead Array. CD28 and CD95 surface expression and T regulatory cell percentage were determined by flow cytometry. RESULTS We observed that PA is toxic to lymphocytes above 50 μM. PA promoted a decrease of lymphocyte proliferation stimulated by ConA in both concentrations. PA also decreased CD28 externalization and increased CD95. On the other hand, OA did not alter these parameters. In the same way, PA reduced IL6, IFN-gamma, TNF-alpha and IL17A production in both concentration and IL2 only at 50 μM (in the presence of ConA). OA promoted IFN-gamma reduction in both concentrations and an increase of IL-2, IL4 and IL10 at 25 μM. Both fatty acids decreased the percentage of T regulatory cells. CONCLUSION In conclusion, PA promoted a suppressive effect on lymphocyte proliferation characterized by a decrease of Th1 and Th17 response, and co-stimulatory molecule (CD28). However, OA increased lymphocyte proliferation through IL2 production and Th2 response. These results also show a more suppressive effect of PA on lymphocytes in comparison to OA.
Collapse
Affiliation(s)
- M E P Passos
- Institute of Physical Activity and Sport Sciences, Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, Rua Galvão Bueno, 868, Liberdade, CEP: 01506 000, São Paulo, SP, Brazil.
| | - H H O Alves
- Institute of Physical Activity and Sport Sciences, Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, Rua Galvão Bueno, 868, Liberdade, CEP: 01506 000, São Paulo, SP, Brazil
| | - C M Momesso
- Institute of Physical Activity and Sport Sciences, Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, Rua Galvão Bueno, 868, Liberdade, CEP: 01506 000, São Paulo, SP, Brazil
| | - F G Faria
- Institute of Physical Activity and Sport Sciences, Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, Rua Galvão Bueno, 868, Liberdade, CEP: 01506 000, São Paulo, SP, Brazil
| | - G Murata
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - M F Cury-Boaventura
- Institute of Physical Activity and Sport Sciences, Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, Rua Galvão Bueno, 868, Liberdade, CEP: 01506 000, São Paulo, SP, Brazil
| | - E Hatanaka
- Institute of Physical Activity and Sport Sciences, Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, Rua Galvão Bueno, 868, Liberdade, CEP: 01506 000, São Paulo, SP, Brazil
| | - S Massao-Hirabara
- Institute of Physical Activity and Sport Sciences, Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, Rua Galvão Bueno, 868, Liberdade, CEP: 01506 000, São Paulo, SP, Brazil
| | - R Gorjão
- Institute of Physical Activity and Sport Sciences, Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, Rua Galvão Bueno, 868, Liberdade, CEP: 01506 000, São Paulo, SP, Brazil.
| |
Collapse
|
46
|
Stearoyl-CoA desaturase 1 deficiency reduces lipid accumulation in the heart by activating lipolysis independently of peroxisome proliferator-activated receptor α. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:2029-2037. [PMID: 27751891 DOI: 10.1016/j.bbalip.2016.10.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 09/19/2016] [Accepted: 10/13/2016] [Indexed: 12/26/2022]
Abstract
Stearoyl-CoA desaturase 1 (SCD1) has recently been shown to be a critical control point in the regulation of cardiac metabolism and function. Peroxisome proliferator-activated receptor α (PPARα) is an important regulator of myocardial fatty acid uptake and utilization. The present study used SCD1 and PPARα double knockout (SCD1-/-/PPARα-/-) mice to test the hypothesis that PPARα is involved in metabolic changes in the heart that are caused by SCD1 downregulation/inhibition. SCD1 deficiency decreased the intracellular content of free fatty acids, triglycerides, and ceramide in the heart of SCD1-/- and SCD1-/-/PPARα-/- mice. SCD1 ablation in PPARα-/- mice decreased diacylglycerol content in cardiomyocytes. These results indicate that the reduction of fat accumulation in the heart associated with SCD1 deficiency occurs independently of the PPARα pathway. To elucidate the mechanism of the observed changes, we treated HL-1 cardiomyocytes with the SCD1 inhibitor A939572 and/or PPARα inhibitor GW6471. SCD1 inhibition decreased the level of lipogenic proteins and increased lipolysis, reflected by a decrease in the content of adipose triglyceride lipase inhibitor G0S2 and a decrease in the ratio of phosphorylated hormone-sensitive lipase (HSL) at Ser565 to HSL (pHSL[Ser565]/HSL). PPARα inhibition alone did not affect the aforementioned protein levels. Finally, PPARα inhibition decreased the phosphorylation level of 5'-adenosine monophosphate-activated protein kinase, indicating lower mitochondrial fatty acid oxidation. In summary, SCD1 ablation/inhibition decreased cardiac lipid content independently of the action of PPARα by reducing lipogenesis and activating lipolysis. The present data suggest that SCD1 is an important component in maintaining proper cardiac lipid metabolism.
Collapse
|
47
|
de Sá RDCDC, Crisma AR, Cruz MM, Martins AR, Masi LN, do Amaral CL, Curi R, Alonso-Vale MIC. Fish oil prevents changes induced by a high-fat diet on metabolism and adipokine secretion in mice subcutaneous and visceral adipocytes. J Physiol 2016; 594:6301-6317. [PMID: 27558442 DOI: 10.1113/jp272541] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 06/22/2016] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS Fish oil (FO), rich in omega-3 polyunsaturated fatty acids, has beneficial effects on changes induced by obesity and partially prevents associated comorbidities. The effects of FO on adipocytes from different adipose tissue depots in high-fat (HF) diet induced obese mice have not been uninvestigated. This is the first study to examine the effects of FO on changes in metabolism and adipokine production in adipocytes from s.c. (inguinal; ING) or visceral (retroperitoneal; RP) white adipose depots in a HF diet-induced obese mice. Unlike most studies performed previously, FO supplementation was initiated 4 weeks before the induction of obesity. HF diet caused marked changes in ING (glucose uptake and secretion of adiponectin, tumour necrosis factor-α and interleukin-6 in ING) and RP (lipolysis, de novo lipogenesis and secretion of pro-inflammatory cytokines) adipose depots. Previous and concomitant FO administration prevented the changes in ING and RP adipocytes induced by the HF diet. ABSTRACT In the present study, we investigated the effect of fish oil (FO) on metabolism and adipokine production by adipocytes from s.c. (inguinal; ING) and visceral (retroperitoneal; RP) white adipose depots in high-fat (HF) diet-induced obese mice. Mice were divided into CO (control diet), CO+FO, HF and HF+FO groups. The HF group presented higher body weight, glucose intolerance, insulin resistance, higher plasma total and low-density lipoprotein cholesterol levels, and greater weights of ING and RP adipose depots accompanied by hypertrophy of the adipocytes. FO exerted anti-obesogenic effects associated with beneficial effects on dyslipidaemia and insulin resistance in mice fed a HF diet (HF+FO group). HF raised RP adipocyte lipolysis and the production of pro-inflammatory cytokines and reduced de novo synthesis of fatty acids, whereas, in ING adipocytes, it decreased glucose uptake and adiponectin secretion but did not change lipolysis. Therefore, the adipose depots play different roles in HF diet-induced insulin resistance according to their location in the body. Concerning cytokine secretion, adipocytes per se in addition to white adopise tissue infiltrated leukocytes have to be considered in the aetiology of the comorbidities associated with obesity. Evidence is presented showing that previous and concomitant administration of FO can prevent changes in metabolism and the secretion of hormones and cytokines in ING and RP adipocytes induced by HF.
Collapse
Affiliation(s)
- Roberta D C da Cunha de Sá
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo, Diadema, Brazil
| | - Amanda R Crisma
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Maysa M Cruz
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo, Diadema, Brazil
| | - Amanda R Martins
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Laureane N Masi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Catia L do Amaral
- Campus of Exact Science and Technology, State University of Goias, Anapolis, Brazil
| | - R Curi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Maria I C Alonso-Vale
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo, Diadema, Brazil.
| |
Collapse
|
48
|
Kawabata K, Karahashi M, Sakamoto T, Tsuji Y, Yamazaki T, Okazaki M, Mitsumoto A, Kudo N, Kawashima Y. Fatty Acid β-Oxidation Plays a Key Role in Regulating cis-Palmitoleic Acid Levels in the Liver. Biol Pharm Bull 2016; 39:1995-2008. [DOI: 10.1248/bpb.b16-00470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | | | | | - Yukiho Tsuji
- School of Pharmaceutical Sciences, Josai University
| | | | - Mari Okazaki
- School of Pharmaceutical Sciences, Josai University
| | | | - Naomi Kudo
- School of Pharmaceutical Sciences, Josai University
| | | |
Collapse
|
49
|
Morse N. Are some health benefits of palmitoleic acid supplementation due to its effects on 5′ adenosine monophosphate-activated protein kinase (AMPK)? ACTA ACUST UNITED AC 2015. [DOI: 10.1002/lite.201500061] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Nancy Morse
- 9 Horsburgh Dr. Berwick Nova Scotia Canada B0P 1E0
| |
Collapse
|
50
|
Foryst-Ludwig A, Kreissl MC, Benz V, Brix S, Smeir E, Ban Z, Januszewicz E, Salatzki J, Grune J, Schwanstecher AK, Blumrich A, Schirbel A, Klopfleisch R, Rothe M, Blume K, Halle M, Wolfarth B, Kershaw EE, Kintscher U. Adipose Tissue Lipolysis Promotes Exercise-induced Cardiac Hypertrophy Involving the Lipokine C16:1n7-Palmitoleate. J Biol Chem 2015; 290:23603-15. [PMID: 26260790 DOI: 10.1074/jbc.m115.645341] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Indexed: 12/28/2022] Open
Abstract
Endurance exercise training induces substantial adaptive cardiac modifications such as left ventricular hypertrophy (LVH). Simultaneously to the development of LVH, adipose tissue (AT) lipolysis becomes elevated upon endurance training to cope with enhanced energy demands. In this study, we investigated the impact of adipose tissue lipolysis on the development of exercise-induced cardiac hypertrophy. Mice deficient for adipose triglyceride lipase (Atgl) in AT (atATGL-KO) were challenged with chronic treadmill running. Exercise-induced AT lipolytic activity was significantly reduced in atATGL-KO mice accompanied by the absence of a plasma fatty acid (FA) increase. These processes were directly associated with a prominent attenuation of myocardial FA uptake in atATGL-KO and a significant reduction of the cardiac hypertrophic response to exercise. FA serum profiling revealed palmitoleic acid (C16:1n7) as a new molecular co-mediator of exercise-induced cardiac hypertrophy by inducing nonproliferative cardiomyocyte growth. In parallel, serum FA analysis and echocardiography were performed in 25 endurance athletes. In consonance, the serum C16:1n7 palmitoleate level exhibited a significantly positive correlation with diastolic interventricular septum thickness in those athletes. No correlation existed between linoleic acid (18:2n6) and diastolic interventricular septum thickness. Collectively, our data provide the first evidence that adipose tissue lipolysis directly promotes the development of exercise-induced cardiac hypertrophy involving the lipokine C16:1n7 palmitoleate as a molecular co-mediator. The identification of a lipokine involved in physiological cardiac growth may help to develop future lipid-based therapies for pathological LVH or heart failure.
Collapse
Affiliation(s)
- Anna Foryst-Ludwig
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany, the DZHK (German Center for Cardiovascular Research), 10115 Berlin, Germany
| | - Michael C Kreissl
- the Department of Nuclear Medicine, University Hospital Wuerzburg, 97080 Wuerzburg, Germany
| | - Verena Benz
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Sarah Brix
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Elia Smeir
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Zsofia Ban
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Elżbieta Januszewicz
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Janek Salatzki
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Jana Grune
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Anne-Kathrin Schwanstecher
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Annelie Blumrich
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Andreas Schirbel
- the Department of Nuclear Medicine, University Hospital Wuerzburg, 97080 Wuerzburg, Germany
| | - Robert Klopfleisch
- the Department of Veterinary Pathology, College of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany
| | | | - Katharina Blume
- the Department of Prevention, Rehabilitation, and Sports Medicine, Technische Universitaet Muenchen, 80809 Muenchen, Germany
| | - Martin Halle
- the Department of Prevention, Rehabilitation, and Sports Medicine, Technische Universitaet Muenchen, 80809 Muenchen, Germany, the DZHK (German Center for Cardiovascular Research), Munich Heart Alliance, 80809 Munich, Germany
| | - Bernd Wolfarth
- the Department of Sports Medicine, Humboldt University/Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Erin E Kershaw
- the Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, and
| | - Ulrich Kintscher
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany, the DZHK (German Center for Cardiovascular Research), 10115 Berlin, Germany
| |
Collapse
|