1
|
Landau M, Elsabbagh S, Gross H, Fuchs ACD, Schultz ACF, Schultz JE. The membrane domains of mammalian adenylyl cyclases are lipid receptors. eLife 2024; 13:RP101483. [PMID: 39611663 PMCID: PMC11606603 DOI: 10.7554/elife.101483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024] Open
Abstract
The biosynthesis of cyclic 3',5'-adenosine monophosphate (cAMP) by mammalian membrane-bound adenylyl cyclases (mACs) is predominantly regulated by G-protein-coupled receptors (GPCRs). Up to now the two hexahelical transmembrane domains of mACs were considered to fix the enzyme to membranes. Here, we show that the transmembrane domains serve in addition as signal receptors and transmitters of lipid signals that control Gsα-stimulated mAC activities. We identify aliphatic fatty acids and anandamide as receptor ligands of mAC isoforms 1-7 and 9. The ligands enhance (mAC isoforms 2, 3, 7, and 9) or attenuate (isoforms 1, 4, 5, and 6) Gsα-stimulated mAC activities in vitro and in vivo. Substitution of the stimulatory membrane receptor of mAC3 by the inhibitory receptor of mAC5 results in a ligand inhibited mAC5-mAC3 chimera. Thus, we discovered a new class of membrane receptors in which two signaling modalities are at a crossing, direct tonic lipid and indirect phasic GPCR-Gsα signaling regulating the biosynthesis of cAMP.
Collapse
Affiliation(s)
- Marius Landau
- Pharmazeutisches Institut der Universität TübingenTübingenGermany
| | - Sherif Elsabbagh
- Pharmazeutisches Institut der Universität TübingenTübingenGermany
| | - Harald Gross
- Pharmazeutisches Institut der Universität TübingenTübingenGermany
| | | | - Anita CF Schultz
- Pharmazeutisches Institut der Universität TübingenTübingenGermany
| | | |
Collapse
|
2
|
Xia J, Yin S, Yu J, Wang J, Jin X, Wang Y, Liu H, Sun G. Improvement in Glycolipid Metabolism Parameters After Supplementing Fish Oil-Derived Omega-3 Fatty Acids Is Associated with Gut Microbiota and Lipid Metabolites in Type 2 Diabetes Mellitus. Nutrients 2024; 16:3755. [PMID: 39519588 PMCID: PMC11547733 DOI: 10.3390/nu16213755] [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: 10/04/2024] [Revised: 10/24/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES This study aimed to investigate the effects of fish oil-derived omega-3 polyunsaturated fatty acids (omega-3 PUFAs) on gut microbiota and serum lipid metabolites in T2DM. METHODS In a three-month, randomized, double-blind, placebo-controlled study, 110 T2DM patients received either fish oil (n = 55) or corn oil (n = 55) capsules daily. Serum lipids, glycemic parameters, gut microbiota diversity, and lipidomics were assessed. RESULTS This study found that fish oil-derived omega-3 PUFAs intervention did not significantly lower the fasting plasma glucose levels when compared with the baseline level (p > 0.05). However, serum fasting blood glucose (p = 0.039), glycosylated hemoglobin levels (p = 0.048), HOMA-IR (p = 0.022), total cholesterol (p < 0.001), triglyceride (p = 0.034), LDL cholesterol (p = 0.048), and non-HDL levels (p = 0.046) were significantly lower in the fish oil group compared with the corn oil group after three months of intervention. Also, it altered glycerophospholipid metabolism and gut microbiota. After three months, the fish oil group showed a significantly lower abundance of Desulfobacterota compared with the corn oil control group (p = 0.003), with reduced levels of Colidextribacter (p = 0.002), Ralstonia (p = 0.021), and Klebsiella (p = 0.013). Conversely, the abundance of Limosilactobacillus (p = 0.017), Lactobacillus (p = 0.011), and Haemophilus (p = 0.018) increased significantly. In addition, relevant glycolipid metabolism indicators showed significant correlations with the altered profiles of serum lipid metabolites, intestinal bacteria, and fungi. CONCLUSIONS This study highlights the impact of fish oil-derived omega-3 PUFAs on intestinal microbiota structure and function in patients with type 2 diabetes. The observed decrease in pathogenic bacterial species and the enhancement of beneficial species may have significant implications for gut health and systemic inflammation, both of which are pivotal in managing diabetes. Further research is warranted to comprehensively elucidate the long-term benefits and underlying mechanisms of these microbiota alterations.
Collapse
Affiliation(s)
- Jiayue Xia
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Shiyu Yin
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Junhui Yu
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Jiongnan Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Xingyi Jin
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Yuanyuan Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Hechun Liu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Guiju Sun
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| |
Collapse
|
3
|
Katare PB, Dalmao-Fernandez A, Mengeste AM, Navabakbar F, Hamarsland H, Ellefsen S, Berge RK, Bakke HG, Nyman TA, Kase ET, Rustan AC, Thoresen GH. Krill oil supplementation in vivo promotes increased fuel metabolism and protein synthesis in cultured human skeletal muscle cells. Front Nutr 2024; 11:1452768. [PMID: 39555189 PMCID: PMC11565515 DOI: 10.3389/fnut.2024.1452768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 10/07/2024] [Indexed: 11/19/2024] Open
Abstract
Introduction Krill oil is a dietary supplement derived from Antarctic krill; a small crustacean found in the ocean. Krill oil is a rich source of omega-3 fatty acids, specifically eicosapentaenoic acid and docosahexaenoic acid, as well as the antioxidant astaxanthin. The aim of this study was to investigate the effects of krill oil supplementation, compared to placebo oil (high oleic sunflower oil added astaxanthin), in vivo on energy metabolism and substrate turnover in human skeletal muscle cells. Methods Skeletal muscle cells (myotubes) were obtained before and after a 7-week krill oil or placebo oil intervention, and glucose and oleic acid metabolism and leucine accumulation, as well as effects of different stimuli in vitro, were studied in the myotubes. The functional data were combined with proteomic and transcriptomic analyses. Results In vivo intervention with krill oil increased oleic acid oxidation and leucine accumulation in skeletal muscle cells, however no effects were observed on glucose metabolism. The krill oil-intervention-induced increase in oleic acid oxidation correlated negatively with changes in serum low-density lipoprotein (LDL) concentration. In addition, myotubes were also exposed to krill oil in vitro. The in vitro study revealed that 24 h of krill oil treatment increased both glucose and oleic acid metabolism in myotubes, enhancing energy substrate utilization. Transcriptomic analysis comparing myotubes obtained before and after krill oil supplementation identified differentially expressed genes associated with e.g., glycolysis/gluconeogenesis, metabolic pathways and calcium signaling pathway, while proteomic analysis demonstrated upregulation of e.g., LDL-receptor in myotubes obtained after the krill oil intervention. Conclusion These findings suggest that krill oil intervention promotes increased fuel metabolism and protein synthesis in human skeletal muscle cells, with potential implications for metabolic health.
Collapse
Affiliation(s)
- Parmeshwar B. Katare
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Andrea Dalmao-Fernandez
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Abel M. Mengeste
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Farnaz Navabakbar
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Håvard Hamarsland
- Section for Health and Exercise Physiology, Faculty of Social and Health Sciences, Inland Norway University of Applied Sciences, Lillehammer, Norway
| | - Stian Ellefsen
- Section for Health and Exercise Physiology, Faculty of Social and Health Sciences, Inland Norway University of Applied Sciences, Lillehammer, Norway
- Innlandet Hospital Trust, Lillehammer, Norway
| | - Rolf K. Berge
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Sciences, University of Bergen, Bergen, Norway
| | - Hege G. Bakke
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Tuula Anneli Nyman
- Department of Immunology, Institute of Clinical Medicine, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Eili Tranheim Kase
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Arild C. Rustan
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - G. Hege Thoresen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
4
|
Akram MZ, Sureda EA, Corion M, Comer L, Everaert N. Linking gastrointestinal tract structure, function, and gene expression signatures to growth variability in broilers: a novel interpretation for flock uniformity. Poult Sci 2024; 103:104158. [PMID: 39173569 PMCID: PMC11387703 DOI: 10.1016/j.psj.2024.104158] [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: 06/09/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 08/24/2024] Open
Abstract
Variation in body weight (BW) within broiler flocks is a significant challenge in poultry production. Investigating differences in gut-related parameters between low (LBW) and high BW (HBW) chicks may provide insights into the underlying causes of BW heterogeneity. 908 day-old male broiler chicks were reared until d 7 and then ranked into LBW and HBW groups. Thereafter, performance parameters were compared between BW groups periodically. On d 7, 14, and 38, visceral organ characteristics, intestinal permeability, and duodenal and ileal histomorphology were examined. Expression profiles were analyzed for 79 ileal genes related to gut barrier function, immune function, nutrient transport, gut hormones, nutrient receptors, metabolism, and oxidation using high-throughput qPCR. Student's t-tests were performed to compare measurements. Multivariate statistics, including partial least square regression (PLSR) analysis, were applied to identify combinations of key genes discriminating BW groups, offering predictive capability for phenotypic variations. The HBW group remained heavier at each timepoint, which could be explained by higher feed intake. The HBW group had shorter relative small intestine length but higher villus height and villi height/crypt depth ratios. The LBW group demonstrated increased intestinal permeability on d 38. The LBW group showed upregulation of immune response genes including TNF-α on d 7 and CYP450 on d 38, while the HBW group showed higher AHSA1 and HSPA4 expressions on d 7. The LBW group had upregulation of the metabolism genes mTOR and EIF4EBP1 on d 7 and the satiety-induced hormone cholecystokinin on d 14, while the HBW group tended to increase expression of the hunger hormone ghrelin on d 38. Genes related to gut barrier function, nutrient transport, and oxidation categories were consistently upregulated in the HBW group. PLSR models revealed 4, 12, and 11 sets of key genes highly predictive of BW phenotypes on d 7, 14, and 38, respectively. These findings suggest that growth rates are linked to the intestinal size, structure, and function of broiler chickens, offering insights into the underlying mechanisms regulating BW.
Collapse
Affiliation(s)
- Muhammad Zeeshan Akram
- Nutrition and Animal-Microbiota Ecosystems Laboratory, Department of Biosystems, KU Leuven, 3000-Heverlee, Belgium; Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Ester Arévalo Sureda
- Nutrition and Animal-Microbiota Ecosystems Laboratory, Department of Biosystems, KU Leuven, 3000-Heverlee, Belgium
| | - Matthias Corion
- Nutrition and Animal-Microbiota Ecosystems Laboratory, Department of Biosystems, KU Leuven, 3000-Heverlee, Belgium
| | - Luke Comer
- Nutrition and Animal-Microbiota Ecosystems Laboratory, Department of Biosystems, KU Leuven, 3000-Heverlee, Belgium
| | - Nadia Everaert
- Nutrition and Animal-Microbiota Ecosystems Laboratory, Department of Biosystems, KU Leuven, 3000-Heverlee, Belgium.
| |
Collapse
|
5
|
Cui Y, Auclair H, He R, Zhang Q. GPCR-mediated regulation of beige adipocyte formation: Implications for obesity and metabolic health. Gene 2024; 915:148421. [PMID: 38561165 DOI: 10.1016/j.gene.2024.148421] [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: 01/11/2024] [Revised: 03/10/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Obesity and its associated complications pose a significant burden on health. The non-shivering thermogenesis (NST) and metabolic capacity properties of brown adipose tissue (BAT), which are distinct from those of white adipose tissue (WAT), in combating obesity and its related metabolic diseases has been well documented. However, beige adipose tissue, the third and relatively novel type of adipose tissue, which emerges in extensive presence of WAT and shares similar favorable metabolic properties with BAT, has garnered considerable attention in recent years. In this review, we focused on the role of G protein-coupled receptors (GPCRs), the largest receptor family and the most successful class of drug targets in humans, in the induction of beige adipocytes. More importantly, we highlight researchers' clinical treatment attempts to ameliorate obesity and other related metabolic diseases through the formation and activation of beige adipose tissue. In summary, this review provides valuable insights into the formation of beige adipose tissue and the involvement of GPCRs, based on the latest advancements in scientific research.
Collapse
Affiliation(s)
- Yuanxu Cui
- Animal Zoology Department, Kunming Medical University, Kunming, China; Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, China
| | - Hugo Auclair
- Faculty of Medicine, François-Rabelais University, Tours, France
| | - Rong He
- Animal Zoology Department, Kunming Medical University, Kunming, China
| | - Qiang Zhang
- Animal Zoology Department, Kunming Medical University, Kunming, China.
| |
Collapse
|
6
|
Wargent ET, Kępczyńska MA, Kaspersen MH, Ulven ER, Arch JRS, Ulven T, Stocker CJ. Chronic administration of hydrolysed pine nut oil to mice improves insulin sensitivity and glucose tolerance and increases energy expenditure via a free fatty acid receptor 4-dependent mechanism. Br J Nutr 2024; 132:13-20. [PMID: 38751244 DOI: 10.1017/s0007114524000965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2024]
Abstract
A healthy diet is at the forefront of measures to prevent type 2 diabetes. Certain vegetable and fish oils, such as pine nut oil (PNO), have been demonstrated to ameliorate the adverse metabolic effects of a high-fat diet. The present study investigates the involvement of the free fatty acid receptors 1 (FFAR1) and 4 (FFAR4) in the chronic activity of hydrolysed PNO (hPNO) on high-fat diet-induced obesity and insulin resistance. Male C57BL/6J wild-type, FFAR1 knockout (-/-) and FFAR4-/- mice were placed on 60 % high-fat diet for 3 months. Mice were then dosed hPNO for 24 d, during which time body composition, energy intake and expenditure, glucose tolerance and fasting plasma insulin, leptin and adiponectin were measured. hPNO improved glucose tolerance and decreased plasma insulin in the wild-type and FFAR1-/- mice, but not the FFAR4-/- mice. hPNO also decreased high-fat diet-induced body weight gain and fat mass, whilst increasing energy expenditure and plasma adiponectin. None of these effects on energy balance were statistically significant in FFAR4-/- mice, but it was not shown that they were significantly less than in wild-type mice. In conclusion, chronic hPNO supplementation reduces the metabolically detrimental effects of high-fat diet on obesity and insulin resistance in a manner that is dependent on the presence of FFAR4.
Collapse
Affiliation(s)
- Edward Taynton Wargent
- Institute of Translational Medicine, Clore Laboratory, University of Buckingham, Buckingham, MK18 1EG, UK
| | - Małgorzata A Kępczyńska
- Institute of Translational Medicine, Clore Laboratory, University of Buckingham, Buckingham, MK18 1EG, UK
| | - Mads H Kaspersen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, 5230 Odense, Denmark
| | - Elisabeth Rexen Ulven
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100Copenhagen, Denmark
| | - Jonathan R S Arch
- Institute of Translational Medicine, Clore Laboratory, University of Buckingham, Buckingham, MK18 1EG, UK
| | - Trond Ulven
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, 5230 Odense, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100Copenhagen, Denmark
| | | |
Collapse
|
7
|
Mantzourani C, Mesimeri ID, Kokotou MG. Free Fatty Acid Determination in Broccoli Tissues Using Liquid Chromatography-High-Resolution Mass Spectrometry. Molecules 2024; 29:754. [PMID: 38398506 PMCID: PMC10891939 DOI: 10.3390/molecules29040754] [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: 12/23/2023] [Revised: 01/23/2024] [Accepted: 02/04/2024] [Indexed: 02/25/2024] Open
Abstract
Broccoli (Brassica oleracea L. var. italica Plenck) is a widely consumed vegetable, very popular due to its various nutritional and bioactive components. Since studies on the lipid components of broccoli have been limited so far, the aim of the present work was the study of free fatty acids (FFAs) present in different broccoli parts, aerial and underground. The direct determination of twenty-four FFAs in broccoli tissues (roots, leaves, and florets) was carried out, using a liquid chromatography-high-resolution mass spectrometry (LC-HRMS) method in a 10 min single run. Linolenic acid was found to be the most abundant FFA in all different broccoli parts in quantities ranging from 0.76 to 1.46 mg/g, followed by palmitic acid (0.17-0.22 mg/g) and linoleic acid (0.06-0.08 mg/g). To extend our knowledge on broccoli's bioactive components, for the first time, the existence of bioactive oxidized fatty acids, namely hydroxy and oxo fatty acids, was explored in broccoli tissues adopting an HRMS-based lipidomics approach. 16- and 2-hydroxypalmitic acids were detected in all parts of broccoli studied, while ricinoleic acid was detected for the first time as a component of broccoli.
Collapse
Affiliation(s)
- Christiana Mantzourani
- Laboratory of Chemistry, Department of Food Science and Human Nutrition, Agricultural University of Athens, 11855 Athens, Greece
| | - Irene-Dimitra Mesimeri
- Laboratory of Chemistry, Department of Food Science and Human Nutrition, Agricultural University of Athens, 11855 Athens, Greece
| | - Maroula G Kokotou
- Laboratory of Chemistry, Department of Food Science and Human Nutrition, Agricultural University of Athens, 11855 Athens, Greece
| |
Collapse
|
8
|
Durand G, Charrier P, Bes S, Bernard L, Lamothe V, Gruffat D, Bonnet M. Gene expression of free fatty acids-sensing G protein-coupled receptors in beef cattle. J Anim Sci 2024; 102:skae114. [PMID: 38659415 DOI: 10.1093/jas/skae114] [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: 01/23/2024] [Accepted: 04/24/2024] [Indexed: 04/26/2024] Open
Abstract
Many physiological functions are regulated by free fatty acids (FFA). Recently, the discovery of FFA-specific G protein-coupled receptors (FFARs) has added to the complexity of their actions at the cellular level. The study of FFAR in cattle is still in its earliest stages focusing mainly on dairy cows. In this study, we set out to map the expression of genes encoding FFARs in 6 tissues of beef cattle. We also investigated the potential effect of dietary forage nature on FFAR gene expression. To this end, 16 purebred Charolais bulls were fed a grass silage ration or a maize silage ration (n = 8/group) with a forage/concentrate ratio close to 60:40 for 196 d. The animals were then slaughtered at 485 ± 42 d and liver, spleen, ileum, rectum, perirenal adipose tissue (PRAT), and Longissimus Thoracis muscle were collected. FFAR gene expression was determined by real-time quantitative PCR. Our results showed that of the five FFARs investigated, FFAR1, FFAR2, FFAR3, and GPR84 are expressed (Ct < 30) in all six tissues, whereas FFAR4 was only expressed (Ct < 30) in PRAT, ileum, and rectum. In addition, our results showed that the nature of the forage, i.e., grass silage or maize silage, had no effect on the relative abundance of FFAR in any of the tissues studied (P value > 0.05). Taken together, these results open new perspectives for studying the physiological role of these receptors in beef cattle, particularly in nutrient partitioning during growth.
Collapse
Affiliation(s)
- Guillaume Durand
- INRAE, Université Clermont Auvergne, VetagroSup, UMRH, 63122 Saint-Genès-Champanelle, France
- Bordeaux Sciences Agro, 33170 Gradignan, France
| | | | - Sébastien Bes
- INRAE, Université Clermont Auvergne, VetagroSup, UMRH, 63122 Saint-Genès-Champanelle, France
| | - Laurence Bernard
- INRAE, Université Clermont Auvergne, VetagroSup, UMRH, 63122 Saint-Genès-Champanelle, France
| | | | - Dominique Gruffat
- INRAE, Université Clermont Auvergne, VetagroSup, UMRH, 63122 Saint-Genès-Champanelle, France
| | - Muriel Bonnet
- INRAE, Université Clermont Auvergne, VetagroSup, UMRH, 63122 Saint-Genès-Champanelle, France
| |
Collapse
|
9
|
Moonwiriyakit A, Yimnual C, Noitem R, Dinsuwannakol S, Sontikun J, Kaewin S, Worakajit N, Soontornniyomkij V, Muanprasat C. GPR120/FFAR4 stimulation attenuates airway remodeling and suppresses IL-4- and IL-13-induced airway epithelial injury via inhibition of STAT6 and Akt. Biomed Pharmacother 2023; 168:115774. [PMID: 37924784 DOI: 10.1016/j.biopha.2023.115774] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/12/2023] [Accepted: 10/20/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Airway remodeling is associated with severity and treatment insensitivity in asthma. This study aimed to investigate the effects of G protein-coupled receptor 120 (GPR120) stimulation on alleviating allergic inflammation and remodeling of airway epithelium. RESEARCH DESIGN AND METHODS Ovalbumin (OVA)-challenged BALB/c mice and type-2-cytokine (IL-4 and IL-13)-exposed 16HBE human bronchial epithelial cells were treated with GSK137647A, a selective GPR120 agonist. Markers of allergic inflammation and airway remodeling were determined. RESULTS GSK137647A attenuated inflammation and mucus secretion in airway epithelium of OVA-challenged mice. Stimulation of GPR120 in 16HBE suppressed expression of asthma-associated cytokines and cytokine-induced expression of pathogenic mucin-MUC5AC. These effects were abolished by co-treatment with AH7614, a GPR120 antagonist. Moreover, GPR120 stimulation in 16HBE cells reduced expression of fibrotic markers including fibronectin protein and ACTA2 mRNA and inhibited epithelial barrier leakage induced by type-2 inflammation via rescuing expression of zonula occludens-1 protein. Furthermore, GPR120 stimulation prevented the cytokine-induced airway epithelial remodeling via suppression of STAT6 and Akt phosphorylation. CONCLUSIONS Our findings suggest that GPR120 activation alleviates allergic inflammation and remodeling of airway epithelium partly through inhibition of STAT6 and Akt. GPR120 may represent a novel therapeutic target for diseases associated with remodeling of airway epithelium, including asthma.
Collapse
Affiliation(s)
- Aekkacha Moonwiriyakit
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Chantapol Yimnual
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Rattikarn Noitem
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand; Translational Medicine Graduate Program, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Sasiwimol Dinsuwannakol
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Jenjira Sontikun
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Suchada Kaewin
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand; Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Nichakorn Worakajit
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand; Translational Medicine Graduate Program, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Virawudh Soontornniyomkij
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Chatchai Muanprasat
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand.
| |
Collapse
|
10
|
Zhao X, Ahn D, Nam G, Kwon J, Song S, Kang MJ, Ahn H, Chung SJ. Identification of Crocetin as a Dual Agonist of GPR40 and GPR120 Responsible for the Antidiabetic Effect of Saffron. Nutrients 2023; 15:4774. [PMID: 38004168 PMCID: PMC10675071 DOI: 10.3390/nu15224774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 11/04/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Crocin, a glycoside of crocetin, has been known as the principal component responsible for saffron's antidiabetic, anticancer, and anti-inflammatory effects. Crocetin, originating from the hydrolytic cleavage of crocin in biological systems, was subjected to ligand-based virtual screening in this investigation. Subsequent biochemical analysis unveiled crocetin, not crocin, as a novel dual GPR40 and GPR120 agonist, demonstrating a marked preference for GPR40 and GPR120 over peroxisome proliferator-activated receptors (PPAR)γ. This compound notably enhanced insulin and GLP-1 secretion from pancreatic β-cells and intestinal neuroendocrine cells, respectively, presenting a dual mechanism of action in glucose-lowering effects. Docking simulations showed that crocetin emulates the binding characteristics of natural ligands through hydrogen bonds and hydrophobic interactions, whereas crocin's hindered fit within the binding pocket is attributed to steric constraints. Collectively, for the first time, this study unveils crocetin as the true active component of saffron, functioning as a GPR40/120 agonist with potential implications in antidiabetic interventions.
Collapse
Affiliation(s)
- Xiaodi Zhao
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; (X.Z.); (G.N.); (J.K.); (S.S.); (M.J.K.); (H.A.)
| | - Dohee Ahn
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea;
| | - Gibeom Nam
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; (X.Z.); (G.N.); (J.K.); (S.S.); (M.J.K.); (H.A.)
| | - Jihee Kwon
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; (X.Z.); (G.N.); (J.K.); (S.S.); (M.J.K.); (H.A.)
| | - Songyi Song
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; (X.Z.); (G.N.); (J.K.); (S.S.); (M.J.K.); (H.A.)
| | - Min Ji Kang
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; (X.Z.); (G.N.); (J.K.); (S.S.); (M.J.K.); (H.A.)
| | - Hyejin Ahn
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; (X.Z.); (G.N.); (J.K.); (S.S.); (M.J.K.); (H.A.)
| | - Sang J. Chung
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; (X.Z.); (G.N.); (J.K.); (S.S.); (M.J.K.); (H.A.)
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea;
| |
Collapse
|
11
|
Wang Y, Liu H, Zhang Z. Recent Advance in Regulatory Effect of GRP120 on Bone Metabolism. Aging Dis 2023; 14:1714-1727. [PMID: 37196107 PMCID: PMC10529742 DOI: 10.14336/ad.2023.0216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/16/2023] [Indexed: 05/19/2023] Open
Abstract
The link between fatty acids and bone metabolism is complex and can be direct and indirect. This link has been reported in different types of bone cells and various stages of bone metabolism. G-protein coupled receptor 120 (GPR120), also called free fatty acid receptor 4 (FFAR4), is a member of the recently discovered G protein-coupled receptor family that can interact with both long-chain saturated fatty acids (C14-C18) and long-chain unsaturated fatty acids (C16-C22). Research shows that GPR120 regulates processes in different types of bone cells, directly or indirectly affecting bone metabolism. Our research reviewed the literature on the effects of GPR120 on bone marrow mesenchymal stem cells (BMMSCs), osteoblasts, osteoclasts, and chondrocytes, focusing on the research findings regarding the mechanism by which GPR120 alters specific bone metabolic diseases-osteoporosis and osteoarthritis. The data reviewed here provide a basis for clinical and basic research into the role of GPR120 on bone metabolic diseases.
Collapse
Affiliation(s)
| | - Haixia Liu
- Institute of Basic Theory for Chinese Medicine, Chinese Academy of Chinese Medical Sciences, Beijing, China.
| | - Zhiguo Zhang
- Institute of Basic Theory for Chinese Medicine, Chinese Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
12
|
Zhang S, Roth BL. Sensing unsaturated fatty acids: insights from GPR120 signaling. Cell Res 2023; 33:657-658. [PMID: 37142674 PMCID: PMC10474016 DOI: 10.1038/s41422-023-00814-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023] Open
Affiliation(s)
- Shicheng Zhang
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bryan L Roth
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
13
|
Primeaux SD, Dubin R, Greenway FL. Orlistat mouth rinse: Using the tongue to deliver antiobesity medication in a double-blind randomized crossover pilot trial. Diabetes Obes Metab 2023; 25:2236-2242. [PMID: 37132340 PMCID: PMC10330229 DOI: 10.1111/dom.15101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/11/2023] [Accepted: 04/15/2023] [Indexed: 05/04/2023]
Abstract
AIM To investigate the effects of an orlistat mouth rinse on the intake of a high-fat meal. METHODS A double-blind, balanced order, crossover study was conducted in participants (n = 10, body mass index 25-30 kg/m2 ) assigned to receive placebo or orlistat (24 mg/mL) prior to a high-fat meal. Participants were divided into low- or high-fat consumers based on calories consumed from fat following placebo administration. RESULTS The orlistat mouth rinse decreased total and fat calories consumed during the high-fat meal in high-fat consumers, and did not alter calories consumed in low-fat consumers (P < 0.05). CONCLUSIONS Orlistat decreases long-chain fatty acid (LCFA) absorption by inhibiting lipases that breakdown triglycerides. Orlistat mouth rinse decreased fat intake in high-fat consumers, suggesting that orlistat inhibited the detection of LCFAs from the high-fat test meal. Lingual delivery of orlistat is predicted to eliminate the risk of oil incontinence and promote weight loss in individuals who prefer fat.
Collapse
Affiliation(s)
- Stefany D Primeaux
- Department of Physiology, LSU Health-NO, New Orleans, Louisiana, USA
- Joint Diabetes, Endocrinology & Metabolism Program, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Robert Dubin
- Florida State University College of Medicine, Venice, Florida, USA
| | - Frank L Greenway
- Clinical Trials Department, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| |
Collapse
|
14
|
Li Y, Lu Z, Kirkwood CL, Kirkwood KL, Wank SA, Li AJ, Lopes-Virella MF, Huang Y. GPR40 deficiency worsens metabolic syndrome-associated periodontitis in mice. J Periodontal Res 2023; 58:575-587. [PMID: 36807310 PMCID: PMC10182248 DOI: 10.1111/jre.13107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 01/13/2023] [Accepted: 01/30/2023] [Indexed: 02/20/2023]
Abstract
BACKGROUND AND OBJECTIVE G protein-coupled receptor 40 (GPR40) is a receptor for medium- and long-chain free fatty acids (FFAs). GPR40 activation improves type 2 diabetes mellitus (T2DM), metabolic syndrome (MetS), and the complications of T2DM and MetS. Periodontitis, a common oral inflammatory disease initiated by periodontal pathogens, is another complication of T2DM and MetS. Since FFAs play a key role in the pathogenesis of MetS which exacerbates periodontal inflammation and GPR40 is a FFA receptor with anti-inflammatory properties, it is important to define the role of GPR40 in MetS-associated periodontitis. MATERIALS AND METHODS We induced MetS and periodontitis by high-fat diet and periodontal injection of lipopolysaccharide (LPS), respectively, in wild-type and GPR40-deficient mice and determined alveolar bone loss and periodontal inflammation using micro-computed tomography, histology, and osteoclast staining. We also performed in vitro study to determine the role of GPR40 in the expression of proinflammatory genes. RESULTS The primary outcome of the study is that GPR40 deficiency increased alveolar bone loss and enhanced osteoclastogenesis in control mice and the mice with both MetS and periodontitis. GPR40 deficiency also augmented periodontal inflammation in control mice and the mice with both MetS and periodontitis. Furthermore, GPR40 deficiency led to increased plasma lipids and insulin resistance in control mice but had no effect on the metabolic parameters in mice with MetS alone. For mice with both MetS and periodontitis, GPR40 deficiency increased insulin resistance. Finally, in vitro studies with macrophages showed that deficiency or inhibition of GPR40 upregulated proinflammatory genes while activation of GPR40 downregulated proinflammatory gene expression stimulated synergistically by LPS and palmitic acid. CONCLUSION GPR40 deficiency worsens alveolar bone loss and periodontal inflammation in mice with both periodontitis and MetS, suggesting that GPR40 plays a favorable role in MetS-associated periodontitis. Furthermore, GPR40 deficiency or inhibition in macrophages further upregulated proinflammatory and pro-osteoclastogenic genes induced by LPS and palmitic acid, suggesting that GPR40 has anti-inflammatory and anti-osteoclastogenic properties.
Collapse
Affiliation(s)
- Yanchun Li
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of Medicine, College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Zhongyang Lu
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of Medicine, College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Cameron L. Kirkwood
- Departments of Oral Biology, School of Dental Medicine, University at Buffalo
| | - Keith L. Kirkwood
- Departments of Oral Biology, School of Dental Medicine, University at Buffalo
- Department of Head & Neck/Plastic & Reconstructive Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Stephen A. Wank
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - Ai-Jun Li
- Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Maria F. Lopes-Virella
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of Medicine, College of Medicine, Medical University of South Carolina, Charleston, South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Yan Huang
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of Medicine, College of Medicine, Medical University of South Carolina, Charleston, South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
15
|
Zu Y, Pahlavani M, Ramalingam L, Jayarathne S, Andrade J, Scoggin S, Festuccia WT, Kalupahana NS, Moustaid-Moussa N. Temperature-Dependent Effects of Eicosapentaenoic Acid (EPA) on Browning of Subcutaneous Adipose Tissue in UCP1 Knockout Male Mice. Int J Mol Sci 2023; 24:ijms24108708. [PMID: 37240054 DOI: 10.3390/ijms24108708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Uncoupling protein 1 (UCP1) plays a central role in thermogenic tissues by uncoupling cellular respiration to dissipate energy. Beige adipocytes, an inducible form of thermogenic cells in subcutaneous adipose tissue (SAT), have become a major focus in obesity research. We have previously shown that eicosapentaenoic acid (EPA) ameliorated high-fat diet (HFD)-induced obesity by activating brown fat in C57BL/6J (B6) mice at thermoneutrality (30 °C), independently of UCP1. Here, we investigated whether ambient temperature (22 °C) impacts EPA effects on SAT browning in wild-type (WT) and UCP1 knockout (KO) male mice and dissected underlying mechanisms using a cell model. We observed resistance to diet-induced obesity in UCP1 KO mice fed HFD at ambient temperature, with significantly higher expression of UCP1-independent thermogenic markers, compared to WT mice. These markers included the fibroblast growth factor 21 (FGF21) and sarco/endoplasmic reticulum Ca2+-ATPase 2b (SERCA2b), suggesting the indispensable role of temperature in beige fat reprogramming. Surprisingly, although EPA induced thermogenic effects in SAT-derived adipocytes harvested from both KO and WT mice, EPA only increased thermogenic gene and protein expression in the SAT of UCP1 KO mice housed at ambient temperature. Collectively, our findings indicate that the thermogenic effects of EPA, which are independent of UCP1, occur in a temperature-dependent manner.
Collapse
Affiliation(s)
- Yujiao Zu
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| | - Mandana Pahlavani
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| | - Shasika Jayarathne
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| | - Jose Andrade
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| | - Shane Scoggin
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| | - William T Festuccia
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Nishan S Kalupahana
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Department of Physiology, Faculty of Medicine, University of Peradeniya, Peradeniya 20400, Sri Lanka
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
16
|
Mao C, Xiao P, Tao XN, Qin J, He QT, Zhang C, Guo SC, Du YQ, Chen LN, Shen DD, Yang ZS, Zhang HQ, Huang SM, He YH, Cheng J, Zhong YN, Shang P, Chen J, Zhang DL, Wang QL, Liu MX, Li GY, Guo Y, Xu HE, Wang C, Zhang C, Feng S, Yu X, Zhang Y, Sun JP. Unsaturated bond recognition leads to biased signal in a fatty acid receptor. Science 2023; 380:eadd6220. [PMID: 36862765 DOI: 10.1126/science.add6220] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 02/16/2023] [Indexed: 03/04/2023]
Abstract
Individual free fatty acids (FAs) play important roles in metabolic homeostasis, many through engagement with more than 40G protein-coupled receptors. Searching for receptors to sense beneficial omega-3 FAs of fish oil enabled the identification of GPR120, which is involved in a spectrum of metabolic diseases. Here, we report six cryo-electron microscopy structures of GPR120 in complex with FA hormones or TUG891 and Gi or Giq trimers. Aromatic residues inside the GPR120 ligand pocket were responsible for recognizing different double-bond positions of these FAs and connect ligand recognition to distinct effector coupling. We also investigated synthetic ligand selectivity and the structural basis of missense single-nucleotide polymorphisms. We reveal how GPR120 differentiates rigid double bonds and flexible single bonds. The knowledge gleaned here may facilitate rational drug design targeting to GPR120.
Collapse
Affiliation(s)
- Chunyou Mao
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Peng Xiao
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xiao-Na Tao
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
- The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Jiao Qin
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qing-Tao He
- Department of Orthopedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Chao Zhang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Sheng-Chao Guo
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Ya-Qin Du
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Li-Nan Chen
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Dan-Dan Shen
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhi-Shuai Yang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Han-Qiong Zhang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Shen-Ming Huang
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Yong-Hao He
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Jie Cheng
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Ya-Ni Zhong
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Pan Shang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Jun Chen
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Dao-Lai Zhang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Qian-Lang Wang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Mei-Xia Liu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Guo-Yu Li
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yongyuan Guo
- Department of Orthopedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - H Eric Xu
- CAS Key Laboratory of Receptor Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Chuanxin Wang
- The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Shiqing Feng
- The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Yan Zhang
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
- MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China
- Research and Development Center for E-Learning, Ministry of Education, Beijing 100816, China
| | - Jin-Peng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
17
|
Sun J, Li M, Xiong Y, Zhai L, Zhao J. Oxidative Stress Mediated by N6-Methyladenosine Methylation Contributes to High-Fat Diet Induced Male Reproductive Dysfunction. Mol Nutr Food Res 2023; 67:e2101052. [PMID: 36738079 DOI: 10.1002/mnfr.202101052] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 10/26/2022] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To determine the mechanism of oxidative stress mediated by N6-methyladenosine (m6A) methylation contributing to high fat diet-induced reproductive dysfunction. RESULTS In vivo, compared with those in the Control group, the sperm count and sperm motility decrease significantly; the testosterone, luteinizing hormone levels, hyaluronidase, acrosomal enzyme levels, and total antioxidant capacity decrease significantly; malondialdehyde increases significantly in the DIO and DIO-R groups. The expression of nuclear factor erythroid 2-related factor 2 (Nrf2), superoxide dismutase 1 (SOD1), and NAD(P)H quinone dehydrogenase 1 (NQO1) decreases significantly in the DIO and DIO-R groups; m6A levels in testis tissue in the DIO and DIO-R groups increase; the enrichment of m6A-modified Nrf2 mRNA in testis in the DIO group and DIO-R group increases significantly. Also the m6A regulatory proteins increase significantly in the DIO group and DIO-R group. In vitro, compared to palmitic acid treated cells, the reactive oxygen species (ROS) level significantly decreases in STM2457, S-Adenosylhomocysteine treated cells and YTHDC2, YTHDF2 gene silence cells; however, Nrf2 expression increases in all treated cells. In addition, m6A expression decreases. CONCLUSIONS Oxidative stress mediates by methylation of m6A may contribute to high fat diet-induced male reproductive dysfunction.
Collapse
Affiliation(s)
- Jingyi Sun
- Department of Pharmacology, Shenyang Pharmaceutical University, No.103, Wenhua Rd, Shenhe Dist, Shenyang, 110016, China
| | - Mujiao Li
- Department of Pharmacology, Shenyang Pharmaceutical University, No.103, Wenhua Rd, Shenhe Dist, Shenyang, 110016, China
| | - Yuting Xiong
- Department of Pharmacology, Shenyang Pharmaceutical University, No.103, Wenhua Rd, Shenhe Dist, Shenyang, 110016, China
| | - Lingling Zhai
- Department of Maternal and Child Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei District, Shenyang, 110122, China
| | - Jian Zhao
- Department of Pharmacology, Shenyang Pharmaceutical University, No.103, Wenhua Rd, Shenhe Dist, Shenyang, 110016, China
| |
Collapse
|
18
|
Karmokar PF, Moniri NH. Oncogenic signaling of the free-fatty acid receptors FFA1 and FFA4 in human breast carcinoma cells. Biochem Pharmacol 2022; 206:115328. [PMID: 36309079 DOI: 10.1016/j.bcp.2022.115328] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 12/14/2022]
Abstract
Globally, breast cancer is the most frequent type of cancer in women, and most breast cancer-associated deaths are due to metastasis and recurrence of the disease. Dietary habits, specifically dietary fat intake is a crucial risk factor involved in breast cancer development and progression. Decades of research has revealed that free-fatty acids (FFA) modulate carcinogenic processes through fatty acid metabolism and lipid peroxidation. The ground-breaking discovery of free-fatty acid receptors, which are members of the G-protein coupled receptor (GPCR) superfamily, has led to the realization that FFA can also act via these receptors to modulate carcinogenic effects. The long-chain free-fatty acid receptors FFA1 (previously termed GPR40) and FFA4 (previously termed GPR120) are activated by mono- and polyunsaturated fatty acids including ω-3, 6, and 9 fatty acids. Initial enthusiasm towards the study of these receptors focused on their insulin secretagogue and sensitization effects, and the downstream associated metabolic regulation. However, recent studies have demonstrated that abnormal expression and/or aberrant FFA1/FFA4 signaling are evident in human breast carcinomas, suggesting that FFA receptors could be a promising target in the treatment of breast cancer. The current review discusses the diverse roles of FFA1 and FFA4 in the regulation of cell proliferation, migration, invasion, and chemotherapy resistance in human breast carcinoma cells and tissue.
Collapse
Affiliation(s)
- Priyanka F Karmokar
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University Health Sciences Center, Mercer University, Atlanta, GA 30341, USA
| | - Nader H Moniri
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University Health Sciences Center, Mercer University, Atlanta, GA 30341, USA; Department of Biomedical Sciences, School of Medicine, Mercer University Health Sciences Center, Mercer University, Macon, GA 31207, USA.
| |
Collapse
|
19
|
Lu Z, Li Y, Li AJ, Syn WK, Wank SA, Lopes-Virella MF, Huang Y. Loss of GPR40 in LDL receptor-deficient mice exacerbates high-fat diet-induced hyperlipidemia and nonalcoholic steatohepatitis. PLoS One 2022; 17:e0277251. [PMID: 36331958 PMCID: PMC9635748 DOI: 10.1371/journal.pone.0277251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
GPR40, a G protein-coupled receptor for free fatty acids (FFAs), is considered as a therapeutic target for type 2 diabetes mellitus (T2DM) since GPR40 activation in pancreatic beta cells enhances glucose-stimulated insulin secretion. Nonalcoholic fatty liver disease (NAFLD) is a common complication of T2DM or metabolic syndrome (MetS). However, the role of GPR40 in NAFLD associated with T2DM or MetS has not been well established. Given that it is known that cholesterol and FFAs are critically involved in the pathogenesis of nonalcoholic steatohepatitis (NASH) and LDL receptor (LDLR)-deficient mice are a good animal model for human hyperlipidemia including high cholesterol and FFAs, we generated GPR40 and LDLR double knockout (KO) mice in this study to determine the effect of GPR40 KO on hyperlipidemia-promoted NASH. We showed that GPR40 KO increased plasma levels of cholesterol and FFAs in high-fat diet (HFD)-fed LDLR-deficient mice. We also showed that GPR40 KO exacerbated HFD-induced hepatic steatosis, inflammation and fibrosis. Further study demonstrated that GPR40 KO led to upregulation of hepatic CD36 and genes involved in lipogenesis, fatty acid oxidation, fibrosis and inflammation. Finally, our in vitro mechanistic studies showed that while CD36 was involved in upregulation of proinflammatory molecules in macrophages by palmitic acid (PA) and lipopolysaccharide (LPS), GPR40 activation in macrophages exerts anti-inflammatory effects. Taken together, this study demonstrated for the first time that loss of GPR40 in LDLR-deficient mice exacerbated HFD-induced hyperlipidemia, hepatic steatosis, inflammation and fibrosis potentially through a CD36-dependent mechanism, suggesting that GPR40 may play a beneficial role in hyperlipidemia-associated NASH in LDLR-deficient mice.
Collapse
Affiliation(s)
- Zhongyang Lu
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Yanchun Li
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Ai-Jun Li
- Programs in Neuroscience, Washington State University, Pullman, Washington, United States of America
| | - Wing-Kin Syn
- Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, United States of America
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, Euskal Herriko Unibertsitatea/Universidad del País Vasco, Leioa, Spain
| | - Stephen A. Wank
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, United States of America
| | - Maria F. Lopes-Virella
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, United States of America
| | - Yan Huang
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
20
|
Liquid Chromatography-Mass Spectrometry (LC-MS) Derivatization-Based Methods for the Determination of Fatty Acids in Biological Samples. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27175717. [PMID: 36080484 PMCID: PMC9458108 DOI: 10.3390/molecules27175717] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/24/2022]
Abstract
Fatty acids (FAs) play pleiotropic roles in living organisms, acting as signaling molecules and gene regulators. They are present in plants and foods and may affect human health by food ingestion. As a consequence, analytical methods for their determination in biological fluids, plants and foods have attracted high interest. Undoubtedly, mass spectrometry (MS) has become an indispensable technique for the analysis of FAs. Due to the inherent poor ionization efficiency of FAs, their chemical derivatization prior to analysis is often employed. Usually, the derivatization of the FA carboxyl group aims to charge reversal, allowing detection and quantification in positive ion mode, thus, resulting in an increase in sensitivity in determination. Another approach is the derivatization of the double bond of unsaturated FAs, which aims to identify the double bond location. The present review summarizes the various classes of reagents developed for FA derivatization and discusses their applications in the liquid chromatography-MS (LC-MS) analysis of FAs in various matrices, including plasma and feces. In addition, applications for the determination of eicosanoids and fatty acid esters of hydroxy fatty acids (FAHFAs) are discussed.
Collapse
|
21
|
Görs PE, Wittenhofer P, Ayala-Cabrera JF, Meckelmann SW. Potential of atmospheric pressure ionization sources for the analysis of free fatty acids in clinical and biological samples by gas chromatography-mass spectrometry. Anal Bioanal Chem 2022; 414:6621-6634. [PMID: 35851410 PMCID: PMC9411222 DOI: 10.1007/s00216-022-04223-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/17/2022] [Accepted: 07/05/2022] [Indexed: 11/26/2022]
Abstract
Because of the central role of fatty acids in biological systems, their accurate quantification is still important. However, the impact of the complex matrix of biologically and clinically relevant samples such as plasma, serum, or cells makes the analysis still challenging, especially, when free non-esterified fatty acids have to be quantified. Here we developed and characterized a novel GC-MS method using pentafluorobenzyl bromide as a derivatization agent and compared different ionization techniques such as atmospheric pressure chemical ionization (APCI), atmospheric pressure chemical photoionization (APPI), and negative ion chemical ionization (NICI). The GC-APCI-MS showed the lowest limits of detection from 30 to 300 nM for a broad range of fatty acids and a similar response for various fatty acids from a chain length of 10 to 20 carbon atoms. This allows the number of internal standards necessary for accurate quantification to be reduced. Moreover, the use of pentafluorobenzyl bromide allows the direct derivatization of free fatty acids making them accessible for GC-MS analysis without labor-intense sample pretreatment.
Collapse
Affiliation(s)
- Paul E Görs
- Applied Analytical Chemistry, University of Duisburg-Essen, Universitätsstrasse 5, 45141, Essen, Germany
| | - Pia Wittenhofer
- Applied Analytical Chemistry, University of Duisburg-Essen, Universitätsstrasse 5, 45141, Essen, Germany
| | - Juan F Ayala-Cabrera
- Applied Analytical Chemistry, University of Duisburg-Essen, Universitätsstrasse 5, 45141, Essen, Germany
| | - Sven W Meckelmann
- Applied Analytical Chemistry, University of Duisburg-Essen, Universitätsstrasse 5, 45141, Essen, Germany.
| |
Collapse
|
22
|
Du YQ, Sha XY, Cheng J, Wang J, Lin JY, An WT, Pan W, Zhang LJ, Tao XN, Xu YF, Jia YL, Yang Z, Xiao P, Liu M, Sun JP, Yu X. Endogenous Lipid-GPR120 Signaling Modulates Pancreatic Islet Homeostasis to Different Extents. Diabetes 2022; 71:1454-1471. [PMID: 35472681 DOI: 10.2337/db21-0794] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 04/04/2022] [Indexed: 11/13/2022]
Abstract
Long-chain fatty acids (LCFAs) are not only energy sources but also serve as signaling molecules. GPR120, an LCFA receptor, plays key roles in maintaining metabolic homeostasis. However, whether endogenous ligand-GPR120 circuits exist and how such circuits function in pancreatic islets are unclear. Here, we found that endogenous GPR120 activity in pancreatic δ-cells modulated islet functions. At least two unsaturated LCFAs, oleic acid (OA) and linoleic acid (LA), were identified as GPR120 agonists within pancreatic islets. These two LCFAs promoted insulin secretion by inhibiting somatostatin secretion and showed bias activation of GPR120 in a model system. Compared with OA, LA exerted higher potency in promoting insulin secretion, which is dependent on β-arrestin2 function. Moreover, GPR120 signaling was impaired in the diabetic db/db model, and replenishing OA and LA improved islet function in both the db/db and streptozotocin-treated diabetic models. Consistently, the administration of LA improved glucose metabolism in db/db mice. Collectively, our results reveal that endogenous LCFA-GPR120 circuits exist and modulate homeostasis in pancreatic islets. The contributions of phenotype differences caused by different LCFA-GPR120 circuits within islets highlight the roles of fine-tuned ligand-receptor signaling networks in maintaining islet homeostasis.
Collapse
Affiliation(s)
- Ya-Qin Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Xue-Ying Sha
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Jie Cheng
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Jin Wang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Jing-Yu Lin
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Wen-Tao An
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Wei Pan
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Li-Jun Zhang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Xiao-Na Tao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Yun-Fei Xu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ying-Li Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Zhao Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Peng Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, MI
| | - Jin-Peng Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| |
Collapse
|
23
|
Kokotou MG, Mantzourani C, Batsika CS, Mountanea OG, Eleftheriadou I, Kosta O, Tentolouris N, Kokotos G. Lipidomics Analysis of Free Fatty Acids in Human Plasma of Healthy and Diabetic Subjects by Liquid Chromatography-High Resolution Mass Spectrometry (LC-HRMS). Biomedicines 2022; 10:biomedicines10051189. [PMID: 35625925 PMCID: PMC9138513 DOI: 10.3390/biomedicines10051189] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/17/2022] [Accepted: 05/17/2022] [Indexed: 11/16/2022] Open
Abstract
Targeted analytical methods for the determination of free fatty acids (FFAs) in human plasma are of high interest because they may help in identifying biomarkers for diseases and in monitoring the progress of a disease. The determination of FFAs is of particular importance in the case of metabolic disorders because FFAs have been associated with diabetes. We present a liquid chromatography-high resolution mass spectrometry (LC-HRMS) method, which allows the simultaneous determination of 74 FFAs in human plasma. The method is fast (10-min run) and straightforward, avoiding any derivatization step and tedious sample preparation. A total of 35 standard saturated and unsaturated FFAs, as well as 39 oxygenated (either hydroxy or oxo) saturated FFAs, were simultaneously detected and quantified in plasma samples from 29 subjects with type 2 diabetes mellitus (T2D), 14 with type 1 diabetes mellitus (T1D), and 28 healthy subjects. Alterations in the levels of medium-chain FFAs (C6:0 to C10:0) were observed between the control group and T2D and T1D patients.
Collapse
Affiliation(s)
- Maroula G. Kokotou
- Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (M.G.K.); (C.M.); (C.S.B.); (O.G.M.)
- Laboratory of Chemistry, Department of Food Science and Human Nutrition, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece; (I.E.); (N.T.)
| | - Christiana Mantzourani
- Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (M.G.K.); (C.M.); (C.S.B.); (O.G.M.)
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece; (I.E.); (N.T.)
| | - Charikleia S. Batsika
- Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (M.G.K.); (C.M.); (C.S.B.); (O.G.M.)
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece; (I.E.); (N.T.)
| | - Olga G. Mountanea
- Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (M.G.K.); (C.M.); (C.S.B.); (O.G.M.)
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece; (I.E.); (N.T.)
| | - Ioanna Eleftheriadou
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece; (I.E.); (N.T.)
- Diabetes Center, First Department of Propaedeutic and Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 15772 Athens, Greece;
| | - Ourania Kosta
- Diabetes Center, First Department of Propaedeutic and Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 15772 Athens, Greece;
| | - Nikolaos Tentolouris
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece; (I.E.); (N.T.)
- Diabetes Center, First Department of Propaedeutic and Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 15772 Athens, Greece;
| | - George Kokotos
- Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (M.G.K.); (C.M.); (C.S.B.); (O.G.M.)
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece; (I.E.); (N.T.)
- Correspondence: ; Tel.: +30-210-7274462
| |
Collapse
|
24
|
Niu B, Lu D, Zheng Z, Yuan S, Pang G. Replacing arginine 99 with leucine to study the kinetics of interconnected allosteric interactions between FFAR4 and naturally occurring fatty acids. Food Chem 2022; 382:132323. [PMID: 35144186 DOI: 10.1016/j.foodchem.2022.132323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/23/2022] [Accepted: 01/30/2022] [Indexed: 11/04/2022]
Abstract
The long-chain fatty acid receptor FFAR4 is the main G-protein-coupled receptor in the body for detecting long-chain fatty acids. It has been shown that Arg99 may be an important residue for fatty acid recognition and for the activation of hFFAR4, though direct evidence is still lacking. In this study, Arg99 on hFFAR4 was substituted with leucine by genetic manipulation, and a double-layer gold nanoparticle biosensor based on hFFAR4 (Arg99 → Leu) was constructed. The interconnected allosteric interaction between 11 naturally occurring fatty acid ligands and the receptor was determined. The results showed that Arg99 is the key residue on hFFAR4 for the recognition of the carboxyl group on fatty acids. This study offered direct quantitative evidence for the role played by different residues in receptor-ligand recognition and interconnected allosterism, providing a new approach for investigating the mechanisms and kinetics of interconnected receptor-ligand allosterism.
Collapse
Affiliation(s)
- Bo Niu
- College of Biotechnology & Food Science, Tianjin University of Commerce, Tianjin, China
| | - Dingqiang Lu
- College of Biotechnology & Food Science, Tianjin University of Commerce, Tianjin, China; Tianjin Key Laboratory of Food Biotechnology, Tianjin, China.
| | - Ziqing Zheng
- College of Biotechnology & Food Science, Tianjin University of Commerce, Tianjin, China
| | - Shuai Yuan
- College of Biotechnology & Food Science, Tianjin University of Commerce, Tianjin, China
| | - Guangchang Pang
- College of Biotechnology & Food Science, Tianjin University of Commerce, Tianjin, China; Tianjin Key Laboratory of Food Biotechnology, Tianjin, China.
| |
Collapse
|
25
|
Ito T, Yamamoto Y, Yamagishi N, Kanai Y. Stomach secretes estrogen in response to the blood triglyceride levels. Commun Biol 2021; 4:1364. [PMID: 34876651 PMCID: PMC8651635 DOI: 10.1038/s42003-021-02901-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 11/23/2021] [Indexed: 11/29/2022] Open
Abstract
Mammals receive body energy information to maintain energy homeostasis. Ghrelin, insulin, leptin and vagal afferents transmit the status of fasting, blood glucose, body fat, and food intake, respectively. Estrogen also inhibits feeding behavior and lipogenesis, but increases body fat mass. However, how blood triglyceride levels are monitored and the physiological roles of estrogen from the perspective of lipid homeostasis remain unsettled. Here, we show that stomach secretes estrogen in response to the blood triglyceride levels. Estrogen-secreting gastric parietal cells predominantly use fatty acids as an energy source. Blood estrogen levels increase as blood triglyceride levels rise in a stomach-dependent manner. Estrogen levels in stomach tissues increase as blood triglyceride levels rise, and isolated gastric gland epithelium produces estrogen in a fatty acid-dependent manner. We therefore propose that stomach monitors and controls blood triglyceride levels using estrogen, which inhibits feeding behavior and lipogenesis, and promotes triglyceride uptake by adipocytes.
Collapse
Affiliation(s)
- Takao Ito
- Cell Biology and Anatomy, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yuta Yamamoto
- Cell Biology and Anatomy, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Naoko Yamagishi
- Cell Biology and Anatomy, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yoshimitsu Kanai
- Cell Biology and Anatomy, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan.
| |
Collapse
|
26
|
Liu T, Ji RL, Tao YX. Naturally occurring mutations in G protein-coupled receptors associated with obesity and type 2 diabetes mellitus. Pharmacol Ther 2021; 234:108044. [PMID: 34822948 DOI: 10.1016/j.pharmthera.2021.108044] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/15/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022]
Abstract
G protein-coupled receptors (GPCRs) are the largest family of membrane receptors involved in the regulation of almost all known physiological processes. Dysfunctions of GPCR-mediated signaling have been shown to cause various diseases. The prevalence of obesity and type 2 diabetes mellitus (T2DM), two strongly associated disorders, is increasing worldwide, with tremendous economical and health burden. New safer and more efficacious drugs are required for successful weight reduction and T2DM treatment. Multiple GPCRs are involved in the regulation of energy and glucose homeostasis. Mutations in these GPCRs contribute to the development and progression of obesity and T2DM. Therefore, these receptors can be therapeutic targets for obesity and T2DM. Indeed some of these receptors, such as melanocortin-4 receptor and glucagon-like peptide 1 receptor, have provided important new drugs for treating obesity and T2DM. This review will focus on the naturally occurring mutations of several GPCRs associated with obesity and T2DM, especially incorporating recent large genomic data and insights from structure-function studies, providing leads for future investigations.
Collapse
Affiliation(s)
- Ting Liu
- Department of Anatomy, Physiology and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL 36849, United States
| | - Ren-Lei Ji
- Department of Anatomy, Physiology and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL 36849, United States
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL 36849, United States.
| |
Collapse
|
27
|
van Daal MT, Folkerts G, Garssen J, Braber S. Pharmacological Modulation of Immune Responses by Nutritional Components. Pharmacol Rev 2021; 73:198-232. [PMID: 34663688 DOI: 10.1124/pharmrev.120.000063] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The incidence of noncommunicable diseases (NCDs) has increased over the last few decades, and one of the major contributors to this is lifestyle, especially diet. High intake of saturated fatty acids and low intake of dietary fiber is linked to an increase in NCDs. Conversely, a low intake of saturated fatty acids and a high intake of dietary fiber seem to have a protective effect on general health. Several mechanisms have been identified that underlie this phenomenon. In this review, we focus on pharmacological receptors, including the aryl hydrocarbon receptor, binding partners of the retinoid X receptor, G-coupled protein receptors, and toll-like receptors, which can be activated by nutritional components and their metabolites. Depending on the nutritional component and the receptors involved, both proinflammatory and anti-inflammatory effects occur, leading to an altered immune response. These insights may provide opportunities for the prevention and treatment of NCDs and their inherent (sub)chronic inflammation. SIGNIFICANCE STATEMENT: This review summarizes the reported effects of nutritional components and their metabolites on the immune system through manipulation of specific (pharmacological) receptors, including the aryl hydrocarbon receptor, binding partners of the retinoid X receptor, G-coupled protein receptors, and toll-like receptors. Nutritional components, such as vitamins, fibers, and unsaturated fatty acids are able to resolve inflammation, whereas saturated fatty acids tend to exhibit proinflammatory effects. This may aid decision makers and scientists in developing strategies to decrease the incidence of noncommunicable diseases.
Collapse
Affiliation(s)
- Marthe T van Daal
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG, Utrecht, The Netherlands (M.T.v.D., G.F., J.G., S.B.); and Danone Nutricia Research, 3584 CT, Utrecht, The Netherlands (J.G.)
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG, Utrecht, The Netherlands (M.T.v.D., G.F., J.G., S.B.); and Danone Nutricia Research, 3584 CT, Utrecht, The Netherlands (J.G.)
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG, Utrecht, The Netherlands (M.T.v.D., G.F., J.G., S.B.); and Danone Nutricia Research, 3584 CT, Utrecht, The Netherlands (J.G.)
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG, Utrecht, The Netherlands (M.T.v.D., G.F., J.G., S.B.); and Danone Nutricia Research, 3584 CT, Utrecht, The Netherlands (J.G.)
| |
Collapse
|
28
|
Ochiai K, Hirooka R, Sakaino M, Takeuchi S, Hira T. 2-Arachidonoyl glycerol potently induces cholecystokinin secretion in murine enteroendocrine STC-1 cells via cannabinoid receptor CB1. Lipids 2021; 56:603-611. [PMID: 34533218 DOI: 10.1002/lipd.12323] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/09/2021] [Accepted: 09/01/2021] [Indexed: 11/05/2022]
Abstract
Cholecystokinin (CCK) is a peptide hormone secreted from enteroendocrine cells and regulates the exocrine pancreas, gastric motility, and appetite. Dietary triacylglycerols are hydrolyzed to fatty acids (FA) and 2-monoacylglycerols (2-MAG) in the small intestine. Although it is well known that FA stimulate CCK secretion, whether 2-MAG have the CCK-releasing activity remains unclear. We examined the CCK-releasing activity of four commercially available 2-MAG in a murine CCK-producing cell line, STC-1, and the molecular mechanism underlying 2-MAG-induced CCK secretion. CCK released from the cells was measured using ELISA. Among four 2-MAG (2-palmitoyl, 2-oleoyl, 2-linoleoyl, and 2-arachidonoyl monoacylglycerols) examined, 2-arachidonoyl glycerol (2-AG) potently stimulated CCK secretion in a dose-dependent manner. Structurally related compounds, such as 2-arachidonoyl glycerol ether and 1-arachidonoyl glycerol, did not stimulate CCK secretion. Both arachidonic acid and 2-AG stimulated CCK secretion at 100 μM, but only 2-AG did at 50 μM. 2-AG-induced CCK secretion but not arachidonic acid-induced CCK secretion was attenuated by treatment with a cannabinoid receptor 1 (CB1) antagonist. These results indicate that a specific 2-MAG, 2-AG, directly stimulates CCK secretion via CB1.
Collapse
Affiliation(s)
- Keita Ochiai
- Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | | | | | | | - Tohru Hira
- Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| |
Collapse
|
29
|
Secor JD, Fligor SC, Tsikis ST, Yu LJ, Puder M. Free Fatty Acid Receptors as Mediators and Therapeutic Targets in Liver Disease. Front Physiol 2021; 12:656441. [PMID: 33897464 PMCID: PMC8058363 DOI: 10.3389/fphys.2021.656441] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/15/2021] [Indexed: 12/29/2022] Open
Abstract
Free fatty acid receptors (FFARs) are a class of G protein-coupled receptors (GPCRs) that have wide-ranging effects on human physiology. The four well-characterized FFARs are FFAR1/GPR40, FFAR2/GPR43, FFAR3/GPR41, and FFAR4/GPR120. Short-chain (<6 carbon) fatty acids target FFAR2/GPR43 and FFAR3/GPR41. Medium- and long-chain fatty acids (6-12 and 13-21 carbon, respectively) target both FFAR1/GPR40 and FFAR4/GPR120. Signaling through FFARs has been implicated in non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), intestinal failure-associated liver disease (IFALD), and a variety of other liver disorders. FFARs are now regarded as targets for therapeutic intervention for liver disease, diabetes, obesity, hyperlipidemia, and metabolic syndrome. In this review, we provide an in-depth, focused summary of the role FFARs play in liver health and disease.
Collapse
Affiliation(s)
- Jordan D. Secor
- Vascular Biology Program and Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | | | | | | | | |
Collapse
|
30
|
Granados JC, Nigam AK, Bush KT, Jamshidi N, Nigam SK. A key role for the transporter OAT1 in systemic lipid metabolism. J Biol Chem 2021; 296:100603. [PMID: 33785360 PMCID: PMC8102404 DOI: 10.1016/j.jbc.2021.100603] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/22/2021] [Accepted: 03/26/2021] [Indexed: 01/06/2023] Open
Abstract
Organic anion transporter 1 (OAT1/SLC22A6) is a drug transporter with numerous xenobiotic and endogenous substrates. The Remote Sensing and Signaling Theory suggests that drug transporters with compatible ligand preferences can play a role in “organ crosstalk,” mediating overall organismal communication. Other drug transporters are well known to transport lipids, but surprisingly little is known about the role of OAT1 in lipid metabolism. To explore this subject, we constructed a genome-scale metabolic model using omics data from the Oat1 knockout mouse. The model implicated OAT1 in the regulation of many classes of lipids, including fatty acids, bile acids, and prostaglandins. Accordingly, serum metabolomics of Oat1 knockout mice revealed increased polyunsaturated fatty acids, diacylglycerols, and long-chain fatty acids and decreased ceramides and bile acids when compared with wildtype controls. Some aged knockout mice also displayed increased lipid droplets in the liver when compared with wildtype mice. Chemoinformatics and machine learning analyses of these altered lipids defined molecular properties that form the structural basis for lipid-transporter interactions, including the number of rings, positive charge/volume, and complexity of the lipids. Finally, we obtained targeted serum metabolomics data after short-term treatment of rodents with the OAT-inhibiting drug probenecid to identify potential drug–metabolite interactions. The treatment resulted in alterations in eicosanoids and fatty acids, further supporting our metabolic reconstruction predictions. Consistent with the Remote Sensing and Signaling Theory, the data support a role of OAT1 in systemic lipid metabolism.
Collapse
Affiliation(s)
- Jeffry C Granados
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Anisha K Nigam
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - Kevin T Bush
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Neema Jamshidi
- Department of Radiological Sciences, University of California Los Angeles, Los Angeles, California, USA
| | - Sanjay K Nigam
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA; Department of Medicine, University of California San Diego, La Jolla, California, USA.
| |
Collapse
|
31
|
Understanding the appetite modulation pathways: The role of the FFA1 and FFA4 receptors. Biochem Pharmacol 2021; 186:114503. [PMID: 33711286 DOI: 10.1016/j.bcp.2021.114503] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/27/2021] [Accepted: 03/02/2021] [Indexed: 11/24/2022]
Abstract
Pharmaconutrition is an area of current interest, especially concerning the advances in the pharmacology of nutrient-sensing receptors, as have been accomplished in the last 20 years. The family of free fatty acid (FFA) receptors is composed of four members, sequentially named as FFA1 to FFA4, which are activated by the short to long-chain fatty acids. The affinity of the FFA1 and FFA4 receptors for the omega-3 polyunsaturated fatty acids prompted pre-clinical and clinical investigations regarding their involvement in metabolic diseases. The main studies have been focused on the receptors' expression analyses, the featuring of knockout mice, and the assessment of selective synthetic ligands. These clearly have indicated a relevant role for FFA1 and FFA4 in the peripheral and central circuits for the regulation of energetic metabolism. This review article aimed to discuss the relevance of the FFA1 and FFA4 receptors in appetite-related complications, mainly related to obesity, cancer cachexia, and anorexia in the elderly, emphasizing whether their pharmacological modulation might be useful for the management of these disorders.
Collapse
|
32
|
Chen GC, Arthur R, Qin LQ, Chen LH, Mei Z, Zheng Y, Li Y, Wang T, Rohan TE, Qi Q. Association of Oily and Nonoily Fish Consumption and Fish Oil Supplements With Incident Type 2 Diabetes: A Large Population-Based Prospective Study. Diabetes Care 2021; 44:672-680. [PMID: 33431419 PMCID: PMC7896269 DOI: 10.2337/dc20-2328] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To evaluate associations of oily and nonoily fish consumption and fish oil supplements with incident type 2 diabetes (T2D). RESEARCH DESIGN AND METHODS We included 392,287 middle-aged and older participants (55.0% women) in the UK Biobank who were free of diabetes, major cardiovascular disease, and cancer and had information on habitual intake of major food groups and use of fish oil supplements at baseline (2006-2010). Of these, 163,706 participated in one to five rounds of 24-h dietary recalls during 2009-2012. RESULTS During a median 10.1 years of follow-up, 7,262 incident cases of T2D were identified. Compared with participants who reported never consumption of oily fish, the multivariable-adjusted hazard ratios of T2D were 0.84 (95% CI 0.78-0.91), 0.78 (0.72-0.85), and 0.78 (0.71-0.86) for those who reported <1 serving/week, weekly, and ≥2 servings/week of oily fish consumption, respectively (P-trend < 0.001). Consumption of nonoily fish was not associated with risk of T2D (P-trend = 0.45). Participants who reported regular fish oil use at baseline had a 9% (95% CI 4-14%) lower risk of T2D compared with nonusers. Baseline regular users of fish oil who also reported fish oil use during at least one of the 24-h dietary recalls had an 18% (8-27%) lower risk of T2D compared with constant nonusers. CONCLUSIONS Our findings suggest that consumption of oily fish but not nonoily fish was associated with a lower risk of T2D. Use of fish oil supplements, especially constant use over time, was also associated with a lower risk of T2D.
Collapse
Affiliation(s)
- Guo-Chong Chen
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Rhonda Arthur
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Li-Qiang Qin
- Department of Nutrition and Food Hygiene, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Li-Hua Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Zhendong Mei
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Yan Zheng
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Yang Li
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Tao Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY .,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
33
|
Grundmann M, Bender E, Schamberger J, Eitner F. Pharmacology of Free Fatty Acid Receptors and Their Allosteric Modulators. Int J Mol Sci 2021; 22:ijms22041763. [PMID: 33578942 PMCID: PMC7916689 DOI: 10.3390/ijms22041763] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 12/19/2022] Open
Abstract
The physiological function of free fatty acids (FFAs) has long been regarded as indirect in terms of their activities as educts and products in metabolic pathways. The observation that FFAs can also act as signaling molecules at FFA receptors (FFARs), a family of G protein-coupled receptors (GPCRs), has changed the understanding of the interplay of metabolites and host responses. Free fatty acids of different chain lengths and saturation statuses activate FFARs as endogenous agonists via binding at the orthosteric receptor site. After FFAR deorphanization, researchers from the pharmaceutical industry as well as academia have identified several ligands targeting allosteric sites of FFARs with the aim of developing drugs to treat various diseases such as metabolic, (auto)inflammatory, infectious, endocrinological, cardiovascular, and renal disorders. GPCRs are the largest group of transmembrane proteins and constitute the most successful drug targets in medical history. To leverage the rich biology of this target class, the drug industry seeks alternative approaches to address GPCR signaling. Allosteric GPCR ligands are recognized as attractive modalities because of their auspicious pharmacological profiles compared to orthosteric ligands. While the majority of marketed GPCR drugs interact exclusively with the orthosteric binding site, allosteric mechanisms in GPCR biology stay medically underexploited, with only several allosteric ligands currently approved. This review summarizes the current knowledge on the biology of FFAR1 (GPR40), FFAR2 (GPR43), FFAR3 (GPR41), FFAR4 (GPR120), and GPR84, including structural aspects of FFAR1, and discusses the molecular pharmacology of FFAR allosteric ligands as well as the opportunities and challenges in research from the perspective of drug discovery.
Collapse
Affiliation(s)
- Manuel Grundmann
- Research and Early Development, Bayer Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany;
- Correspondence:
| | - Eckhard Bender
- Drug Discovery Sciences, Bayer Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany; (E.B.); (J.S.)
| | - Jens Schamberger
- Drug Discovery Sciences, Bayer Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany; (E.B.); (J.S.)
| | - Frank Eitner
- Research and Early Development, Bayer Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany;
| |
Collapse
|
34
|
Son SE, Kim NJ, Im DS. Development of Free Fatty Acid Receptor 4 (FFA4/GPR120) Agonists in Health Science. Biomol Ther (Seoul) 2021; 29:22-30. [PMID: 33372166 PMCID: PMC7771848 DOI: 10.4062/biomolther.2020.213] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 11/28/2020] [Accepted: 11/30/2020] [Indexed: 12/14/2022] Open
Abstract
Till the 21st century, fatty acids were considered as merely building blocks for triglycerides, phospholipids, or cholesteryl esters. However, the discovery of G protein-coupled receptors (GPCRs) for free fatty acids at the beginning of the 21st century challenged that idea and paved way for a new field of research, merged into the field of receptor pharmacology for intercellular lipid mediators. Among the GPCRs for free fatty acids, free fatty acid receptor 4 (FFA4, also known as GPR120) recognizes long-chain polyunsaturated fatty acids such as DHA and EPA. It is significant in drug discovery because it regulates obesity-induced metaflammation and GLP-1 secretion. Our study reviews information on newly developed FFA4 agonists and their application in pathophysiologic studies and drug discovery. It also offers a potency comparison of the FFA4 agonists in an AP-TGF-α shedding assay.
Collapse
Affiliation(s)
- So-Eun Son
- Department of Pharmacy, College of Pharmacy, and Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Nam-Jung Kim
- Department of Pharmacy, College of Pharmacy, and Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Dong-Soon Im
- Department of Pharmacy, College of Pharmacy, and Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
35
|
López-Méndez I, Méndez-Maldonado K, Manzo-Francisco LA, Juárez-Hernández E, Uribe M, Barbero-Becerra VJ. G protein-coupled receptors: Key molecules in metabolic associated fatty liver disease development. Nutr Res 2020; 87:70-79. [PMID: 33601216 DOI: 10.1016/j.nutres.2020.12.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/07/2020] [Accepted: 12/20/2020] [Indexed: 02/08/2023]
Abstract
Metabolic associated fatty liver disease (MAFLD) is a range of hepatic disorders with progression to steatohepatitis with risk of development of fibrosis, cirrhosis, and hepatocellular carcinoma. MAFLD is strongly related to metabolic disorders of active fatty acids, which seem to be selective according to their specific ligand of G protein-coupled receptors (GPRs) located in immune response cells. An approach to study the pathophysiological mechanisms of MAFLD could be through the expression of active fatty acids ligands. The expression of GPRs is associated with obesity, microbiota environment, and dietary characteristics in patients with MAFLD. More specifically, GPR41, GPR43, GPR20, and GPR120 have been associated with alteration of lipid metabolism in hepatic and intestinal cells, and consequently they have a key role in metabolic diseases. We observed that GPR120 is not expressed in nonoverweight/obese patients, regardless of the presence of MAFLD; meanwhile the expression of GPR41 is increased in patients with lean MAFLD. GPRs role in liver disease is intriguing and a field of research opportunity. More studies are necessary to define the role of active fatty acids in the development of metabolic diseases.
Collapse
Affiliation(s)
- Iván López-Méndez
- Transplants and Hepatology Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
| | - Karla Méndez-Maldonado
- Cellular Physiology Institute, Neurosciences Division & Physiology and Pharmacology Department, Veterinary and Zootechnics Faculty, UNAM, Mexico City, Mexico
| | | | - Eva Juárez-Hernández
- Translational Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
| | - Misael Uribe
- Gastrointestinal and Obesity Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
| | | |
Collapse
|
36
|
Husted AS, Ekberg JH, Tripp E, Nissen TAD, Meijnikman S, O'Brien SL, Ulven T, Acherman Y, Bruin SC, Nieuwdorp M, Gerhart-Hines Z, Calebiro D, Dragsted LO, Schwartz TW. Autocrine negative feedback regulation of lipolysis through sensing of NEFAs by FFAR4/GPR120 in WAT. Mol Metab 2020; 42:101103. [PMID: 33091626 PMCID: PMC7683346 DOI: 10.1016/j.molmet.2020.101103] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/11/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES Long-chain fatty acids (LCFAs) released from adipocytes inhibit lipolysis through an unclear mechanism. We hypothesized that the LCFA receptor, FFAR4 (GPR120), which is highly expressed in adipocytes, may be involved in this feedback regulation. METHODS AND RESULTS Liquid chromatography mass spectrometry (LC-MS) analysis of conditioned media from isoproterenol-stimulated primary cultures of murine and human adipocytes demonstrated that most of the released non-esterified free fatty acids (NEFAs) are known agonists for FFAR4. In agreement with this, conditioned medium from isoproterenol-treated adipocytes stimulated signaling strongly in FFAR4 transfected COS-7 cells as opposed to non-transfected control cells. In transfected 3T3-L1 cells, FFAR4 agonism stimulated Gi- and Go-mini G protein binding more strongly than Gq, effects which were blocked by the selective FFAR4 antagonist AH7614. In primary cultures of murine white adipocytes, the synthetic, selective FFAR4 agonist CpdA inhibited isoproterenol-induced intracellular cAMP accumulation in a manner similar to the antilipolytic control agent nicotinic acid acting through another receptor, HCAR2. In vivo, oral gavage with the synthetic, specific FFAR4 agonist CpdB decreased the level of circulating NEFAs in fasting lean mice to a similar degree as nicotinic acid. In agreement with the identified anti-lipolytic effect of FFAR4, plasma NEFAs and glycerol were increased in FFAR4-deficient mice as compared to littermate controls despite having elevated insulin levels, and cAMP accumulation in primary adipocyte cultures was augmented by treatment with the FFAR4 antagonist conceivably by blocking the stimulatory tone of endogenous NEFAs on FFAR4. CONCLUSIONS In white adipocytes, FFAR4 functions as an NEFA-activated, autocrine, negative feedback regulator of lipolysis by decreasing cAMP though Gi-mediated signaling.
Collapse
Affiliation(s)
- Anna Sofie Husted
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark.
| | - Jeppe H Ekberg
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark.
| | - Emma Tripp
- Institute of Metabolism and Systems Research and Center of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham B15 2TT, United Kingdom.
| | - Tinne A D Nissen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark.
| | - Stijn Meijnikman
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands.
| | - Shannon L O'Brien
- Institute of Metabolism and Systems Research and Center of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham B15 2TT, United Kingdom.
| | - Trond Ulven
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark.
| | - Yair Acherman
- Department of Surgery, Spaarne Hospital, Hoofddorp, the Netherlands.
| | - Sjoerd C Bruin
- Department of Surgery, Spaarne Hospital, Hoofddorp, the Netherlands.
| | - Max Nieuwdorp
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands.
| | - Zach Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark.
| | - Davide Calebiro
- Institute of Metabolism and Systems Research and Center of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham B15 2TT, United Kingdom.
| | - Lars O Dragsted
- Department of Nutrition, Exercise, and Sports, Section of Preventive and Clinical Nutrition, University of Copenhagen, Rolighedsvej 30, Frederiksberg C, 1958, Denmark.
| | - Thue W Schwartz
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark.
| |
Collapse
|
37
|
Freitas RDS, Muradás TC, Dagnino APA, Rost FL, Costa KM, Venturin GT, Greggio S, da Costa JC, Campos MM. Targeting FFA1 and FFA4 receptors in cancer-induced cachexia. Am J Physiol Endocrinol Metab 2020; 319:E877-E892. [PMID: 32893672 DOI: 10.1152/ajpendo.00509.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Free fatty acid (FFA) receptors FFA1 and FFA4 are omega-3 molecular targets in metabolic diseases; however, their function in cancer cachexia remains unraveled. We assessed the role of FFA1 and FFA4 receptors in the mouse model of cachexia induced by Lewis lung carcinoma (LLC) cell implantation. Naturally occurring ligands such as α-linolenic acid (ALA) and docosahexaenoic acid (DHA), the synthetic FFA1/FFA4 agonists GW9508 and TUG891, or the selective FFA1 GW1100 or FFA4 AH7614 antagonists were tested. FFA1 and FFA4 expression and other cachexia-related parameters were evaluated. GW9508 and TUG891 decreased tumor weight in LLC-bearing mice. Regarding cachexia-related end points, ALA, DHA, and the preferential FFA1 agonist GW9508 rescued body weight loss. Skeletal muscle mass was reestablished by ALA treatment, but this was not reflected in the fiber cross-sectional areas (CSA) measurement. Otherwise, TUG891, GW1100, or AH7614 reduced the muscle fiber CSA. Treatments with ALA, GW9508, GW1100, or AH7614 restored white adipose tissue (WAT) depletion. As for inflammatory outcomes, ALA improved anemia, whereas GW9508 reduced splenomegaly. Concerning behavioral impairments, ALA and GW9508 rescued locomotor activity, whereas ALA improved motor coordination. Additionally, DHA improved grip strength. Notably, GW9508 restored abnormal brain glucose metabolism in different brain regions. The GW9508 treatment increased leptin levels, without altering uncoupling protein-1 downregulation in visceral fat. LLC-cachectic mice displayed FFA1 upregulation in subcutaneous fat, but not in visceral fat or gastrocnemius muscle, whereas FFA4 was unaltered. Overall, the present study shed new light on FFA1 and FFA4 receptors' role in metabolic disorders, indicating FFA1 receptor agonism as a promising strategy in mitigating cancer cachexia.
Collapse
Affiliation(s)
- Raquel D S Freitas
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Centro de Pesquisas em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Thaís C Muradás
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Centro de Pesquisas em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Ana Paula A Dagnino
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Centro de Pesquisas em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernanda L Rost
- Centro de Pesquisas em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Kesiane M Costa
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Gianina T Venturin
- Centro de Pesquisa Pré-Clínica, Instituto do Cérebro do Rio Grande do Sul (Brain Institute of Rio Grande do Sul - BraIns), Porto Alegre, Brazil
| | - Samuel Greggio
- Centro de Pesquisa Pré-Clínica, Instituto do Cérebro do Rio Grande do Sul (Brain Institute of Rio Grande do Sul - BraIns), Porto Alegre, Brazil
| | - Jaderson C da Costa
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Centro de Pesquisa Pré-Clínica, Instituto do Cérebro do Rio Grande do Sul (Brain Institute of Rio Grande do Sul - BraIns), Porto Alegre, Brazil
| | - Maria M Campos
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Centro de Pesquisas em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-graduação em Odontologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
38
|
Dragano NR, Monfort-Pires M, Velloso LA. Mechanisms Mediating the Actions of Fatty Acids in the Hypothalamus. Neuroscience 2020; 447:15-27. [PMID: 31689488 DOI: 10.1016/j.neuroscience.2019.10.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/02/2019] [Accepted: 10/03/2019] [Indexed: 12/12/2022]
|
39
|
Luscombe VB, Lucy D, Bataille CJR, Russell AJ, Greaves DR. 20 Years an Orphan: Is GPR84 a Plausible Medium-Chain Fatty Acid-Sensing Receptor? DNA Cell Biol 2020; 39:1926-1937. [PMID: 33001759 DOI: 10.1089/dna.2020.5846] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
GPR84 is an inflammation-induced receptor highly expressed on immune cells, yet its endogenous ligand is still unknown. This makes any interpretation of its physiological activity in vivo difficult. However, experiments with potent synthetic agonists have highlighted what the receptor can do, namely, enhance proinflammatory signaling and macrophage effector functions such as phagocytosis. Developing drugs to block these effects has attracted interest from the scientific community with the aim of decreasing disease activity in inflammatory disorders or enhancing inflammation resolution. In this review, we critically reassess the widely held belief that the major role of GPR84 is that of being a medium-chain fatty acid (MCFA) receptor. While MCFAs have been shown to activate GPR84, it remains to be demonstrated that they are present in relevant tissues at appropriate concentrations. In contrast to four other "full-time" free fatty acid receptor subtypes, GPR84 is not expressed by enteroendocrine cells and has limited expression in the gastrointestinal tract. Across multiple tissues and cell types, the highest expression levels of GPR84 are observed hours after exposure to an inflammatory stimulus. These factors obscure the relationship between ligand and receptor in the human body and do not support the exclusive physiological pairing of MCFAs with GPR84. To maximize the chances of developing efficacious drugs for inflammatory diseases, we must advance our understanding of GPR84 and what it does in vivo.
Collapse
Affiliation(s)
- Vincent B Luscombe
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Daniel Lucy
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom.,Department of Chemistry and University of Oxford, Oxford, United Kingdom
| | | | - Angela J Russell
- Department of Chemistry and University of Oxford, Oxford, United Kingdom.,Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - David R Greaves
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
40
|
Wawrzyniak P, Noureddine N, Wawrzyniak M, Lucchinetti E, Krämer SD, Rogler G, Zaugg M, Hersberger M. Nutritional Lipids and Mucosal Inflammation. Mol Nutr Food Res 2020; 65:e1901269. [PMID: 32780927 DOI: 10.1002/mnfr.201901269] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/24/2020] [Indexed: 12/19/2022]
Abstract
Inflammatory bowel disease (IBD) is characterized by chronic relapsing inflammation in the intestine. Given their role in regulation of inflammation, long-chain n-3 polyunsaturated fatty acids (PUFAs) represent a potential supplementary therapeutic approach to current drug regimens used for IBD. Mechanistically, there is ample evidence for an anti-inflammatory and pro-resolution effect of long-chain n-3 PUFAs after they incorporate into cell membrane phospholipids. They disrupt membrane rafts and when released from the membrane suppress inflammatory signaling by activating PPAR-γ and free fatty acid receptor 4; furthermore, they shift the lipid mediator profile from pro-inflammatory eicosanoids to specialized pro-resolving mediators. The allocation of long-chain n-3 PUFAs also leads to a higher microbiome diversity in the gut, increases short-chain fatty acid-producing bacteria, and improves intestinal barrier function by sealing epithelial tight junctions. In line with these mechanistic studies, most epidemiological studies support a beneficial effect of long-chain n-3 PUFAs intake on reducing the incidence of IBD. However, the results from intervention trials on the prevention of relapse in IBD patients show no or only a marginal effect of long-chain n-3 PUFAs supplementation. In light of the current literature, international recommendations are supported that adequate diet-derived n-3 PUFAs might be beneficial in maintaining remission in IBD patients.
Collapse
Affiliation(s)
- Paulina Wawrzyniak
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, 8032, Switzerland.,Children's Research Center, University Children's Hospital Zurich, Zurich, 8032, Switzerland
| | - Nazek Noureddine
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, 8032, Switzerland.,Children's Research Center, University Children's Hospital Zurich, Zurich, 8032, Switzerland.,Center for Integrative Human Physiology, University of Zurich, Zurich, 8057, Switzerland
| | - Marcin Wawrzyniak
- Department of Gastroenterology and Hepatology, University Hospital Zurich and University of Zurich, Zurich, 8091, Switzerland
| | - Eliana Lucchinetti
- Department of Anesthesiology and Pain Medicine and Cardiovascular Research Centre, University of Alberta, Edmonton, T6G 2R3, Canada
| | - Stefanie D Krämer
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, 8093, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich and University of Zurich, Zurich, 8091, Switzerland
| | - Michael Zaugg
- Department of Anesthesiology and Pain Medicine and Cardiovascular Research Centre, University of Alberta, Edmonton, T6G 2R3, Canada.,Department of Pharmacology, University of Alberta, Edmonton, T6G 2R3, Canada
| | - Martin Hersberger
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, 8032, Switzerland.,Children's Research Center, University Children's Hospital Zurich, Zurich, 8032, Switzerland.,Center for Integrative Human Physiology, University of Zurich, Zurich, 8057, Switzerland
| |
Collapse
|
41
|
Tsuchiya Y, Hayashi M, Nagamatsu K, Ono T, Kamakura M, Iwata T, Nakashima T. The key royal jelly component 10-hydroxy-2-decenoic acid protects against bone loss by inhibiting NF-κB signaling downstream of FFAR4. J Biol Chem 2020; 295:12224-12232. [PMID: 32647011 PMCID: PMC7443481 DOI: 10.1074/jbc.ra120.013821] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/03/2020] [Indexed: 12/11/2022] Open
Abstract
The supplementation of royal jelly (RJ) is known to provide a variety of health benefits, including anti-inflammatory and anti-obesity effects. RJ treatment also reportedly protects against bone loss, but no single factor in RJ has yet been identified as an anti-osteoporosis agent. Here we fractionated RJ and identified 10-hydroxy-2-decenoic acid (10H2DA) as a key component involved in inhibiting osteoclastogenesis based on mass spectrometric analysis. We further demonstrated free fatty acid receptor 4 (FFAR4) as directly interacting with 10H2DA; binding of 10H2DA to FFAR4 on osteoclasts inhibited receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL)-induced activation of NF-κB signaling, thereby attenuating the induction of nuclear factor of activated T cells (NFAT) c1, a key transcription factor for osteoclastogenesis. Oral administration of 10H2DA attenuated bone resorption in ovariectomized mice. These results suggest a potential therapeutic approach of targeting osteoclast differentiation by the supplementation of RJ, and specifically 10H2DA, in cases of pathological bone loss such as occur in postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Yosuke Tsuchiya
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mikihito Hayashi
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Agency for Medical Research and Development, Precursory Research for Innovative Medical Care (PRIME), Tokyo, Japan
| | | | - Takehito Ono
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Masaki Kamakura
- Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, Toyama, Japan
| | - Takanori Iwata
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomoki Nakashima
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan.
| |
Collapse
|
42
|
Palmitic Acid Targets Human Testicular Peritubular Cells and Causes a Pro-Inflammatory Response. J Clin Med 2020; 9:jcm9082655. [PMID: 32824411 PMCID: PMC7463762 DOI: 10.3390/jcm9082655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/07/2020] [Accepted: 08/13/2020] [Indexed: 11/18/2022] Open
Abstract
Palmitic acid (PA) is a major fatty acid, derived from diet and endogenous production, which is being linked to inflammation. While such actions of PA at the level of the testis remain difficult to examine, we reasoned that studies in human testicular cells may be instructive. Human testicular peritubular cells (HTPCs) can be isolated from men and cultured. They have contractile properties but also produce Interleukin 6 (IL6), express the inflammasome member NLRP3, and via glia cell line derived neurotrophic factor (GDNF), they contribute to the spermatogonial stem cell niche. We found that PA at 100 µM significantly increased the levels of IL6, while NLRP3 or the related Interleukin 1 beta (IL1beta) were not affected. The contractility marker calponin (CNN1) and the growth factor GDNF were likewise not affected. ELISA studies confirmed the stimulatory PA actions on IL6. Hence, PA derived from diet and/or endogenous sources may be able to foster a pro-inflammatory milieu in the testis. A possible link of these results to diet and high fat intake and obesity is indicated by the about 12-fold elevated testicular levels of IL6 in testes of obese rhesus monkeys (n = 3), fed with a Western Style diet. They had elevated 2–5-fold increased body fat and increased circulating triglyceride levels. Further consequences of PA and obesity for testicular functions remain to be evaluated.
Collapse
|
43
|
Schots PC, Pedersen AM, Eilertsen KE, Olsen RL, Larsen TS. Possible Health Effects of a Wax Ester Rich Marine Oil. Front Pharmacol 2020; 11:961. [PMID: 32676029 PMCID: PMC7333527 DOI: 10.3389/fphar.2020.00961] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 06/12/2020] [Indexed: 12/19/2022] Open
Abstract
The consumption of seafood and the use of fish oil for the production of nutraceuticals and fish feed have increased over the past decades due the high content of long-chain polyunsaturated omega-3 fatty acids. This increase has put pressure on the sustainability of fisheries. One way to overcome the limited supply of fish oil is to harvest lower in the marine food web. Calanus finmarchicus, feeding on phytoplankton, is a small copepod constituting a considerable biomass in the North Atlantic and is a novel source of omega-3 fatty acids. The oil is, however, different from other commercial marine oils in terms of chemistry and, possibly, bioactivity since it contains wax esters. Wax esters are fatty acids that are esterified with alcohols. In addition to the long-chain polyunsaturated omega-3 fatty acids, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), the oil is also rich in stearidonic acid (SDA), long-chain monounsaturated fatty acids, and the long-chain fatty alcohols eicosenol and docosenol. Recent animal studies have indicated anti-inflammatory and anti-obesogenic actions of this copepod oil beyond that provided by EPA and DHA. This review will discuss potential mechanisms behind these beneficial effects of the oil, focusing on the impact of the various components of the oil. The health effects of EPA and DHA are well recognized, whereas long-chain monounsaturated fatty acids and long-chain fatty alcohols have to a large degree been overlooked in relation to human health. Recently, however the fatty alcohols have received interest as potential targets for improved health via conversion to their corresponding fatty acids. Together, the different lipid components of the oil from C. finmarchicus may have potential as nutraceuticals for reducing obesity and obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Pauke Carlijn Schots
- Faculty of Biosciences, Fisheries and Economics, Norwegian College of Fishery Science, UiT The Arctic University of Norway, Tromsø, Norway
| | | | - Karl-Erik Eilertsen
- Faculty of Biosciences, Fisheries and Economics, Norwegian College of Fishery Science, UiT The Arctic University of Norway, Tromsø, Norway
| | - Ragnar Ludvig Olsen
- Faculty of Biosciences, Fisheries and Economics, Norwegian College of Fishery Science, UiT The Arctic University of Norway, Tromsø, Norway
| | - Terje Steinar Larsen
- Cardiovascular Research Group, Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
44
|
Tomita Y, Cakir B, Liu CH, Fu Z, Huang S, Cho SS, Britton WR, Sun Y, Puder M, Hellström A, Talukdar S, Smith LEH. Free fatty acid receptor 4 activation protects against choroidal neovascularization in mice. Angiogenesis 2020; 23:385-394. [PMID: 32140799 DOI: 10.1007/s10456-020-09717-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/24/2020] [Indexed: 02/06/2023]
Abstract
To examine whether free fatty acid receptor 4 (FFAR4) activation can protect against choroidal neovascularization (CNV), which is a common cause of blindness, and to elucidate the mechanism underlying the inhibition, we used the mouse model of laser-induced CNV to mimic angiogenic aspects of age-related macular degeneration (AMD). Laser-induced CNV was compared between groups treated with an FFAR4 agonist or vehicle, and between FFAR4 wild-type (Ffar4+/+) and knock out (Ffar4-/-) mice on a C57BL/6J/6N background. The ex vivo choroid-sprouting assay, including primary retinal pigment epithelium (RPE) and choroid, without retina was used to investigate whether FFAR4 affects choroidal angiogenesis. Western blotting for pNF-ĸB/NF-ĸB and qRT-PCR for Il-6, Il-1β, Tnf-α, Vegf, and Nf-ĸb were used to examine the influence of FFAR4 on inflammation, known to influence CNV. RPE isolated from Ffar4+/+ and Ffar4-/- mice were used to assess RPE contribution to inflammation. The FFAR4 agonist suppressed laser-induced CNV in C57BL/6J mice, and CNV increased in Ffar4-/- compared to Ffar4+/+ mice. We showed that the FFAR4 agonist acted through the FFAR4 receptor. The FFAR4 agonist suppressed mRNA expression of inflammation markers (Il-6, Il-1β) via the NF-ĸB pathway in the retina, choroid, RPE complex. The FFAR4 agonist suppressed neovascularization in the choroid-sprouting ex vivo assay and FFAR4 deficiency exacerbated sprouting. Inflammation markers were increased in primary RPE cells of Ffar4-/- mice compared with Ffar4+/+ RPE. In this mouse model, the FFAR4 agonist suppressed CNV, suggesting FFAR4 to be a new molecular target to reduce pathological angiogenesis in CNV.
Collapse
Affiliation(s)
- Yohei Tomita
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Bertan Cakir
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Chi-Hsiu Liu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Shuo Huang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Steve S Cho
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Boston, MA, 02115, USA
| | - William R Britton
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Ye Sun
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Mark Puder
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Boston, USA
| | - Ann Hellström
- Pediatric Ophthalmology, Sahlgrenska Academy, The Queen Silvia Children's Hospital, Göteborg, Sweden
| | | | - Lois E H Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Boston, MA, 02115, USA.
| |
Collapse
|
45
|
Senatorov IS, Cheshmehkani A, Burns RN, Singh K, Moniri NH. Carboxy-Terminal Phosphoregulation of the Long Splice Isoform of Free-Fatty Acid Receptor-4 Mediates β-Arrestin Recruitment and Signaling to ERK1/2. Mol Pharmacol 2020; 97:304-313. [PMID: 32132133 DOI: 10.1124/mol.119.117697] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 01/31/2020] [Indexed: 12/13/2022] Open
Abstract
Free-fatty acid receptor-4 (FFA4), previously termed GPR120, is a G protein-coupled receptor (GPCR) for medium and long-chained fatty acids, agonism of which can regulate a myriad of metabolic, sensory, inflammatory, and proliferatory signals. Two alternative splice isoforms of FFA4 exist that differ by the presence of an additional 16 amino acids in the longer (FFA4-L) transcript, which has been suggested to be an intrinsically β-arrestin-biased GPCR. Although the shorter isoform (FFA4-S) has been studied more extensively, very little is known about mechanisms of regulation or signaling of the longer isoform. Because β-arrestin recruitment is dependent on receptor phosphorylation, in the current study, we used the endogenous agonist docosahexaenoic acid (DHA) to examine the mechanisms of FFA4-L phosphorylation, as well as DHA-dependent β-arrestin recruitment and DHA-dependent extracellular-signal regulated kinase-1/2 (ERK1/2) signaling in human embryonic kidney 293 cells. Our results reveal differences in basal phosphorylation of the two FFA4 isoforms, and we show that DHA-mediated phosphorylation of FFA4-L is primarily regulated by G protein-coupled receptor kinase 6, whereas protein kinase-C can also contribute to agonist-induced and heterologous phosphorylation. Moreover, our data demonstrate that FFA4-L phosphorylation occurs on the distal C terminus and is directly responsible for recruitment and interactions with β-arrestin-2. Finally, using CRISPR/Cas9 genome-edited cells, our data reveal that unlike FFA4-S, the longer isoform is unable to facilitate phosphorylation of ERK1/2 in cells that are devoid of β-arrestin-1/2. Together, these results are the first to demonstrate phosphoregulation of FFA4-L as well as the effects of loss of phosphorylation sites on β-arrestin recruitment and ERK1/2 activation. SIGNIFICANCE STATEMENT: Free-fatty acid receptor-4 (FFA4) is a cell-surface G protein-coupled receptor for medium and long-chained fatty acids that can be expressed as distinct short (FFA4-S) or long (FFA4-L) isoforms. Although much is known about FFA4-S, the longer isoform remains virtually unstudied. Here, we reveal the mechanisms of docosahexaenoic acid-induced phosphorylation of FFA4-L and subsequent β-arrestin-2 recruitment and extracellular-signal regulated kinase-1/2 activity.
Collapse
Affiliation(s)
- Ilya S Senatorov
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University Health Sciences Center, Mercer University, Atlanta, Georgia
| | - Ameneh Cheshmehkani
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University Health Sciences Center, Mercer University, Atlanta, Georgia
| | - Rebecca N Burns
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University Health Sciences Center, Mercer University, Atlanta, Georgia
| | - Kirti Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University Health Sciences Center, Mercer University, Atlanta, Georgia
| | - Nader H Moniri
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University Health Sciences Center, Mercer University, Atlanta, Georgia
| |
Collapse
|
46
|
Zhu L, Sha L, Li K, Wang Z, Wang T, Li Y, Liu P, Dong X, Dong Y, Zhang X, Wang H. Dietary flaxseed oil rich in omega-3 suppresses severity of type 2 diabetes mellitus via anti-inflammation and modulating gut microbiota in rats. Lipids Health Dis 2020; 19:20. [PMID: 32028957 PMCID: PMC7006389 DOI: 10.1186/s12944-019-1167-4] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 12/09/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is closely associated with hyperglycemia, abnormal lipid profiles, chronic low-grade inflammation and gut dysbiosis. Dietary intervention plays a crucial role in the control of diabetes. Flaxseed oil (FO), a plant-derived omega-3 (ω-3) polyunsaturated fatty acids (PUFAs), is rich in α-linolenic acid (ALA) which has been proved to benefit for chronic metabolic disease. However, the exact effects of dietary FO on T2DM remains largely unclear. METHODS In the present study, SD rats were randomly allocated into four groups: pair-fed (PF) with corn oil (CO) group (PF/CO); DM with CO group (DM/CO); PF with FO group (PF/FO); DM with FO group (DM/FO). A diabetic rat model was generated by a single intraperitoneal injection of streptozotocin-nicotinamide (STZ-NA). After 5 weeks of intervention, rats were euthanized and associated indications were investigated. RESULTS Dietary FO significantly reduced fasting blood glucose (FBG), glycated hemoglobin (GHb), blood lipid, plasma lipopolysaccharide (LPS), interleukin (IL)-1β, tumor necrosis factor (TNF)-α, IL-6, IL-17A and malondialdehyde (MDA), compared to control group, respectively. Moreover, body mass (BM) and superoxide dismutase (SOD) in DM/FO group were dramatically increased respectively, compared with those in DM/CO group. But insulin (INS) and homeostasis model assessment of insulin resistance (HOMA-IR) remained no significant difference between DM/CO group and DM/FO group. Sequencing analysis of gut microbiota showed a reduction in the relative abundance of Firmicutes and Blautia, as well as a reduction in the ratio of Bacteroidetes-Firmicutes in DM/FO group compared to DM/CO group. An elevation in the relative abundance of Bacteroidetes and Alistipes were detected in DM/FO group. Acetic acid, propionic acid and butyric acid belonging to short chain fatty acids (SCFAs) as gut microbiota metabolites, were dramatically increased after FO intervention. Correlation analysis revealed that the relative abundance of Firmicutes and Blautia were positively correlated with IL-1β, TNF-α, IL-6, IL-17A or LPS, respectively. Additionally, Bacteroidetes and Alistipes were negatively correlated with LPS. CONCLUSIONS Taken together, dietary FO ameliorated T2DM via suppressing inflammation and modulating gut microbiota, which may potentially contribute to dietary control of diabetes.
Collapse
Affiliation(s)
- Lili Zhu
- Department of Pathogenic Biology and Medical Immunology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Liping Sha
- Endocrinology Department, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Ke Li
- Endocrinology Department, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750002, Ningxia, China
| | - Zhen Wang
- Clinical Medical College, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Ting Wang
- Department of Pathogenic Biology and Medical Immunology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yiwei Li
- Department of Pathogenic Biology and Medical Immunology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Ping Liu
- Endocrinology Department, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Xiaoying Dong
- Endocrinology Department, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Youping Dong
- Endocrinology Department, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Xiaoxia Zhang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| | - Hao Wang
- Department of Pathogenic Biology and Medical Immunology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
47
|
Pharmacological potential of novel agonists for FFAR4 on islet and enteroendocrine cell function and glucose homeostasis. Eur J Pharm Sci 2019; 142:105104. [PMID: 31669388 DOI: 10.1016/j.ejps.2019.105104] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 09/27/2019] [Accepted: 10/10/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND To investigate the metabolic effects of FFAR4-selective agonists on islet and enteroendocrine cell hormone release and the combined therapeutic effectiveness with DPP-IV inhibitors. METHODS Insulinotropic activity and specificity of FFAR4 agonists were determined in clonal pancreatic BRIN-BD11 cells. Expression of FFAR4 was assessed by qPCR and western blotting following agonist treatment in BRIN-BD11 cells and by immunohistochemistry in mouse islets. Acute in-vivo effects of agonists was investigated after intraperitoneal (i.p.) or oral administration in lean and HFF-obese diabetic mice. RESULTS GSK137647 (10-11-10-4 M) and Compound-A (10-10-10-4 M) stimulated insulin secretion at 5.6 mM (p < 0.05-p < 0.001) and 16.7 mM (p < 0.05-p < 0.001) glucose in BRIN-BD11 cells, with no cytotoxicity effects as assessed by MTT. FFAR4 antagonist (AH-7614) abolished the insulintropic effect of GSK137647 (p < 0.05-p < 0.001), whilst FFAR1 antagonist (GW1100) had no effect. Incubation of BRIN-BD11 cells with GSK137647 and Compound-A increased FFAR4 (p < 0.01) gene expression at 16.7 mM glucose, with a corresponding increase in FFAR4 (p < 0.01) protein concentrations. FFAR4 upregulation was attenuated under normoglycaemic conditions. Immunohistochemistry demonstrated co-localisation of FFAR4 and insulin in mouse islets. Orally administered GSK137647 or Compound-A (0.1 µmol/kgBW) improved glucose tolerance (p < 0.001), increased plasma insulin (p < 0.001), GLP-1 (p < 0.05), GIP (p < 0.05) and induced satiety (p < 0.001) in HFF mice, with glucose-lowering effects enhanced in combination with DPP-IV inhibitor (Sitagliptin) (p < 0.05). CONCLUSIONS Specific FFAR4 agonism improves glucose tolerance through insulin and incretin secretion, with enhanced DPP-IV inhibition in combination with Sitagliptin. GENERAL SIGNIFICANCE These findings have for the first time demonstrated that selective FFAR4 activation regulates both islet and enteroendocrine hormone function with agonist combinational therapy, presenting a promising strategy for the treatment of type-2-diabetes.
Collapse
|
48
|
Gaudet DA, El-Desoky D, Poret JM, Braymer HD, Primeaux SD. Expression of neural markers of gustatory signaling are differentially altered by continuous and intermittent feeding patterns. Physiol Behav 2019; 212:112719. [PMID: 31634524 DOI: 10.1016/j.physbeh.2019.112719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/06/2019] [Accepted: 10/17/2019] [Indexed: 12/15/2022]
Abstract
Food intake patterns are regulated by signals from the gustatory neural circuit, a complex neural network that begins at the tongue and continues to homeostatic and hedonic brain regions involved in eating behavior. The goal of the current study was to investigate the short-term effects of continuous access to a high fat diet (HFD) versus limited access to dietary fat on the gustatory neural circuit. Male Sprague-Dawley rats were fed a chow diet, a HFD (56% kcal from fat), or provided limited, daily (2 h/day) or limited, intermittent (2 h/day, 3 times/week) access to vegetable shortening for 2 weeks. Real time PCR was used to determine mRNA expression of markers of fat sensing/signaling (e.g. CD36) on the circumvallate papillae, markers of homeostatic eating in the mediobasal hypothalamus (MBH) and markers of hedonic eating in the nucleus accumbens (NAc). Continuous HFD increased mRNA levels of lingual CD36 and serotonin signaling, altered markers of homeostatic and hedonic eating. Limited, intermittent access to dietary fat selectively altered the expression of genes associated with the regulation of dopamine signaling. Overall, these data suggest that short-term, continuous access to HFD leads to altered fat taste and decreased expression of markers of homeostatic and hedonic eating. Limited, intermittent access, or binge-like, consumption of dietary fat led to an overall increase in markers of hedonic eating, without altering expression of lingual fat sensors or homeostatic eating. These data suggest that there are differential effects of meal patterns on gustatory neurocircuitry which may regulate the overconsumption of fat and lead to obesity.
Collapse
Affiliation(s)
- Darryl A Gaudet
- Department of Physiology, LSU Health Sciences Center, New Orleans, LA 70112, USA
| | - Dalia El-Desoky
- Department of Physiology, LSU Health Sciences Center, New Orleans, LA 70112, USA
| | - Jonquil M Poret
- Department of Physiology, LSU Health Sciences Center, New Orleans, LA 70112, USA
| | | | - Stefany D Primeaux
- Department of Physiology, LSU Health Sciences Center, New Orleans, LA 70112, USA; Joint Diabetes, Endocrinology & Metabolism Program, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| |
Collapse
|
49
|
Park H, He A, Lodhi IJ. Lipid Regulators of Thermogenic Fat Activation. Trends Endocrinol Metab 2019; 30:710-723. [PMID: 31422871 PMCID: PMC6779522 DOI: 10.1016/j.tem.2019.07.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 12/16/2022]
Abstract
The global prevalence of obesity continues to increase, suggesting a need for alternative treatment approaches. Targeting brown fat function to promote energy expenditure represents one such approach. Brown adipocytes and the related beige adipocytes oxidize fatty acids and glucose to generate heat and are activated by cold exposure or consumption of high-calorie diets. Alternative, more practical means to activate thermogenic fat are needed. Here, we review emerging data suggesting new roles for lipids in activating thermogenesis that extend beyond their serving as a fuel source for heat generation. Lipids have also been implicated in mediating interorgan communication, crosstalk between organelles, and cellular signaling regulating thermogenesis. Understanding how lipids regulate thermogenesis could identify innovative therapeutic interventions for obesity.
Collapse
Affiliation(s)
- Hongsuk Park
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anyuan He
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
50
|
Mäkelä AM, Hohtola E, Miinalainen IJ, Autio JA, Schmitz W, Niemi KJ, Hiltunen JK, Autio KJ. Mitochondrial 2,4-dienoyl-CoA reductase (Decr) deficiency and impairment of thermogenesis in mouse brown adipose tissue. Sci Rep 2019; 9:12038. [PMID: 31427678 PMCID: PMC6700156 DOI: 10.1038/s41598-019-48562-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 08/07/2019] [Indexed: 12/17/2022] Open
Abstract
A large number of studies have demonstrated significance of polyunsaturated fatty acids (PUFAs) for human health. However, many aspects on signals translating PUFA-sensing into body homeostasis have remained enigmatic. To shed light on PUFA physiology, we have generated a mouse line defective in mitochondrial dienoyl-CoA reductase (Decr), which is a key enzyme required for β-oxidation of PUFAs. Previously, we have shown that these mice, whose oxidation of saturated fatty acid is intact but break-down of unsaturated fatty acids is blunted, develop severe hypoglycemia during metabolic stresses and fatal hypothermia upon acute cold challenge. In the current work, indirect calorimetry and thermography suggested that cold intolerance of Decr−/− mice is due to failure in maintaining appropriate heat production at least partly due to failure of brown adipose tissue (BAT) thermogenesis. Magnetic resonance imaging, electron microscopy, mass spectrometry and biochemical analysis showed attenuation in activation of lipolysis despite of functional NE-signaling and inappropriate expression of genes contributing to thermogenesis in iBAT when the Decr−/− mice were exposed to cold. We hypothesize that the failure in turning on BAT thermogenesis occurs due to accumulation of unsaturated long-chain fatty acids or their metabolites in Decr−/− mice BAT suppressing down-stream propagation of NE-signaling.
Collapse
Affiliation(s)
- Anne M Mäkelä
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Esa Hohtola
- Department of Ecology and Genetics, University of Oulu, Oulu, Finland
| | | | - Joonas A Autio
- Center for Biosystems Dynamics Research, RIKEN, Kobe, Japan.,Medical Research Center, University of Oulu and Oulu University Hospital, Oulu, Finland
| | | | - Kalle J Niemi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - J Kalervo Hiltunen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Kaija J Autio
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
| |
Collapse
|