1
|
Seguella L, Corpetti C, Lu J, Pesce M, Franzin SB, Palenca I, Zilli A, Vincenzi M, Caprioli D, Paytuví-Gallart A, Sanseverino W, Rurgo S, Sarnelli G, Esposito G. Oleoylethanolamide-producing Lactobacillus paracasei F19 improves metabolic and behavioral disorders by restoring intestinal permeability and microbiota-gut-brain axis in high-fat diet-induced obese male mice. Brain Behav Immun 2025; 127:25-44. [PMID: 39988008 DOI: 10.1016/j.bbi.2025.02.014] [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: 08/23/2024] [Revised: 02/04/2025] [Accepted: 02/17/2025] [Indexed: 02/25/2025] Open
Abstract
Metabolic and mood disorders elicited by chronic exposure of high-fat diet (HFD) are often associated with intestinal dysbiosis and persistent low-grade inflammation in the small intestine. This leads to remodeling of the epithelial barrier with disruption of the neuroepithelial circuits that control energy homeostasis by the gut-brain axis. Therefore, therapies that restoreintestinal microbial niche and barrier function are promising candidates to counter peripheral metabolic challenges that affect behaviors controlled by the brain. The endogenous oleoylethanolamine (OEA) was found to shape the intestinal microbiota profile towards a "lean-like phenotype", ameliorating pathological profiles of metabolic diseases. Further, OEA displays beneficial effects in several cognitive paradigms and preserves the epithelial barrier integrity, acting as an intestinal "gate-keeper". Here, we developed an "intestinal OEA factory" for the in-situ and controlled release of OEA by using a probiotic-based delivery system. We engineered the Lactobacillus paracasei F19 (LP) to express the human N-acylphosphatidylethanolamine-preferring phospholipase D (NAPEpld) gene and to produce OEA in response to dietary ultra-low oleate supply. We treated 12-week HFD male mice with oleate-probiotic formulations and assessed their impact on metabolic and behavioral dysfunctions, and microbiota-gut-brain signaling after 8 weeks of treatment. NAPE-expressing LP (pNAPE-LP) led to significant reduced weight loss and improved metabolic dysfunction in HFD-treated mice. Further, a parallel improvement in depressive- and anxiety-like phenotypes was associated with the duodenal barrier function retrieval, the restoration of the Firmicutes/Bacteroidetes ratio, and an increase in beneficial bacteria, such as Lactobacillus, Prevotella, and Parabacteroides. The HFD-driven changes both in the enteric and central nervous system were prevented by pNAPE-LP/oleate treatment. Collectively, our data suggest that these effects were mediated by the oleate-dependent release of OEA by pNAPE-LP since no significant effects were observed in HFD mice treated with the native probiotic alone (pLP). This oleate-regulated delivery system of OEA is a safe and efficient probiotic-based strategy for the treatment of metabolic syndrome and related behavioral disorders.
Collapse
Affiliation(s)
- Luisa Seguella
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Chiara Corpetti
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Jie Lu
- Department of Anatomy and Cell Biology, China Medical University, N0.77 Puhe Road, Shenbei New District, Shenyang City, Liaoning Province, PR China.
| | - Marcella Pesce
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy.
| | - Silvia Basili Franzin
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Irene Palenca
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Aurora Zilli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Martina Vincenzi
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Daniele Caprioli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | | | - Walter Sanseverino
- Sequentia Biotech SL, Carrer del Dr. Trueta, 179, 08005 Barcelona, Spain.
| | - Sara Rurgo
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy.
| | - Giovanni Sarnelli
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy.
| | - Giuseppe Esposito
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| |
Collapse
|
2
|
McKnight AD, Alhadeff AL. Nutrient detection pathways for food reinforcement and satiation. Curr Opin Neurobiol 2025; 92:103040. [PMID: 40349609 DOI: 10.1016/j.conb.2025.103040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 04/08/2025] [Accepted: 04/13/2025] [Indexed: 05/14/2025]
Abstract
Ingested food is broken down into macronutrient components that are detected by gut-brain signaling pathways that play a vital role in feeding behavior. These specialized mechanisms both promote food intake (via appetition and food reinforcement pathways) and terminate food intake (via satiation pathways). Despite recent significant advances in our understanding of gut-brain signaling and behavior, questions remain about the distinct mechanisms mediating food reinforcement and satiation. Here, we review the receptors/transporters and gut-brain pathways that contribute to nutrient sensing and feeding behavior, and highlight key knowledge gaps that will guide future research on the complex gut-brain systems that influence food intake.
Collapse
Affiliation(s)
- Aaron D McKnight
- Monell Chemical Senses Center, Philadelphia, 19104, PA, United States; Department of Neuroscience, University of Pennsylvania, Philadelphia, 19104, PA, United States
| | - Amber L Alhadeff
- Monell Chemical Senses Center, Philadelphia, 19104, PA, United States; Department of Neuroscience, University of Pennsylvania, Philadelphia, 19104, PA, United States.
| |
Collapse
|
3
|
Chiarugi S, Margheriti F, De Lorenzi V, Martino E, Margheritis EG, Moscardini A, Marotta R, Chaves-Sanjuan A, Del Seppia C, Federighi G, Lapi D, Bandiera T, Rapposelli S, Scuri R, Bolognesi M, Garau G. NAPE-PLD is target of thiazide diuretics. Cell Chem Biol 2025; 32:449-462.e5. [PMID: 39999832 DOI: 10.1016/j.chembiol.2025.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/01/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025]
Abstract
Thiazide and thiazide-like diuretics are among the most efficacious and used drugs for the treatment of hypertension, edema, and major cardiovascular outcomes. Despite more then than six decades of clinical use, the molecular target and mechanism of action by which these drugs cure hypertension after long-term use have remained mysterious. Here we report the discovery and validation of a previously unknown renal and extrarenal target of these antihypertensives, the membrane-associated phospholipase N-acylphosphatidylethanolamine-specific phospholipase D (NAPE-PLD) of the endocannabinoid system. Structural and functional insights, together with preclinical studies in hypertensive rats, disclose the molecular and physiological basis by which thiazides cause acute diuresis and, at the same time, the distinctive chronic reduction of vascular resistance. Our results shed light on the mechanism of treatment of hypertension and will be useful for developing more efficacious medications for the management of vascular risk factors, as well as associated leukoencephalopathies and myelin disorders.
Collapse
Affiliation(s)
- Sara Chiarugi
- BioStructures Lab, Istituto Italiano di Tecnologia (IIT@NEST), Piazza San Silvestro 12, 56127 Pisa, Italy; Laboratorio NEST, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Francesco Margheriti
- BioStructures Lab, Istituto Italiano di Tecnologia (IIT@NEST), Piazza San Silvestro 12, 56127 Pisa, Italy; Laboratorio NEST, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Valentina De Lorenzi
- BioStructures Lab, Istituto Italiano di Tecnologia (IIT@NEST), Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Elisa Martino
- BioStructures Lab, Istituto Italiano di Tecnologia (IIT@NEST), Piazza San Silvestro 12, 56127 Pisa, Italy; Laboratorio NEST, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | | | - Aldo Moscardini
- Laboratorio NEST, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Roberto Marotta
- Electron Microscopy Facility, Istituto Italiano di Tecnologia (IIT) Via Morego 30, 16163 Genova, Italy
| | - Antonio Chaves-Sanjuan
- Department of BioSciences, University of Milano, Via Celoria 26, 20133 Milano, Italy; Pediatric Research Center Fondazione R.E. Invernizzi and NOLIMITS Center, Università degli Studi di Milano, Milan, Italy
| | | | - Giuseppe Federighi
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Via San Zeno 31, 56126 Pisa, Italy
| | - Dominga Lapi
- Department of Biology, University of Pisa, Via Alessandro Volta 4, 56127 Pisa, Italy
| | - Tiziano Bandiera
- D3 PharmaChemistry, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy
| | - Simona Rapposelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Rossana Scuri
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Via San Zeno 31, 56126 Pisa, Italy
| | - Martino Bolognesi
- Department of BioSciences, University of Milano, Via Celoria 26, 20133 Milano, Italy; Pediatric Research Center Fondazione R.E. Invernizzi and NOLIMITS Center, Università degli Studi di Milano, Milan, Italy
| | - Gianpiero Garau
- BioStructures Lab, Istituto Italiano di Tecnologia (IIT@NEST), Piazza San Silvestro 12, 56127 Pisa, Italy; Laboratorio NEST, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy; Laboratori Aliveda, Viale Karol Wojtyla 19, 56042 Crespina Lorenzana, Pisa, Italy.
| |
Collapse
|
4
|
Idahosa SO, Diarra R, Ranu HK, Nasiri RH, Higuchi S. Evidence and Mechanism of Bile Acid-Mediated Gut-Brain Axis in Anxiety and Depression. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:163-173. [PMID: 39566821 DOI: 10.1016/j.ajpath.2024.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 09/19/2024] [Accepted: 10/29/2024] [Indexed: 11/22/2024]
Abstract
Bidirectional communication between the brain and gastrointestinal tract, called the gut-brain axis, is linked with our emotions. Intestinal lipids, hormones, and molecules, such as bile acids (BAs), impact our mood, motivation, and emotions via the gut-brain axis. BAs are synthesized from cholesterol in the liver and serve as a regulator of lipid metabolism and hormonal secretion in the intestine. Human studies have indicated that the alteration of plasma BA levels is associated with depression and anxiety. Several possible mechanisms, such as BA receptor-dependent and receptor-independent mechanisms, have been reported for emotional control. Animal studies have indicated that the deletion of BA receptors shows behavioral abnormalities. BAs regulate gut hormones, glucagon-like peptide-1 secretion, bioactive lipids, oleoylethanolamide, and the immune system function, which influences neural activities. Thus, BAs act as an emotional regulator. This review aims to summarize the following: clinical evidence of BA concentration linked to mental disorders, including depression and anxiety; and animal studies of BA-related signaling correlated with its neurobehavioral effect supporting its mechanism. We will also discuss future research required for further neurobehavioral treatment.
Collapse
Affiliation(s)
- Sydney O Idahosa
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Rokia Diarra
- Department of Biology, St. John's University, Queens, New York
| | - Hernoor K Ranu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Raidah H Nasiri
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Sei Higuchi
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York.
| |
Collapse
|
5
|
Goodman MT, Lombardi C, Torrens A, Bresee C, Saloman JL, Li L, Yang Y, Fisher WE, Fogel EL, Forsmark CE, Conwell DL, Hart PA, Park WG, Topazian M, Vege SS, Van Den Eeden SK, Bellin MD, Andersen DK, Serrano J, Yadav D, Pandol SJ, Piomelli D, on behalf of the Consortium for the Study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer (CPDPC). Association of Serum Endocannabinoid Levels with Pancreatitis and Pancreatitis-Related Pain. Cannabis Cannabinoid Res 2025; 10:60-70. [PMID: 39291350 PMCID: PMC11947650 DOI: 10.1089/can.2024.0079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
Background and Aims: This investigation examined the association of pancreatitis and pancreatitis-related pain with serum levels of two endocannabinoid molecules such as anandamide (AEA) and 2-arachidonoylglycerol (2-AG) and two paracannabinoid molecules such as oleoylethanolamide (OEA) and palmitoylethanolamide (PEA). Methods: A case-control study was conducted within the Prospective Evaluation of Chronic Pancreatitis for Epidemiological and Translational Studies, including participants with no pancreas disease (N = 56), chronic abdominal pain of suspected pancreatic origin or indeterminate chronic pancreatitis (CP) (N = 22), acute pancreatitis (N = 33), recurrent acute pancreatitis (N = 57), and definite CP (N = 63). Results: Circulating AEA concentrations were higher in women than in men (p = 0.0499), and PEA concentrations were higher in obese participants than those who were underweight/normal or overweight (p = 0.003). Asymptomatic controls with no pancreatic disease had significantly (p = 0.03) lower concentrations of AEA compared with all disease groups combined. The highest concentrations of AEA were observed in participants with acute pancreatitis, followed by those with recurrent acute pancreatitis, chronic abdominal pain/indeterminant CP, and definite CP. Participants with pancreatitis reporting abdominal pain in the past year had significantly (p = 0.04) higher concentrations of AEA compared with asymptomatic controls. Levels of 2-AG were significantly lower (p = 0.02) among participants reporting abdominal pain in the past week, and pain intensity was inversely associated with concentrations of 2-AG and OEA. Conclusions: Endocannabinoid levels may be associated with stage of pancreatitis, perhaps through activation of the CB1 receptor. Validation of our findings would support the investigation of novel therapeutics, including cannabinoid receptor-1 antagonists, in this patient population.
Collapse
Affiliation(s)
- Marc T. Goodman
- Prevention and Control Program, Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Christina Lombardi
- Prevention and Control Program, Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Alexa Torrens
- Department and Anatomy and Neurobiology, University of California, Irvine, California, USA
| | - Catherine Bresee
- Department of Biostatistics and Bioinformatics Research Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jami L. Saloman
- Center for Pain Research, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Liang Li
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yunlong Yang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - William E. Fisher
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Evan L. Fogel
- Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Christopher E. Forsmark
- Division of Gastroenterology, Hepatology, and Nutrition, University of Florida, Gainesville, Florida, USA
| | - Darwin L. Conwell
- Department of Internal Medicine, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Phil A. Hart
- Division of Gastroenterology, Hepatology, & Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Walter G. Park
- Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University Medical Center, Stanford, California, USA
| | | | - Santhi S. Vege
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Melena D. Bellin
- Division of Pediatric Endocrinology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Dana K. Andersen
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jose Serrano
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Dhiraj Yadav
- Department of Medicine Division of Gastroenterology, Hepatology & Nutrition University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Stephen J. Pandol
- Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Daniele Piomelli
- Department and Anatomy and Neurobiology, University of California, Irvine, California, USA
- Department of Biological Chemistry, University of California, Irvine, California, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, California, USA
| | - on behalf of the Consortium for the Study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer (CPDPC)
- Prevention and Control Program, Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department and Anatomy and Neurobiology, University of California, Irvine, California, USA
- Department of Biostatistics and Bioinformatics Research Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Center for Pain Research, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
- Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Division of Gastroenterology, Hepatology, and Nutrition, University of Florida, Gainesville, Florida, USA
- Department of Internal Medicine, University of Kentucky College of Medicine, Lexington, Kentucky, USA
- Division of Gastroenterology, Hepatology, & Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
- Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University Medical Center, Stanford, California, USA
- Mayo Clinic, Rochester, Minnesota, USA
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
- Division of Pediatric Endocrinology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota, USA
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Medicine Division of Gastroenterology, Hepatology & Nutrition University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biological Chemistry, University of California, Irvine, California, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, California, USA
| |
Collapse
|
6
|
Patel C, Patel R, Kesharwani A, Rao L, Jain NS. Central cholinergic transmission modulates endocannabinoid-induced marble-burying behavior in mice. Behav Brain Res 2025; 476:115252. [PMID: 39278464 DOI: 10.1016/j.bbr.2024.115252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/30/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
Central cholinergic system and endocannabinoid, anandamide exhibits anti-compulsive-like behavior in mice. However, the role of the central cholinergic system in the anandamide-induced anti-compulsive-like behavior is still unexplored. Therefore, the present study assessed the role of central cholinergic transmission in the anandamide-induced anti-compulsive activity using a marble-burying behavior (MBB) model in mice. The modulation in the anandamide-induced effect on MBB was evaluated using mice with altered central cholinergic transmission achieved by pretreatment (i.c.v.) with various cholinergic agents like acetylcholine (ACh), acetylcholinesterase inhibitor (AChEI), neostigmine, nicotine, mAChR antagonist, atropine, and nAChR antagonist, mecamylamine. The influence of anandamide treatment on the brain AChE activity was also evaluated. The results revealed that i.c.v. injection of anandamide (10, 20 µg/mouse, i.c.v.) dose-dependently reduced MBB in mice. Moreover, anandamide in all the tested doses inhibited the brain AChE activity indicating the role of an enhanced central cholinergic transmission in its anti-compulsive-like effect . Furthermore, the anti-compulsive-like effect of anandamide (20 µg/mouse, i.c.v.) was found to be enhanced in mice centrally pre-treated with, ACh (0.1 µg/mouse, i.c.v.) or AChEI, neostigmine (0.3 µg/mouse, i.c.v.). In addition, the anandamide-induced anti-compulsive-like effect was significantly increased in mice pre-treated with a low dose of nicotine (0.1 µg/mouse, i.c.v.) while, it was attenuated by the higher dose of nicotine (2 µg/mouse, i.c.v.). On the other hand, the anandamide (20 µg/mouse, i.c.v.) induced anti-compulsive-like effect was found to be diminished in mice pre-treated with mAChR antagonist, atropine (0.1, 0.5 µg/mouse, i.c.v.) and pre-injection of nAChR antagonist, mecamylamine (0.1, 0.5 µg/mouse, i.c.v.) potentiated the anandamide induced anti-compulsive-like response in mice. Thus, the present investigation delineates the modulatory role of an enhanced central cholinergic transmission in the anandamide-induced anti-compulsive-like behavior in mice by inhibition of brain AChE or via muscarinic and nicotinic receptors mediated mechanism.
Collapse
Affiliation(s)
- Chhatrapal Patel
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya, Koni, Bilaspur, Chhattisgarh, India
| | - Richa Patel
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya, Koni, Bilaspur, Chhattisgarh, India
| | - Anuradha Kesharwani
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya, Koni, Bilaspur, Chhattisgarh, India
| | - Laxmi Rao
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya, Koni, Bilaspur, Chhattisgarh, India
| | - Nishant Sudhir Jain
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya, Koni, Bilaspur, Chhattisgarh, India.
| |
Collapse
|
7
|
Wang L, Cheng C, Yu X, Guo L, Wan X, Xu J, Xiang X, Yang J, Kang J, Deng Q. Conversion of α-linolenic acid into n-3 long-chain polyunsaturated fatty acids: bioavailability and dietary regulation. Crit Rev Food Sci Nutr 2024:1-33. [PMID: 39686568 DOI: 10.1080/10408398.2024.2442064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
N-3 long-chain polyunsaturated fatty acids (n-3 LCPUFAs) are essential for physiological requirements and disease prevention throughout life but are not adequately consumed worldwide. Dietary supplementation with plant-derived α-linolenic acid (ALA) has the potential to rebalance the fatty acid profile and enhance health benefits but faces challenges such as high β-oxidation consumption, low hepatic conversion efficiency, and high oxidative susceptibility under stress. This review focuses on the metabolic fate and potential regulatory targets of ALA-containing lipids in vivo, specifically the pathway from the gastrointestinal tract to the lymph, blood circulation, and liver. We propose a hypothesis that positively regulates the conversion of ALA into n-3 LCPUFAs based on the model of "fast" or "slow" absorption, transport, and hepatic metabolic fate. Furthermore, the potential effects of dietary nutrients on the metabolic conversion of ALA into n-3 LCPUFAs are discussed. The conversion of ALA is differentially regulated by structured lipids, phospholipids, other lipids, carbohydrates, specific proteins, amino acids, polyphenols, vitamins, and minerals. Future research should focus on designing a steady-state and precise delivery system for ALA, coupled with specific nutrients or phytochemicals, to effectively improve its metabolic conversion and ultimately achieve synergistic regulation of nutrition and health effects.
Collapse
Affiliation(s)
- Lei Wang
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| | - Chen Cheng
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| | - Xiao Yu
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou, Henan, China
| | - Liang Guo
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xia Wan
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| | - Jiqu Xu
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| | - Xia Xiang
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| | - Jing Yang
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| | - Jingxuan Kang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Qianchun Deng
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| |
Collapse
|
8
|
Xu T, Zhang C, Yang Y, Huang L, Liu Q, Li L, Zeng Q, Li Z. Role of Milk Intake in Modulating Serum Lipid Profiles and Gut Metabolites. Metabolites 2024; 14:688. [PMID: 39728469 DOI: 10.3390/metabo14120688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Milk is one of the main sources of nutrition in people's daily diet, but the fat in milk raises health concerns in consumers. Here, we aimed to elucidate the impact of Buffalo milk and Holstein cow milk consumption on blood lipid health through metabolomics analysis. METHODS Golden hamsters were administered Murrah Buffalo milk (BM) or Holstein cow milk (HM), and the body weight and serum lipid indicators were tested and recorded. The hamsters receiving equal amounts of physiological saline were used as the negative control (NC). Serum and fecal samples were collected, and LC-MS was used to identify the metabolites in the samples. RESULTS The results showed that both the BM and HM groups exhibited a significant reduction in body weight compared to that of the NC group from day 9, and the serum TG, TC, and LDL-C levels were significantly lower than those of the NC group. Further analysis identified 564 and 567 metabolites in the serum and fecal samples shared in the BM and HM groups and significantly different from those in the NC group, which were mainly enriched in the pathways related to lipid metabolism, such as fatty acid biosynthesis, arachidonic acid metabolism, and primary bile acid biosynthesis. Correlation analysis further suggested that milk intake can increase the levels of Muramic Acid, Oleoyl Ethanolamide, Seratrodast, Chenodeoxycholic Acid, Docosahexaenoic Acid Ethyl Ester, and Deoxycholic Acid in the serum and gut microbiota, which may affect TG, TC, HDL-C, and LDL-C in the serum, and thereby benefit the body's lipid health. CONCLUSIONS The results further confirmed that milk intake has a beneficial effect on blood lipid health by altering multiple metabolites in the serum and the gut. This study provides novel evidence that milk consumption is beneficial to health and is a reference for guiding people to a healthy diet.
Collapse
Affiliation(s)
- Ting Xu
- Guangxi Zhuang Autonomous Region Buffalo Milk Quality and Safety Control Technology Engineering Research Center, Guangxi Buffalo Research Institute, Chinese Academy of Agricultural Sciences, Nanning 530001, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Chang Zhang
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Yufeng Yang
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Liang Huang
- School of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qingyou Liu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Ling Li
- Guangxi Zhuang Autonomous Region Buffalo Milk Quality and Safety Control Technology Engineering Research Center, Guangxi Buffalo Research Institute, Chinese Academy of Agricultural Sciences, Nanning 530001, China
| | - Qingkun Zeng
- Guangxi Zhuang Autonomous Region Buffalo Milk Quality and Safety Control Technology Engineering Research Center, Guangxi Buffalo Research Institute, Chinese Academy of Agricultural Sciences, Nanning 530001, China
| | - Zhipeng Li
- Guangxi Zhuang Autonomous Region Buffalo Milk Quality and Safety Control Technology Engineering Research Center, Guangxi Buffalo Research Institute, Chinese Academy of Agricultural Sciences, Nanning 530001, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| |
Collapse
|
9
|
Cheng J, Venkatesh S, Ke K, Barratt MJ, Gordon JI. A human gut Faecalibacterium prausnitzii fatty acid amide hydrolase. Science 2024; 386:eado6828. [PMID: 39446943 PMCID: PMC11572954 DOI: 10.1126/science.ado6828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 08/14/2024] [Indexed: 10/26/2024]
Abstract
Undernutrition in Bangladeshi children is associated with disruption of postnatal gut microbiota assembly; compared with standard therapy, a microbiota-directed complementary food (MDCF) substantially improved their ponderal and linear growth. Here, we characterize a fatty acid amide hydrolase (FAAH) from a growth-associated intestinal strain of Faecalibacterium prausnitzii cultured from these children. This enzyme, expressed and purified from Escherichia coli, hydrolyzes a variety of N-acylamides, including oleoylethanolamide (OEA), neurotransmitters, and quorum sensing N-acyl homoserine lactones; it also synthesizes a range of N-acylamides, notably N-acyl amino acids. Treating germ-free mice with N-oleoylarginine and N-oleolyhistidine, major products of FAAH OEA metabolism, markedly affected expression of intestinal immune function pathways. Administering MDCF to Bangladeshi children considerably reduced fecal OEA, a satiety factor whose levels were negatively correlated with abundance and expression of their F. prausnitzii FAAH. This enzyme, structurally and catalytically distinct from mammalian FAAH, is positioned to regulate levels of a variety of bioactive molecules.
Collapse
Affiliation(s)
- Jiye Cheng
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- The Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Siddarth Venkatesh
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- The Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Ke Ke
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- The Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Michael J. Barratt
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- The Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Jeffrey I. Gordon
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- The Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| |
Collapse
|
10
|
Zomeño MD, Malcampo M, Pérez-Vega KA, Pastor A, López-Roura M, Arrufat B, Atarés S, Ramos SJ, Alonso D, Subirana I, Muñoz-Aguayo D, Blanchart G, Gaixas S, Cabañero M, Tello S, Konstantinidou V, Hernando-Redondo J, Goday A, Castañer O, Schröder H, Fitó M. Effect on Satiety-Related Biomarkers of Bar Snacks Containing Chickpea Flour and Pork Protein. Nutrients 2024; 16:3180. [PMID: 39339780 PMCID: PMC11434683 DOI: 10.3390/nu16183180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/20/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
This project aims to establish the acceptability and satiety of a hybrid snack containing plant protein and a small percentage of animal protein compared to a meat-based snack. DESIGN Randomised, crossover, double-blind, controlled post-prandial trial involving 24 participants (18-30 years), with two interventions: (a) a hybrid snack containing plant protein derived from chickpeas and 6.6% lean high-quality pork meat; and (b) a meat-based snack containing 90% lean pork meat. METHODS General, life-style, sensory acceptability questionnaire, and the following laboratory analyses were performed: lipid profile, endocannabinoids, and related compounds. RESULTS Sensory questionnaires showed in general good acceptability for both bars. Additionally, there was a greater increase in glycemia at 30, 60, and 90 min after consuming the hybrid snack compared to the meat-based snack, with no changes in the lipid profile. Regarding the endocannabinoid compounds and related compounds, the compound N-palmitoleoyl ethanolamine in the acylethanolamide group showed higher levels overall following the consumption of the hybrid snack compared to the meat-based snack, particularly at 2 h. CONCLUSIONS The hybrid snack was associated with changes in endocannabinoid-like compounds. Therefore, it may provide a lasting satiating effect, while complementing the protein profile of plant-based foods with the quality of animal protein.
Collapse
Affiliation(s)
- María-Dolores Zomeño
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; (M.-D.Z.); (M.M.); (K.A.P.-V.); (A.P.); (M.L.-R.); (I.S.); (D.M.-A.); (G.B.); (M.C.); (J.H.-R.); (A.G.); (H.S.); (M.F.)
- Consortium Center for Biomedical Research Network (CIBER), M.P. Pathophysiology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Blanquerna School of Health Sciences, Universitat Ramon Llull, 08025 Barcelona, Spain
| | - Mireia Malcampo
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; (M.-D.Z.); (M.M.); (K.A.P.-V.); (A.P.); (M.L.-R.); (I.S.); (D.M.-A.); (G.B.); (M.C.); (J.H.-R.); (A.G.); (H.S.); (M.F.)
| | - Karla Alejandra Pérez-Vega
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; (M.-D.Z.); (M.M.); (K.A.P.-V.); (A.P.); (M.L.-R.); (I.S.); (D.M.-A.); (G.B.); (M.C.); (J.H.-R.); (A.G.); (H.S.); (M.F.)
- Consortium Center for Biomedical Research Network (CIBER), M.P. Pathophysiology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Antoni Pastor
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; (M.-D.Z.); (M.M.); (K.A.P.-V.); (A.P.); (M.L.-R.); (I.S.); (D.M.-A.); (G.B.); (M.C.); (J.H.-R.); (A.G.); (H.S.); (M.F.)
- Consortium Center for Biomedical Research Network (CIBER), M.P. Pathophysiology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Maria López-Roura
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; (M.-D.Z.); (M.M.); (K.A.P.-V.); (A.P.); (M.L.-R.); (I.S.); (D.M.-A.); (G.B.); (M.C.); (J.H.-R.); (A.G.); (H.S.); (M.F.)
| | - Begoña Arrufat
- Fertinagro Biotech S.L., 44195 Teruel, Spain; (B.A.); (S.A.)
| | - Sergio Atarés
- Fertinagro Biotech S.L., 44195 Teruel, Spain; (B.A.); (S.A.)
| | | | - David Alonso
- Naturuel S.L., 44002 Teruel, Spain; (S.J.R.); (D.A.)
| | - Isaac Subirana
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; (M.-D.Z.); (M.M.); (K.A.P.-V.); (A.P.); (M.L.-R.); (I.S.); (D.M.-A.); (G.B.); (M.C.); (J.H.-R.); (A.G.); (H.S.); (M.F.)
- Blanquerna School of Health Sciences, Universitat Ramon Llull, 08025 Barcelona, Spain
- Consortium Center for Biomedical Research Network (CIBER), M.P. Cardiovascular Diseases (CIBERcv), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Daniel Muñoz-Aguayo
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; (M.-D.Z.); (M.M.); (K.A.P.-V.); (A.P.); (M.L.-R.); (I.S.); (D.M.-A.); (G.B.); (M.C.); (J.H.-R.); (A.G.); (H.S.); (M.F.)
- Consortium Center for Biomedical Research Network (CIBER), M.P. Pathophysiology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Gemma Blanchart
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; (M.-D.Z.); (M.M.); (K.A.P.-V.); (A.P.); (M.L.-R.); (I.S.); (D.M.-A.); (G.B.); (M.C.); (J.H.-R.); (A.G.); (H.S.); (M.F.)
| | - Sònia Gaixas
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; (M.-D.Z.); (M.M.); (K.A.P.-V.); (A.P.); (M.L.-R.); (I.S.); (D.M.-A.); (G.B.); (M.C.); (J.H.-R.); (A.G.); (H.S.); (M.F.)
| | - Marta Cabañero
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; (M.-D.Z.); (M.M.); (K.A.P.-V.); (A.P.); (M.L.-R.); (I.S.); (D.M.-A.); (G.B.); (M.C.); (J.H.-R.); (A.G.); (H.S.); (M.F.)
- Consortium Center for Biomedical Research Network (CIBER), M.P. Epidemiology and Public Health (CIBEResp), Carlos III Health Institute, 28029 Madrid, Spain
| | - Susanna Tello
- Consortium Center for Biomedical Research Network (CIBER), M.P. Pathophysiology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | | | - Javier Hernando-Redondo
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; (M.-D.Z.); (M.M.); (K.A.P.-V.); (A.P.); (M.L.-R.); (I.S.); (D.M.-A.); (G.B.); (M.C.); (J.H.-R.); (A.G.); (H.S.); (M.F.)
- Consortium Center for Biomedical Research Network (CIBER), M.P. Pathophysiology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Albert Goday
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; (M.-D.Z.); (M.M.); (K.A.P.-V.); (A.P.); (M.L.-R.); (I.S.); (D.M.-A.); (G.B.); (M.C.); (J.H.-R.); (A.G.); (H.S.); (M.F.)
- Consortium Center for Biomedical Research Network (CIBER), M.P. Pathophysiology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Department of Medicine, Autonomous University of Barcelona, 08193 Barcelona, Spain
| | - Olga Castañer
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; (M.-D.Z.); (M.M.); (K.A.P.-V.); (A.P.); (M.L.-R.); (I.S.); (D.M.-A.); (G.B.); (M.C.); (J.H.-R.); (A.G.); (H.S.); (M.F.)
- Consortium Center for Biomedical Research Network (CIBER), M.P. Epidemiology and Public Health (CIBEResp), Carlos III Health Institute, 28029 Madrid, Spain
| | - Helmut Schröder
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; (M.-D.Z.); (M.M.); (K.A.P.-V.); (A.P.); (M.L.-R.); (I.S.); (D.M.-A.); (G.B.); (M.C.); (J.H.-R.); (A.G.); (H.S.); (M.F.)
- Consortium Center for Biomedical Research Network (CIBER), M.P. Epidemiology and Public Health (CIBEResp), Carlos III Health Institute, 28029 Madrid, Spain
| | - Montserrat Fitó
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; (M.-D.Z.); (M.M.); (K.A.P.-V.); (A.P.); (M.L.-R.); (I.S.); (D.M.-A.); (G.B.); (M.C.); (J.H.-R.); (A.G.); (H.S.); (M.F.)
- Consortium Center for Biomedical Research Network (CIBER), M.P. Pathophysiology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain;
| |
Collapse
|
11
|
Akakura M, Watari I, Watanabe M, Jiratchaya S, Ono T. Expression of Glucagon-Like Peptide-1 Receptors in the Submandibular Gland of Mice and Its Implications in Type 2 Diabetes. Cureus 2024; 16:e70465. [PMID: 39479116 PMCID: PMC11524602 DOI: 10.7759/cureus.70465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2024] [Indexed: 11/02/2024] Open
Abstract
Introduction Type 2 diabetes mellitus (T2DM) not only affects the pancreas directly involved in glucose metabolism but also impairs salivary gland function. Glucagon-like peptide-1 (GLP-1) is a gastrointestinal hormone that lowers postprandial blood glucose levels by stimulating insulin secretion from the pancreas. Previous studies have revealed the presence of GLP-1 receptors (GLP-1R) in salivary glands. However, the effect of diabetes on salivary gland GLP-1R remains unclear. This study aimed to observe the impact of T2DM on GLP-1R in the submandibular gland (SMG). Materials and methods Twenty-five-week-old mice were randomly divided into four groups (n=5 each): 11w and 13w control groups (CON), and 11w and 13w diabetes mellitus groups (DM). After a five-day adaptation period, the DM group mice were subjected to a high-fat diet, while the CON group received a standard diet. The DM group mice were then induced into a state of T2DM by a single low-dose streptozotocin injection at nine weeks of age. Oral glucose tolerance tests (OGTT) were conducted to evaluate mouse glucose tolerance. At 11 and 13 weeks of age, SMG was excised under general anesthesia, and the morphology of SMG was evaluated by hematoxylin-eosin staining, while the distribution and expression of GLP-1R were assessed by immunohistochemical staining. The data obtained were subjected to the Shapiro-Wilk test to confirm normal distribution, the t-test for the OGTT results, and statistical analysis for other results by one-way analysis of variance. Results Consistent with previous reports, the mice in the DM group showed higher body weight and lower glucose tolerance. Histological analysis revealed an increase in the acinar area and a decrease in the ductal area of the SMG in the DM group. Although there was no significant decrease in the cell count regarding the ductal area, a tendency toward luminal dilation was observed. Interestingly, the expression pattern of GLP-1R was limited to the ductal portion of the SMG, with a decrease in anti-GLP-1R-positive areas observed in the DM group compared to the CON group. While there was no significant difference in anti-GLP-1R-positive areas between the CON11w and CON13w groups, the DM13w group exhibited a significant decrease compared to the DM11w group. These data suggest that diabetes induces both structural changes in the SMG and a reduction in GLP-1R expression, particularly in the ductal regions. Conclusions We found that the expression level of GLP-1R in SMG was decreased in the DM group mice. This data demonstrates the potential relationship between T2DM and GLP-1R expression. Moreover, there was an indication of a temporal decrease in anti-GLP-1R positive areas over time. This result may suggest the involvement of salivary gland GLP-1R in the mechanism of impaired SMG function caused by T2DM, potentially mediated through the decrease in blood GLP-1 levels.
Collapse
Affiliation(s)
- Masato Akakura
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, JPN
| | - Ippei Watari
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, JPN
| | - Minami Watanabe
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, JPN
| | - Srisutha Jiratchaya
- Department of Orthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, THA
| | - Takashi Ono
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, JPN
| |
Collapse
|
12
|
Jieu B, Sykorova EB, Rohleder C, Marcolini E, Hoffmann AE, Koethe D, Leweke FM, Couttas TA. Alterations to sphingolipid metabolism from antipsychotic administration in healthy volunteers are restored following the use of cannabidiol. Psychiatry Res 2024; 339:116005. [PMID: 38950483 DOI: 10.1016/j.psychres.2024.116005] [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: 01/25/2024] [Revised: 05/06/2024] [Accepted: 06/01/2024] [Indexed: 07/03/2024]
Abstract
Randomized clinical trials substantiate cannabidiol (CBD) as a next-generation antipsychotic, effective in alleviating positive and negative symptoms associated with psychosis, while minimising the adverse effects seen with established treatments. Although the mechanisms remain debated, CBD is known to induce drug-responsive changes in lipid-based retrograde neurotransmitters. Lipid aberrations are also frequently observed with antipsychotics, which may contribute to their efficacy or increase the risk of undesirables, including metabolic dysfunction, obesity and dyslipidaemia. Our study investigated CBD's impact following lipid responses triggered by interaction with second-generation antipsychotics (SGA) in a randomized phase I safety study. Untargeted mass spectrometry assessed the lipidomic profiles of human sera, collected from 38 healthy volunteers. Serum samples were obtained prior to commencement of any medication (t = 0), 3 days after consecutive administration of one of the five, placebo-controlled, treatment arms designed to achieve steady-state concentrations of each SGA (amisulpride, 150 mg/day; quetiapine, 300 mg/day; olanzapine 10 mg/day; risperidone, 3 mg/day), and after six successive days of SGA treatment combined with CBD (800 mg/day). Receiver operating characteristics (ROC) refined 3712 features to a putative list of 15 lipids significantly altered (AUC > 0.7), classified into sphingolipids (53 %), glycerolipids (27 %) and glycerophospholipids (20 %). Targeted mass spectrometry confirmed reduced sphingomyelin and ceramide levels with antipsychotics, which mapped along their catabolic pathway and were restored by CBD. These sphingolipids inversely correlated with body weight after olanzapine, quetiapine, and risperidone treatment, where CBD appears to have arrested or attenuated these effects. Herein, we propose CBD may alleviate aberrant sphingolipid metabolism and that further investigation into sphingolipids as markers for monitoring side effects of SGAs and efficacy of CBD is warranted.
Collapse
Affiliation(s)
- Beverly Jieu
- Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - Eliska B Sykorova
- Dept. of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Cathrin Rohleder
- Brain and Mind Centre, The University of Sydney, Sydney, Australia; Dept. of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Endosane Pharmaceuticals GmbH, Berlin, Germany
| | - Elisabeth Marcolini
- Dept. of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Anna E Hoffmann
- Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - Dagmar Koethe
- Brain and Mind Centre, The University of Sydney, Sydney, Australia; Dept. of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - F Markus Leweke
- Brain and Mind Centre, The University of Sydney, Sydney, Australia; Dept. of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Endosane Pharmaceuticals GmbH, Berlin, Germany
| | | |
Collapse
|
13
|
González-Portilla M, Montagud-Romero S, Mellado S, de Fonseca FR, Pascual M, Rodríguez-Arias M. Region-Specific Gene Expression Changes Associated with Oleoylethanolamide-Induced Attenuation of Alcohol Self-Administration. Int J Mol Sci 2024; 25:9002. [PMID: 39201687 PMCID: PMC11354326 DOI: 10.3390/ijms25169002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/10/2024] [Accepted: 08/17/2024] [Indexed: 09/03/2024] Open
Abstract
Oleoylethanolamide (OEA) is a lipid with anti-inflammatory activity that modulates multiple reward-related behaviors. Previous studies have shown that OEA treatment reduces alcohol self-administration (SA) while inhibiting alcohol-induced inflammatory signaling. Nevertheless, the specific mechanisms that OEA targets to achieve these effects have not been widely explored. Here, we tested the effects of OEA treatment during alcohol SA, extinction or previous to cue-induced reinstatement of alcohol seeking. In addition, we measured gene expression changes in the striatum and hippocampus of relevant receptors for alcohol consumption (Drd1, Drd2, Cnr1, Oprm) as well as immune-related proteins (Il-6, Il-1β, Tlr4) and the brain-derived neurotrophic factor (Bdnf). Our results confirmed that when administered contingently, systemic OEA administration reduced alcohol SA and attenuated cue-induced reinstatement. Interestingly, we also observed that OEA treatment reduced the number of sessions needed for the extinction of alcohol seeking. Biochemical analyses showed that OEA induced gene expression changes in dopamine and cannabinoid receptors in the striatum and hippocampus. In addition, OEA treatment modulated the long-term immune response and increased Bdnf expression. These results suggest that boosting OEA levels may be an effective strategy for reducing alcohol SA and preventing relapse.
Collapse
Affiliation(s)
- Macarena González-Portilla
- Department of Psychobiology, Faculty of Psychology, Universitat de València, Avda. Blasco Ibáñez 21, 46010 Valencia, Spain; (M.G.-P.); (S.M.-R.)
| | - Sandra Montagud-Romero
- Department of Psychobiology, Faculty of Psychology, Universitat de València, Avda. Blasco Ibáñez 21, 46010 Valencia, Spain; (M.G.-P.); (S.M.-R.)
| | - Susana Mellado
- Department of Physiology, School of Medicine, Universitat de Valencia, Avda. Blasco Ibáñez 15, 46010 Valencia, Spain; (S.M.); (M.P.)
| | - Fernando Rodríguez de Fonseca
- Mental Health Clinical Management Unit, Institute of Biomedical Research of Malaga-IBIMA, Regional University Hospital of Málaga, 29010 Málaga, Spain;
- Atención Primaria, Cronicidad y Promoción de la Salud, Red de Investigación en Atención Primaria de Adicciones (RIAPAD) Rd21/0009/0005/0003, Valencia, Spain
| | - María Pascual
- Department of Physiology, School of Medicine, Universitat de Valencia, Avda. Blasco Ibáñez 15, 46010 Valencia, Spain; (S.M.); (M.P.)
| | - Marta Rodríguez-Arias
- Department of Psychobiology, Faculty of Psychology, Universitat de València, Avda. Blasco Ibáñez 21, 46010 Valencia, Spain; (M.G.-P.); (S.M.-R.)
- Atención Primaria, Cronicidad y Promoción de la Salud, Red de Investigación en Atención Primaria de Adicciones (RIAPAD) Rd21/0009/0005/0003, Valencia, Spain
| |
Collapse
|
14
|
Bai B, Wen Y, Wang J, Wen F, Yan H, Yuan X, Xie J, Zhang R, Xia Q, Wang G. Fatty Acid Desaturase Bmdesat5, Suppressed in the Salivary Glands by Domestication, is Involved in Regulation of Food Intake in Silkworms. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:14177-14190. [PMID: 38875711 DOI: 10.1021/acs.jafc.4c02511] [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: 06/16/2024]
Abstract
Understanding the evolutionary genetics of food intake regulation in domesticated animals has relevance to evolutionary biology, animal improvement, and obesity treatment. Here, we observed that the fatty acid desaturase gene (Bmdesat5), which regulates food intake, is suppressed in domesticated silkworms, but expressed in the salivary glands of the wild silkworm Bombyx mandarina. The content of its catalytic product, cis-vaccenic acid, was related to the expression levels of Bmdesat5 in the salivary glands of domesticated and wild silkworm strains. These two strains also showed significant differences in food intake. Using orally administering cis-vaccenic acid and transgenic-mediated overexpression, we verified that cis-vaccenic acid functions as a satiation signal, regulating food intake and growth in silkworms. Selection analysis showed that Bmdesat5 experienced selection, especially in the potential promoter, 5'-untranslated, and intron regions. This study highlights the importance of the decrement of satiety in silkworm domestication and provides new insights into the potential involvement of salivary glands in the regulation of satiety in animals, by acting as a supplement to gut-brain nutrient signaling.
Collapse
Affiliation(s)
- Bingchuan Bai
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
| | - Yuchan Wen
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
| | - Jing Wang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
| | - Feng Wen
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
| | - Hao Yan
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
| | - Xingli Yuan
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
| | - Jiatong Xie
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
| | - Ruihan Zhang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
| | - Qingyou Xia
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
| | - Genhong Wang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
| |
Collapse
|
15
|
Casselbrant A, Elias E, Hallersund P, Elebring E, Cervin J, Fändriks L, Wallenius V. Intestinal Ketogenesis and Permeability. Int J Mol Sci 2024; 25:6555. [PMID: 38928261 PMCID: PMC11204016 DOI: 10.3390/ijms25126555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Consumption of a high-fat diet (HFD) has been suggested as a contributing factor behind increased intestinal permeability in obesity, leading to increased plasma levels of microbial endotoxins and, thereby, increased systemic inflammation. We and others have shown that HFD can induce jejunal expression of the ketogenic rate-limiting enzyme mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase (HMGCS). HMGCS is activated via the free fatty acid binding nuclear receptor PPAR-α, and it is a key enzyme in ketone body synthesis that was earlier believed to be expressed exclusively in the liver. The function of intestinal ketogenesis is unknown but has been described in suckling rats and mice pups, possibly in order to allow large molecules, such as immunoglobulins, to pass over the intestinal barrier. Therefore, we hypothesized that ketone bodies could regulate intestinal barrier function, e.g., via regulation of tight junction proteins. The primary aim was to compare the effects of HFD that can induce intestinal ketogenesis to an equicaloric carbohydrate diet on inflammatory responses, nutrition sensing, and intestinal permeability in human jejunal mucosa. Fifteen healthy volunteers receiving a 2-week HFD diet compared to a high-carbohydrate diet were compared. Blood samples and mixed meal tests were performed at the end of each dietary period to examine inflammation markers and postprandial endotoxemia. Jejunal biopsies were assessed for protein expression using Western blotting, immunohistochemistry, and morphometric characteristics of tight junctions by electron microscopy. Functional analyses of permeability and ketogenesis were performed in Caco-2 cells, mice, and human enteroids. Ussing chambers were used to analyze permeability. CRP and ALP values were within normal ranges and postprandial endotoxemia levels were low and did not differ between the two diets. The PPARα receptor was ketone body-dependently reduced after HFD. None of the tight junction proteins studied, nor the basal electrical parameters, were different between the two diets. However, the ketone body inhibitor hymeglusin increased resistance in mucosal biopsies. In addition, the tight junction protein claudin-3 was increased by ketone inhibition in human enteroids. The ketone body β-Hydroxybutyrate (βHB) did not, however, change the mucosal transition of the large-size molecular FD4-probe or LPS in Caco-2 and mouse experiments. We found that PPARα expression was inhibited by the ketone body βHB. As PPARα regulates HMGCS expression, the ketone bodies thus exert negative feedback signaling on their own production. Furthermore, ketone bodies were involved in the regulation of permeability on intestinal mucosal cells in vitro and ex vivo. We were not, however, able to reproduce these effects on intestinal permeability in vivo in humans when comparing two weeks of high-fat with high-carbohydrate diet in healthy volunteers. Further, neither the expression of inflammation markers nor the aggregate tight junction proteins were changed. Thus, it seems that not only HFD but also other factors are needed to permit increased intestinal permeability in vivo. This indicates that the healthy gut can adapt to extremes of macro-nutrients and increased levels of intestinally produced ketone bodies, at least during a shorter dietary challenge.
Collapse
Affiliation(s)
- Anna Casselbrant
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 40530 Gothenburg, Sweden; (E.E.); (P.H.); (L.F.); (V.W.)
| | - Erik Elias
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 40530 Gothenburg, Sweden; (E.E.); (P.H.); (L.F.); (V.W.)
| | - Peter Hallersund
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 40530 Gothenburg, Sweden; (E.E.); (P.H.); (L.F.); (V.W.)
| | - Erik Elebring
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 40530 Gothenburg, Sweden; (E.E.); (P.H.); (L.F.); (V.W.)
| | - Jakob Cervin
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, 40530 Gothenburg, Sweden;
| | - Lars Fändriks
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 40530 Gothenburg, Sweden; (E.E.); (P.H.); (L.F.); (V.W.)
- Department of Surgery, Region Västra Götaland, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
| | - Ville Wallenius
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 40530 Gothenburg, Sweden; (E.E.); (P.H.); (L.F.); (V.W.)
- Department of Surgery, Region Västra Götaland, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
| |
Collapse
|
16
|
Srisutha J, Watari I, Akakura M, Watanabe M, Changsiripun C, Ono T. P2X7R and P2X4R expression of mice submandibular gland in high-fat diet/streptozotocin-induced type 2 diabetes. Sci Rep 2024; 14:10855. [PMID: 38740782 PMCID: PMC11091137 DOI: 10.1038/s41598-024-60519-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/24/2024] [Indexed: 05/16/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a chronic inflammatory disease that can compromise the functioning of various organs, including the salivary glands (SG). The purinergic system is one of the most important inflammatory pathways in T2DM condition, and P2X7R and P2X4R are the primary purinergic receptors in SG that regulate inflammatory homeostasis. This study aimed to evaluate P2X7R and P2X4R expression, and morphological changes in the submandibular gland (SMG) in T2DM. Twenty-four 5-week-old mice were randomly assigned to control (CON) and diabetes mellitus (DM) groups (n = 12 each). Body weight, diet, and blood glucose levels were monitored weekly. The histomorphology of the SMG and the expression of the P2X7R, and P2X7R was evaluated by immunohistochemistry (IHC) staining and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) at 11 and 13 weeks of age. Our findings indicate a significant increase in food consumption, body weight, and blood glucose levels in the DM group. Although a significant increase in P2X7R and P2X4R expression was observed in the DM groups, the receptor location remained unchanged. We also observed a significant increase in the acinar area in the DM13w group, and a significant decrease in the ductal area in the DM11w and DM13w groups. Targeting purinergic receptors may offer novel therapeutic methods for diabetic complications.
Collapse
MESH Headings
- Animals
- Mice
- Blood Glucose/metabolism
- Body Weight
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/chemically induced
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diet, High-Fat/adverse effects
- Mice, Inbred C57BL
- Receptors, Purinergic P2X4/metabolism
- Receptors, Purinergic P2X4/genetics
- Receptors, Purinergic P2X7/metabolism
- Receptors, Purinergic P2X7/genetics
- Streptozocin
- Submandibular Gland/metabolism
- Submandibular Gland/pathology
Collapse
Affiliation(s)
- Jiratchaya Srisutha
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo city, Tokyo, 113-8510, Japan
- Department of Orthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Ippei Watari
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo city, Tokyo, 113-8510, Japan.
| | - Masato Akakura
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo city, Tokyo, 113-8510, Japan
| | - Minami Watanabe
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo city, Tokyo, 113-8510, Japan
| | - Chidsanu Changsiripun
- Department of Orthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Takashi Ono
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo city, Tokyo, 113-8510, Japan
| |
Collapse
|
17
|
Sangsuriyothai P, Watari I, Serirukchutarungsee S, Satrawaha S, Podyma-Inoue KA, Ono T. Expression of glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide in the rat submandibular gland is influenced by pre- and post-natal high-fat diet exposure. Front Physiol 2024; 15:1357730. [PMID: 38595641 PMCID: PMC11002158 DOI: 10.3389/fphys.2024.1357730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/20/2024] [Indexed: 04/11/2024] Open
Abstract
Background: Incretins, i.e., glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) promote insulin secretion to reduce postprandial blood sugar. Previous studies found incretins in the salivary glands. However, the role of GLP-1 and GIP in the submandibular gland (SMG) is unclear. This study investigates the effects of a high-fat diet (HFD) on the expression of GLP-1 and GIP throughout the development of rat SMG. Methods: Pregnant 11-week-old Wistar rats were divided into two groups: those fed on a standard diet (n = 5) and those fed on a HFD (n = 5). From day 7 of pregnancy and throughout the lactation period, all the rats were fed on either a chow diet or HFD. The newborns were divided into four subgroups (n = 6): standard diet males (SM), HFD males (HM), standard diet females (SF), and HFD females (HF). The SMGs of 3- and 10-week-old rats from each subgroup were collected under general anesthesia. Moreover, body weight, food intake, and fasting blood sugar were measured. The mRNA expression of GLP-1 and GIP was quantified, and the localization was observed using immunohistochemistry (p < 0.05). Results: GLP-1 mRNA expression was statistically significantly more upregulated in HM than in HF at 3 weeks. Moreover, GLP-1 mRNA expression was significantly higher in HM than in both SM and HF at 10 weeks. Although a decreasing trend was observed in GIP mRNA expression in both 3- and 10-week-old rats fed on a HFD, a significant difference between HM and SM only occurred at 3 weeks. Furthermore, the GIP mRNA expression of HM was lower than that of HF at 10 weeks. Immunohistochemical staining revealed GLP-1 and GIP expression mainly in the SMG duct system. Moreover, vacuolated cytoplasm in the duct was observed in rats fed on a HFD. Conclusion: Exposure to HFD during pre- and post-natal periods increased GLP-1 mRNA expression in the SMGs of male rats. However, GIP expression decreased following the HFD in male newborns. Furthermore, a decreasing trend of GIP mRNA expression was observed in male newborns after HFD feeding. Sex influenced incretin hormones secretion and obesity-related conditions. HFD during pre- and post-natal periods reprograms the epigenome, contributing to subsequent disease development.
Collapse
Affiliation(s)
- Pornchanok Sangsuriyothai
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Department of Orthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Ippei Watari
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Saranya Serirukchutarungsee
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Department of Pedodontics and Preventive Dentistry, Faculty of Dentistry, Srinakharinwirot University, Bangkok, Thailand
| | - Sirichom Satrawaha
- Department of Orthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Katarzyna Anna Podyma-Inoue
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takashi Ono
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
18
|
Chen G, Staffen N, Wu Z, Xu X, Pan J, Inoue A, Shi T, Gmeiner P, Du Y, Xu J. Structural and functional characterization of the endogenous agonist for orphan receptor GPR3. Cell Res 2024; 34:262-265. [PMID: 38287118 PMCID: PMC10907609 DOI: 10.1038/s41422-023-00919-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 12/20/2023] [Indexed: 01/31/2024] Open
Affiliation(s)
- Geng Chen
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Nico Staffen
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, Erlangen, Germany
| | - Zhangsong Wu
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Xinyu Xu
- Beijing Advanced Innovation Center for Structural Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Jinheng Pan
- Mass Spectrometry & Metabolomics Core Facility, Biomedical Research Core Facilities, Westlake University, Hangzhou, Zhejiang, China
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Tingyi Shi
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, Erlangen, Germany.
| | - Yang Du
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, Chinese University of Hong Kong, Shenzhen, Guangdong, China.
| | - Jun Xu
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, Chinese University of Hong Kong, Shenzhen, Guangdong, China.
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
19
|
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
|
20
|
Bitencourt Brito P, Dalcin Teixeira M, Lehtonen Rodrigues de Souza R, Furtado-Alle L, Viater Tureck L. Olive oil increases the LIPC expression when associated with an Eastern pattern diet: An experimental study with Wistar rats. Gene 2023; 887:147738. [PMID: 37625559 DOI: 10.1016/j.gene.2023.147738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/05/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023]
Abstract
Some nutrigenomic effects of extra virgin olive oil (EVOO) are described in the literature; however, it is unknown whether its interaction with lipid-related genes is independent of the combined diet. In this sense, our objective was to investigate whether EVOO consumption associated with Western or Eastern human-based chow modulates the expression of APOE, APOB, and LIPC genes in rats. In view of this, the hypothesis is that the consumption of olive oil may not have the same nutrigenomic effects, depending on the diet consumed. For this study, 56 female rats were randomly divided into four groups: Western diet with EVOO (WS), Western-diet control (WC), Eastern-diet with EVOO (ES), and Eastern-diet control (EC). After 15 weeks, the animals were anesthetized with an intraperitoneal injection of chloral hydrate 15% (1.5 mL/kg) and euthanized by guillotining, and adipose tissue, liver, and blood were extracted. Triglycerides, cholesterol, and glucose levels were obtained following standard protocols, and relative gene expressions were calculated using the ΔΔCt method after quantitative PCR. The EVOO consumption was associated with LIPC gene expression increase in the liver only in animals fed the Eastern diet, compared to EC and WS animals. The EVOO consumption, combined with the Eastern diet, was associated with decreased triglyceride levels compared to WC. Although final weight and weight gain were similar between groups, WS animals had lower daily energy consumption. Conclusion: Given these results, the authors suggested that the EVOO nutrigenomic effects were restricted to an Eastern human-based diet.
Collapse
Affiliation(s)
- Priscila Bitencourt Brito
- Polymorphism and Linkage Laboratory, Department of Genetics, Federal University of Paraná, Curitiba, Paraná State, Brazil
| | - Mayza Dalcin Teixeira
- Polymorphism and Linkage Laboratory, Department of Genetics, Federal University of Paraná, Curitiba, Paraná State, Brazil
| | | | - Lupe Furtado-Alle
- Polymorphism and Linkage Laboratory, Department of Genetics, Federal University of Paraná, Curitiba, Paraná State, Brazil
| | - Luciane Viater Tureck
- Polymorphism and Linkage Laboratory, Department of Genetics, Federal University of Paraná, Curitiba, Paraná State, Brazil.
| |
Collapse
|
21
|
Ellermann M. Emerging mechanisms by which endocannabinoids and their derivatives modulate bacterial populations within the gut microbiome. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2023; 3:11359. [PMID: 38389811 PMCID: PMC10880783 DOI: 10.3389/adar.2023.11359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 11/28/2023] [Indexed: 02/24/2024]
Abstract
Bioactive lipids such as endocannabinoids serve as important modulators of host health and disease through their effects on various host functions including central metabolism, gut physiology, and immunity. Furthermore, changes to the gut microbiome caused by external factors such as diet or by disease development have been associated with altered endocannabinoid tone and disease outcomes. These observations suggest the existence of reciprocal relationships between host lipid signaling networks and bacterial populations that reside within the gut. Indeed, endocannabinoids and their congeners such as N-acylethanolamides have been recently shown to alter bacterial growth, functions, physiology, and behaviors, therefore introducing putative mechanisms by which these bioactive lipids directly modulate the gut microbiome. Moreover, these potential interactions add another layer of complexity to the regulation of host health and disease pathogenesis that may be mediated by endocannabinoids and their derivatives. This mini review will summarize recent literature that exemplifies how N-acylethanolamides and monoacylglycerols including endocannabinoids can impact bacterial populations in vitro and within the gut microbiome. We also highlight exciting preclinical studies that have engineered gut bacteria to synthesize host N-acylethanolamides or their precursors as potential strategies to treat diseases that are in part driven by aberrant lipid signaling, including obesity and inflammatory bowel diseases.
Collapse
Affiliation(s)
- Melissa Ellermann
- Department of Biological Sciences, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
22
|
Takemoto K, Kato H, Higashino K. Involvement of the vagus nerve in the anorectic effect of monoacylglycerol acyltransferase 2 inhibition in mice. Obes Sci Pract 2023; 9:601-608. [PMID: 38090688 PMCID: PMC10712405 DOI: 10.1002/osp4.693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/13/2023] [Accepted: 06/22/2023] [Indexed: 10/16/2024] Open
Abstract
Background Many of the drugs used for obesity treatment have adverse effects on the central nervous system. Therefore, novel treatments, such as peripherally acting drugs, are needed. Monoacylglycerol acyltransferase 2 (MGAT2), highly expressed in the small intestine, catalyzes the first step of triacylglycerol re-synthesis. MGAT2 inhibition suppresses food intake in high-fat diet (HFD)-fed mice, but the mechanisms remain unclear. Here, the involvement of the vagus nerve in MGAT2 inhibition-induced feeding suppression was investigated. Methods Fasted mice were administered an MGAT2 inhibitor. Food intake was measured after HFD re-feeding, and the effect of capsaicin pretreatment on changes in food intake was evaluated. The number of c-fos-positive cells in the nucleus tractus solitarius and levels of appetite regulators were determined after HFD re-feeding or lipid gavage. Results The anorectic effect of the MGAT2 inhibitor was abolished when vagus nerve function was interrupted by capsaicin. MGAT2 inhibition increased the number of c-fos-positive cells in the nucleus tractus solitarius and elevated intestinal oleoylethanolamide, plasma peptide tyrosine-tyrosine and plasma glucagon-like peptide-1 levels. Conclusion MGAT2 inhibition suppresses feeding behavior via peripheral vagus nerve signaling and may serve as a novel anti-obesity strategy with a low risk of unexpected central nervous system-related adverse effects.
Collapse
Affiliation(s)
- Kosuke Takemoto
- Drug Discovery & Disease Research LaboratoryShionogi & Co., Ltd.OsakaJapan
- Laboratory of Veterinary PathologyJoint Faculty of Veterinary MedicineYamaguchi UniversityYamaguchiJapan
| | - Hideaki Kato
- Drug Discovery & Disease Research LaboratoryShionogi & Co., Ltd.OsakaJapan
| | - Kenichi Higashino
- Drug Discovery & Disease Research LaboratoryShionogi & Co., Ltd.OsakaJapan
| |
Collapse
|
23
|
Tovar R, de Ceglia M, Ubaldi M, Rodríguez-Pozo M, Soverchia L, Cifani C, Rojo G, Gavito A, Hernandez-Folgado L, Jagerovic N, Ciccocioppo R, Baixeras E, Rodríguez de Fonseca F, Decara J. Administration of Linoleoylethanolamide Reduced Weight Gain, Dyslipidemia, and Inflammation Associated with High-Fat-Diet-Induced Obesity. Nutrients 2023; 15:4448. [PMID: 37892524 PMCID: PMC10609991 DOI: 10.3390/nu15204448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/27/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Acylethanolamides (NAEs) are bioactive lipids derived from diet fatty acids that modulate important homeostatic functions, including appetite, fatty acid synthesis, mitochondrial respiration, inflammation, and nociception. Among the naturally circulating NAEs, the pharmacology of those derived from either arachidonic acid (Anandamide), oleic acid (OEA), and palmitic acid (PEA) have been extensively characterized in diet-induced obesity. For the present work, we extended those studies to linoleoylethanolamide (LEA), one of the most abundant NAEs found not only in plasma and body tissues but also in foods such as cereals. In our initial study, circulating concentrations of LEA were found to be elevated in overweight humans (body mass index (BMI, Kg/m2) > 25) recruited from a representative population from the south of Spain, together with AEA and the endocannabinoid 2-Arachidonoyl glycerol (2-AG). In this population, LEA concentrations correlated with the circulating levels of cholesterol and triglycerides. In order to gain insight into the pharmacology of LEA, we administered it for 14 days (10 mg/kg i.p. daily) to obese male Sprague Dawley rats receiving a cafeteria diet or a standard chow diet for 12 consecutive weeks. LEA treatment resulted in weight loss and a reduction in circulating triglycerides, cholesterol, and inflammatory markers such as Il-6 and Tnf-alpha. In addition, LEA reduced plasma transaminases and enhanced acetyl-CoA-oxidase (Acox) and Uncoupling protein-2 (Ucp2) expression in the liver of the HFD-fed animals. Although the liver steatosis induced by the HFD was not reversed by LEA, the overall data suggest that LEA contributes to the homeostatic signals set in place in response to diet-induced obesity, potentially contributing with OEA to improve lipid metabolism after high fat intake. The anti-inflammatory response associated with its administration suggests its potential for use as a nutrient supplement in non-alcoholic steatohepatitis.
Collapse
Affiliation(s)
- Rubén Tovar
- Grupo de Neuropsicofarmacología, Instituto IBIMA-Plataforma BIONAND, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Avda, Carlos Haya 82, Sótano, 29010 Málaga, Spain; (R.T.); (M.d.C.); (M.R.-P.); (A.G.); (E.B.)
| | - Marialuisa de Ceglia
- Grupo de Neuropsicofarmacología, Instituto IBIMA-Plataforma BIONAND, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Avda, Carlos Haya 82, Sótano, 29010 Málaga, Spain; (R.T.); (M.d.C.); (M.R.-P.); (A.G.); (E.B.)
| | - Massimo Ubaldi
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy; (M.U.); (L.S.); (C.C.); (R.C.)
| | - Miguel Rodríguez-Pozo
- Grupo de Neuropsicofarmacología, Instituto IBIMA-Plataforma BIONAND, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Avda, Carlos Haya 82, Sótano, 29010 Málaga, Spain; (R.T.); (M.d.C.); (M.R.-P.); (A.G.); (E.B.)
| | - Laura Soverchia
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy; (M.U.); (L.S.); (C.C.); (R.C.)
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy; (M.U.); (L.S.); (C.C.); (R.C.)
| | - Gema Rojo
- Department of Endocrinology and Nutrition, Hospital Regional Universitario de Málaga, Instituto IBIMA-Plataforma BIONAND, 29010 Málaga, Spain;
| | - Ana Gavito
- Grupo de Neuropsicofarmacología, Instituto IBIMA-Plataforma BIONAND, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Avda, Carlos Haya 82, Sótano, 29010 Málaga, Spain; (R.T.); (M.d.C.); (M.R.-P.); (A.G.); (E.B.)
| | - Laura Hernandez-Folgado
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas, Avenida Juan de la Cierva, 28006 Madrid, Spain; (L.H.-F.); (N.J.)
| | - Nadine Jagerovic
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas, Avenida Juan de la Cierva, 28006 Madrid, Spain; (L.H.-F.); (N.J.)
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy; (M.U.); (L.S.); (C.C.); (R.C.)
| | - Elena Baixeras
- Grupo de Neuropsicofarmacología, Instituto IBIMA-Plataforma BIONAND, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Avda, Carlos Haya 82, Sótano, 29010 Málaga, Spain; (R.T.); (M.d.C.); (M.R.-P.); (A.G.); (E.B.)
| | - Fernando Rodríguez de Fonseca
- Grupo de Neuropsicofarmacología, Instituto IBIMA-Plataforma BIONAND, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Avda, Carlos Haya 82, Sótano, 29010 Málaga, Spain; (R.T.); (M.d.C.); (M.R.-P.); (A.G.); (E.B.)
- Unidad Clínica de Neurología, Hospital Regional Universitario de Málaga, Instituto IBMA-Plataforma BIONAND, 29010 Málaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology [NEURO-RECA], 29010 Malaga, Spain
| | - Juan Decara
- Grupo de Neuropsicofarmacología, Instituto IBIMA-Plataforma BIONAND, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Avda, Carlos Haya 82, Sótano, 29010 Málaga, Spain; (R.T.); (M.d.C.); (M.R.-P.); (A.G.); (E.B.)
| |
Collapse
|
24
|
Astarita G, Kelly RS, Lasky-Su J. Metabolomics and lipidomics strategies in modern drug discovery and development. Drug Discov Today 2023; 28:103751. [PMID: 37640150 PMCID: PMC10543515 DOI: 10.1016/j.drudis.2023.103751] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/09/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
Metabolomics and lipidomics have an increasingly pivotal role in drug discovery and development. In the context of drug discovery, monitoring changes in the levels or composition of metabolites and lipids relative to genetic variations yields functional insights, bolstering human genetics and (meta)genomic methodologies. This approach also sheds light on potential novel targets for therapeutic intervention. In the context of drug development, metabolite and lipid biomarkers contribute to enhanced success rates, promising a transformative impact on precision medicine. In this review, we deviate from analytical chemist-focused perspectives, offering an overview tailored to drug discovery. We provide introductory insight into state-of-the-art mass spectrometry (MS)-based metabolomics and lipidomics techniques utilized in drug discovery and development, drawing from the collective expertise of our research teams. We comprehensively outline the application of metabolomics and lipidomics in advancing drug discovery and development, spanning fundamental research, target identification, mechanisms of action, and the exploration of biomarkers.
Collapse
Affiliation(s)
- Giuseppe Astarita
- Georgetown University, Washington, DC, USA; Arkuda Therapeutics, Watertown, MA, USA.
| | - Rachel S Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
25
|
Forte N, Roussel C, Marfella B, Lauritano A, Villano R, De Leonibus E, Salviati E, Khalilzadehsabet T, Giorgini G, Silvestri C, Piscitelli F, Mollica MP, Di Marzo V, Cristino L. Olive oil-derived endocannabinoid-like mediators inhibit palatable food-induced reward and obesity. Commun Biol 2023; 6:959. [PMID: 37735539 PMCID: PMC10514336 DOI: 10.1038/s42003-023-05295-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 08/29/2023] [Indexed: 09/23/2023] Open
Abstract
N-oleoylglycine (OlGly), a lipid derived from the basic component of olive oil, oleic acid, and N-oleoylalanine (OlAla) are endocannabinoid-like mediators. We report that OlGly and OlAla, by activating the peroxisome proliferator-activated receptor alpha (PPARα), reduce the rewarding properties of a highly palatable food, dopamine neuron firing in the ventral tegmental area, and the obesogenic effect of a high-fat diet rich in lard (HFD-L). An isocaloric olive oil HFD (HFD-O) reduced body weight gain compared to the HFD-L, in a manner reversed by PPARα antagonism, and enhanced brain and intestinal OlGly levels and gut microbial diversity. OlGly or OlAla treatment of HFD-L mice resulted in gut microbiota taxonomic changes partly similar to those induced by HFD-O. We suggest that OlGly and OlAla control body weight by counteracting highly palatable food overconsumption, and possibly rebalancing the gut microbiota, and provide a potential new mechanism of action for the obeso-preventive effects of olive oil-rich diets.
Collapse
Affiliation(s)
- Nicola Forte
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, 80078, Pozzuoli (NA), Italy
| | - Charlène Roussel
- Heart and Lung Research Institute of Université Laval, Québec City, QC, Canada
- Institute for Nutrition and Functional Foods, Centre NUTRISS, Université Laval, Québec City, QC, Canada
| | - Brenda Marfella
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, 80078, Pozzuoli (NA), Italy
- Department of Biology, University of Naples Federico II, 80126, Naples, Italy
| | - Anna Lauritano
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, 80078, Pozzuoli (NA), Italy
| | - Rosaria Villano
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, 80078, Pozzuoli (NA), Italy
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
- Institute of Biochemistry and Cell Biology, Consiglio Nazionale delle Ricerche (CNR), Monterotondo Scalo, Rome, Italy
| | | | - Tina Khalilzadehsabet
- Heart and Lung Research Institute of Université Laval, Québec City, QC, Canada
- Institute for Nutrition and Functional Foods, Centre NUTRISS, Université Laval, Québec City, QC, Canada
| | - Giada Giorgini
- Heart and Lung Research Institute of Université Laval, Québec City, QC, Canada
- Institute for Nutrition and Functional Foods, Centre NUTRISS, Université Laval, Québec City, QC, Canada
| | - Cristoforo Silvestri
- Heart and Lung Research Institute of Université Laval, Québec City, QC, Canada
- Institute for Nutrition and Functional Foods, Centre NUTRISS, Université Laval, Québec City, QC, Canada
| | - Fabiana Piscitelli
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, 80078, Pozzuoli (NA), Italy
| | - Maria Pina Mollica
- Department of Biology, University of Naples Federico II, 80126, Naples, Italy
- Centro Servizi Metrologici e Tecnologici Avanzati (CeSMA), Complesso Universitario di Monte Sant'Angelo, 80126, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, 80138, Naples, Italy
| | - Vincenzo Di Marzo
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, 80078, Pozzuoli (NA), Italy.
- Heart and Lung Research Institute of Université Laval, Québec City, QC, Canada.
- Institute for Nutrition and Functional Foods, Centre NUTRISS, Université Laval, Québec City, QC, Canada.
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Québec City, QC, 61V0AG, Canada.
| | - Luigia Cristino
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, 80078, Pozzuoli (NA), Italy.
| |
Collapse
|
26
|
Maccarrone M, Di Marzo V, Gertsch J, Grether U, Howlett AC, Hua T, Makriyannis A, Piomelli D, Ueda N, van der Stelt M. Goods and Bads of the Endocannabinoid System as a Therapeutic Target: Lessons Learned after 30 Years. Pharmacol Rev 2023; 75:885-958. [PMID: 37164640 PMCID: PMC10441647 DOI: 10.1124/pharmrev.122.000600] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 05/12/2023] Open
Abstract
The cannabis derivative marijuana is the most widely used recreational drug in the Western world and is consumed by an estimated 83 million individuals (∼3% of the world population). In recent years, there has been a marked transformation in society regarding the risk perception of cannabis, driven by its legalization and medical use in many states in the United States and worldwide. Compelling research evidence and the Food and Drug Administration cannabis-derived cannabidiol approval for severe childhood epilepsy have confirmed the large therapeutic potential of cannabidiol itself, Δ9-tetrahydrocannabinol and other plant-derived cannabinoids (phytocannabinoids). Of note, our body has a complex endocannabinoid system (ECS)-made of receptors, metabolic enzymes, and transporters-that is also regulated by phytocannabinoids. The first endocannabinoid to be discovered 30 years ago was anandamide (N-arachidonoyl-ethanolamine); since then, distinct elements of the ECS have been the target of drug design programs aimed at curing (or at least slowing down) a number of human diseases, both in the central nervous system and at the periphery. Here a critical review of our knowledge of the goods and bads of the ECS as a therapeutic target is presented to define the benefits of ECS-active phytocannabinoids and ECS-oriented synthetic drugs for human health. SIGNIFICANCE STATEMENT: The endocannabinoid system plays important roles virtually everywhere in our body and is either involved in mediating key processes of central and peripheral diseases or represents a therapeutic target for treatment. Therefore, understanding the structure, function, and pharmacology of the components of this complex system, and in particular of key receptors (like cannabinoid receptors 1 and 2) and metabolic enzymes (like fatty acid amide hydrolase and monoacylglycerol lipase), will advance our understanding of endocannabinoid signaling and activity at molecular, cellular, and system levels, providing new opportunities to treat patients.
Collapse
Affiliation(s)
- Mauro Maccarrone
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Vincenzo Di Marzo
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Jürg Gertsch
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Uwe Grether
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Allyn C Howlett
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Tian Hua
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Alexandros Makriyannis
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Daniele Piomelli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Natsuo Ueda
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Mario van der Stelt
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| |
Collapse
|
27
|
Seramur ME, Sink S, Cox AO, Furdui CM, Key CCC. ABHD4 regulates adipocyte differentiation in vitro but does not affect adipose tissue lipid metabolism in mice. J Lipid Res 2023; 64:100405. [PMID: 37352974 PMCID: PMC10400869 DOI: 10.1016/j.jlr.2023.100405] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 05/02/2023] [Accepted: 06/10/2023] [Indexed: 06/25/2023] Open
Abstract
Alpha/beta hydrolase domain-containing protein 4 (ABHD4) catalyzes the deacylation of N-acyl phosphatidyl-ethanolamine (NAPE) and lyso-NAPE to produce glycerophospho-N-acyl ethanolamine (GP-NAE). Through a variety of metabolic enzymes, NAPE, lyso-NAPE, and GP-NAE are ultimately converted into NAE, a group of bioactive lipids that control many physiological processes including inflammation, cognition, food intake, and lipolysis (i.e., oleoylethanolamide or OEA). In a diet-induced obese mouse model, adipose tissue Abhd4 gene expression positively correlated with adiposity. However, it is unknown whether Abhd4 is a causal or a reactive gene to obesity. To fill this knowledge gap, we generated an Abhd4 knockout (KO) 3T3-L1 pre-adipocyte. During adipogenic stimulation, Abhd4 KO pre-adipocytes had increased adipogenesis and lipid accumulation, suggesting Abhd4 is responding to (a reactive gene), not contributing to (not a causal gene), adiposity, and may serve as a mechanism for protecting against obesity. However, we did not observe any differences in adiposity and metabolic outcomes between whole-body Abhd4 KO or adipocyte-specific Abhd4 KO mice and their littermate control mice (both male and female) on chow or a high-fat diet. This might be because we found that deletion of Abhd4 did not affect NAE such as OEA production, even though Abhd4 was highly expressed in adipose tissue and correlated with fasting adipose OEA levels and lipolysis. These data suggest that ABHD4 regulates adipocyte differentiation in vitro but does not affect adipose tissue lipid metabolism in mice despite nutrient overload, possibly due to compensation from other NAPE and NAE metabolic enzymes.
Collapse
Affiliation(s)
- Mary E Seramur
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Sandy Sink
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Anderson O Cox
- Wake Forest Baptist Comprehensive Cancer Center Proteomics and Metabolomics Shared Resource, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Cristina M Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Chia-Chi Chuang Key
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest University School of Medicine, Winston Salem, NC, USA.
| |
Collapse
|
28
|
Higuchi S, Wood C, Nasiri RH, Giddla LJ, Molina V, Diarra R, DiPatrizio NV, Kawamura A, Haeusler RA. The 16α-hydroxylated Bile Acid, Pythocholic Acid Decreases Food Intake and Increases Oleoylethanolamide in Male Mice. Endocrinology 2023; 164:bqad116. [PMID: 37490843 PMCID: PMC10407715 DOI: 10.1210/endocr/bqad116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 07/27/2023]
Abstract
Modulation of bile acid (BA) structure is a potential strategy for obesity and metabolic disease treatment. BAs act not only as signaling molecules involved in energy expenditure and glucose homeostasis, but also as regulators of food intake. The structure of BAs, particularly the position of the hydroxyl groups of BAs, impacts food intake partly by intestinal effects: (1) modulating the activity of N-acyl phosphatidylethanolamine phospholipase D, which produces the anorexigenic bioactive lipid oleoylethanolamide (OEA) or (2) regulating lipid absorption and the gastric emptying-satiation pathway. We hypothesized that 16α-hydroxylated BAs uniquely regulate food intake because of the long intermeal intervals in snake species in which these BAs are abundant. However, the effects of 16α-hydroxylated BAs in mammals are completely unknown because they are not naturally found in mammals. To test the effect of 16α-hydroxylated BAs on food intake, we isolated the 16α-hydroxylated BA pythocholic acid from ball pythons (Python regius). Pythocholic acid or deoxycholic acid (DCA) was given by oral gavage in mice. DCA is known to increase N-acyl phosphatidylethanolamine phospholipase D activity better than other mammalian BAs. We evaluated food intake, OEA levels, and gastric emptying in mice. We successfully isolated pythocholic acid from ball pythons for experimental use. Pythocholic acid treatment significantly decreased food intake in comparison to DCA treatment, and this was associated with increased jejunal OEA, but resulted in no change in gastric emptying or lipid absorption. The exogenous BA pythocholic acid is a novel regulator of food intake and the satiety signal for OEA in the mouse intestine.
Collapse
Affiliation(s)
- Sei Higuchi
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Courtney Wood
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Raidah H Nasiri
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Leela J Giddla
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Valentina Molina
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Rokia Diarra
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| | - Nicholas V DiPatrizio
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Akira Kawamura
- Department of Chemistry, Hunter College of CUNY, New York, NY 10065, USA
| | - Rebecca A Haeusler
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| |
Collapse
|
29
|
Anavi-Cohen S, Tsybina-Shimshilashvili N, Zandani G, Hovav R, Sela N, Nyska A, Madar Z. Effects of high oleic acid peanuts on mice's liver and adipose tissue metabolic parameters and gut microbiota composition. Front Nutr 2023; 10:1205377. [PMID: 37575334 PMCID: PMC10415107 DOI: 10.3389/fnut.2023.1205377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/28/2023] [Indexed: 08/15/2023] Open
Abstract
This study aimed to investigate the effects of two types of peanuts, regular Hanoch (HN) and a new high-oleic cultivar., Hanoch-Oleic (HO), on metabolic parameters and gut microbiota composition. Male C57BL/6 mice were fed with a normal diet (ND) or ND supplemented with HN (NDh) or HO (NDo). Following 18 weeks of diet regimen, the NDo group exhibited reduced body weight and peri-gonadal adipose-to-body weight ratio, paralleled to lesser food consumption. Although blood levels of total cholesterol, HDL-cholesterol, free fatty acids, and liver enzyme levels did not differ between groups, decreased insulin sensitivity was found in the NDh group. Within adipose tissue, the expression of lipolytic and lipogenic enzymes was higher, while those related to lipid oxidation were lower in the NDh group compared to the NDo group. Additionally, HO peanuts consumption promoted the establishment of a healthy microbiota, with an enhanced abundance of Bifidobacterium, Lactobacillus, and Coprococcus genera. In conclusion, the inclusion of the HO peanut cultivar., rather than the conventional peanut cultivar., in a balanced diet was related to better metabolic outcomes and was linked to a favorable microbiota profile.
Collapse
Affiliation(s)
| | | | - Gil Zandani
- The Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Ran Hovav
- Department of Field Crops and Vegetables Research, Plant Sciences Institute, Agricultural Research Organization, Rishon LeZion, Israel
| | - Noa Sela
- Department of Plant Pathology and Weed Research, Volcani Center, Rishon LeZion, Israel
| | - Abraham Nyska
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Zecharia Madar
- Peres Academic Center, Rehovot, Israel
- The Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
30
|
Darcey VL, Guo J, Courville AB, Gallagher I, Avery JA, Simmons WK, Ingeholm JE, Herscovitch P, Martin A, Hall KD. Dietary fat restriction affects brain reward regions in a randomized crossover trial. JCI Insight 2023; 8:e169759. [PMID: 37345661 PMCID: PMC10371234 DOI: 10.1172/jci.insight.169759] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/10/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUNDWeight-loss diets often target dietary fat or carbohydrates, macronutrients that are sensed via distinct gut-brain pathways and differentially affect peripheral hormones and metabolism. However, the effects of such diet changes on the human brain are unclear. METHODSWe investigated whether selective isocaloric reductions in dietary fat or carbohydrates altered dopamine D2/3 receptor binding potential (D2BP) and neural activity in brain-reward regions in response to visual food cues in 17 inpatient adults with obesity as compared with a eucaloric baseline diet using a randomized crossover design. RESULTSOn the fifth day of dietary fat restriction, but not carbohydrate restriction, both D2BP and neural activity to food cues were decreased in brain-reward regions. After the reduced-fat diet, ad libitum intake shifted toward foods high in both fat and carbohydrates. CONCLUSIONThese results suggest that dietary fat restriction increases tonic dopamine in brain-reward regions and affects food choice in ways that may hamper diet adherence. TRIAL REGISTRATIONClinicalTrials.gov NCT00846040 FUNDING. NIDDK 1ZIADK013037.
Collapse
Affiliation(s)
- Valerie L Darcey
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, Bethesda, Maryland, USA
| | - Juen Guo
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, Bethesda, Maryland, USA
| | - Amber B Courville
- Human Energy and Body Weight Regulation Core, National Institute of Diabetes & Digestive & Kidney Diseases, Bethesda, Maryland, USA
| | - Isabelle Gallagher
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, Bethesda, Maryland, USA
| | - Jason A Avery
- Laboratory of Brain and Cognition, National Institute of Mental Health, Rockland, Maryland, USA
| | - W Kyle Simmons
- Biomedical Imaging Center, Oklahoma State University, Stillwater, Oklahoma, USA
| | - John E Ingeholm
- Laboratory of Brain and Cognition, National Institute of Mental Health, Rockland, Maryland, USA
| | - Peter Herscovitch
- Clinical Center Positron Emission Tomography Department, NIH, Bethesda, Maryland, USA
| | - Alex Martin
- Laboratory of Brain and Cognition, National Institute of Mental Health, Rockland, Maryland, USA
| | - Kevin D Hall
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, Bethesda, Maryland, USA
| |
Collapse
|
31
|
van Galen KA, Schrantee A, Ter Horst KW, la Fleur SE, Booij J, Constable RT, Schwartz GJ, DiLeone RJ, Serlie MJ. Brain responses to nutrients are severely impaired and not reversed by weight loss in humans with obesity: a randomized crossover study. Nat Metab 2023:10.1038/s42255-023-00816-9. [PMID: 37308722 DOI: 10.1038/s42255-023-00816-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/04/2023] [Indexed: 06/14/2023]
Abstract
Post-ingestive nutrient signals to the brain regulate eating behaviour in rodents, and impaired responses to these signals have been associated with pathological feeding behaviour and obesity. To study this in humans, we performed a single-blinded, randomized, controlled, crossover study in 30 humans with a healthy body weight (females N = 12, males N = 18) and 30 humans with obesity (females N = 18, males N = 12). We assessed the effect of intragastric glucose, lipid and water (noncaloric isovolumetric control) infusions on the primary endpoints cerebral neuronal activity and striatal dopamine release, as well as on the secondary endpoints plasma hormones and glucose, hunger scores and caloric intake. To study whether impaired responses in participants with obesity would be partially reversible with diet-induced weight loss, imaging was repeated after 10% diet-induced weight loss. We show that intragastric glucose and lipid infusions induce orosensory-independent and preference-independent, nutrient-specific cerebral neuronal activity and striatal dopamine release in lean participants. In contrast, participants with obesity have severely impaired brain responses to post-ingestive nutrients. Importantly, the impaired neuronal responses are not restored after diet-induced weight loss. Impaired neuronal responses to nutritional signals may contribute to overeating and obesity, and ongoing resistance to post-ingestive nutrient signals after significant weight loss may in part explain the high rate of weight regain after successful weight loss.
Collapse
Affiliation(s)
- Katy A van Galen
- Amsterdam UMC, location AMC, Department of Radiology and Nuclear Medicine, Amsterdam, the Netherlands
| | - Anouk Schrantee
- Amsterdam UMC, location AMC, Department of Radiology and Nuclear Medicine, Amsterdam, the Netherlands
| | - Kasper W Ter Horst
- Amsterdam University Medical Centers (UMC), location AMC, Department of Endocrinology and Metabolism and Amsterdam Gastroenterology Metabolism Endocrinology Institute, Amsterdam, the Netherlands
| | - Susanne E la Fleur
- Amsterdam University Medical Centers (UMC), location AMC, Department of Endocrinology and Metabolism and Amsterdam Gastroenterology Metabolism Endocrinology Institute, Amsterdam, the Netherlands
- Amsterdam UMC, location AMC, Department of Clinical Chemistry, Laboratory of Endocrinology, Amsterdam, the Netherlands
| | - Jan Booij
- Amsterdam UMC, location AMC, Department of Radiology and Nuclear Medicine, Amsterdam, the Netherlands
| | - R Todd Constable
- Yale University School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, CT, USA
| | - Gary J Schwartz
- Albert Einstein College of Medicine, Fleischer Institute for Diabetes and Metabolism, Bronx, NY, USA
| | - Ralph J DiLeone
- Yale University School of Medicine, Department of Psychiatry, New Haven, CT, USA
| | - Mireille J Serlie
- Amsterdam University Medical Centers (UMC), location AMC, Department of Endocrinology and Metabolism and Amsterdam Gastroenterology Metabolism Endocrinology Institute, Amsterdam, the Netherlands.
- Yale University School of Medicine, Department of Endocrinology, New Haven, CT, USA.
| |
Collapse
|
32
|
Serirukchutarungsee S, Watari I, Narukawa M, Podyma-Inoue KA, Sangsuriyothai P, Ono T. Two-generation exposure to a high-fat diet induces the change of salty taste preference in rats. Sci Rep 2023; 13:5742. [PMID: 37029190 PMCID: PMC10082214 DOI: 10.1038/s41598-023-31662-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 03/15/2023] [Indexed: 04/09/2023] Open
Abstract
High-fat diet (HFD) leads to multiple complications, including taste alteration. This study observed the effect of a two-generation exposure to an HFD on the peripheral taste system in offspring. Ten pregnant Wistar rats were assigned a standard diet (SD) (n = 5) or HFD (n = 5) from day 7 of pregnancy through the lactation. Thirty-six male and female 3-week-old offspring were measured for body weight and blood glucose level, and the circumvallate papillae were collected. The other twenty-four 3-week-old offspring were weaned on the same diet as their mothers and raised individually. The taste preference behaviors were studied using the two-bottle taste preference test and analyzed five basic tastes (sweet, bitter, umami, sour, and salty). The expressions of epithelial sodium channel alpha subunit (ENaCα) and angiotensin II receptor type 1 (AT1) in the circumvallate papilla were analyzed by immunohistochemical staining and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). We found increased body weight and salty taste preference of offspring from the HFD group in both sexes. Correspondingly, the AT1 level of the taste bud cells significantly increased in 3-week-old female offspring from the HFD group. An increase in AT1 levels may be a risk factor for changes in salty taste preference.
Collapse
Affiliation(s)
- Saranya Serirukchutarungsee
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo City, Tokyo, 113-8510, Japan
- Department of Pedodontics and Preventive Dentistry, Faculty of Dentistry, Srinakharinwirot University, Bangkok, Thailand
| | - Ippei Watari
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo City, Tokyo, 113-8510, Japan.
| | - Masataka Narukawa
- Department of Food and Nutrition, Kyoto Women's University, Kyoto, Japan
| | - Katarzyna Anna Podyma-Inoue
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Pornchanok Sangsuriyothai
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo City, Tokyo, 113-8510, Japan
- Department of Orthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Takashi Ono
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo City, Tokyo, 113-8510, Japan
| |
Collapse
|
33
|
De Filippo C, Costa A, Becagli MV, Monroy MM, Provensi G, Passani MB. Gut microbiota and oleoylethanolamide in the regulation of intestinal homeostasis. Front Endocrinol (Lausanne) 2023; 14:1135157. [PMID: 37091842 PMCID: PMC10113643 DOI: 10.3389/fendo.2023.1135157] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/22/2023] [Indexed: 04/08/2023] Open
Abstract
A vast literature strongly suggests that the endocannabinoid (eCB) system and related bioactive lipids (the paracannabinoid system) contribute to numerous physiological processes and are involved in pathological conditions such as obesity, type 2 diabetes, and intestinal inflammation. The gut paracannabinoid system exerts a prominent role in gut physiology as it affects motility, permeability, and inflammatory responses. Another important player in the regulation of host metabolism is the intestinal microbiota, as microorganisms are indispensable to protect the intestine against exogenous pathogens and potentially harmful resident microorganisms. In turn, the composition of the microbiota is regulated by intestinal immune responses. The intestinal microbial community plays a fundamental role in the development of the innate immune system and is essential in shaping adaptive immunity. The active interplay between microbiota and paracannabinoids is beginning to appear as potent regulatory system of the gastrointestinal homeostasis. In this context, oleoylethanolamide (OEA), a key component of the physiological systems involved in the regulation of dietary fat consumption, energy homeostasis, intestinal motility, and a key factor in modulating eating behavior, is a less studied lipid mediator. In the small intestine namely duodenum and jejunum, levels of OEA change according to the nutrient status as they decrease during food deprivation and increase upon refeeding. Recently, we and others showed that OEA treatment in rodents protects against inflammatory events and changes the intestinal microbiota composition. In this review, we briefly define the role of OEA and of the gut microbiota in intestinal homeostasis and recapitulate recent findings suggesting an interplay between OEA and the intestinal microorganisms.
Collapse
Affiliation(s)
- Carlotta De Filippo
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche, Pisa, Italy
| | - Alessia Costa
- Dipartimento di Scienze della Salute, Università di Firenze, Firenze, Italy
| | | | - Mariela Mejia Monroy
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche, Pisa, Italy
| | - Gustavo Provensi
- Dipartimento di Neurofarba, Università di Firenze, Firenze, Italy
- *Correspondence: Maria Beatrice Passani, ; Gustavo Provensi,
| | - Maria Beatrice Passani
- Dipartimento di Scienze della Salute, Università di Firenze, Firenze, Italy
- *Correspondence: Maria Beatrice Passani, ; Gustavo Provensi,
| |
Collapse
|
34
|
Santa-María C, López-Enríquez S, Montserrat-de la Paz S, Geniz I, Reyes-Quiroz ME, Moreno M, Palomares F, Sobrino F, Alba G. Update on Anti-Inflammatory Molecular Mechanisms Induced by Oleic Acid. Nutrients 2023; 15:nu15010224. [PMID: 36615882 PMCID: PMC9824542 DOI: 10.3390/nu15010224] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
In 2010, the Mediterranean diet was recognized by UNESCO as an Intangible Cultural Heritage of Humanity. Olive oil is the most characteristic food of this diet due to its high nutraceutical value. The positive effects of olive oil have often been attributed to its minor components; however, its oleic acid (OA) content (70-80%) is responsible for its many health properties. OA is an effective biomolecule, although the mechanism by which OA mediates beneficial physiological effects is not fully understood. OA influences cell membrane fluidity, receptors, intracellular signaling pathways, and gene expression. OA may directly regulate both the synthesis and activities of antioxidant enzymes. The anti-inflammatory effect may be related to the inhibition of proinflammatory cytokines and the activation of anti-inflammatory ones. The best-characterized mechanism highlights OA as a natural activator of sirtuin 1 (SIRT1). Oleoylethanolamide (OEA), derived from OA, is an endogenous ligand of the peroxisome proliferator-activated receptor alpha (PPARα) nuclear receptor. OEA regulates dietary fat intake and energy homeostasis and has therefore been suggested to be a potential therapeutic agent for the treatment of obesity. OEA has anti-inflammatory and antioxidant effects. The beneficial effects of olive oil may be related to the actions of OEA. New evidence suggests that oleic acid may influence epigenetic mechanisms, opening a new avenue in the exploration of therapies based on these mechanisms. OA can exert beneficial anti-inflammatory effects by regulating microRNA expression. In this review, we examine the cellular reactions and intracellular processes triggered by OA in T cells, macrophages, and neutrophils in order to better understand the immune modulation exerted by OA.
Collapse
Affiliation(s)
- Consuelo Santa-María
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Seville, 41012 Seville, Spain
- Correspondence: (C.S.-M.); (S.L.-E.)
| | - Soledad López-Enríquez
- Departamento de Bioquímica Médica, Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Seville, 41009 Seville, Spain
- Correspondence: (C.S.-M.); (S.L.-E.)
| | - Sergio Montserrat-de la Paz
- Departamento de Bioquímica Médica, Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Seville, 41009 Seville, Spain
| | - Isabel Geniz
- Distrito Sanitario Seville Norte y Aljarafe, Servicio Andaluz de Salud, 41008 Seville, Spain
| | - María Edith Reyes-Quiroz
- Departamento de Bioquímica Médica, Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Seville, 41009 Seville, Spain
| | - Manuela Moreno
- Departamento de Farmacia y Nutrición, Hospital Costa del Sol, 29603 Málaga, Spain
| | - Francisca Palomares
- Departamento de Bioquímica Médica, Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Seville, 41009 Seville, Spain
| | - Francisco Sobrino
- Departamento de Bioquímica Médica, Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Seville, 41009 Seville, Spain
| | - Gonzalo Alba
- Departamento de Bioquímica Médica, Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Seville, 41009 Seville, Spain
| |
Collapse
|
35
|
Igarashi M, Hayakawa T, Tanabe H, Watanabe K, Nishida A, Kimura I. Intestinal GPR119 activation by microbiota-derived metabolites impacts feeding behavior and energy metabolism. Mol Metab 2022; 67:101649. [PMID: 36462626 PMCID: PMC9771719 DOI: 10.1016/j.molmet.2022.101649] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
OBJECTIVE The gastrointestinal tract affects physiological activities and behavior by secreting hormones and generating signals through the activation of nutrient sensors. GPR119, a lipid sensor, is indirectly involved in the secretion of incretins, such as glucagon-like peptide-1 and glucose-dependent insulinotropic peptide, by enteroendocrine cells, while it directly stimulates insulin secretion by pancreatic beta cells. Since GPR119 has the potential to modulate metabolic homeostasis in obesity and diabetes, it has attracted interest as a therapeutic target. However, previous studies have shown that the deletion of Gpr119 in mice does not affect glucose homeostasis and appetite in either basal or high-fat diet-fed conditions. Therefore, the present study aimed to explore the role of GPR119 signaling system in energy metabolism and feeding behavior in mice. METHODS Gpr119 knockout (KO) mice were generated using CRISPR-Cas9 gene-editing technology, and their feeding behavior and energy metabolism were evaluated and compared with those of wild type (WT) mice. RESULTS Upon inducing metabolic stress via food deprivation, Gpr119 KO mice exhibited lower blood glucose levels and a higher body weight reduction compared to WT mice. Although food intake in WT and KO mice were similar under free-feeding conditions, Gpr119 KO mice exhibited increased food intake when they were refed after 24 h of food deprivation. Further, food-deprived Gpr119 KO mice presented shorter post-meal intervals and lower satiety for second and later meals during refeeding, resulting in increased food intake. Associated with this meal pattern, levels of oleoylethanolamide (OEA), an endogenous agonist of GPR119, in the luminal contents of the distal gastrointestinal tract were elevated within 2 h after refeeding. The large-intestinal infusion of OEA prolonged post-meal intervals and increased satiety in the first meal, but not the second meal. On the other hand, infusion of oleic acid increased cecal OEA levels at 2 h from the beginning of infusion, while prolonging post-meal intervals and increasing satiety on the meals that occurred approximately 2 h after the infusion. Cecal OEA levels were low in antibiotic-treated mice, suggesting that the gut microbiota partially synthesizes OEA from oleic acid. CONCLUSIONS Collectively, our results indicate that the activation of gastrointestinal GPR119 by microbiota-produced OEA derived from oleic acid is associated with satiety control and energy homeostasis under energy shortage conditions.
Collapse
Affiliation(s)
- Miki Igarashi
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai, Fuchu-City, Tokyo 183-8509, Japan; Advanced Clinical Research Center, Institute of Neurological Disorders, 255 Furusawa-Tsuko, Asao-ku, Kanagawa 215-0026, Japan.
| | - Tetsuhiko Hayakawa
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai, Fuchu-City, Tokyo 183-8509, Japan
| | - Haruka Tanabe
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Keita Watanabe
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Akari Nishida
- Laboratory of Molecular Neurobiology, Graduate School of Pharmaceutical Sciences, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Ikuo Kimura
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai, Fuchu-City, Tokyo 183-8509, Japan; Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; Laboratory of Molecular Neurobiology, Graduate School of Pharmaceutical Sciences, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
36
|
Lockridge A, Hanover JA. A nexus of lipid and O-Glcnac metabolism in physiology and disease. Front Endocrinol (Lausanne) 2022; 13:943576. [PMID: 36111295 PMCID: PMC9468787 DOI: 10.3389/fendo.2022.943576] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Although traditionally considered a glucose metabolism-associated modification, the O-linked β-N-Acetylglucosamine (O-GlcNAc) regulatory system interacts extensively with lipids and is required to maintain lipid homeostasis. The enzymes of O-GlcNAc cycling have molecular properties consistent with those expected of broad-spectrum environmental sensors. By direct protein-protein interactions and catalytic modification, O-GlcNAc cycling enzymes may provide both acute and long-term adaptation to stress and other environmental stimuli such as nutrient availability. Depending on the cell type, hyperlipidemia potentiates or depresses O-GlcNAc levels, sometimes biphasically, through a diversity of unique mechanisms that target UDP-GlcNAc synthesis and the availability, activity and substrate selectivity of the glycosylation enzymes, O-GlcNAc Transferase (OGT) and O-GlcNAcase (OGA). At the same time, OGT activity in multiple tissues has been implicated in the homeostatic regulation of systemic lipid uptake, storage and release. Hyperlipidemic patterns of O-GlcNAcylation in these cells are consistent with both transient physiological adaptation and feedback uninhibited obesogenic and metabolic dysregulation. In this review, we summarize the numerous interconnections between lipid and O-GlcNAc metabolism. These links provide insights into how the O-GlcNAc regulatory system may contribute to lipid-associated diseases including obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Amber Lockridge
- Laboratory of Cell and Molecular Biology, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - John A. Hanover
- Laboratory of Cell and Molecular Biology, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
37
|
Diao X, Ye F, Zhang M, Ren X, Tian X, Lu J, Sun X, Hou Z, Chen X, Li F, Zhuang J, Ding H, Peng C, Rastinejad F, Luo C, Wu D. Identification of oleoylethanolamide as an endogenous ligand for HIF-3α. Nat Commun 2022; 13:2529. [PMID: 35534502 PMCID: PMC9085743 DOI: 10.1038/s41467-022-30338-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 04/26/2022] [Indexed: 12/15/2022] Open
Abstract
Hypoxia-inducible factors (HIFs) are α/β heterodimeric transcription factors modulating cellular responses to the low oxygen condition. Among three HIF-α isoforms, HIF-3α is the least studied to date. Here we show that oleoylethanolamide (OEA), a physiological lipid known to regulate food intake and metabolism, binds selectively to HIF-3α. Through crystallographic analysis of HIF-3 α/β heterodimer in both apo and OEA-bound forms, hydrogen-deuterium exchange mass spectrometry (HDX-MS), molecular dynamics (MD) simulations, and biochemical and cell-based assays, we unveil the molecular mechanism of OEA entry and binding to the PAS-B pocket of HIF-3α, and show that it leads to enhanced heterodimer stability and functional modulation of HIF-3. The identification of HIF-3α as a selective lipid sensor is consistent with recent human genetic findings linking HIF-3α with obesity, and demonstrates that endogenous metabolites can directly interact with HIF-α proteins to modulate their activities, potentially as a regulatory mechanism supplementary to the well-known oxygen-dependent HIF-α hydroxylation.
Collapse
Affiliation(s)
- Xiaotong Diao
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, 266237, Qingdao, China
| | - Fei Ye
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 310053, Hangzhou, China
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 310018, Hangzhou, China
| | - Meina Zhang
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, 266237, Qingdao, China
| | - Xintong Ren
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, 266237, Qingdao, China
| | - Xiaoxu Tian
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, 201210, Shanghai, China
| | - Jingping Lu
- Target Discovery Institute, NDM Research Building, University of Oxford, Old Road Campus, Oxford, OX3 7FZ, UK
| | - Xiangnan Sun
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, 266237, Qingdao, China
| | - Zeng Hou
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 310053, Hangzhou, China
- Drug Discovery and Design Center, the Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Xiaoyu Chen
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, 266237, Qingdao, China
| | - Fengwei Li
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, 266237, Qingdao, China
| | - Jingjing Zhuang
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, 266237, Qingdao, China
| | - Hong Ding
- Drug Discovery and Design Center, the Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Chao Peng
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, 201210, Shanghai, China
| | - Fraydoon Rastinejad
- Target Discovery Institute, NDM Research Building, University of Oxford, Old Road Campus, Oxford, OX3 7FZ, UK.
| | - Cheng Luo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 310053, Hangzhou, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 310053, Hangzhou, China.
- Drug Discovery and Design Center, the Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.
| | - Dalei Wu
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, 266237, Qingdao, China.
| |
Collapse
|
38
|
Sionov RV, Steinberg D. Anti-Microbial Activity of Phytocannabinoids and Endocannabinoids in the Light of Their Physiological and Pathophysiological Roles. Biomedicines 2022; 10:biomedicines10030631. [PMID: 35327432 PMCID: PMC8945038 DOI: 10.3390/biomedicines10030631] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/06/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022] Open
Abstract
Antibiotic resistance has become an increasing challenge in the treatment of various infectious diseases, especially those associated with biofilm formation on biotic and abiotic materials. There is an urgent need for new treatment protocols that can also target biofilm-embedded bacteria. Many secondary metabolites of plants possess anti-bacterial activities, and especially the phytocannabinoids of the Cannabis sativa L. varieties have reached a renaissance and attracted much attention for their anti-microbial and anti-biofilm activities at concentrations below the cytotoxic threshold on normal mammalian cells. Accordingly, many synthetic cannabinoids have been designed with the intention to increase the specificity and selectivity of the compounds. The structurally unrelated endocannabinoids have also been found to have anti-microbial and anti-biofilm activities. Recent data suggest for a mutual communication between the endocannabinoid system and the gut microbiota. The present review focuses on the anti-microbial activities of phytocannabinoids and endocannabinoids integrated with some selected issues of their many physiological and pharmacological activities.
Collapse
|
39
|
Clark TD, Crean AJ, Senior AM. Obesogenic diets induce anxiety in rodents: A systematic review and meta-analysis. Obes Rev 2022; 23:e13399. [PMID: 34811885 DOI: 10.1111/obr.13399] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/27/2021] [Accepted: 10/31/2021] [Indexed: 01/04/2023]
Abstract
Obesity and mood disorders have been linked in a positive feedback loop. However, due to the bidirectional relationship between obesity and mental health, it is not clear whether anxiety is correlated with or caused by consumption of obesogenic diets. Here, we present a meta-analysis on the effects of dietary manipulation on rodent behavior in the elevated plus maze and open field test, the most common tests of anxiety-like behavior in animal models. The main dataset examined effects of obesogenic diets on time spent in the open and movement around the mazes. Auxiliary datasets examined effects of caloric restriction and protein restriction. Obesogenic diets had a negative effect on the time spent in the open overall. Effects were stronger in males than females. No effect of diet on locomotion was found, indicating that increases in anxiety-like behavior are not due to altered activity levels. No consistent effect of caloric restriction was observed, whereas protein restriction was associated with an increase in the time spent in the open. We show that obesogenic diets have a causative effect on anxiety-like behaviors in rodents, and not via a change in activity. Further studies are required to determine why obesogenic diets induce these behavioral changes.
Collapse
Affiliation(s)
- Thomas D Clark
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Angela J Crean
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Alistair M Senior
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
40
|
Lebrun LJ, Moreira S, Tavernier A, Niot I. Postprandial consequences of lipid absorption in the onset of obesity: Role of intestinal CD36. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159154. [DOI: 10.1016/j.bbalip.2022.159154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 10/18/2022]
|
41
|
Abstract
The endocannabinoid system is found in most, if not all, mammalian organs and is involved in a variety of physiological functions, ranging from the control of synaptic plasticity in the brain to the modulation of smooth muscle motility in the gastrointestinal tract. This signaling complex consists of G protein-coupled cannabinoid receptors, endogenous ligands for those receptors (endocannabinoids) and enzymes/transporters responsible for the formation and deactivation of these ligands. There are two subtypes of cannabinoid receptors, CB1 and CB2, and two major endocannabinoids, arachidonoylethanolamide (anandamide) and 2-arachidonoyl-sn-glycerol (2-AG), which are produced upon demand through cleavage of distinct phospholipid precursors. All molecular components of the endocannabinoid system are represented in the adipose organ, where endocannabinoid signals are thought to regulate critical homeostatic processes, including adipogenesis, lipogenesis and thermogenesis. Importantly, obesity was found to be associated with excess endocannabinoid activity in visceral fat depots, and the therapeutic potential of normalizing such activity by blocking CB1 receptors has been the focus of substantial preclinical and clinical research. Results have been mixed thus far, mostly owing to the emergence of psychiatric side effects rooted in the protective functions served by brain endocannabinoids in mood and affect regulation. Further studies about the roles played by the endocannabinoid system in the adipose organ will offer new insights into the pathogenesis of obesity and might help identify new ways to leverage this signaling complex for therapeutic benefit.
Collapse
Affiliation(s)
- Kwang-Mook Jung
- Department of Anatomy and Neurobiology, University of California, Irvine, 3101 Gillespie NRF, Irvine, CA, 92697-1275, USA
| | - Lin Lin
- Department of Anatomy and Neurobiology, University of California, Irvine, 3101 Gillespie NRF, Irvine, CA, 92697-1275, USA
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California, Irvine, 3101 Gillespie NRF, Irvine, CA, 92697-1275, USA.
- Department of Pharmacology, University of California, Irvine, Irvine, CA, 92697, USA.
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
42
|
Luo Z, Yu Z, Yin D. Obesogenic effect of erythromycin on Caenorhabditis elegans through over-eating and lipid metabolism disturbances. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 294:118615. [PMID: 34863891 DOI: 10.1016/j.envpol.2021.118615] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/08/2021] [Accepted: 11/29/2021] [Indexed: 06/13/2023]
Abstract
Environmental obesogens contributed significantly to the obesity prevalence. Recently, antibiotics joined the list of environmental obesogens, while the underlying mechanisms remained to be explored. In the present study, effects of erythromycin (ERY), one widely used macrolide antibiotic, were measured on C. elegans to investigate the obesogenic mechanism. Results showed that ERY at 0.1 μg/L significantly increased the fat content by 17.4% more than the control and also stimulated triacylglycerol (TAG) levels by 25.7% more than the control. Regarding the obesogenic mechanisms, ERY provoked over-eating by stimulation on the pharyngeal pumping and reduction on the satiety quiescence percentage and duration. Such effects were resulted from stimulation on the neurotransmitters including serotonin (5-HT), dopamine (DA) and acetylcholine (ACh). The nervous responses involved the up-regulation of Gsα (e.g., ser-7, gsa-1, acy-1 and kin-2) signaling pathway and the down-regulation of TGFβ (daf-7) but not via cGMP-dependent regulations (e.g., egl-4). Moreover, ERY stimulated the activities of fatty acid synthase (FAS) and glycerol-3-phosphateacyl transferases (GPAT) that catalyze lipogenesis, while ERY inhibited those of acyl-CoA synthetase (ACS), carnitine palmitoyl transferase (CPT) and acyl-CoA oxidase (ACO) that catalyze lipolysis. The unbalance between lipogenesis and lipolysis resulted in the fat accumulation which was consistent with up-regulation on mgl-1 and mgl-3 which are the down-steam of TGFβ regulation. Such consistence supported the close connection between nervous regulation and lipid metabolism. In addition, ERY also disturbed insulin which connects lipid with glucose in metabolism.
Collapse
Affiliation(s)
- Zhili Luo
- State Key Laboratory of Pollution Control and Resource Reuse, Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, PR China; Jiaxing Tongji Institute for Environment, Jiaxing, Zhejiang Province, 3014051, PR China; Shanghai Institute of Pollution Control and Ecological Security, Shanghai, 200092, PR China
| | - Zhenyang Yu
- State Key Laboratory of Pollution Control and Resource Reuse, Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, PR China; Jiaxing Tongji Institute for Environment, Jiaxing, Zhejiang Province, 3014051, PR China; Shanghai Institute of Pollution Control and Ecological Security, Shanghai, 200092, PR China.
| | - Daqiang Yin
- State Key Laboratory of Pollution Control and Resource Reuse, Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, PR China; Shanghai Institute of Pollution Control and Ecological Security, Shanghai, 200092, PR China
| |
Collapse
|
43
|
Lisuzzo A, Bonelli F, Sgorbini M, Nocera I, Cento G, Mazzotta E, Turini L, Martini M, Salari F, Morgante M, Badon T, Fiore E. Differences of the Plasma Total Lipid Fraction from Pre-Foaling to Post-Foaling Period in Donkeys. Animals (Basel) 2022; 12:ani12030304. [PMID: 35158628 PMCID: PMC8833319 DOI: 10.3390/ani12030304] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 12/16/2022] Open
Abstract
Simple Summary An association between increased metabolic demands and reduced dry matter intake is observed from late gestation to early lactation in donkeys. Furthermore, little is known about the nutritional and energy requirements of this period in animals. Changes in energy metabolism make donkeys more susceptible to metabolic diseases such as hyperlipemia, which is characterized by the mobilization of fatty acids from adipose tissue. A better knowledge of this period could improve animal husbandry, well-being, and health. The aim of this study was to analyze the plasma total lipid fraction, to highlight metabolic changes from the pre-foaling to post-foaling periods, using the gas chromatography technique. Our findings reveal a greater risk of metabolic disease in late gestation to early lactation in donkeys. Abstract The period from late gestation to early lactation is characterized by changes in energy metabolism. The aim of the current study was to analyze the plasma total lipid fraction using gas chromatography (GC) analysis, in order to highlight metabolic changes from the pre-foaling to post-foaling periods. Eleven pluriparous dairy jennies (mean age of 11.88 ± 3.79 years) belonging to the Amiata donkey breed were enrolled. Blood sampling was performed at 15 days before foaling (T0), and 15 (T1), 30 (T2), 60 (T3), and 90 (T4) days after foaling, for biochemical and GC analysis. A total of 37 fatty acids were identified in plasma samples: 4 medium chain (MCFA), 24 long chain (LCFA), and 9 very-long chain (VLCFA) fatty acids. Among them, 20 fatty acids changed significantly, and two fatty acid showed a trend toward significance. Furthermore, the LCFA, saturated, unsaturated, monounsaturated, and polyunsaturated ω-3 fatty acids changed significantly during the study period. The main alterations were between T0 and the other time points and appeared to be related to lipid metabolism, cellular structure and function, and inflammatory and immune responses. Our findings reveal greater energy requirements at the end of gestation compared to early lactation in donkeys.
Collapse
Affiliation(s)
- Anastasia Lisuzzo
- Department of Animal Medicine, Production and Health, University of Padova, Viale dell’Università 16, 35020 Legnaro, Italy; (A.L.); (G.C.); (E.M.); (M.M.); (T.B.)
| | - Francesca Bonelli
- Department of Veterinary Science, University of Pisa, Viale delle Piagge 2, 56124 Pisa, Italy; (F.B.); (M.S.); (I.N.); (L.T.); (M.M.); (F.S.)
| | - Micaela Sgorbini
- Department of Veterinary Science, University of Pisa, Viale delle Piagge 2, 56124 Pisa, Italy; (F.B.); (M.S.); (I.N.); (L.T.); (M.M.); (F.S.)
| | - Irene Nocera
- Department of Veterinary Science, University of Pisa, Viale delle Piagge 2, 56124 Pisa, Italy; (F.B.); (M.S.); (I.N.); (L.T.); (M.M.); (F.S.)
| | - Giulia Cento
- Department of Animal Medicine, Production and Health, University of Padova, Viale dell’Università 16, 35020 Legnaro, Italy; (A.L.); (G.C.); (E.M.); (M.M.); (T.B.)
| | - Elisa Mazzotta
- Department of Animal Medicine, Production and Health, University of Padova, Viale dell’Università 16, 35020 Legnaro, Italy; (A.L.); (G.C.); (E.M.); (M.M.); (T.B.)
| | - Luca Turini
- Department of Veterinary Science, University of Pisa, Viale delle Piagge 2, 56124 Pisa, Italy; (F.B.); (M.S.); (I.N.); (L.T.); (M.M.); (F.S.)
| | - Mina Martini
- Department of Veterinary Science, University of Pisa, Viale delle Piagge 2, 56124 Pisa, Italy; (F.B.); (M.S.); (I.N.); (L.T.); (M.M.); (F.S.)
| | - Federica Salari
- Department of Veterinary Science, University of Pisa, Viale delle Piagge 2, 56124 Pisa, Italy; (F.B.); (M.S.); (I.N.); (L.T.); (M.M.); (F.S.)
| | - Massimo Morgante
- Department of Animal Medicine, Production and Health, University of Padova, Viale dell’Università 16, 35020 Legnaro, Italy; (A.L.); (G.C.); (E.M.); (M.M.); (T.B.)
| | - Tamara Badon
- Department of Animal Medicine, Production and Health, University of Padova, Viale dell’Università 16, 35020 Legnaro, Italy; (A.L.); (G.C.); (E.M.); (M.M.); (T.B.)
| | - Enrico Fiore
- Department of Animal Medicine, Production and Health, University of Padova, Viale dell’Università 16, 35020 Legnaro, Italy; (A.L.); (G.C.); (E.M.); (M.M.); (T.B.)
- Correspondence:
| |
Collapse
|
44
|
Igarashi M, Iwasa K, Hayakawa T, Tsuduki T, Kimura I, Maruyama K, Yoshikawa K. Dietary oleic acid contributes to the regulation of food intake through the synthesis of intestinal oleoylethanolamide. Front Endocrinol (Lausanne) 2022; 13:1056116. [PMID: 36733808 PMCID: PMC9886573 DOI: 10.3389/fendo.2022.1056116] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/07/2022] [Indexed: 01/18/2023] Open
Abstract
INTRODUCTION Among the fatty acid ethanolamides (FAEs), oleoylethanolamide (OEA), linoleoylethanolamide (LEA), and palmitoylethanolamide (PEA) are reported to be involved in feeding regulation. In particular, OEA is well characterized as a satiety signal. Following food consumption, OEA is synthesized from oleic acid (OA) via an N-acyl phosphatidylethanolamine-specific phospholipase D-dependent pathway in the gastroenterocytes, and OEA induces satiety by recruiting sensory fibers. Thus, we hypothesized that dietary OA is an important satiety-inducing molecule. However, there has been no direct demonstration of the effect of dietary OA on satiety induction without the influence of the endogenous biosynthesis of OA from stearic acid (SA) or other FAEs. METHODS In this study, we used two experimental diets to test our hypothesis: (i) an OA diet (OAD; 38.4 mg of OA/g and 7.2 mg of SA/g) and (ii) a low OA diet (LOAD; 3.1 mg of OA/g and 42.4 mg of SA/g). RESULTS Relative to mice fed the OAD, mice fed the LOAD for two weeks exhibited reduced levels of jejunal OEA but not jejunal LEA and PEA. The LOAD-fed mice showed an increase in food intake and body weight gain. Moreover, LOAD-induced increase in food intake was immediately observed after the switch from the OAD, whereas these effects were diminished by the switch back to the OAD. Furthermore, treatment with OA and OEA diminished the effects of LOAD on food intake. CONCLUSION Collectively, these results show that dietary OA is a key factor in the reduction of food intake and increase in satiety mediated by OEA signaling.
Collapse
Affiliation(s)
- Miki Igarashi
- Advanced Clinical Research Center, Institute of Neurological Disorders, Kawasaki, Japan
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
- *Correspondence: Miki Igarashi,
| | - Kensuke Iwasa
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Tetsuhiko Hayakawa
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Tsuyoshi Tsuduki
- Department of Bioscience and Biotechnology for Future Bioindustries, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Ikuo Kimura
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Kei Maruyama
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Keisuke Yoshikawa
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| |
Collapse
|
45
|
Gish A, Wiart JF, Turpin E, Allorge D, Gaulier JM. État de l’art et intérêt des dosages plasmatiques des substances endocannabinoïdes et endocannabinoïdes-like. TOXICOLOGIE ANALYTIQUE ET CLINIQUE 2021. [DOI: 10.1016/j.toxac.2021.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
46
|
Schumann L, Wilken-Schmitz A, Trautmann S, Vogel A, Schreiber Y, Hahnefeld L, Gurke R, Geisslinger G, Tegeder I. Increased Fat Taste Preference in Progranulin-Deficient Mice. Nutrients 2021; 13:4125. [PMID: 34836380 PMCID: PMC8623710 DOI: 10.3390/nu13114125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/22/2021] [Accepted: 11/12/2021] [Indexed: 11/25/2022] Open
Abstract
Progranulin deficiency in mice is associated with deregulations of the scavenger receptor signaling of CD36/SCARB3 in immune disease models, and CD36 is a dominant receptor in taste bud cells in the tongue and contributes to the sensation of dietary fats. Progranulin-deficient mice (Grn-/-) are moderately overweight during middle age. We therefore asked if there was a connection between progranulin/CD36 in the tongue and fat taste preferences. By using unbiased behavioral analyses in IntelliCages and Phenomaster cages we showed that progranulin-deficient mice (Grn-/-) developed a strong preference of fat taste in the form of 2% milk over 0.3% milk, and for diluted MCTs versus tap water. The fat preference in the 7d-IntelliCage observation period caused an increase of 10% in the body weight of Grn-/- mice, which did not occur in the wildtype controls. CD36 expression in taste buds was reduced in Grn-/- mice at RNA and histology levels. There were no differences in the plasma or tongue lipids of various classes including sphingolipids, ceramides and endocannabinoids. The data suggest that progranulin deficiency leads to a lower expression of CD36 in the tongue resulting in a stronger urge for fatty taste and fatty nutrition.
Collapse
Affiliation(s)
- Lana Schumann
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
| | - Annett Wilken-Schmitz
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
| | - Sandra Trautmann
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
| | - Alexandra Vogel
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
| | - Yannick Schreiber
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt, Germany
| | - Lisa Hahnefeld
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt, Germany
| | - Robert Gurke
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), 60596 Frankfurt, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
| |
Collapse
|
47
|
Ulu A, Burr A, Heires AJ, Pavlik J, Larsen T, Perez PA, Bravo C, DiPatrizio NV, Baack M, Romberger DJ, Nordgren TM. A high docosahexaenoic acid diet alters lung inflammation and recovery following repetitive exposure to aqueous organic dust extracts. J Nutr Biochem 2021; 97:108797. [PMID: 34126202 PMCID: PMC8725620 DOI: 10.1016/j.jnutbio.2021.108797] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 12/15/2022]
Abstract
Agricultural workers, especially those who work in swine confinement facilities, are at increased risk for developing pulmonary diseases including asthma, chronic obstructive pulmonary disease, and chronic bronchitis due to exposures to fumes, vapors, and organic dust. Repetitive exposure to agricultural dust leads to unresolved inflammation, a common underlying mechanism that worsens lung disease. Besides occupational exposure to dusts, diet also significantly contributes to inflammation and disease progression. Since DHA (docosahexaenoic acid), a polyunsaturated omega-3 fatty acid and its bioactive metabolites have key roles in inflammation resolution, we rationalized that individuals chronically exposed to organic dusts can benefit from dietary modifications. Here, we evaluated the role of DHA in modifying airway inflammation in a murine model of repetitive exposure to an aqueous extract of agricultural dust (three-week exposure to swine confinement dust extract, HDE) and after a one-week resolution/recovery period. We found that mice fed a high DHA diet had significantly increased bronchoalveolar lavage fluid (BALF) levels of DHA-derived resolvins and lower TNFα along with altered plasma levels of endocannabinoids and related lipid mediators. Following the one-week recovery we identified significantly reduced BALF cellularity and cytokine/chemokine release along with increased BALF amphiregulin and resolvins in DHA diet-fed versus control diet-fed mice challenged with HDE. We further report observations on the effects of repetitive HDE exposure on lung Ym1+ and Arg-1+ macrophages. Overall, our findings support a protective role for DHA and identify DHA-derived resolvins and endocannabinoids among the potential mediators of DHA in altering airway inflammation in chronic agricultural dust exposure.
Collapse
Affiliation(s)
- Arzu Ulu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
| | - Abigail Burr
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
| | - Art J Heires
- Pulmonary, Critical Care, Sleep and Allergy Division, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jacqueline Pavlik
- Pulmonary, Critical Care, Sleep and Allergy Division, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Tricia Larsen
- Children's Health Research Center, Sanford Research, Sioux Falls, South Dakota, USA
| | - Pedro A Perez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
| | - Carissa Bravo
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
| | - Nicholas V DiPatrizio
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
| | - Michelle Baack
- Children's Health Research Center, Sanford Research, Sioux Falls, South Dakota, USA; Division of Neonatology, University of South Dakota-Sanford School of Medicine, Sioux Falls, South Dakota, USA
| | - Debra J Romberger
- VA Nebraska-Western Iowa Healthcare System, Omaha, Nebraska, USA; Pulmonary, Critical Care, Sleep and Allergy Division, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Tara M Nordgren
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA; Pulmonary, Critical Care, Sleep and Allergy Division, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| |
Collapse
|
48
|
Burman A, Kaji I. Luminal Chemosensory Cells in the Small Intestine. Nutrients 2021; 13:nu13113712. [PMID: 34835968 PMCID: PMC8620795 DOI: 10.3390/nu13113712] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/23/2022] Open
Abstract
In addition to the small intestine's well-known function of nutrient absorption, the small intestine also plays a major role in nutrient sensing. Similar to taste sensors seen on the tongue, GPCR-coupled nutrient sensors are expressed throughout the intestinal epithelium and respond to nutrients found in the lumen. These taste receptors respond to specific ligands, such as digested carbohydrates, fats, and proteins. The activation of nutrient sensors in the intestine allows for the induction of signaling pathways needed for the digestive system to process an influx of nutrients. Such processes include those related to glucose homeostasis and satiety. Defects in intestinal nutrient sensing have been linked to a variety of metabolic disorders, such as type 2 diabetes and obesity. Here, we review recent updates in the mechanisms related to intestinal nutrient sensors, particularly in enteroendocrine cells, and their pathological roles in disease. Additionally, we highlight the emerging nutrient sensing role of tuft cells and recent work using enteroids as a sensory organ model.
Collapse
Affiliation(s)
- Andreanna Burman
- Cell and Developmental Biology and Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA;
| | - Izumi Kaji
- Epithelial Biology Center and Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Correspondence:
| |
Collapse
|
49
|
Berland C, Small DM, Luquet S, Gangarossa G. Dietary lipids as regulators of reward processes: multimodal integration matters. Trends Endocrinol Metab 2021; 32:693-705. [PMID: 34148784 DOI: 10.1016/j.tem.2021.05.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/08/2021] [Accepted: 05/24/2021] [Indexed: 02/03/2023]
Abstract
The abundance of energy-dense and palatable diets in the modern food environment tightly contributes to the obesity pandemic. The reward circuit participates to the regulation of body homeostasis by integrating energy-related signals with neural substrates encoding cognitive and motivational components of feeding behaviors. Obesity and lipid-rich diets alter dopamine (DA) transmission leading to reward dysfunctions and food overconsumption. Recent reports indicate that dietary lipids can act, directly and indirectly, as functional modulators of the DA circuit. This raises the possibility that nutritional or genetic conditions affecting 'lipid sensing' mechanisms might lead to maladaptations of the DA system. Here, we discuss the most recent findings connecting dietary lipid sensing with DA signaling and its multimodal influence on circuits regulating food-reward processes.
Collapse
Affiliation(s)
- Chloé Berland
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France; Department of Medicine, The Naomi Berrie Diabetes Center, Columbia University, New York, NY 10032, USA
| | - Dana M Small
- Department of Psychiatry, and the Modern Diet and Physiology Research Center, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Serge Luquet
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France.
| | | |
Collapse
|
50
|
A Duet Between Histamine and Oleoylethanolamide in the Control of Homeostatic and Cognitive Processes. Curr Top Behav Neurosci 2021; 59:389-410. [PMID: 34410679 DOI: 10.1007/7854_2021_236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In ballet, a pas de deux (in French it means "step of two") is a duet in which the two dancers perform ballet steps together. The suite of dances shares a common theme of partnership. How could we better describe the fine interplay between oleoylethanolamide (OEA) and histamine, two phylogenetically ancient molecules controlling metabolic, homeostatic and cognitive processes? Contrary to the pas de deux though, the two dancers presumably never embrace each other as a dancing pair but execute their "virtuoso solo" constantly exchanging interoceptive messages presumably via vagal afferents, the blood stream, the neuroenteric system. With one exception, which is in the control of liver ketogenesis, as in hepatocytes, OEA biosynthesis strictly depends on the activation of histaminergic H1 receptors. In this review, we recapitulate our main findings that evidence the interplay of histamine and OEA in the control of food consumption and eating behaviour, in the consolidation of emotional memory and mood, and finally, in the synthesis of ketone bodies. We will also summarise some of the putative underlying mechanisms for each scenario.
Collapse
|