1
|
La H, Kim J, Kim DH, Kim SH, Singh P, Nam G, Moon K, Kim I, Kim IS. Discovery of 1,4-Disubstituted Cyclohexene Analogues as Selective GPR119 Agonists for the Treatment of Type 2 Diabetes. J Med Chem 2025; 68:4619-4634. [PMID: 39853173 DOI: 10.1021/acs.jmedchem.4c02655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2025]
Abstract
GPR119 has emerged as a promising target for treating type 2 diabetes and associated obesity, as its stimulation induces the secretion of glucagon-like peptide-1 and glucose-dependent insulinotropic peptide in the intestinal tract as well as the glucose-dependent release of insulin in pancreatic β-cells. We describe the design and synthesis of novel GPR119 agonists containing a 1,4-disubstituted cyclohexene scaffold. Compound 21b displayed nanomolar potency (EC50 = 3.8 nM) for hGPR119 activation and demonstrated a hypoglycemic efficacy of 17.0% in an oral glucose tolerance test. The hypoglycemic effect of compound 21b, compared to sitagliptin, a DPP-4 inhibitor, showed the relatively higher efficacy in both FATZO and db/db mice. Additionally, compound 21b exhibited a significant reduction in body weight in a female diet-induced obese rat model, comparable to that of metformin. Furthermore, in vivo pharmacokinetic experiments revealed that compound 21b is a potential candidate for the treatment of type 2 diabetes and obesity.
Collapse
Affiliation(s)
- Hyunhwa La
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Medicinal Chemistry Research Team, New Drug Discovery Lab., Hyundai Pharmaceutical Co. Ltd., Suwon 16229, Republic of Korea
| | - Jinwoong Kim
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, Republic of Korea
- Medicinal Chemistry Research Team, New Drug Discovery Lab., Hyundai Pharmaceutical Co. Ltd., Suwon 16229, Republic of Korea
| | - Dae-Hoon Kim
- Nonclinical Research Team, New Drug Discovery Lab., Hyundai Pharmaceutical Co. Ltd., Yongin 17089, Republic of Korea
| | - Seong-Heon Kim
- Medicinal Chemistry Research Team, New Drug Discovery Lab., Hyundai Pharmaceutical Co. Ltd., Suwon 16229, Republic of Korea
- Nonclinical Research Team, New Drug Discovery Lab., Hyundai Pharmaceutical Co. Ltd., Yongin 17089, Republic of Korea
| | - Pargat Singh
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Gibeom Nam
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Kyeongwon Moon
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ikyon Kim
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, Republic of Korea
| | - In Su Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
2
|
Ostadrahimi A, Khajebishak Y, Moradi F, Payahoo L. The effect of Oleoylethanolamide supplementation on lipid profile, fasting blood sugar and dietary habits in obese people: a randomized double-blind placebo-control trial. BMC Endocr Disord 2024; 24:210. [PMID: 39379951 PMCID: PMC11460158 DOI: 10.1186/s12902-024-01738-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/18/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Abnormalities in biochemical parameters and changes in eating habits are considered complications of obesity. Oleoylethanolamide (OEA), an endocannabinoid-like compound, has been shown to have protective effects on many metabolic disorders. Given this evidence, the present study aimed to assess the effects of OEA on lipid profile parameters, fasting blood sugar (FBS), and dietary habits in healthy obese people. METHODS In this randomized, double-blind, placebo-controlled clinical trial, which was carried out in 2016 in Tabriz, Iran, 60 obese people were enrolled in the study based on inclusion criteria. The intervention group consumed 125 mg of OEA capsules, and the placebo group received the same amount of starch twice for 8 weeks. Blood samples (5 mL) were taken at baseline and the end of the study in a fasting state. Serum concentrations of FBS, triglycerides (TGs), high-density lipoprotein cholesterol (HDL-C), and total cholesterol (TC) were measured by enzymatic methods using commercial kits. The low-density lipoprotein cholesterol (LDL-C) concentration was obtained using the Friede-Wald formula. To assess dietary habits, a food frequency questionnaire (147 items) was used at baseline and the end of the study. A value less than < 0.05 was considered to indicate statistical significance. RESULTS The TG concentration decreased significantly in the intervention group (mean (SD): 166.29 (70.01) mg/dL to 142.22 (48.05) mg/dL, p = 0.047). Changes in the placebo group were not significant (p > 0.05). After adjusting for baseline values and demographic characteristics, the difference in TG between groups remained significant (p = 0.044). Changes in other biochemical parameters were not significant. There was no significant difference between or within groups in terms of food groups. CONCLUSION OEA, as a complementary agent, plays a protective role in TG regulation. However, future studies with longer durations are needed to explore the impact of OEA on regulating dietary habits and to identify the mechanisms related to metabolic abnormalities in obese people. TRIAL REGISTRATION The study was registered in the Iranian Registry of Clinical Trials (IRCT) center as IRCT201607132017N30 with URL. www.IRCT.IR in date 03/10/2016.
Collapse
Affiliation(s)
- Alireza Ostadrahimi
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yaser Khajebishak
- Nutrition Sciences, Department of Nutrition, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Fardin Moradi
- Student Research Committee, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Laleh Payahoo
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
- Nutrition Sciences, Medicinal Plants Research Center, Maragheh University of Medical Sciences, Maragheh, Iran.
| |
Collapse
|
3
|
Engin AB, Engin A. The Checkpoints of Intestinal Fat Absorption in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:73-95. [PMID: 39287849 DOI: 10.1007/978-3-031-63657-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
In this chapter, intestinal lipid transport, which plays a central role in fat homeostasis and the development of obesity in addition to the mechanisms of fatty acids and monoacylglycerol absorption in the intestinal lumen and reassembly of these within the enterocyte was described. A part of the resynthesized triglycerides (triacylglycerols; TAG) is repackaged in the intestine to form the hydrophobic core of chylomicrons (CMs). These are delivered as metabolic fuels, essential fatty acids, and other lipid-soluble nutrients, from enterocytes to the peripheral tissues following detachment from the endoplasmic reticulum membrane. Moreover, the attitudes of multiple receptor functions in dietary lipid uptake, synthesis, and transport are highlighted. Additionally, intestinal fatty acid binding proteins (FABPs), which increase the cytosolic flux of fatty acids via intermembrane transfer in enterocytes, and the functions of checkpoints for receptor-mediated fatty acid signaling are debated. The importance of the balance between storage and secretion of dietary fat by enterocytes in determining the physiological fate of dietary fat, including regulation of blood lipid concentrations and energy balance, is mentioned. Consequently, promising checkpoints regarding how intestinal fat processing affects lipid homeostatic mechanisms and lipid stores in the body and the prevention of obesity-lipotoxicity due to excessive intestinal lipid absorption are evaluated. In this context, dietary TAG digestion, pharmacological inhibition of TAG hydrolysis, the regulation of long-chain fatty acid uptake traffic into adipocytes, intracellular TAG resynthesis, the enlargement of cytoplasmic lipid droplets in enterocytes and constitutional alteration of their proteome, CD36-mediated conversion of diet-derived fatty acid into cellular lipid messengers and their functions are discussed.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey
| |
Collapse
|
4
|
Rahman SMK, Uyama T, Hussain Z, Ueda N. Roles of Endocannabinoids and Endocannabinoid-like Molecules in Energy Homeostasis and Metabolic Regulation: A Nutritional Perspective. Annu Rev Nutr 2021; 41:177-202. [PMID: 34115519 DOI: 10.1146/annurev-nutr-043020-090216] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The endocannabinoid system is involved in signal transduction in mammals. It comprises principally G protein-coupled cannabinoid receptors and their endogenous agonists, called endocannabinoids, as well as the enzymes and transporters responsible for the metabolism of endocannabinoids. Two arachidonic acid-containing lipid molecules, arachidonoylethanolamide (anandamide) and 2-arachidonoylglycerol, function as endocannabinoids. N-acylethanolamines and monoacylglycerols, in which the arachidonic acid chain is replaced with a saturated or monounsaturated fatty acid, are not directly involved in the endocannabinoid system but exhibit agonistic activities for other receptors. These endocannabinoid-like molecules include palmitoylethanolamide, oleoylethanolamide (OEA), and 2-oleoylglycerol. Endocannabinoids stimulate feeding behavior and the anabolism of lipids and glucose, while OEA suppresses appetite. Both central and peripheral systems are included in these nutritional and metabolic contexts. Therefore, they have potential in the treatment and prevention of obesity. We outline the structure, metabolism, and biological activities of endocannabinoids and related molecules, and focus on their involvement in energy homeostasis and metabolic regulation. Expected final online publication date for the Annual Review of Nutrition, Volume 41 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- S M Khaledur Rahman
- Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa 761-0793, Japan; , , .,Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore-7408, Bangladesh
| | - Toru Uyama
- Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa 761-0793, Japan; , ,
| | - Zahir Hussain
- Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa 761-0793, Japan; , , .,Department of Pharmaceutical Sciences, School of Pharmacy, Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA;
| | - Natsuo Ueda
- Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa 761-0793, Japan; , ,
| |
Collapse
|
5
|
Klockars A, Levine AS, Head MA, Perez-Leighton CE, Kotz CM, Olszewski PK. Impact of Gut and Metabolic Hormones on Feeding Reward. Compr Physiol 2021; 11:1425-1447. [PMID: 33577129 DOI: 10.1002/cphy.c190042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Ingestion of food activates a cascade of endocrine responses (thereby reflecting a contemporaneous feeding status) that include the release of hormones from the gastrointestinal (GI) tract, such as cholecystokinin (CCK), glucagonlike peptide YY (PYY), peptide PP, and oleoylethanolamide, as well as suppression of ghrelin secretion. The pancreas and adipose tissue, on the other hand, release hormones that serve as a measure of the current metabolic state or the long-term energy stores, that is, insulin, leptin, and adiponectin. It is well known and intuitively understandable that these hormones target either directly (by crossing the blood-brain barrier) or indirectly (e.g., via vagal input) the "homeostatic" brainstem-hypothalamic pathways involved in the regulation of appetite. The current article focuses on yet another target of the metabolic and GI hormones that is critical in inducing changes in food intake, namely, the reward system. We discuss the physiological basis of this functional interaction, its importance in the control of appetite, and the impact that disruption of this crosstalk has on energy intake in select physiological and pathophysiological states. We conclude that metabolic and GI hormones have a capacity to strengthen or weaken a response of the reward system to a given food, and thus, they are fundamental in ensuring that feeding reward is plastic and dependent on the energy status of the organism. © 2021 American Physiological Society. Compr Physiol 11:1425-1447, 2021.
Collapse
Affiliation(s)
- Anica Klockars
- Faculty of Science and Engineering, University of Waikato, Hamilton, New Zealand
| | - Allen S Levine
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, Minnesota, USA
| | - Mitchell A Head
- Faculty of Science and Engineering, University of Waikato, Hamilton, New Zealand
| | | | - Catherine M Kotz
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, Minnesota, USA.,Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Pawel K Olszewski
- Faculty of Science and Engineering, University of Waikato, Hamilton, New Zealand.,Department of Food Science and Nutrition, University of Minnesota, St. Paul, Minnesota, USA.,Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
6
|
Tutunchi H, Naeini F, Saghafi-Asl M, Farrin N, Monshikarimi A, Ostadrahimi A. Effects of oleoylethanolamide supplementation on atherogenic indices and hematological parameters in patients with nonalcoholic fatty liver disease: A clinical trial. Health Promot Perspect 2020; 10:373-382. [PMID: 33312933 PMCID: PMC7722997 DOI: 10.34172/hpp.2020.56] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 10/10/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Non-alcoholic fatty liver disease (NAFLD) is the most frequent cause of chronic liver disease in the world. The current interventional trial aimed to evaluate the effects of supplementation with oleoylethanolamide (OEA) in combination with weight loss intervention on some atherogenic indices as well as hematological parameters in patients newly diagnosed with NAFLD. Methods: In this triple-blinded, randomized, placebo-controlled clinical trial, 76 obese patients with NAFLD confirmed by ultra-sonographic findings were randomly assigned to receive a weight reduction diet plus either 250 mg OEA (n=38) or placebo (n=38) for 12 weeks. Atherogenic factors including total cholesterol/high-density lipoprotein cholesterol (HDL-C),low-density lipoprotein cholesterol (LDL-C)/HDL-C, triglyceride (TG)/HDL-C, non-HDL-C/HDL-C ratios and non-HDL-C level, as well as hematological parameters were assessed before and after intervention. Results : After adjustment for potential confounding factors, between group analyses demonstrated a significantly lower LDL-C/HDL-C, TG/HDL-C, and non-HDL-C/HDL-C ratios in the OEA group compared to the placebo, post-intervention (95% confidence interval [CI]:0.06 to 0.85, P = 0.024; 95% CI: -2.06 to -0.05, P = 0.039; 95% CI: -1.05 to -0.02, P = 0.042,respectively). Additionally, OEA supplementation could significantly decrease the levels of red blood cell distribution width (RDW) compared to the placebo at the endpoint after considering potential confounding variables (95% CI: -0.56 to -0.003, P = 0.041). No significant differences were found between the two study groups in terms of other hematological parameters. Conclusion: The results of the current study indicated that OEA supplementation had beneficial effects on LDL-C/HDL-C, TG/HDL-C, and non-HDL-C/HDL-C ratios as well as RDW in obese patients with NAFLD. Trial Registration: IRCT20110530006652N2; https://www.irct.ir/trial/37228.
Collapse
Affiliation(s)
- Helda Tutunchi
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Naeini
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Saghafi-Asl
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Farrin
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Monshikarimi
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Ostadrahimi
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
7
|
Tutunchi H, Ostadrahimi A, Saghafi-Asl M, Hosseinzadeh-Attar MJ, Shakeri A, Asghari-Jafarabadi M, Roshanravan N, Farrin N, Naemi M, Hasankhani M. Oleoylethanolamide supplementation in obese patients newly diagnosed with non-alcoholic fatty liver disease: Effects on metabolic parameters, anthropometric indices, and expression of PPAR-α, UCP1, and UCP2 genes. Pharmacol Res 2020; 156:104770. [PMID: 32217148 DOI: 10.1016/j.phrs.2020.104770] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 12/18/2022]
Abstract
The effects of oleoylethanolamide (OEA) on NAFLD are yet to be examined in human. The objective of the present study was to examine the effects of OEA supplementation along with weight loss intervention on the expression of PPAR-α, uncoupling proteins 1and 2 (UCP1 and UCP2) genes in the peripheral blood mononuclear cells (PBMCs), metabolic parameters, and anthropometric indices among obese patients with NAFLD. In this triple-blind placebo-controlled randomized clinical trial, 76 obese patients newly diagnosed with NAFLD were randomly allocated into either OEA or placebo group along with calorie-restricted diets for 12 weeks. At pre-and post-intervention phase, mRNA expression levels of PPAR-α, UCP1, and UCP2 genes in the PBMCs, serum levels of metabolic parameters as well as diet and appetite sensations were assessed. There was a significant increase in the expression levels of PPAR-α, UCP1, and UCP2 genes in the PBMCs, compared to the placebo at the endpoint. A significant decrease in the anthropometric indices, energy and carbohydrate intakes, glycemic parameters, except for hemoglobin A1c concentration was also observed in the OEA group, compared to the placebo group. OEA treatment significantly resulted in decreased serum levels of triglyceride (TG), alanine aminotransferase (ALT), aspartate aminotransferase (AST), ALT/AST, increased serum levels of high-density lipoprotein cholesterol (HDL-C), and improved appetite sensations. Importantly, a significant improvement in TG, ALT, AST, ALT/AST, HDL-C levels as well as appetite sensations by OEA were under the influence of body mass index (BMI). Although liver steatosis severity was significantly reduced in both groups, the between-group differences did not reach statistical significance (P = 0.061). In conclusion, the present study, for the first time, revealed that OEA supplementation significantly improved anthropometric and metabolic risk factors related to NAFLD.
Collapse
Affiliation(s)
- Helda Tutunchi
- Student Research Committee, Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Alireza Ostadrahimi
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Maryam Saghafi-Asl
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad-Javad Hosseinzadeh-Attar
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran.
| | - Abolhasan Shakeri
- Department of Radiology, Imam Reza Teaching Hospital, Clinical Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Neda Roshanravan
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Nazila Farrin
- Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Naemi
- Student Research Committee, Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Hasankhani
- Student Research Committee, Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Tutunchi H, Saghafi-Asl M, Ostadrahimi A. A systematic review of the effects of oleoylethanolamide, a high-affinity endogenous ligand of PPAR-α, on the management and prevention of obesity. Clin Exp Pharmacol Physiol 2020; 47:543-552. [PMID: 31868943 DOI: 10.1111/1440-1681.13238] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 11/23/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023]
Abstract
Along with an increase in overweight and obesity among all age groups, the development of efficacious and safe anti-obesity strategies for patients, as well as health systems, is critical. Oleoylethanolamide (OEA), a high-affinity endogenous ligand of nuclear receptor peroxisome proliferator-activated receptor alpha (PPAR-α), plays important physiological and metabolic actions. OEA is derived from oleic acid, a monounsaturated fatty acid, which has beneficial effects on body composition and regional fat distribution. The role of OEA in the modulation of food consumption and weight management makes it an attractive molecule requiring further exploration in obesogenic environments. This systematic review was conducted to assess the effects of OEA on the obesity management, with emphasizing on its physiological roles and possible mechanisms of action in energy homeostasis. We searched PubMed/Medline, Google Scholar, ScienceDirect, Scopus, ProQuest, and EMBASE up until September 2019. Out of 712 records screened, 30 articles met the study criteria. The evidence reviewed here indicates that OEA, an endocannabinoid-like compound, leads to satiation or meal termination through PPAR-α activation and fatty acid translocase (FAT)/CD36. Additionally, the lipid-amide OEA stimulates fatty acid uptake, lipolysis, and beta-oxidation, and also promotes food intake control. OEA also exerts satiety-inducing effects by activating the hedonic dopamine pathways and increasing homeostatic oxytocin and brain histamine. In conclusion, OEA may be a key component of the physiological system involved in the regulation of dietary fat consumption and energy homeostasis; therefore, it is suggested as a possible therapeutic agent for the management of obesity.
Collapse
Affiliation(s)
- Helda Tutunchi
- Student Research Committee, Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Saghafi-Asl
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Ostadrahimi
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
9
|
De Luca L, Ferracane R, Vitaglione P. Food database of N-acyl-phosphatidylethanolamines, N-acylethanolamines and endocannabinoids and daily intake from a Western, a Mediterranean and a vegetarian diet. Food Chem 2019; 300:125218. [PMID: 31351254 DOI: 10.1016/j.foodchem.2019.125218] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 07/09/2019] [Accepted: 07/20/2019] [Indexed: 12/14/2022]
Abstract
The contents of N-acylphosphatidylethanolamines (NAPEs), N-acylethanolamines (NAEs) and endocannabinoids (ECs) in 43 food products were assessed and daily intakes, based on consumption of Mediterranean, vegetarian and Western diets, were simulated. NAPEs and NAEs were more abundant in plant-based foods than in animal food products; NAPEs were in the ranges 0-4032 vs 4-398 µg/g dw and NAEs were in the ranges 0-35 vs 0.1-0.7 µg/g dw, respectively while ECs were in the range 0-0.1 vs 0-34 µg/g dw. Daily intakes of NAPEs and NAEs were higher from Mediterranean (263 and 0.25 mg/day) and vegetarian (242 and 0.28 mg/day) diets than the Western diet (163 and 0.08 mg/day). Conversely, ECs intakes were higher from Western and Mediterranean diets (0.17 mg/day) than the vegetarian diet (0.01 mg/day). Future studies will evaluate the physiological role of dietary NAPEs, NAEs and ECs in humans.
Collapse
Affiliation(s)
- Lucia De Luca
- Department of Agricultural Sciences, University of Naples "Federico II", 80055 Portici, Italy
| | - Rosalia Ferracane
- Department of Agricultural Sciences, University of Naples "Federico II", 80055 Portici, Italy
| | - Paola Vitaglione
- Department of Agricultural Sciences, University of Naples "Federico II", 80055 Portici, Italy.
| |
Collapse
|
10
|
Tutunchi H, Ostadrahimi A, Saghafi-Asl M, Maleki V. The effects of oleoylethanolamide, an endogenous PPAR-α agonist, on risk factors for NAFLD: A systematic review. Obes Rev 2019; 20:1057-1069. [PMID: 31111657 DOI: 10.1111/obr.12853] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/04/2019] [Accepted: 03/04/2019] [Indexed: 12/15/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease. Recently, some novel compounds have been investigated for the prevention and treatment of NAFLD. Oleoylethanolamide (OEA), an endogenous PPAR-α agonist, has exhibited a plethora of pharmacological properties for the treatment of obesity and other obesity-associated metabolic complications. This systematic review was performed with a focus on the effects of OEA on the risk factors for NAFLD. PubMed, Scopus, Embase, ProQuest, and Google Scholar databases were searched up to December 2018 using relevant keywords. All articles written in English evaluating the effects of OEA on the risk factors for NAFLD were eligible for the review. The evidence reviewed in this article illustrates that OEA regulates multiple biological processes associated with NAFLD, including lipid metabolism, inflammation, oxidative stress, and energy homeostasis through different mechanisms. In summary, many beneficial effects of OEA have led to the understanding that OEA may be an effective therapeutic strategy for the management of NAFLD. Although a wide range of studies have demonstrated the most useful effects of OEA on NAFLD and the associated risk factors, further clinical trials, from both in vivo studies and in vitro experiments, are warranted to verify these outcomes.
Collapse
Affiliation(s)
- Helda Tutunchi
- Student Research Committee, Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Ostadrahimi
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Saghafi-Asl
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Maleki
- Student Research Committee, Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
11
|
Fedele S, Arnold M, Krieger JP, Wolfstädter B, Meyer U, Langhans W, Mansouri A. Oleoylethanolamide-induced anorexia in rats is associated with locomotor impairment. Physiol Rep 2019; 6. [PMID: 29388342 PMCID: PMC5817840 DOI: 10.14814/phy2.13517] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 11/01/2017] [Indexed: 11/24/2022] Open
Abstract
The endogenous peroxisome proliferator‐activated receptor alpha (PPAR‐α) agonist Oleoylethanolamide (OEA) inhibits eating in rodents, mainly by delaying the onset of meals. The underlying mechanisms of OEA‐induced anorexia, however, remain unclear. Animals treated with high OEA doses were shown to display signs of discomfort and impaired locomotion. Therefore, we first examined whether the impaired locomotion may contribute to OEA's anorectic effect. Second, it is controversial whether abdominal vagal afferents are necessary for OEA's anorectic effect. Thus, we explored alternative peripheral neural pathways mediating IP OEA's anorectic effect by performing a celiac‐superior mesenteric ganglionectomy (CGX) or a subdiaphragmatic vagal deafferentation (SDA) alone or in combination. Exogenously administered OEA at a commonly used dose (10 mg/kg BW, IP) concurrently reduced food intake and compromised locomotor activity. Attempts to dissociate both phenomena using the dopamine D2/D3 receptor agonist Quinpirole (1 mg/kg BW, SC) failed because Quinpirole antagonized both, OEA‐induced locomotor impairment and delay in eating onset. CGX attenuated the prolongation of the latency to eat by IP OEA, but neither SDA nor CGX prevented IP OEA‐induced locomotor impairment. Our results indicate that IP OEA's anorectic effect may be secondary to impaired locomotion rather than due to physiological satiety. They further confirm that vagal afferents do not mediate exogenous OEA's anorectic effects, but suggest a role for spinal afferents in addition to an alternative, nonneuronal signaling route.
Collapse
Affiliation(s)
- Shahana Fedele
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Myrtha Arnold
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | | | - Bernd Wolfstädter
- Laboratorium für Organische Chemie, ETH Zurich, Zürich, Switzerland.,Laboratory of Translational Nutrition Biology, ETH Zurich, Schwerzenbach, Switzerland
| | - Urs Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Abdelhak Mansouri
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| |
Collapse
|
12
|
Pan X, Schwartz GJ, Hussain MM. Oleoylethanolamide differentially regulates glycerolipid synthesis and lipoprotein secretion in intestine and liver. J Lipid Res 2018; 59:2349-2359. [PMID: 30369486 DOI: 10.1194/jlr.m089250] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/26/2018] [Indexed: 01/13/2023] Open
Abstract
Dietary fat absorption takes place in the intestine, and the liver mobilizes endogenous fat to other tissues by synthesizing lipoproteins that require apoB and microsomal triglyceride transfer protein (MTP). Dietary fat triggers the synthesis of oleoylethanolamide (OEA), a regulatory fatty acid that signals satiety to reduce food intake mainly by enhancing neural PPARα activity, in enterocytes. We explored OEA's roles in the assembly of lipoproteins in WT and Ppara -/- mouse enterocytes and hepatocytes, Caco-2 cells, and human liver-derived cells. In differentiated Caco-2 cells, OEA increased synthesis and secretion of triacylglycerols, apoB secretion in chylomicrons, and MTP expression in a dose-dependent manner. OEA also increased MTP activity and triacylglycerol secretion in WT and knockout primary enterocytes. In contrast to its intestinal cell effects, OEA reduced synthesis and secretion of triacylglycerols, apoB secretion, and MTP expression and activity in human hepatoma Huh-7 and HepG2 cells. Also, OEA reduced MTP expression and triacylglycerol secretion in WT, but not knockout, primary hepatocytes. These studies indicate differential effects of OEA on lipid synthesis and lipoprotein assembly: in enterocytes, OEA augments glycerolipid synthesis and lipoprotein assembly independent of PPARα. Conversely, in hepatocytes, OEA reduces MTP expression, glycerolipid synthesis, and lipoprotein secretion through PPARα-dependent mechanisms.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY .,Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, NY
| | - Gary J Schwartz
- Departments of Medicine and Neuroscience, Albert Einstein College of Medicine, Bronx, NY
| | - M Mahmood Hussain
- Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY .,Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, NY.,Veterans Affairs New York Harbor Healthcare System, Brooklyn, NY
| |
Collapse
|
13
|
Brown JD, McAnally D, Ayala JE, Burmeister MA, Morfa C, Smith L, Ayala JE. Oleoylethanolamide modulates glucagon-like peptide-1 receptor agonist signaling and enhances exendin-4-mediated weight loss in obese mice. Am J Physiol Regul Integr Comp Physiol 2018; 315:R595-R608. [PMID: 29949410 PMCID: PMC6230892 DOI: 10.1152/ajpregu.00459.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 06/20/2018] [Accepted: 06/20/2018] [Indexed: 02/08/2023]
Abstract
Long-acting glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) agonists (GLP-1RA), such as exendin-4 (Ex4), promote weight loss. On the basis of a newly discovered interaction between GLP-1 and oleoylethanolamide (OEA), we tested whether OEA enhances GLP-1RA-mediated anorectic signaling and weight loss. We analyzed the effect of GLP-1+OEA and Ex4+OEA on canonical GLP-1R signaling and other proteins/pathways that contribute to the hypophagic action of GLP-1RA (AMPK, Akt, mTOR, and glycolysis). We demonstrate that OEA enhances canonical GLP-1R signaling when combined with GLP-1 but not with Ex4. GLP-1 and Ex4 promote phosphorylation of mTOR pathway components, but OEA does not enhance this effect. OEA synergistically enhanced GLP-1- and Ex4-stimulated glycolysis but did not augment the hypophagic action of GLP-1 or Ex4 in lean or diet-induced obese (DIO) mice. However, the combination of Ex4+OEA promoted greater weight loss in DIO mice than Ex4 or OEA alone during a 7-day treatment. This was due in part to transient hypophagia and increased energy expenditure, phenotypes also observed in Ex4-treated DIO mice. Thus, OEA augments specific GLP-1RA-stimulated signaling but appears to work in parallel with Ex4 to promote weight loss in DIO mice. Elucidating cooperative mechanisms underlying Ex4+OEA-mediated weight loss could, therefore, be leveraged toward more effective obesity therapies.
Collapse
Affiliation(s)
- Jacob D Brown
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona , Orlando, Florida
| | - Danielle McAnally
- Cardiovascular Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona , Orlando, Florida
| | - Jennifer E Ayala
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona , Orlando, Florida
| | - Melissa A Burmeister
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona , Orlando, Florida
| | - Camilo Morfa
- Cardiovascular Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona , Orlando, Florida
| | - Layton Smith
- Cardiovascular Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona , Orlando, Florida
| | - Julio E Ayala
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona , Orlando, Florida
| |
Collapse
|
14
|
Laleh P, Yaser K, Abolfazl B, Shahriar A, Mohammad AJ, Nazila F, Alireza O. Oleoylethanolamide increases the expression of PPAR-Α and reduces appetite and body weight in obese people: A clinical trial. Appetite 2018; 128:44-49. [PMID: 29787831 DOI: 10.1016/j.appet.2018.05.129] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/16/2018] [Accepted: 05/16/2018] [Indexed: 12/11/2022]
Abstract
Obesity is a crucial public health problem worldwide and is considered as the main cause of many chronic diseases. The present study evaluated the effects of Oleoylethanolamide (OEA) supplementation on proximal proliferator-activated receptor-α (PPAR-α) gene expression, appetite sensations, and anthropometric measurements in obese people. This randomized, double-blind, placebo-controlled clinical trial was carried out on 60 healthy obese people in Tabriz, Iran, in 2016. The eligible subjects were divided into an intervention group (who received two 125 mg OEA capsules daily) and a placebo group (who received the same amount of starches) and treated for 60 days. Anthropometric measurements and body composition were assessed in a fasting state at baseline and at the end of the study. The visual analogue scales (VAS) were used to assess appetite sensations. Quantitative real-time PCR analysis targeting the 16S rRNA gene of PPAR-α was done. Analysis was done on 56 participants who continued intervention until the end of the study. A significant increase in PPAR-α gene expression was observed in the intervention group (p < 0.001). Weight, body mass index, waist circumference, and fat percent decreased significantly at the end of the study in the intervention group (all p < 0.01). Hunger, the desire to eat, and cravings for sweet foods decreased significantly and fullness increased significantly by the end of study in the intervention group at the end of study (all p < 0.01). The fullness item increased significantly by the end of study in the intervention group (p < 0.001). Use of OEA as a complementary approach could be effective in suppressing appetite and modulating energy balance in obese people.
Collapse
Affiliation(s)
- Payahoo Laleh
- Talented Student Center, Student Research Committee, Nutrition Research Center, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khajebishak Yaser
- Student Research Committee, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Barzegari Abolfazl
- Student Research Committee, School of Advanced Biomedical Sciences, Tabriz University of Medical Science, Tabriz, Iran
| | - Alipour Shahriar
- Connective Tissue Disease Research Center, Department of Molecular Medicine, Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asghari Jafarabadi Mohammad
- Road Traffic Injury Research Center, Department of Statistics and Epidemiology, Faculty of Health, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farrin Nazila
- Nutrition Research Center, Faculty of Nutrition and Food Science, Department of Nutrition, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ostadrahimi Alireza
- Nutrition Research Center, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
15
|
Cifarelli V, Abumrad NA. Intestinal CD36 and Other Key Proteins of Lipid Utilization: Role in Absorption and Gut Homeostasis. Compr Physiol 2018; 8:493-507. [PMID: 29687890 PMCID: PMC6247794 DOI: 10.1002/cphy.c170026] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Several proteins have been implicated in fatty acid (FA) transport by enterocytes including the scavenger receptor CD36 (SR-B2), the scavenger receptor B1 (SR-B1) a member of the CD36 family and the FA transport protein 4 (FATP4). Here, we review the regulation of enterocyte FA uptake and its function in lipid absorption including prechylomicron formation, assembly and transport. Emphasis is given to CD36, which is abundantly expressed along the digestive tract of rodents and humans and has been the most studied. We also address the pleiotropic functions of CD36 that go beyond lipid absorption and metabolism to include recent evidence of its impact on intestinal homeostasis and barrier maintenance. Areas of progress involving contribution of membrane phospholipid remodeling and of cytosolic FA-binding proteins, FABP1 and FABP2 to fat absorption will be covered. © 2018 American Physiological Society. Compr Physiol 8:493-507, 2018.
Collapse
Affiliation(s)
- Vincenza Cifarelli
- Department of Internal Medicine, Center for Human Nutrition, Washington University School of Medicine, St Louis, Missouri, USA
| | - Nada A. Abumrad
- Department of Internal Medicine, Center for Human Nutrition, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
16
|
Sihag J, Jones PJH. Oleoylethanolamide: The role of a bioactive lipid amide in modulating eating behaviour. Obes Rev 2018; 19:178-197. [PMID: 29124885 DOI: 10.1111/obr.12630] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/19/2017] [Accepted: 09/19/2017] [Indexed: 12/13/2022]
Abstract
Fatty acid ethanolamides are lipid mediators that regulate a plethora of physiological functions. One such bioactive lipid mediator, oleoylethanolamide (OEA), is a potent agonist of the peroxisome proliferator-activated receptor-alpha (PPAR-α), which modulates increased expression of the fatty acid translocase CD36 that enables the regulation of feeding behaviour. Consumption of dietary fat rich in oleic acid activates taste receptors in the gut activating specific enzymes that lead to the formation of OEA. OEA further combines with PPAR-α to enable fat oxidation in the liver, resulting in enhanced energy production. Evidence suggests that sustained ingestion of a high-fat diet abolishes the anorexic signal of OEA. Additionally, malfunction of the enterocyte that transforms oleic acid produced during fat digestion into OEA might be responsible for reduced satiety and hyperphagia, resulting in overweight and obesity. Thus, OEA anorectic signalling may be an essential element of the physiology and metabolic system regulating dietary fat intake and obesity. The evidence reviewed in this article indicates that intake of oleic acid, and thereby the resulting OEA imparting anorexic properties, is dependent on CD36, PPAR-α, enterocyte fat sensory receptors, histamine, oxytocin and dopamine; leading to increased fat oxidation and enhanced energy expenditure to induce satiety and increase feeding latency; and that a disruption in any of these systems will cease/curb fat-induced satiety.
Collapse
Affiliation(s)
- J Sihag
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, Winnipeg, Manitoba, Canada
| | - P J H Jones
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
17
|
Abstract
Hypothalamic integration of gastrointestinal and adipose tissue-derived hormones serves as a key element of neuroendocrine control of food intake. Leptin, adiponectin, oleoylethanolamide, cholecystokinin, and ghrelin, to name a few, are in a constant "cross talk" with the feeding-related brain circuits that encompass hypothalamic populations synthesizing anorexigens (melanocortins, CART, oxytocin) and orexigens (Agouti-related protein, neuropeptide Y, orexins). While this integrated neuroendocrine circuit successfully ensures that enough energy is acquired, it does not seem to be equally efficient in preventing excessive energy intake, especially in the obesogenic environment in which highly caloric and palatable food is constantly available. The current review presents an overview of intricate mechanisms underlying hypothalamic integration of energy balance-related peripheral endocrine input. We discuss vulnerabilities and maladaptive neuroregulatory processes, including changes in hypothalamic neuronal plasticity that propel overeating despite negative consequences.
Collapse
|
18
|
Demenis C, McLaughlin J, Smith CP. Sulfated Cholecystokinin-8 Promotes CD36-Mediated Fatty Acid Uptake into Primary Mouse Duodenal Enterocytes. Front Physiol 2017; 8:660. [PMID: 28919867 PMCID: PMC5586203 DOI: 10.3389/fphys.2017.00660] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/18/2017] [Indexed: 11/13/2022] Open
Abstract
Cholecystokinin (CCK) is an archetypal incretin hormone secreted by intestinal enteroendocrine cells (EEC) in response to ingested nutrients. The aim of this study was to determine whether CCK modulates enterocyte fatty acid uptake by primary mouse duodenal cells. Exposure of primary mouse duodenal cells to 10 pM sulfated CCK-8 caused a two fold increase in dodecanoic acid fatty acid (FA) uptake. The selective CCK A receptor antagonist loxiglumide (100 μM) completely abolished the CCK-8 induced FA uptake. The CD36 fatty acid translocase-specific inhibitor sulfo-N-succinimidyl oleate (1 μM) also completely inhibited CCK-8 induced FA uptake, as did treatment with 200 μM phloretin. Together these data show CCK induces FA uptake into duodenal enterocytes; this action involves the CCK-RA receptor and is carrier mediated by CD36.
Collapse
Affiliation(s)
- Claire Demenis
- School of Medical Sciences, University of ManchesterManchester, United Kingdom
| | - John McLaughlin
- School of Medical Sciences, University of ManchesterManchester, United Kingdom
| | - Craig P Smith
- School of Medical Sciences, University of ManchesterManchester, United Kingdom
| |
Collapse
|
19
|
Liu X, Kris-Etherton PM, West SG, Lamarche B, Jenkins DJA, Fleming JA, McCrea CE, Pu S, Couture P, Connelly PW, Jones PJH. Effects of canola and high-oleic-acid canola oils on abdominal fat mass in individuals with central obesity. Obesity (Silver Spring) 2016; 24:2261-2268. [PMID: 27804268 PMCID: PMC5119743 DOI: 10.1002/oby.21584] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 05/09/2016] [Accepted: 05/17/2016] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To determine the effect of diets low in saturated fatty acids and high in monounsaturated fatty acids (MUFA) or polyunsaturated fatty acids on body composition in participants at risk for metabolic syndrome (MetS). METHODS This study was a randomized, crossover, controlled feeding study. Participants (n = 101, ages 49.5 ± 1.2, BMI 29.4 ± 0.4 kg/m2 ) were randomized to five isocaloric diets containing treatment oils: Canola, CanolaOleic, CanolaDHA, Corn/Safflower, and Flax/Safflower. Each diet period was 4 weeks followed by a 2- to 4-week washout period. RESULTS Canola (3.1 kg, P = 0.026) and CanolaOleic oil diets (3.09 kg, P = 0.03) reduced android fat mass compared with the Flax/Saff oil diet (3.2 kg), particularly in men. The decrease in abdominal fat mass was correlated with the reduction in blood pressure after the Canola (systolic blood pressure: r = 0.26, P = 0.062; diastolic blood pressure: r = 0.38, P = 0.0049) and CanolaOleic oil diets (systolic blood pressure: r = 0.39 P = 0.004; diastolic blood pressure: r = 0.45, P = 0.0006). The decrease in abdominal fat mass also was associated with a reduction in triglyceride levels after the CanolaOleic oil diet (r = 0.42, P = 0.002). CONCLUSIONS Diets high in MUFA (compared with PUFA) reduced central obesity with an accompanying improvement in MetS risk factors. Diets high in MUFA may be beneficial for treating and perhaps preventing MetS.
Collapse
Affiliation(s)
- Xiaoran Liu
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Penny M Kris-Etherton
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA.
| | - Sheila G West
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Benoît Lamarche
- Institute of Nutrition and Functional Foods, Laval University, Quebec City, Quebec, Canada
| | - David J A Jenkins
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer A Fleming
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Cindy E McCrea
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Shuaihua Pu
- Richardson Center for Functional Foods and Nutraceuticals, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Patrick Couture
- Institute of Nutrition and Functional Foods, Laval University, Quebec City, Quebec, Canada
| | - Philip W Connelly
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Peter J H Jones
- Richardson Center for Functional Foods and Nutraceuticals, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
20
|
Stochastic sensors designed for assessment of biomarkers specific to obesity. J Pharm Biomed Anal 2016; 128:280-285. [PMID: 27288757 DOI: 10.1016/j.jpba.2016.05.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 05/22/2016] [Accepted: 05/23/2016] [Indexed: 12/15/2022]
Abstract
Two stochastic sensors based on the following oleamides: 1-adamantyloleamide and N,N-dimethyl-N-(2-oleylamidoethyl)amine physically immobilized on graphite paste were designed. The sensors were able to determine simultaneously from the whole blood of Wistar rats three biomarkers specific to obesity: leptin, interleukin-6 (IL-6) and plasminogen activator inhibitor 1 (PAI-1). The whole blood samples were obtained from Wistar rats treated with oleoylethanolamide (OEA), (Z)-N-[(1S)-2-hidroxy-1-(phenylmethyl) ethyl]-9octadecenamide (OLA), and with the aqueous solution of 1% Tween 80 used as solvent for oleamides formulations (control samples). The proposed sensors were very sensitive and reliable for the assay of obesity biomarkers in whole blood of rats.
Collapse
|
21
|
The satiety factor oleoylethanolamide impacts hepatic lipid and glucose metabolism in goldfish. J Comp Physiol B 2016; 186:1009-1021. [DOI: 10.1007/s00360-016-1009-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/25/2016] [Accepted: 06/03/2016] [Indexed: 12/14/2022]
|
22
|
Abstract
Fat is a vital macronutrient, and its intake is closely monitored by an array of molecular sensors distributed throughout the alimentary canal. In the mouth, dietary fat constituents such as mono- and diunsaturated fatty acids give rise to taste signals that stimulate food intake, in part by enhancing the production of lipid-derived endocannabinoid messengers in the gut. As fat-containing chyme enters the small intestine, it causes the formation of anorexic lipid mediators, such as oleoylethanolamide, which promote satiety. These anatomically and functionally distinct responses may contribute to the homeostatic control and, possibly, the pathological dysregulation of food intake.
Collapse
Affiliation(s)
| | - Daniele Piomelli
- Departments of Anatomy and Neurobiology
- Department of Pharmacology, and
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, California, USA
- Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| |
Collapse
|
23
|
Role of G protein-coupled orphan receptors in intestinal inflammation: novel targets in inflammatory bowel diseases. Inflamm Bowel Dis 2015; 21:666-73. [PMID: 25461621 DOI: 10.1097/mib.0000000000000258] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A large number of proteins were classified into the family of G protein-coupled receptors (GPCRs). Based on their characteristic serpentine domain, they are called 7 TM receptors. Presently, their ligands and physiological functions remain unknown. In this review, we summarize what is known on these receptors and discuss the potential use of these orphan GPCRs (GPRs) in the induction or maintenance of remission in inflammatory bowel diseases. We focus on GPRs 30, 41, 43, 55, 119, and 120, where scientific evidence supports a potential role in intestinal inflammation.
Collapse
|
24
|
Karimian Azari E, Ramachandran D, Weibel S, Arnold M, Romano A, Gaetani S, Langhans W, Mansouri A. Vagal afferents are not necessary for the satiety effect of the gut lipid messenger oleoylethanolamide. Am J Physiol Regul Integr Comp Physiol 2014; 307:R167-78. [DOI: 10.1152/ajpregu.00067.2014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The endogenous lipid messenger oleoylethanolamide (OEA) inhibits eating and modulates fat metabolism supposedly through the activation of peroxisome proliferator-activated receptor-α (PPARα) and vagal sensory fibers. We tested in adult male rats whether OEA stimulates fatty acid oxidation (FAO) and ketogenesis and whether it increases plasma levels of the satiating gut peptides glucagon-like peptide-1 (GLP-1) and peptide YY (PYY). We also explored whether OEA still inhibits eating after subdiaphragmatic vagal deafferentation (SDA). We found that intraperitoneally injected OEA (10 mg/kg body wt) reduced ( P < 0.05) food intake mainly by increasing meal latency and that this effect was stronger in rats fed a 60% high-fat diet (HFD) than in chow-fed rats. OEA increased ( P < 0.05) postprandial plasma nonesterified fatty acids and β-hydroxybutyrate (BHB) in the hepatic portal vein (HPV) and vena cava (VC) 30 min after injection, which was more pronounced in HFD- than in chow-fed rats. OEA also increased the protein expression of the key ketogenetic enzyme, mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase, in the jejunum of HFD-fed rats, but not in the liver or duodenum of either diet group. Furthermore, OEA decreased GLP-1 and PYY concentrations ( P < 0.05) in the HPV and VC 30 min after administration. Finally, OEA reduced food intake in SDA and sham-operated rats similarly. Our findings indicate that neither intact abdominal vagal afferents nor prandial increases in GLP-1 or PYY are necessary for the satiety effect of OEA. The enhanced FAO and ketogenesis raise the possibility of an involvement of intestine-derived BHB in OEA's satiety effect under certain conditions.
Collapse
Affiliation(s)
| | - Deepti Ramachandran
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland; and
| | - Sandra Weibel
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland; and
| | - Myrtha Arnold
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland; and
| | - Adele Romano
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Silvana Gaetani
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland; and
| | - Abdelhak Mansouri
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland; and
| |
Collapse
|
25
|
Hansen HS. Role of anorectic N-acylethanolamines in intestinal physiology and satiety control with respect to dietary fat. Pharmacol Res 2014; 86:18-25. [PMID: 24681513 DOI: 10.1016/j.phrs.2014.03.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 03/17/2014] [Accepted: 03/18/2014] [Indexed: 02/06/2023]
Abstract
Anandamide is a well-known agonist for the cannabinoid receptors. Along with endogenous anandamide other non-endocannabinoid N-acylethanolamines are also formed, apparently in higher amounts. These include mainly oleoylethanolamide (OEA), palmitoyelethanolamide (PEA) and linoleoylethanolamide (LEA), and they have biological activity by themselves being anorectic and anti-inflammatory. It appears that the major effect of dietary fat on the level of these molecules is in the gastrointestinal system, where OEA, PEA and LEA in the enterocytes may function as homeostatic signals, which are decreased by prolonged consumption of a high-fat diet. These lipid amides appear to mediate their signaling activity via activation of PPARα in the enterocyte followed by activation of afferent vagal fibers leading to the brain. Through this mechanism OEA, PEA and LEA may both reduce the consumption of a meal as well as increase the reward value of the food. Thus, they may function as homeostatic intestinal signals involving hedonic aspects that contribute to the regulation of the amounts of dietary fat to be ingested.
Collapse
Affiliation(s)
- Harald S Hansen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| |
Collapse
|
26
|
Fan A, Wu X, Wu H, Li L, Huang R, Zhu Y, Qiu Y, Fu J, Ren J, Zhu C. Atheroprotective effect of oleoylethanolamide (OEA) targeting oxidized LDL. PLoS One 2014; 9:e85337. [PMID: 24465540 PMCID: PMC3896367 DOI: 10.1371/journal.pone.0085337] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 12/04/2013] [Indexed: 01/11/2023] Open
Abstract
Dietary fat-derived lipid oleoylethanolamide (OEA) has shown to modulate lipid metabolism through a peroxisome proliferator-activated receptor-alpha (PPAR-α)-mediated mechanism. In our study, we further demonstrated that OEA, as an atheroprotective agent, modulated the atherosclerotic plaques development. In vitro studies showed that OEA antagonized oxidized LDL (ox-LDL)-induced vascular endothelial cell proliferation and vascular smooth muscle cell migration, and suppressed lipopolysaccharide (LPS)-induced LDL modification and inflammation. In vivo studies, atherosclerosis animals were established using balloon-aortic denudation (BAD) rats and ApoE(-/-) mice fed with high-caloric diet (HCD) for 17 or 14 weeks respectively, and atherosclerotic plaques were evaluated by oil red staining. The administration of OEA (5 mg/kg/day, intraperitoneal injection, i.p.) prevented or attenuated the formation of atherosclerotic plaques in HCD-BAD rats or HCD-ApoE(-/-) mice. Gene expression analysis of vessel tissues from these animals showed that OEA induced the mRNA expressions of PPAR-α and downregulated the expression of M-CFS, an atherosclerotic marker, and genes involved in oxidation and inflammation, including iNOS, COX-2, TNF-α and IL-6. Collectively, our results suggested that OEA exerted a pharmacological effect on modulating atherosclerotic plaque formation through the inhibition of LDL modification in vascular system and therefore be a potential candidate for anti-atherosclerosis drug.
Collapse
Affiliation(s)
- Angran Fan
- Department of Medical Science, Medical College, Xiamen University, Xiamen, Fujian, China
| | - Xiaofeng Wu
- Department of Medical Science, Medical College, Xiamen University, Xiamen, Fujian, China
| | - Huijuan Wu
- Department of Medical Science, Medical College, Xiamen University, Xiamen, Fujian, China
| | - Long Li
- Department of Medical Science, Medical College, Xiamen University, Xiamen, Fujian, China
| | - Rui Huang
- Department of Medical Science, Medical College, Xiamen University, Xiamen, Fujian, China
| | - Yueyong Zhu
- Division of Liver Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yan Qiu
- Department of Medical Science, Medical College, Xiamen University, Xiamen, Fujian, China
| | - Jin Fu
- Department of Medical Science, Medical College, Xiamen University, Xiamen, Fujian, China
| | - Jie Ren
- Department of Medical Science, Medical College, Xiamen University, Xiamen, Fujian, China
| | - Chenggang Zhu
- School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
27
|
Abstract
The absorptive epithelium of the proximal small intestine converts oleic acid released during fat digestion into oleoylethanolamide (OEA), an endogenous high-affinity agonist of peroxisome proliferator-activated receptor-α (PPAR-α). OEA interacts with this receptor to cause a state of satiety characterized by prolonged inter-meal intervals and reduced feeding frequency. The two main branches of the autonomic nervous system, sympathetic and parasympathetic, contribute to this effect: the former by enabling OEA mobilization in the gut and the latter by relaying the OEA signal to brain structures, such as the hypothalamus, that are involved in feeding regulation. OEA signaling may be a key component of the physiological system devoted to the monitoring of dietary fat intake, and its dysfunction might contribute to overweight and obesity.
Collapse
Affiliation(s)
- Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California, Irvine, CA 92612, USA.
| |
Collapse
|
28
|
Naughton SS, Mathai ML, Hryciw DH, McAinch AJ. Fatty Acid modulation of the endocannabinoid system and the effect on food intake and metabolism. Int J Endocrinol 2013; 2013:361895. [PMID: 23762050 PMCID: PMC3677644 DOI: 10.1155/2013/361895] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 04/25/2013] [Accepted: 05/07/2013] [Indexed: 01/26/2023] Open
Abstract
Endocannabinoids and their G-protein coupled receptors (GPCR) are a current research focus in the area of obesity due to the system's role in food intake and glucose and lipid metabolism. Importantly, overweight and obese individuals often have higher circulating levels of the arachidonic acid-derived endocannabinoids anandamide (AEA) and 2-arachidonoyl glycerol (2-AG) and an altered pattern of receptor expression. Consequently, this leads to an increase in orexigenic stimuli, changes in fatty acid synthesis, insulin sensitivity, and glucose utilisation, with preferential energy storage in adipose tissue. As endocannabinoids are products of dietary fats, modification of dietary intake may modulate their levels, with eicosapentaenoic and docosahexaenoic acid based endocannabinoids being able to displace arachidonic acid from cell membranes, reducing AEA and 2-AG production. Similarly, oleoyl ethanolamide, a product of oleic acid, induces satiety, decreases circulating fatty acid concentrations, increases the capacity for β -oxidation, and is capable of inhibiting the action of AEA and 2-AG in adipose tissue. Thus, understanding how dietary fats alter endocannabinoid system activity is a pertinent area of research due to public health messages promoting a shift towards plant-derived fats, which are rich sources of AEA and 2-AG precursor fatty acids, possibly encouraging excessive energy intake and weight gain.
Collapse
Affiliation(s)
- Shaan S. Naughton
- Biomedical and Lifestyle Diseases Unit, College of Health and Biomedicine, Victoria University, P.O. Box 14428, Melbourne, VIC 8001, Australia
| | - Michael L. Mathai
- Biomedical and Lifestyle Diseases Unit, College of Health and Biomedicine, Victoria University, P.O. Box 14428, Melbourne, VIC 8001, Australia
- Florey Neuroscience Institutes, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Deanne H. Hryciw
- Department of Physiology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Andrew J. McAinch
- Biomedical and Lifestyle Diseases Unit, College of Health and Biomedicine, Victoria University, P.O. Box 14428, Melbourne, VIC 8001, Australia
- *Andrew J. McAinch:
| |
Collapse
|
29
|
Abstract
Intestinal lipid transport plays a central role in fat homeostasis. Here we review the pathways regulating intestinal absorption and delivery of dietary and biliary lipid substrates, principally long-chain fatty acid, cholesterol, and other sterols. We discuss the regulation and functions of CD36 in fatty acid absorption, NPC1L1 in cholesterol absorption, as well as other lipid transporters including FATP4 and SRB1. We discuss the pathways of intestinal sterol efflux via ABCG5/G8 and ABCA1 as well as the role of the small intestine in high-density lipoprotein (HDL) biogenesis and reverse cholesterol transport. We review the pathways and genetic regulation of chylomicron assembly, the role of dominant restriction points such as microsomal triglyceride transfer protein and apolipoprotein B, and the role of CD36, l-FABP, and other proteins in formation of the prechylomicron complex. We will summarize current concepts of regulated lipoprotein secretion (including HDL and chylomicron pathways) and include lessons learned from families with genetic mutations in dominant pathways (i.e., abetalipoproteinemia, chylomicron retention disease, and familial hypobetalipoproteinemia). Finally, we will provide an integrative view of intestinal lipid homeostasis through recent findings on the role of lipid flux and fatty acid signaling via diverse receptor pathways in regulating absorption and production of satiety factors.
Collapse
Affiliation(s)
- Nada A Abumrad
- Center for Human Nutrition and Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | |
Collapse
|
30
|
Naville D, Duchampt A, Vigier M, Oursel D, Lessire R, Poirier H, Niot I, Bégeot M, Besnard P, Mithieux G. Link between intestinal CD36 ligand binding and satiety induced by a high protein diet in mice. PLoS One 2012; 7:e30686. [PMID: 22295104 PMCID: PMC3266275 DOI: 10.1371/journal.pone.0030686] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 12/21/2011] [Indexed: 01/09/2023] Open
Abstract
CD36 is a ubiquitous membrane glycoprotein that binds long-chain fatty acids. The presence of a functional CD36 is required for the induction of satiety by a lipid load and its role as a lipid receptor driving cellular signal has recently been demonstrated. Our project aimed to further explore the role of intestinal CD36 in the regulation of food intake. Duodenal infusions of vehicle or sulfo-N-succinimidyl-oleate (SSO) was performed prior to acute infusions of saline or Intralipid (IL) in mice. Infusion of minute quantities of IL induced a decrease in food intake (FI) compared to saline. Infusion of SSO had the same effect but no additive inhibitory effect was observed in presence of IL. No IL- or SSO-mediated satiety occurred in CD36-null mice. To determine whether the CD36-mediated hypophagic effect of lipids was maintained in animals fed a satietogen diet, mice were subjected to a High-Protein diet (HPD). Concomitantly with the satiety effect, a rise in intestinal CD36 gene expression was observed. No satiety effect occurred in CD36-null mice. HPD-fed WT mice showed a diminished FI compared to control mice, after saline duodenal infusion. But there was no further decrease after lipid infusion. The lipid-induced decrease in FI observed on control mice was accompanied by a rise in jejunal oleylethanolamide (OEA). Its level was higher in HPD-fed mice than in controls after saline infusion and was not changed by lipids. Overall, we demonstrate that lipid binding to intestinal CD36 is sufficient to produce a satiety effect. Moreover, it could participate in the satiety effect induced by HPD. Intestine can modulate FI by several mechanisms including an increase in OEA production and CD36 gene expression. Furthermore, intestine of mice adapted to HPD have a diminished capacity to modulate their food intake in response to dietary lipids.
Collapse
|
31
|
Abstract
Obesity will continue to be one of the leading causes of chronic disease unless the ongoing rise in the prevalence of this condition is reversed. Accumulating morbidity figures and a shortage of effective drugs have generated substantial research activity with several molecular targets being investigated. However, pharmacological modulation of body weight is extremely complex, since it is essentially a battle against one of the strongest human instincts and highly efficient mechanisms of energy uptake and storage. This review provides an overview of the different molecular strategies intended to lower body weight or adipose tissue mass. Weight-loss drugs in development include molecules intended to reduce the absorption of lipids from the GI tract, various ways to limit food intake, and compounds that increase energy expenditure or reduce adipose tissue size. A number of new preparations, including combinations of the existing drugs topiramate plus phentermine, bupropion plus naltrexone, and the selective 5-HT(2C) agonist lorcaserin have recently been filed for approval. Behind these leading candidates are several other potentially promising compounds and combinations currently undergoing phase II and III testing. Some interesting targets further on the horizon are also discussed.
Collapse
Affiliation(s)
- Renger F Witkamp
- Division of Human Nutrition, Wageningen University, P.O. Box 8129, 6700 EV Wageningen, The Netherlands.
| |
Collapse
|
32
|
Thabuis C, Destaillats F, Lambert DM, Muccioli GG, Maillot M, Harach T, Tissot-Favre D, Martin JC. Lipid transport function is the main target of oral oleoylethanolamide to reduce adiposity in high-fat-fed mice. J Lipid Res 2011; 52:1373-82. [PMID: 21515921 DOI: 10.1194/jlr.m013391] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We evaluated the biological basis of reduced fat gain by oleoylethanolamide (OEA) in high-fat-fed mice and sought to determine how degradation of OEA affected its efficiency by comparing its effects to those of KDS-5104, a nonhydrolyzable lipid OEA analog. Mice were given OEA or KDS-5104 by the oral route (100 mg/kg body weight). Sixty-eight variables per mouse, describing six biological processes (lipid transport, lipogenesis, energy intake, energy expenditure, endocannabinoid signaling, and glucose metabolism), spanning gene expression of biochemical and physiological parameters were examined to determine the primary target whereby OEA reduces fat gain. Although KDS-5104 but not OEA was resistant to fatty acid amide hydrolase hydrolysis, OEA was degraded by an unidentified hydrolysis system in the liver. Nevertheless, both compounds equally decreased body fat pads after 5 weeks (20%; P < 0.05). The six biological functions constructed from the 68 initial variables predicted up to 58% of adipose fat variations. Lipid transport appeared central to the explanation for body fat deposition (16%; P < 0.0001), in which decreased expression of the FAT/CD36 gene was the component most related to adipose depots. Lipid transport appears to be a determinant player in the OEA fat-lowering response, with adipose tissue FAT/CD36 expression being the most relevant bioindicator of OEA action.
Collapse
Affiliation(s)
- Clémentine Thabuis
- INRA (Institut National de la Recherche Agronomique), UMR1260 Nutriments Lipidiques et Préventiondes Maladies Métaboliques, Marseille, France
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Dipasquale P, Romano A, Cianci S, Righetti L, Gaetani S. Oleoylethanolamide: a new player in energy metabolism control. Role in food intake. ACTA ACUST UNITED AC 2010. [DOI: 10.1016/j.ddmec.2011.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
34
|
Pavón FJ, Serrano A, Romero-Cuevas M, Alonso M, Rodríguez de Fonseca F. Oleoylethanolamide: a new player in peripheral control of energy metabolism. Therapeutic implications. ACTA ACUST UNITED AC 2010. [DOI: 10.1016/j.ddmec.2011.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
35
|
Covasa M. Deficits in gastrointestinal responses controlling food intake and body weight. Am J Physiol Regul Integr Comp Physiol 2010; 299:R1423-39. [PMID: 20861277 DOI: 10.1152/ajpregu.00126.2010] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The gastrointestinal tract serves as a portal sensing incoming nutrients and relays mechanical and chemosensory signals of a meal to higher brain centers. Prolonged consumption of dietary fat causes adaptive changes within the alimentary, metabolic, and humoral systems that promote a more efficient process for energy metabolism from this rich source, leading to storage of energy in the form of adipose tissue. Furthermore, prolonged ingestion of dietary fats exerts profound effects on responses to signals involved in termination of a meal. This article reviews the effects of ingested fat on gastrointestinal motility, hormone release, and neuronal substrates. It focuses on changes in sensitivity to satiation signals resulting from chronic ingestion of high-fat diet, which may lead to disordered appetite and dysregulation of body weight.
Collapse
Affiliation(s)
- Mihai Covasa
- L'Institute National de la Recherche Agronomique, Centre de Recherche, Microbiologie de l'Alimentation au service de la Santé Humaine (MICALIS), Neurobiology of Ingestive Behavior, Jouy-en-Josas, France.
| |
Collapse
|
36
|
The fat-induced satiety factor oleoylethanolamide suppresses feeding through central release of oxytocin. J Neurosci 2010. [PMID: 20554860 DOI: 10.1523/jneur osci.0036-10.2010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Oleoylethanolamide (OEA) is a biologically active lipid amide that is released by small-intestinal enterocytes during the absorption of dietary fat and inhibits feeding by engaging the nuclear receptor, peroxisome proliferator-activated receptor-alpha (PPAR-alpha). Previous studies have shown that the anorexic effects of systemically administered OEA require the activation of sensory afferents of the vagus nerve. The central circuits involved in mediating OEA-induced hypophagia remain unknown. In the present study, we report the results of in situ hybridization and immunohistochemistry experiments in rats and mice, which show that systemic injections of OEA (5-10 mg kg(-1), intraperitoneal) enhance expression of the neuropeptide oxytocin in magnocellular neurons of the paraventricular nucleus (PVN) and supraoptic nucleus (SON) of the hypothalamus. No such effect is observed with other hypothalamic neuropeptides, including vasopressin, thyrotropin-releasing hormone and pro-opiomelanocortin. The increase in oxytocin expression elicited by OEA was absent in mutant PPAR-alpha-null mice. Pharmacological blockade of oxytocin receptors in the brain by intracerebroventricular infusion of the selective oxytocin antagonist, L-368,899, prevented the anorexic effects of OEA. The results suggest that OEA suppresses feeding by activating central oxytocin transmission.
Collapse
|
37
|
Gaetani S, Fu J, Cassano T, Dipasquale P, Romano A, Righetti L, Cianci S, Laconca L, Giannini E, Scaccianoce S, Mairesse J, Cuomo V, Piomelli D. The fat-induced satiety factor oleoylethanolamide suppresses feeding through central release of oxytocin. J Neurosci 2010; 30:8096-101. [PMID: 20554860 PMCID: PMC2900249 DOI: 10.1523/jneurosci.0036-10.2010] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Revised: 04/15/2010] [Accepted: 04/20/2010] [Indexed: 11/21/2022] Open
Abstract
Oleoylethanolamide (OEA) is a biologically active lipid amide that is released by small-intestinal enterocytes during the absorption of dietary fat and inhibits feeding by engaging the nuclear receptor, peroxisome proliferator-activated receptor-alpha (PPAR-alpha). Previous studies have shown that the anorexic effects of systemically administered OEA require the activation of sensory afferents of the vagus nerve. The central circuits involved in mediating OEA-induced hypophagia remain unknown. In the present study, we report the results of in situ hybridization and immunohistochemistry experiments in rats and mice, which show that systemic injections of OEA (5-10 mg kg(-1), intraperitoneal) enhance expression of the neuropeptide oxytocin in magnocellular neurons of the paraventricular nucleus (PVN) and supraoptic nucleus (SON) of the hypothalamus. No such effect is observed with other hypothalamic neuropeptides, including vasopressin, thyrotropin-releasing hormone and pro-opiomelanocortin. The increase in oxytocin expression elicited by OEA was absent in mutant PPAR-alpha-null mice. Pharmacological blockade of oxytocin receptors in the brain by intracerebroventricular infusion of the selective oxytocin antagonist, L-368,899, prevented the anorexic effects of OEA. The results suggest that OEA suppresses feeding by activating central oxytocin transmission.
Collapse
Affiliation(s)
- Silvana Gaetani
- Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, 00185 Rome, Italy
| | - Jin Fu
- Department of Pharmacology, University of California, Irvine, Irvine, California 92697-4625
- Unit of Drug Discovery and Development, Italian Institute of Technology, 16163 Genoa, Italy, and
| | - Tommaso Cassano
- Department of Biomedical Sciences, University of Foggia, 71100 Foggia, Italy
| | - Pasqua Dipasquale
- Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, 00185 Rome, Italy
| | - Adele Romano
- Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, 00185 Rome, Italy
| | - Laura Righetti
- Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, 00185 Rome, Italy
| | - Silvia Cianci
- Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, 00185 Rome, Italy
| | - Leonardo Laconca
- Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, 00185 Rome, Italy
- Department of Biomedical Sciences, University of Foggia, 71100 Foggia, Italy
| | - Elisa Giannini
- Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, 00185 Rome, Italy
| | - Sergio Scaccianoce
- Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, 00185 Rome, Italy
| | - Jérôme Mairesse
- Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, 00185 Rome, Italy
| | - Vincenzo Cuomo
- Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, 00185 Rome, Italy
| | - Daniele Piomelli
- Department of Pharmacology, University of California, Irvine, Irvine, California 92697-4625
- Unit of Drug Discovery and Development, Italian Institute of Technology, 16163 Genoa, Italy, and
| |
Collapse
|
38
|
André A, Gonthier MP. The endocannabinoid system: its roles in energy balance and potential as a target for obesity treatment. Int J Biochem Cell Biol 2010; 42:1788-801. [PMID: 20541029 DOI: 10.1016/j.biocel.2010.06.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Revised: 05/28/2010] [Accepted: 06/02/2010] [Indexed: 01/28/2023]
Abstract
Obesity and cardiometabolic risk continue to be major public health concerns. A better understanding of the physiopathological mechanisms leading to obesity may help to identify novel therapeutic targets. The endocannabinoid system discovered in the early 1990s is believed to influence body weight regulation and cardiometabolic risk factors. This article aims to review the literature on the endocannabinoid system including the biological roles of its major components, namely, the cannabinoid receptors, their endogenous ligands the endocannabinoids and the ligand-metabolising enzymes. The review also discusses evidence that the endocannabinoid system constitutes a new physiological pathway occurring in the central nervous system and peripheral tissues that has a key role in the control of food intake and energy expenditure, insulin sensitivity, as well as glucose and lipid metabolism. Based on the important finding that there is a close association between obesity and the hyperactivity of the endocannabinoid system, interest in blocking stimulation of this pathway to aid weight loss and reduce cardiometabolic risk factor development has become an important area of research. Among the pharmacological strategies proposed, the antagonism of the cannabinoid receptors has been particularly investigated and several clinical trials have been conducted. One challenging pharmacological task will be to target the endocannabinoid system in a more selective, and hence, safe way. As the management of obesity also requires lifestyle modifications in terms of healthy eating and physical activity, the targeting of the endocannabinoid system may represent a novel approach for a multifactorial therapeutic strategy.
Collapse
Affiliation(s)
- Aurore André
- Groupe d'Etude sur l'Inflammation Chronique et l'Obésité, Laboratoire de Biochimie et Génétique Moléculaire, Université de La Réunion, La Réunion, France
| | | |
Collapse
|
39
|
Izzo AA, Sharkey KA. Cannabinoids and the gut: new developments and emerging concepts. Pharmacol Ther 2010; 126:21-38. [PMID: 20117132 DOI: 10.1016/j.pharmthera.2009.12.005] [Citation(s) in RCA: 309] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 12/24/2009] [Indexed: 12/11/2022]
Abstract
Cannabis has been used to treat gastrointestinal (GI) conditions that range from enteric infections and inflammatory conditions to disorders of motility, emesis and abdominal pain. The mechanistic basis of these treatments emerged after the discovery of Delta(9)-tetrahydrocannabinol as the major constituent of Cannabis. Further progress was made when the receptors for Delta(9)-tetrahydrocannabinol were identified as part of an endocannabinoid system, that consists of specific cannabinoid receptors, endogenous ligands and their biosynthetic and degradative enzymes. Anatomical, physiological and pharmacological studies have shown that the endocannabinoid system is widely distributed throughout the gut, with regional variation and organ-specific actions. It is involved in the regulation of food intake, nausea and emesis, gastric secretion and gastroprotection, GI motility, ion transport, visceral sensation, intestinal inflammation and cell proliferation in the gut. Cellular targets have been defined that include the enteric nervous system, epithelial and immune cells. Molecular targets of the endocannabinoid system include, in addition to the cannabinoid receptors, transient receptor potential vanilloid 1 receptors, peroxisome proliferator-activated receptor alpha receptors and the orphan G-protein coupled receptors, GPR55 and GPR119. Pharmacological agents that act on these targets have been shown in preclinical models to have therapeutic potential. Here, we discuss cannabinoid receptors and their localization in the gut, the proteins involved in endocannabinoid synthesis and degradation and the presence of endocannabinoids in the gut in health and disease. We focus on the pharmacological actions of cannabinoids in relation to GI disorders, highlighting recent data on genetic mutations in the endocannabinoid system in GI disease.
Collapse
Affiliation(s)
- Angelo A Izzo
- Department of Experimental Pharmacology, University of Naples Federico II and Endocannabinoid Research Group, Naples, Italy.
| | | |
Collapse
|
40
|
Izzo AA, Piscitelli F, Capasso R, Marini P, Cristino L, Petrosino S, Di Marzo V. Basal and fasting/refeeding-regulated tissue levels of endogenous PPAR-alpha ligands in Zucker rats. Obesity (Silver Spring) 2010; 18:55-62. [PMID: 19521349 DOI: 10.1038/oby.2009.186] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
N-oleoylethanolamine (OEA) and N-palmitoylethanolamine (PEA) are endogenous lipids that activate peroxisome proliferator-activated receptor-alpha with high and intermediate potency, and exert anorectic and anti-inflammatory actions in rats, respectively. We investigated OEA and PEA tissue level regulation by the nutritional status in lean and obese rats. OEA and PEA levels in the brainstem, duodenum, liver, pancreas, and visceral (VAT) or subcutaneous (SAT) adipose tissues of 7-week-old wild-type (WT) and Zucker rats, fed ad libitum or following overnight food deprivation, with and without refeeding, were measured by liquid chromatography-mass spectrometry. In WT rats, duodenal OEA, but not PEA, levels were reduced by food deprivation and restored by refeeding, whereas the opposite was observed for OEA in the pancreas, and for both mediators in the liver and SAT. In ad lib fed Zucker rats, PEA and OEA levels were up to tenfold higher in the duodenum, slightly higher in the brainstem, and lower in the other tissues. Fasting/refeeding-induced changes in OEA levels were maintained in the duodenum, liver, and SAT, and lost in the pancreas, whereas fasting upregulated this compound also in the VAT. The observed changes in OEA levels in WT rats are relevant to the actions of this mediator on satiety, hepatic and adipocyte metabolism, and insulin release. OEA dysregulation in Zucker rats might counteract hyperphagia in the duodenum, but contribute to hyperinsulinemia in the pancreas, and to fat accumulation in adipose tissues and liver. Changes in PEA levels might be relevant to the inflammatory state of Zucker rats.
Collapse
Affiliation(s)
- Angelo A Izzo
- Department of Experimental Pharmacology, University of Naples Federico II, Naples, Italy
| | | | | | | | | | | | | |
Collapse
|
41
|
Guijarro A, Fu J, Astarita G, Piomelli D. CD36 gene deletion decreases oleoylethanolamide levels in small intestine of free-feeding mice. Pharmacol Res 2009; 61:27-33. [PMID: 19778614 DOI: 10.1016/j.phrs.2009.09.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Revised: 09/11/2009] [Accepted: 09/12/2009] [Indexed: 02/07/2023]
Abstract
Oleoylethanolamide (OEA) is an endogenous lipid mediator that decreases food intake and enhances lipid catabolism. Dietary fat stimulates OEA mobilization in the proximal small intestine, through a mechanism that requires the participation of the membrane glycoprotein CD36 (fatty acid translocase, FAT). CD36 is highly expressed in small-intestinal enterocytes and is involved in fatty acid uptake and intracellular signaling. Here, we analyze the impact of genetic CD36 deletion on OEA production in various mouse tissues under free-feeding conditions and at different times of the light/dark cycle. CD36 ablation decreases OEA levels in jejunum and plasma during the dark phase, when mice consume most of their daily food. CD36 deletion is also associated with reduced OEA levels in kidney, but not in other tissues including duodenum, stomach, adrenals, white and brown fat, heart, liver, pancreas, skeletal muscle and brain. The results underscore the important role of CD36 in jejunal OEA production linked to feeding.
Collapse
Affiliation(s)
- Ana Guijarro
- Department of Pharmacology, University of California, Irvine, CA 92697-4625, USA
| | | | | | | |
Collapse
|
42
|
Campolongo P, Roozendaal B, Trezza V, Cuomo V, Astarita G, Fu J, McGaugh JL, Piomelli D. Fat-induced satiety factor oleoylethanolamide enhances memory consolidation. Proc Natl Acad Sci U S A 2009; 106:8027-31. [PMID: 19416833 PMCID: PMC2683095 DOI: 10.1073/pnas.0903038106] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Indexed: 01/08/2023] Open
Abstract
The ability to remember contexts associated with aversive and rewarding experiences provides a clear adaptive advantage to animals foraging in the wild. The present experiments investigated whether hormonal signals released during feeding might enhance memory of recently experienced contextual information. Oleoylethanolamide (OEA) is an endogenous lipid mediator that is released when dietary fat enters the small intestine. OEA mediates fat-induced satiety by engaging type-alpha peroxisome proliferator-activated receptors (PPAR-alpha) in the gut and recruiting local afferents of the vagus nerve. Here we show that post-training administration of OEA in rats improves retention in the inhibitory avoidance and Morris water maze tasks. These effects are blocked by infusions of lidocaine into the nucleus tractus solitarii (NTS) and by propranolol infused into the basolateral complex of the amygdala (BLA). These findings suggest that the memory-enhancing signal generated by OEA activates the brain via afferent autonomic fibers and stimulates noradrenergic transmission in the BLA. The actions of OEA are mimicked by PPAR-alpha agonists and abolished in mutant mice lacking PPAR-alpha. The results indicate that OEA, acting as a PPAR-alpha agonist, facilitates memory consolidation through noradrenergic activation of the BLA, a mechanism that is also critically involved in memory enhancement induced by emotional arousal.
Collapse
Affiliation(s)
- Patrizia Campolongo
- Department of Pharmacology, University of California, Irvine, CA 92697-4625
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA 92657-3800
- Department of Physiology and Pharmacology, University of Rome ‘La Sapienza’, Piazzale Aldo Moro 5, 00185 Roma, Italy; and
| | - Benno Roozendaal
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA 92657-3800
| | - Viviana Trezza
- Department of Physiology and Pharmacology, University of Rome ‘La Sapienza’, Piazzale Aldo Moro 5, 00185 Roma, Italy; and
| | - Vincenzo Cuomo
- Department of Physiology and Pharmacology, University of Rome ‘La Sapienza’, Piazzale Aldo Moro 5, 00185 Roma, Italy; and
| | - Giuseppe Astarita
- Department of Pharmacology, University of California, Irvine, CA 92697-4625
| | - Jin Fu
- Department of Pharmacology, University of California, Irvine, CA 92697-4625
| | - James L. McGaugh
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA 92657-3800
| | - Daniele Piomelli
- Department of Pharmacology, University of California, Irvine, CA 92697-4625
- Drug Discovery and Development, Italian Institute of Technology, Via Morego 30, 16163 Genoa, Italy
| |
Collapse
|
43
|
Cluny NL, Keenan CM, Lutz B, Piomelli D, Sharkey KA. The identification of peroxisome proliferator-activated receptor alpha-independent effects of oleoylethanolamide on intestinal transit in mice. Neurogastroenterol Motil 2009; 21:420-9. [PMID: 19140957 DOI: 10.1111/j.1365-2982.2008.01248.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oleoylethanolamide (OEA) is an endogenous lipid produced in the intestine that mediates satiety by activation of peroxisome proliferator-activated receptor alpha (PPARalpha). OEA inhibits gastric emptying and intestinal motility, but the mechanism of action remains to be determined. We investigated whether OEA inhibits intestinal motility by activation of PPARalpha. PPARalpha immunoreactivity was examined in whole mount preparations of mouse gastrointestinal (GI) tract. The effect of OEA on motility was assessed in wildtype, PPARalpha, cannabinoid CB(1) receptor and CB(2) receptor gene-deficient mice and in a model of accelerated GI transit. In addition, the effect of OEA on motility was assessed in mice injected with the PPARalpha antagonist GW6471, transient receptor potential vanilloid 1 antagonist SB366791 or the glucagon-like peptide 1 antagonist exendin-3(9-39) amide. PPARalpha immunoreactivity was present in neurons in the myenteric and submucosal plexuses throughout the GI tract. OEA inhibited upper GI transit in a dose-dependent manner, but was devoid of an effect on whole gut transit or colonic propulsion. OEA-induced inhibition of motility was still present in PPARalpha, CB(1) and CB(2) receptor gene-deficient mice and in the presence of GW6471, SB366791 and exendin-3(9-39) amide, suggesting neither PPARalpha nor the cannabinoids and other likely receptors are involved in mediating the effects of OEA. OEA blocked stress-induced accelerated upper GI transit at a dose that had no effect on physiological transit. We show that PPARalpha is found in the enteric nervous system, but our results suggest that PPARalpha is not involved in the suppression of motility by OEA.
Collapse
Affiliation(s)
- N L Cluny
- Department of Physiology and Biophysics, Hotchkiss Brain Institute and Snyder Institute of Infection, Immunity and Inflammation, University of Calgary, Calgary, AB, Canada
| | | | | | | | | |
Collapse
|
44
|
Borrelli F, Izzo AA. Role of acylethanolamides in the gastrointestinal tract with special reference to food intake and energy balance. Best Pract Res Clin Endocrinol Metab 2009; 23:33-49. [PMID: 19285259 DOI: 10.1016/j.beem.2008.10.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Acylethanolamides (AEs) are a group of lipids occurring in both plants and animals. The best-studied AEs are the endocannabinoid anandamide (AEA), the anti-inflammatory compound palmitoylethanolamide (PEA), and the potent anorexigenic molecule oleoylethanolamide (OEA). AEs are biosynthesized in the gastrointestinal tract, and their levels may change in response to noxious stimuli, food deprivation or diet-induced obesity. The biological actions of AEs within the gut are not limited to the modulation of food intake and energy balance. For example, AEs exert potential beneficial effects in the regulation of intestinal motility, secretion, inflammation and cellular proliferation. Molecular targets of AEs, which have been identified in the gastrointestinal tract, include cannabinoid CB(1) and CB(2) receptors (activated by AEA), transient receptor potential vanilloid type 1 (TRPV1, activated by AEA and OEA), the nuclear receptor peroxisome proliferators-activated receptor-alpha (PPAR-alpha, activated by OEA and, to a less extent, by PEA), and the orphan G-coupled receptors GPR119 (activated by OEA) and GPR55 (activated by PEA and, with lower potency, by AEA and OEA). Modulation of AE levels in the gut may provide new pharmacological strategies not only for the treatment of feeding disorders but also for the prevention or cure of widespread intestinal diseases such as inflammatory bowel disease and colon cancer.
Collapse
Affiliation(s)
- Francesca Borrelli
- Department of Experimental Pharmacology, University of Naples Federico II and Endocannabinoid Research Group, via D. Montesano 49, 80131 Naples, Italy
| | | |
Collapse
|
45
|
Schwartz GJ, Fu J, Astarita G, Li X, Gaetani S, Campolongo P, Cuomo V, Piomelli D. The lipid messenger OEA links dietary fat intake to satiety. Cell Metab 2008; 8:281-288. [PMID: 18840358 PMCID: PMC2572640 DOI: 10.1016/j.cmet.2008.08.005] [Citation(s) in RCA: 277] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2008] [Revised: 06/27/2008] [Accepted: 08/01/2008] [Indexed: 11/27/2022]
Abstract
The association between fat consumption and obesity underscores the need to identify physiological signals that control fat intake. Previous studies have shown that feeding stimulates small-intestinal mucosal cells to produce the lipid messenger oleoylethanolamide (OEA) which, when administered as a drug, decreases meal frequency by engaging peroxisome proliferator-activated receptors-alpha (PPAR-alpha). Here, we report that duodenal infusion of fat stimulates OEA mobilization in the proximal small intestine, whereas infusion of protein or carbohydrate does not. OEA production utilizes dietary oleic acid as a substrate and is disrupted in mutant mice lacking the membrane fatty-acid transporter CD36. Targeted disruption of CD36 or PPAR-alpha abrogates the satiety response induced by fat. The results suggest that activation of small-intestinal OEA mobilization, enabled by CD36-mediated uptake of dietary oleic acid, serves as a molecular sensor linking fat ingestion to satiety.
Collapse
Affiliation(s)
- Gary J. Schwartz
- Diabetes Research Center, Departments of Medicine and Neuroscience, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY
| | - Jin Fu
- Department of Pharmacology, University of California, Irvine, California
| | - Giuseppe Astarita
- Department of Pharmacology, University of California, Irvine, California
| | - Xiaosong Li
- Diabetes Research Center, Departments of Medicine and Neuroscience, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY
| | - Silvana Gaetani
- Department of Human Physiology and Pharmacology, University of Rome ‘La Sapienza’, Rome, Italy
| | - Patrizia Campolongo
- Department of Human Physiology and Pharmacology, University of Rome ‘La Sapienza’, Rome, Italy
| | - Vincenzo Cuomo
- Department of Human Physiology and Pharmacology, University of Rome ‘La Sapienza’, Rome, Italy
| | - Daniele Piomelli
- Department of Pharmacology, University of California, Irvine, California
- Unit of Drug Discovery and Development, Italian Institute of Technology, Genoa, Italy
| |
Collapse
|
46
|
Capasso R, Izzo AA. Gastrointestinal regulation of food intake: general aspects and focus on anandamide and oleoylethanolamide. J Neuroendocrinol 2008; 20 Suppl 1:39-46. [PMID: 18426498 DOI: 10.1111/j.1365-2826.2008.01686.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The gastrointestinal tract plays a pivotal role in the regulation of food intake and energy balance. Signals from the gastrointestinal tract generally function to limit ingestion in the interest of efficient digestion. These signals may be released into the bloodstream or may activate afferent neurones that carry information to the brain and its cognitive centres, which regulates food intake. The rate at which nutrients become systemically available is also influenced by gastrointestinal motility: a delay in gastric emptying may evoke a satiety effect. Recent evidence suggests that the endocannabinoid anandamide and the related acylethanolamide oleoylethanolamide are produced in the intestine and might regulate feeding behaviour by engaging sensory afferent neurones that converge information to specific areas of the brain. The intestinal levels of these acylethanolamides are inversely correlated to feeding, as food deprivation increases intestinal levels of anandamide (which acts in the gut as a 'hunger signal'), while it decreases the levels of oleoylethanolamide (which acts in the gut as a 'satiety signal'). Additionally, these acylethanolamides, whose gastric levels change in response to diet-induced obesity, alter gastrointestinal motility, which might contribute to their effect on food intake and nutrient absorption.
Collapse
Affiliation(s)
- R Capasso
- Department of Experimental Pharmacology, University of Naples Federico II, Naples, Italy
| | | |
Collapse
|
47
|
Fu J, Kim J, Oveisi F, Astarita G, Piomelli D. Targeted enhancement of oleoylethanolamide production in proximal small intestine induces across-meal satiety in rats. Am J Physiol Regul Integr Comp Physiol 2008; 295:R45-50. [PMID: 18434444 DOI: 10.1152/ajpregu.00126.2008] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Pharmacological administration of the natural lipid amide, oleoylethanolamide (OEA), inhibits food intake in free-feeding rodents by prolonging latency to feed and postmeal interval. This anorexic effect is mediated by activation of type-alpha peroxisome proliferator-activated receptors (PPAR-alpha). Food intake stimulates mucosal cells in duodenum and jejunum to generate OEA, suggesting that this lipid-derived messenger may act as a local satiety hormone. As a test of this hypothesis, here, we examined whether targeted enhancement of OEA production in the small intestine affects feeding behavior in rats. We constructed an adenoviral vector (Ad-NPLD) that directs overexpression of the enzyme N-acylphosphatidylethanolamine (NAPE)-phospholipase D (PLD), which catalyzes the hydrolysis of NAPE to generate OEA. Intraduodenal injection of the Ad-NPLD vector resulted in a time-dependent increase in NAPE-PLD expression and OEA production, which was restricted to the proximal small intestine. No such effect was observed after administration of a control adenoviral vector. Enhanced OEA production in Ad-NPLD-injected animals was temporally associated with increased expression of two PPAR-alpha target genes (PPAR-alpha and CD36) and with decreased food intake. The hypophagic phenotype of Ad-NPLD-injected rats was attributable to increase feeding latency and postmeal interval, rather than decreased meal size. The results suggest that localized changes in OEA production in the small intestine, such as those produced by food intake, are sufficient to induce in rats a state of across-meal satiety similar to that elicited by systemic administration of exogenous OEA.
Collapse
Affiliation(s)
- Jin Fu
- Department of Pharmacology and Center for Drug Discovery, University of California, Irvine, California, USA
| | | | | | | | | |
Collapse
|
48
|
Artmann A, Petersen G, Hellgren LI, Boberg J, Skonberg C, Nellemann C, Hansen SH, Hansen HS. Influence of dietary fatty acids on endocannabinoid and N-acylethanolamine levels in rat brain, liver and small intestine. Biochim Biophys Acta Mol Cell Biol Lipids 2008; 1781:200-12. [DOI: 10.1016/j.bbalip.2008.01.006] [Citation(s) in RCA: 254] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Revised: 12/30/2007] [Accepted: 01/23/2008] [Indexed: 01/30/2023]
|
49
|
Atkinson TJ. Central and peripheral neuroendocrine peptides and signalling in appetite regulation: considerations for obesity pharmacotherapy. Obes Rev 2008; 9:108-20. [PMID: 18257752 DOI: 10.1111/j.1467-789x.2007.00412.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Appetite and satiety are mediated by complex neuroendocrine signalling pathways involving over 40 hormones, neuropeptides, enzymes, other chemical messengers and their receptors. Research efforts continue to expand understanding of the role of signalling molecules between central hypothalamic nuclei and peripheral enteroendocrine cells; and discoveries of novel networks and messengers provide new biological insights on how to manipulate appetite-satiety pathways. Despite the vast array of peptides that are potentially useful for anti-obesity drug development, only four classes of agents are approved: (i) catecholamine stimulants; (ii) serotonin and noradrenaline reuptake inhibitors; (iii) lipase inhibitors; and (iv) more recently cannabinoid-1 receptor antagonists. Clinical effects of these drugs confer modest improvements, and side effects negatively impact long-term treatment course. This paper suggests single target pharmacological interventions are possibly hampered by the myriad of alternate orexigenic peptidic signals that drive hyperphagia, hence a multiple target model or combination treatment approach is proposed to offer greater therapeutic potential in modulating appetite and managing weight.
Collapse
|
50
|
Aviello G, Matias I, Capasso R, Petrosino S, Borrelli F, Orlando P, Romano B, Capasso F, Di Marzo V, Izzo AA. Inhibitory effect of the anorexic compound oleoylethanolamide on gastric emptying in control and overweight mice. J Mol Med (Berl) 2008; 86:413-22. [PMID: 18278475 DOI: 10.1007/s00109-008-0305-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Revised: 12/21/2007] [Accepted: 01/02/2008] [Indexed: 12/13/2022]
Abstract
Gastric emptying regulates food intake. Oleoylethanolamide (OEA), an endogenous acylethanolamide chemically related to the endocannabinoid anandamide, inhibits food intake, but its effect on gastric emptying is unknown. Here, we investigated the effect and the role of OEA on gastric emptying in mice fed either a standard (STD) or a high-fat diet (HFD) for 14 weeks. Gastric emptying was reduced by OEA, but not by its saturated analog, palmitoylethanolamide. The effect of OEA was unaffected by rimonabant (cannabinoid CB1 receptor antagonist), SR144528 (cannabinoid CB2 receptor antagonist), 5'-iodoresiniferatoxin (transient receptor potential vanilloid type 1 antagonist), or MK886 (peroxisome proliferator-activated receptor-alpha) antagonist. Compared to STD mice, HFD mice showed delayed gastric emptying and higher levels of gastric OEA. HFD-induced increase in OEA levels was accompanied by increased expression of the OEA-synthesizing enzyme N-acyl-phosphatidylethanolamine-selective phospholipase D and decreased expression of the OEA-degrading enzyme fatty acid amide hydrolase. These results might suggest that elevation of gastric OEA could possibly contribute to the delayed gastric emptying observed in HFD-fed animals. HFD regulates OEA levels in the stomach through an increase of its biosynthesis and a decrease of its enzymatic degradation. The inhibitory effect of OEA on gastric emptying here observed might underlie part of the anorexic effects of this compound previously reported.
Collapse
Affiliation(s)
- Gabriella Aviello
- Endocannabinoid Research Group, Department of Experimental Pharmacology, University of Naples Federico II, Naples, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|