1
|
Dagbasi A, Fuller A, Hanyaloglu AC, Carroll B, McLaughlin J, Frost G, Holliday A. The role of nutrient sensing dysregulation in anorexia of ageing: The little we know and the much we don't. Appetite 2024; 203:107718. [PMID: 39423861 DOI: 10.1016/j.appet.2024.107718] [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: 04/08/2024] [Revised: 08/01/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
The age-related decline in appetite and food intake - termed "anorexia of ageing" - is implicated in undernutrition in later life and hence provides a public health challenge for our ageing population. Eating behaviour is controlled, in part, by homeostatic mechanisms which sense nutrient status and provide feedback to appetite control regions of the brain. Such feedback signals, propagated by episodic gut hormones, are dysregulated in some older adults. The secretory responses of appetite-related gut hormones to feeding are amplified, inducing a more anorexigenic signal which is associated with reduced appetite and food intake. Such an augmented response would indicate an increase in gut sensitivity to nutrients. Consequently, this review explores the role of gastrointestinal tract nutrient sensing in age-related appetite dysregulation. We review and synthesise evidence for age-related alterations in nutrient sensing which may explain the observed hormonal dysregulation. Drawing on what is known regarding elements of nutrient sensing pathways in animal models, in other tissues of the body, and in certain models of disease, we identify potential causal mechanisms including alterations in enteroendocrine cell number and distribution, dysregulation of cell signalling pathways, and changes in the gut milieu. From identified gaps in evidence, we highlight interesting and important avenues for future research.
Collapse
Affiliation(s)
- Aygul Dagbasi
- Section of Nutrition, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London, W12 0NN, UK
| | - Amy Fuller
- Research Centre for Health and Life Sciences, Institute of Health and Wellbeing, Faculty of Health and Life Science, Coventry University, Coventry, CV1 5FB, UK
| | - Aylin C Hanyaloglu
- Institute of Reproductive and Developmental Biology (IRDB), Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Bernadette Carroll
- School of Biochemistry, University of Bristol, University Walk, Bristol, BS1 8TD, UK
| | - John McLaughlin
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester and Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK
| | - Gary Frost
- Section of Nutrition, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London, W12 0NN, UK
| | - Adrian Holliday
- School of Biomedical, Nutritional, and Sport Science, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK; Human Nutrition and Exercise Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK.
| |
Collapse
|
2
|
Jiang L, Han D, Hao Y, Song Z, Sun Z, Dai Z. Linking serotonin homeostasis to gut function: Nutrition, gut microbiota and beyond. Crit Rev Food Sci Nutr 2024; 64:7291-7310. [PMID: 36861222 DOI: 10.1080/10408398.2023.2183935] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Serotonin (5-HT) produced by enterochromaffin (EC) cells in the digestive tract is crucial for maintaining gut function and homeostasis. Nutritional and non-nutritional stimuli in the gut lumen can modulate the ability of EC cells to produce 5-HT in a temporal- and spatial-specific manner that toning gut physiology and immune response. Of particular interest, the interactions between dietary factors and the gut microbiota exert distinct impacts on gut 5-HT homeostasis and signaling in metabolism and the gut immune response. However, the underlying mechanisms need to be unraveled. This review aims to summarize and discuss the importance of gut 5-HT homeostasis and its regulation in maintaining gut metabolism and immune function in health and disease with special emphasis on different types of nutrients, dietary supplements, processing, and gut microbiota. Cutting-edge discoveries in this area will provide the basis for the development of new nutritional and pharmaceutical strategies for the prevention and treatment of serotonin homeostasis-related gut and systematic disorders and diseases.
Collapse
Affiliation(s)
- Lili Jiang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Youling Hao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Zhuan Song
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Zhiyuan Sun
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| |
Collapse
|
3
|
Dontamsetti KD, Pedrosa‐Suarez LC, Aktar R, Peiris M. Sensing of luminal contents and downstream modulation of GI function. JGH Open 2024; 8:e13083. [PMID: 38779131 PMCID: PMC11109814 DOI: 10.1002/jgh3.13083] [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: 03/19/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Abstract
The luminal environment is rich in macronutrients coming from our diet and resident microbial populations including their metabolites. Together, they have the capacity to modulate unique cell surface receptors, known as G-protein coupled receptors (GPCRs). Along the entire length of the gut epithelium, enteroendocrine cells express GPCRs to interact with luminal contents, such as GPR93 and the calcium sensing receptor to sense proteins, FFA2 and GPR84 to sense fatty acids, and SGLT1 and T1R to sense carbohydrates. Nutrient-receptor interaction causes the release of hormonal stores such as glucagon-like peptide 1, peptide YY, and cholecystokinin, which further regulate gut function. Existing data show the role of luminal components and microbial fermentation products on gut function. However, there is a lack of understanding in the mechanistic interactions between diet-derived luminal components and microbial products and nutrient-sensing receptors and downstream gastrointestinal modulation. This review summarizes current knowledge on various luminal components and describes in detail the range of nutrients and metabolites and their interaction with nutrient receptors in the gut epithelium and the emerging impact on immune cells.
Collapse
Affiliation(s)
- Kiran Devi Dontamsetti
- Centre for Neuroscience, Surgery & Trauma, Blizard Institute, Barts and The London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Laura Camila Pedrosa‐Suarez
- Centre for Neuroscience, Surgery & Trauma, Blizard Institute, Barts and The London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Rubina Aktar
- Centre for Neuroscience, Surgery & Trauma, Blizard Institute, Barts and The London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Madusha Peiris
- Centre for Neuroscience, Surgery & Trauma, Blizard Institute, Barts and The London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| |
Collapse
|
4
|
Aktar R, Rondinelli S, Peiris M. GPR84 in physiology-Many functions in many tissues. Br J Pharmacol 2024; 181:1524-1535. [PMID: 37533166 DOI: 10.1111/bph.16206] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 06/20/2023] [Accepted: 07/17/2023] [Indexed: 08/04/2023] Open
Abstract
Members of the GPCR superfamily have a wide variety of physiological roles and are therefore valuable targets for developing effective medicines. However, within this superfamily are receptors that are less well characterized and remain orphans, including GPR84. This receptor is stimulated by ligands derived from dietary nutrients, specifically medium chain fatty acids (C9-14), and novel synthetic agonists. There are data demonstrating the role of GPR84 in inflammatory pathways, in addition to emerging data suggesting a key role for GPR84 as a nutrient-sensing GPCR involved in metabolism by sensing energy load via nutrient exposure and subsequent signalling leading to modulation of food intake. Exploring GPR84 pharmacology, its localization and what drives its expression has revealed multiple roles for this receptor. Here, we will reflect on these various roles of GRP84 demonstrated thus far, primarily by exploring data from pre-clinical and clinical studies in various physiological systems, with a specific focus on the gastrointestinal tract. LINKED ARTICLES: This article is part of a themed issue GPR84 Pharmacology. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.10/issuetoc.
Collapse
Affiliation(s)
- Rubina Aktar
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Silvia Rondinelli
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Madusha Peiris
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
5
|
Walmsley R, Chong L, Hii MW, Brown RM, Sumithran P. The effect of bariatric surgery on the expression of gastrointestinal taste receptors: A systematic review. Rev Endocr Metab Disord 2024; 25:421-446. [PMID: 38206483 PMCID: PMC10942945 DOI: 10.1007/s11154-023-09865-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/11/2023] [Indexed: 01/12/2024]
Abstract
Gastrointestinal nutrient sensing via taste receptors may contribute to weight loss, metabolic improvements, and a reduced preference for sweet and fatty foods following bariatric surgery. This review aimed to investigate the effect of bariatric surgery on the expression of oral and post-oral gastrointestinal taste receptors and associations between taste receptor alterations and clinical outcomes of bariatric surgery. A systematic review was conducted to capture data from both human and animal studies on changes in the expression of taste receptors in oral or post-oral gastrointestinal tissue following any type of bariatric surgery. Databases searched included Medline, Embase, Emcare, APA PsychInfo, Cochrane Library, and CINAHL. Two human and 21 animal studies were included. Bariatric surgery alters the quantity of many sweet, umami, and fatty acid taste receptors in the gastrointestinal tract. Changes to the expression of sweet and amino acid receptors occur most often in intestinal segments surgically repositioned more proximally, such as the alimentary limb after gastric bypass. Conversely, changes to fatty acid receptors were observed more frequently in the colon than in the small intestine. Significant heterogeneity in the methodology of included studies limited conclusions regarding the direction of change in taste receptor expression induced by bariatric surgeries. Few studies have investigated associations between taste receptor expression and clinical outcomes of bariatric surgery. As such, future studies should look to investigate the relationship between bariatric surgery-induced changes to gut taste receptor expression and function and the impact of surgery on taste preferences, food palatability, and eating behaviour.Registration code in PROSPERO: CRD42022313992.
Collapse
Affiliation(s)
- Rosalind Walmsley
- Department of Medicine, St Vincent's Hospital Melbourne, University of Melbourne, Parkville, VIC, 3052, Australia
- Department of Surgery, St Vincent's Hospital Melbourne, University of Melbourne, Victoria, Australia
| | - Lynn Chong
- Department of Surgery, St Vincent's Hospital Melbourne, University of Melbourne, Victoria, Australia
| | - Michael W Hii
- Department of Surgery, St Vincent's Hospital Melbourne, University of Melbourne, Victoria, Australia
| | - Robyn M Brown
- Department of Pharmacology and Biochemistry, University of Melbourne, Victoria, Australia
| | - Priya Sumithran
- Department of Medicine, St Vincent's Hospital Melbourne, University of Melbourne, Parkville, VIC, 3052, Australia.
- Department of Surgery, Central Clinical School, Monash University, Victoria, Australia.
- Department of Endocrinology and Diabetes, Alfred Health, Victoria, Australia.
| |
Collapse
|
6
|
Ieremias L, Kaspersen MH, Manandhar A, Schultz-Knudsen K, Vrettou CI, Pokhrel R, Heidtmann CV, Jenkins L, Kanellou C, Marsango S, Li Y, Bräuner-Osborne H, Rexen Ulven E, Milligan G, Ulven T. Structure-Activity Relationship Studies and Optimization of 4-Hydroxypyridones as GPR84 Agonists. J Med Chem 2024; 67:3542-3570. [PMID: 38381650 DOI: 10.1021/acs.jmedchem.3c01923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
GPR84 is a putative medium-chain fatty acid receptor that is implicated in regulation of inflammation and fibrogenesis. Studies have indicated that GPR84 agonists may have therapeutic potential in diseases such as Alzheimer's disease, atherosclerosis, and cancer, but there is a lack of quality tool compounds to explore this potential. The fatty acid analogue LY237 (4a) is the most potent GPR84 agonist disclosed to date but has unfavorable physicochemical properties. We here present a SAR study of 4a. Several highly potent agonists were identified with EC50 down to 28 pM, and with SAR generally in excellent agreement with structure-based modeling. Proper incorporation of rings and polar groups resulted in the identification of TUG-2099 (4s) and TUG-2208 (42a), both highly potent GPR84 agonists with lowered lipophilicity and good to excellent solubility, in vitro permeability, and microsomal stability, which will be valuable tools for exploring the pharmacology and therapeutic prospects of GPR84.
Collapse
Affiliation(s)
- Loukas Ieremias
- Department of Drug Design and Pharmacology, Faculty of Health, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Copenhagen, Denmark
| | - Mads H Kaspersen
- Department of Drug Design and Pharmacology, Faculty of Health, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Copenhagen, Denmark
- Department of Physics, Chemistry and Pharmacy, Faculty of Science, University of Southern Denmark, Campusvej 55, 5230 Odense M, Odense, Denmark
| | - Asmita Manandhar
- Department of Drug Design and Pharmacology, Faculty of Health, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Copenhagen, Denmark
| | - Katrine Schultz-Knudsen
- Department of Drug Design and Pharmacology, Faculty of Health, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Copenhagen, Denmark
| | - Christina Ioanna Vrettou
- Department of Drug Design and Pharmacology, Faculty of Health, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Copenhagen, Denmark
| | - Rina Pokhrel
- Department of Drug Design and Pharmacology, Faculty of Health, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Copenhagen, Denmark
| | - Christoffer V Heidtmann
- Department of Physics, Chemistry and Pharmacy, Faculty of Science, University of Southern Denmark, Campusvej 55, 5230 Odense M, Odense, Denmark
| | - Laura Jenkins
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, U.K
| | - Christina Kanellou
- Department of Drug Design and Pharmacology, Faculty of Health, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Copenhagen, Denmark
| | - Sara Marsango
- Department of Drug Design and Pharmacology, Faculty of Health, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Copenhagen, Denmark
| | - Yueming Li
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, U.K
| | - Hans Bräuner-Osborne
- Department of Drug Design and Pharmacology, Faculty of Health, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Copenhagen, Denmark
| | - Elisabeth Rexen Ulven
- Department of Drug Design and Pharmacology, Faculty of Health, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Copenhagen, Denmark
| | - Graeme Milligan
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, U.K
| | - Trond Ulven
- Department of Drug Design and Pharmacology, Faculty of Health, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Copenhagen, Denmark
- Department of Physics, Chemistry and Pharmacy, Faculty of Science, University of Southern Denmark, Campusvej 55, 5230 Odense M, Odense, Denmark
| |
Collapse
|
7
|
Patil M, Casari I, Warne LN, Falasca M. G protein-coupled receptors driven intestinal glucagon-like peptide-1 reprogramming for obesity: Hope or hype? Biomed Pharmacother 2024; 172:116245. [PMID: 38340396 DOI: 10.1016/j.biopha.2024.116245] [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: 09/18/2023] [Revised: 01/23/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
'Globesity' is a foremost challenge to the healthcare system. The limited efficacy and adverse effects of available oral pharmacotherapies pose a significant obstacle in the fight against obesity. The biology of the leading incretin hormone glucagon-like-peptide-1 (GLP-1) has been highly captivated during the last decade owing to its multisystemic pleiotropic clinical outcomes beyond inherent glucoregulatory action. That fostered a pharmaceutical interest in synthetic GLP-1 analogues to tackle type-2 diabetes (T2D), obesity and related complications. Besides, mechanistic insights on metabolic surgeries allude to an incretin-based hormonal combination strategy for weight loss that emerged as a forerunner for the discovery of injectable 'unimolecular poly-incretin-agonist' therapies. Physiologically, intestinal enteroendocrine L-cells (EECs) are the prominent endogenous source of GLP-1 peptide. Despite comprehending the potential of various G protein-coupled receptors (GPCRs) to stimulate endogenous GLP-1 secretion, decades of translational GPCR research have failed to yield regulatory-approved endogenous GLP-1 secretagogue oral therapy. Lately, a dual/poly-GPCR agonism strategy has emerged as an alternative approach to the traditional mono-GPCR concept. This review aims to gain a comprehensive understanding by revisiting the pharmacology of a few potential GPCR-based complementary avenues that have drawn attention to the design of orally active poly-GPCR agonist therapy. The merits, challenges and recent developments that may aid future poly-GPCR drug discovery are critically discussed. Subsequently, we project the mechanism-based therapeutic potential and limitations of oral poly-GPCR agonism strategy to augment intestinal GLP-1 for weight loss. We further extend our discussion to compare the poly-GPCR agonism approach over invasive surgical and injectable GLP-1-based regimens currently in clinical practice for obesity.
Collapse
Affiliation(s)
- Mohan Patil
- Metabolic Signalling Group, Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Ilaria Casari
- Metabolic Signalling Group, Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Leon N Warne
- Little Green Pharma, West Perth, Western Australia 6872, Australia
| | - Marco Falasca
- University of Parma, Department of Medicine and Surgery, Via Volturno 39, 43125 Parma, Italy.
| |
Collapse
|
8
|
Baboumian S, Puma L, Swencionis C, Astbury NM, Ho J, Pantazatos SP, Geliebter A. Binge Eating (BE) and Obesity: Brain Activity and Psychological Measures before and after Roux-En-Y Gastric Bypass (RYGB). Nutrients 2023; 15:3808. [PMID: 37686840 PMCID: PMC10490010 DOI: 10.3390/nu15173808] [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: 08/01/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 09/10/2023] Open
Abstract
Brain activity in response to food cues following Roux-En-Y Gastric Bypass (RYGB) in binge eating (BE) or non-binge eating (NB) individuals is understudied. Here, 15 RYGB (8 BE; 7 NB) and 13 no treatment (NT) (7 BE; 6 NB) women with obesity underwent fMRI imaging while viewing high and low energy density food (HEF and LEF, respectively) and non-food (NF) visual cues. A region of interest (ROI) analysis compared BE participants to NB participants in those undergoing RYGB surgery pre-surgery and 4 months post. Results were corrected for multiple comparisons using liberal (p < 0.006 uncorrected) and stringent (p < 0.05 FDR corrected) thresholds. Four months following RYGB (vs. no treatment (NT) control), both BE and NB participants showed greater reductions in blood oxygen level-dependent (BOLD) signals (a proxy of local brain activity) in the dorsomedial prefrontal cortex in response to HEF (vs. LEF) cues (p < 0.006). BE (vs. NB) participants showed greater increases in the precuneus (p < 0.006) and thalamic regions (p < 0.05 corrected) to food (vs. NF). For RYGB (vs. NT) participants, BE participants, but not NB participants, showed lower BOLD signal in the middle occipital gyrus (p < 0.006), whilst NB participants, but not BE participants, showed lower signal in inferior frontal gyrus (p < 0.006) in response to HEF (vs. LEF). Results suggest distinct neural mechanisms of RGYB in BE and may help lead to improved clinical treatments.
Collapse
Affiliation(s)
- Shaunte Baboumian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, Mount Sinai Morningside, 1111 Amsterdam Ave, New York, NY 10025, USA
| | - Lauren Puma
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, Mount Sinai Morningside, 1111 Amsterdam Ave, New York, NY 10025, USA
| | - Charles Swencionis
- Ferkauf Graduate School of Psychology, Yeshiva University, 500 West 185th Street, New York, NY 10033, USA
| | - Nerys M. Astbury
- Nuffield Department of Primary Care Health Sciences, Medical Sciences Division, University of Oxford, Oxford OX2 6GG, UK
| | - Jennifer Ho
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, Mount Sinai Morningside, 1111 Amsterdam Ave, New York, NY 10025, USA
| | - Spiro P. Pantazatos
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, Department of Psychiatry, Columbia University Irving Medical Center, 1051 Riverside Dr, New York, NY 10032, USA
| | - Allan Geliebter
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, Mount Sinai Morningside, 1111 Amsterdam Ave, New York, NY 10025, USA
| |
Collapse
|
9
|
Lange O, Proczko-Stepaniak M, Mika A. Short-Chain Fatty Acids-A Product of the Microbiome and Its Participation in Two-Way Communication on the Microbiome-Host Mammal Line. Curr Obes Rep 2023:10.1007/s13679-023-00503-6. [PMID: 37208544 DOI: 10.1007/s13679-023-00503-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/29/2023] [Indexed: 05/21/2023]
Abstract
PURPOSE OF REVIEW The review aims to describe short-chain fatty acids (SCFAs) as metabolites of bacteria, their complex influence on whole-body metabolism, and alterations in the SCFA profile in obesity and after bariatric surgery (BS). RECENT FINDINGS The fecal profile of SCFAs in obese patients differs from that of lean patients, as well as their gut microbiota composition. In obese patients, a lower diversity of bacteria is observed, as well as higher concentrations of SCFAs in stool samples. Obesity is now considered a global epidemic and bariatric surgery (BS) is an effective treatment for severe obesity. BS affects the structure and functioning of the digestive system, and also alters gut microbiota and the concentration of fecal SCFAs. Generally, after BS, SCFA levels are lower but levels of branched short-chain fatty acids (BSCFAs) are elevated, the effect of which is not fully understood. Moreover, changes in the profile of circulating SCFAs are little known and this is an area for further research. Obesity seems to be inherently associated with changes in the SCFA profile. It is necessary to better understand the impact of BS on microbiota and the metabolome in both feces and blood as only a small percentage of SCFAs are excreted. Further research may allow the development of a personalized therapeutic approach to the BS patient in terms of diet and prebiotic intervention.
Collapse
Affiliation(s)
- Oliwia Lange
- Department of Environmental Analysis, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland
| | - Monika Proczko-Stepaniak
- Department of General, Endocrine, and Transplant Surgery, Faculty of Medicine, Medical University of Gdansk, Smoluchowskiego 17, 80-214, Gdansk, Poland
| | - Adriana Mika
- Department of Environmental Analysis, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland.
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland.
| |
Collapse
|
10
|
Besa E, Tembo MJ, Mulenga C, Mweetwa M, Choudhry N, Chandwe K, Storer C, Head R, Amadi B, Haritunians T, McGovern D, Kwenda G, Peiris M, Kelly P. Potential determinants of low circulating glucagon-like peptide 2 concentrations in Zambian children with non-responsive stunting. Exp Physiol 2023; 108:568-580. [PMID: 36744850 PMCID: PMC10103869 DOI: 10.1113/ep090492] [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: 01/23/2023] [Accepted: 01/19/2023] [Indexed: 02/07/2023]
Abstract
NEW FINDINGS What is the central question of this study? Non-responsive stunting is characterised by a progressive decline of circulating glucagon-like peptide 2: what are the possible causes of this decline? What is the main finding and its importance? In contrast with the established loss of Paneth and goblet cells in environmental enteropathy, there was no evidence of a parallel loss of enteroendocrine cells as seen by positive tissue staining for chromogranin A. Transcriptomic and genomic analyses showed evidence of genetic transcripts that could account for some of the variability seen in circulating glucagon-like peptide 2 values. ABSTRACT Nutrient sensing determines digestive and hormonal responses following nutrient ingestion. We have previously reported decreased levels of glucagon-like peptide 2 (GLP-2) in children with stunting. Here we demonstrate the presence of enteroendocrine cells in stunted children and explore potential pathways that may be involved in reduced circulating levels of GLP-2. At the time of performing diagnostic endoscopies for non-responsive stunted children, intestinal biopsies were collected for immunofluorescence staining of enteroendocrine cells and transcriptomic analysis. Circulating levels of GLP-2 were also measured and correlated with transcriptomic data. An exploratory genome-wide association study (GWAS) was conducted on DNA samples (n = 158) to assess genetic contribution to GLP-2 variability. Intestinal tissue sections collected from non-responsive stunted children stained positive for chromogranin A (88/89), alongside G-protein-coupled receptors G-protein receptor 119 (75/87), free fatty acid receptor 3 (76/89) and taste 1 receptor 1 (39/45). Transcriptomic analysis found three pathways correlated with circulating GLP-2: sugar metabolism, epithelial transport, and barrier function, which likely reflect downstream events following receptor-ligand interaction. GWAS analysis revealed potential genetic contributions to GLP-2 half-life and receptor binding. Enteroendocrine cell loss was not identified in stunted Zambian children as has been observed for goblet and Paneth cells. Transcriptomic analysis suggests that GLP-2 has pleiotrophic actions on the intestinal mucosa in malnutrition, but further work is needed to dissect pathways leading to perturbations in nutrient sensing.
Collapse
Affiliation(s)
- Ellen Besa
- Tropical Gastroenterology and Nutrition Group, School of MedicineUniversity of ZambiaLusakaZambia
| | - Mizinga Jacqueline Tembo
- Tropical Gastroenterology and Nutrition Group, School of MedicineUniversity of ZambiaLusakaZambia
| | - Chola Mulenga
- Tropical Gastroenterology and Nutrition Group, School of MedicineUniversity of ZambiaLusakaZambia
| | - Monica Mweetwa
- Tropical Gastroenterology and Nutrition Group, School of MedicineUniversity of ZambiaLusakaZambia
| | - Naheed Choudhry
- Blizard Institute, Centre for Neuroscience, Surgery and Trauma, Barts and The London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Kanta Chandwe
- Tropical Gastroenterology and Nutrition Group, School of MedicineUniversity of ZambiaLusakaZambia
| | - Chad Storer
- Genome Technology Access Center at McDonnell Genome InstituteWashington University in St LouisSt LouisMOUSA
| | - Richard Head
- Genome Technology Access Center at McDonnell Genome InstituteWashington University in St LouisSt LouisMOUSA
| | - Beatrice Amadi
- Tropical Gastroenterology and Nutrition Group, School of MedicineUniversity of ZambiaLusakaZambia
| | - Talin Haritunians
- Cedars‐Sinai Medical CenterInflammatory Bowel and Immunobiology Research InstituteLos AngelesCAUSA
| | - Dermot McGovern
- Cedars‐Sinai Medical CenterInflammatory Bowel and Immunobiology Research InstituteLos AngelesCAUSA
| | - Geoffrey Kwenda
- Department of Biomedical Sciences, School of Health SciencesUniversity of ZambiaLusakaZambia
| | - Madusha Peiris
- Blizard Institute, Centre for Neuroscience, Surgery and Trauma, Barts and The London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Paul Kelly
- Tropical Gastroenterology and Nutrition Group, School of MedicineUniversity of ZambiaLusakaZambia
- Blizard Institute, Centre for Neuroscience, Surgery and Trauma, Barts and The London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| |
Collapse
|
11
|
Albaugh VL, He Y, Münzberg H, Morrison CD, Yu S, Berthoud HR. Regulation of body weight: Lessons learned from bariatric surgery. Mol Metab 2023; 68:101517. [PMID: 35644477 PMCID: PMC9938317 DOI: 10.1016/j.molmet.2022.101517] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 05/04/2022] [Accepted: 05/21/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Bariatric or weight loss surgery is currently the most effective treatment for obesity and metabolic disease. Unlike dieting and pharmacology, its beneficial effects are sustained over decades in most patients, and mortality is among the lowest for major surgery. Because there are not nearly enough surgeons to implement bariatric surgery on a global scale, intensive research efforts have begun to identify its mechanisms of action on a molecular level in order to replace surgery with targeted behavioral or pharmacological treatments. To date, however, there is no consensus as to the critical mechanisms involved. SCOPE OF REVIEW The purpose of this non-systematic review is to evaluate the existing evidence for specific molecular and inter-organ signaling pathways that play major roles in bariatric surgery-induced weight loss and metabolic benefits, with a focus on Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG), in both humans and rodents. MAJOR CONCLUSIONS Gut-brain communication and its brain targets of food intake control and energy balance regulation are complex and redundant. Although the relatively young science of bariatric surgery has generated a number of hypotheses, no clear and unique mechanism has yet emerged. It seems increasingly likely that the broad physiological and behavioral effects produced by bariatric surgery do not involve a single mechanism, but rather multiple signaling pathways. Besides a need to improve and better validate surgeries in animals, advanced techniques, including inducible, tissue-specific knockout models, and the use of humanized physiological traits will be necessary. State-of-the-art genetically-guided neural identification techniques should be used to more selectively manipulate function-specific pathways.
Collapse
Affiliation(s)
- Vance L Albaugh
- Translational and Integrative Gastrointestinal and Endocrine Research Laboratory, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Yanlin He
- Brain Glycemic and Metabolism Control Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Heike Münzberg
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Christopher D Morrison
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Sangho Yu
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Hans-Rudolf Berthoud
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
12
|
Nie J, Liao W, Zhang Z, Zhang M, Wen Y, Capanoglu E, Sarker MMR, Zhu R, Zhao C. A 3D co-culture intestinal organoid system for exploring glucose metabolism. Curr Res Food Sci 2022; 6:100402. [DOI: 10.1016/j.crfs.2022.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/02/2022] [Accepted: 11/25/2022] [Indexed: 11/30/2022] Open
|
13
|
Sridhar A, Khan D, Abdelaal M, Elliott JA, Naughton V, Flatt PR, Le Roux CW, Docherty NG, Moffett CR. Differential effects of RYGB surgery and best medical treatment for obesity-diabetes on intestinal and islet adaptations in obese-diabetic ZDSD rats. PLoS One 2022; 17:e0274788. [PMID: 36137097 PMCID: PMC9499270 DOI: 10.1371/journal.pone.0274788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022] Open
Abstract
Modification of gut-islet secretions after Roux-En-Y gastric bypass (RYBG) surgery contributes to its metabolic and anti-diabetic benefits. However, there is limited knowledge on tissue-specific hormone distribution post-RYGB surgery and how this compares with best medical treatment (BMT). In the present study, pancreatic and ileal tissues were excised from male Zucker-Diabetic Sprague Dawley (ZDSD) rats 8-weeks after RYGB, BMT (daily oral dosing with metformin 300mg/kg, fenofibrate 100mg/kg, ramipril 1mg/kg, rosuvastatin 10mg/kg and subcutaneous liraglutide 0.2mg/kg) or sham operation (laparotomy). Insulin, glucagon, somatostatin, PYY, GLP-1 and GIP expression patterns were assessed using immunocytochemistry and analyzed using ImageJ. After RYGB and BMT, body weight and plasma glucose were decreased. Intestinal morphometry was unaltered by RYGB, but crypt depth was decreased by BMT. Intestinal PYY cells were increased by both interventions. GLP-1- and GIP-cell counts were unchanged by RYGB but BMT increased ileal GLP-1-cells and decreased those expressing GIP. The intestinal contents of PYY and GLP-1 were significantly enhanced by RYGB, whereas BMT decreased ileal GLP-1. No changes of islet and beta-cell area or proliferation were observed, but the extent of beta-cell apoptosis and islet integrity calculated using circularity index were improved by both treatments. Significantly decreased islet alpha-cell areas were observed in both groups, while beta- and PYY-cell areas were unchanged. RYGB also induced a decrease in islet delta-cell area. PYY and GLP-1 colocalization with glucagon in islets was significantly decreased in both groups, while co-staining of PYY with glucagon was decreased and that with somatostatin increased. These data characterize significant cellular islet and intestinal adaptations following RYGB and BMT associated with amelioration of obesity-diabetes in ZDSD rats. The differential responses observed and particularly those within islets, may provide important clues to the unique ability of RYGB to cause diabetes remission.
Collapse
Affiliation(s)
- Ananyaa Sridhar
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Dawood Khan
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
- * E-mail:
| | - Mahmoud Abdelaal
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Jessie A. Elliott
- Department of Surgery, Trinity Centre for Health Sciences and St. James’s Hospital, Dublin, Ireland
| | - Violetta Naughton
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Peter R. Flatt
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Carel W. Le Roux
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Neil G. Docherty
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Charlotte R. Moffett
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| |
Collapse
|
14
|
Changes of Short-Chain Fatty Acids and Their Receptors in an Obese Rat Model After Sleeve Gastrectomy. Obes Surg 2022; 32:2649-2657. [PMID: 35648365 DOI: 10.1007/s11695-022-06130-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 05/16/2022] [Accepted: 05/25/2022] [Indexed: 01/19/2023]
Abstract
BACKGROUND Short-chain fatty acids (SCFAs) and gut microbiota have health-related effects and are associated with a wide range of disorders. However, the changes of SCFAs and their receptors after sleeve gastrectomy (SG) remain unclear. This study aimed to examine changes of SCFAs and their receptors after SG in an obese rat model. METHODS Thirty obese Sprague-Dawley rats eating a high-energy diet for 6 weeks were divided into three groups: sham-operated (SO) control, pair-fed (PF) control, and SG group. Six weeks after the surgery, metabolic parameters, SCFA levels in the blood and stool, mRNA and protein expression of SCFA receptors in the ileum and epididymal fat, and gut microbiota were examined. RESULTS Metabolic parameters in the SG group were significantly improved compared with the SO group. Acetic acid levels in the blood and stool were significantly higher in the SG group than the PF group. The butyric acid level in the stool was also significantly higher in the SG group than in the PF group. In the ileum and epididymal fat, mRNA and protein expression of GPR41 was significantly higher in the SG group than in the other two groups, and mRNA and protein expression of GPR43 was significantly higher in the SG group than in the PF group. Increases in the genera Enterococcus, Lactobacillus, Lactococcus, and Clostridium were observed in the stool after SG. CONCLUSIONS SG may activate SCFA pathways through a change in gut microbiota.
Collapse
|
15
|
Almeida PP, Valdetaro L, Thomasi BBDM, Stockler-Pinto MB, Tavares-Gomes AL. High-fat diets on the enteric nervous system: Possible interactions and mechanisms underlying dysmotility. Obes Rev 2022; 23:e13404. [PMID: 34873814 DOI: 10.1111/obr.13404] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/25/2021] [Accepted: 11/15/2021] [Indexed: 01/09/2023]
Abstract
Obesity is a chronic disease that affects various physiological systems. Among them, the gastrointestinal tract appears to be a main target of this disease. High-fat diet (HFD) animal models can help recapitulate the classic signs of obesity and present a series of gastrointestinal alterations, mainly dysmotility. Because intestinal motility is governed by the enteric nervous system (ENS), enteric neurons, and glial cells have been studied in HFD models. Given the importance of the ENS in general gut physiology, this review aims to discuss the relationship between HFD-induced neuroplasticity and gut dysmotility observed in experimental models. Furthermore, we highlight components of the gut environment that might influence enteric neuroplasticity, including gut microbiota, enteric glio-epithelial unit, serotonin release, immune cells, and disturbances such as inflammation and oxidative stress.
Collapse
Affiliation(s)
| | - Luisa Valdetaro
- Postgraduate Program in Neurosciences, Fluminense Federal University, Niterói, Brazil
| | | | - Milena Barcza Stockler-Pinto
- Postgraduate Program in Cardiovascular Sciences, Fluminense Federal University, Niterói, Brazil.,Postgraduate Program in Nutrition Sciences, Fluminense Federal University, Niterói, Brazil
| | | |
Collapse
|
16
|
Enteroendocrine System and Gut Barrier in Metabolic Disorders. Int J Mol Sci 2022; 23:ijms23073732. [PMID: 35409092 PMCID: PMC8998765 DOI: 10.3390/ijms23073732] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/25/2022] [Accepted: 03/26/2022] [Indexed: 02/06/2023] Open
Abstract
With the continuous rise in the worldwide prevalence of obesity and type 2 diabetes, developing therapies regulating body weight and glycemia has become a matter of great concern. Among the current treatments, evidence now shows that the use of intestinal hormone analogs (e.g., GLP1 analogs and others) helps to control glycemia and reduces body weight. Indeed, intestinal endocrine cells produce a large variety of hormones regulating metabolism, including appetite, digestion, and glucose homeostasis. Herein, we discuss how the enteroendocrine system is affected by local environmental and metabolic signals. These signals include those arising from unbalanced diet, gut microbiota, and the host metabolic organs and their complex cross-talk with the intestinal barrier integrity.
Collapse
|
17
|
Abstract
The enteroendocrine system coordinates the physiological response to food intake by regulating rates of digestion, nutrient absorption, insulin secretion, satiation and satiety. Gut hormones with important anorexigenic and/or insulinotropic roles include glucagon-like peptide 1 (GLP-1), peptide YY (PYY3-36), cholecystokinin (CCK) and glucose-dependent insulinotropic peptide (GIP). High BMI or obesogenic diets do not markedly disrupt this enteroendocrine system, which represents a critical target for inducing weight loss and treating co-morbidities in individuals with obesity.
Collapse
|
18
|
Blonde GD, Price RK, le Roux CW, Spector AC. Meal Patterns and Food Choices of Female Rats Fed a Cafeteria-Style Diet Are Altered by Gastric Bypass Surgery. Nutrients 2021; 13:3856. [PMID: 34836110 PMCID: PMC8623594 DOI: 10.3390/nu13113856] [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] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/21/2021] [Accepted: 10/26/2021] [Indexed: 12/28/2022] Open
Abstract
After Roux-en-Y gastric bypass surgery (RYGB), rats tend to reduce consumption of high-sugar and/or high-fat foods over time. Here, we sought to investigate the behavioral mechanisms underlying these intake outcomes. Adult female rats were provided a cafeteria diet comprised of five palatable foodstuffs varying in sugar and fat content and intake was monitored continuously. Rats were then assigned to either RYGB, or one of two control (CTL) groups: sham surgery or a nonsurgical control group receiving the same prophylactic iron treatments as RYGB rats. Post-sur-gically, all rats consumed a large first meal of the cafeteria diet. After the first meal, RYGB rats reduced intake primarily by decreasing the meal sizes relative to CTL rats, ate meals more slowly, and displayed altered nycthemeral timing of intake yielding more daytime meals and fewer nighttime meals. Collectively, these meal patterns indicate that despite being motivated to consume a cafeteria diet after RYGB, rats rapidly learn to modify eating behaviors to consume foods more slowly across the entire day. RYGB rats also altered food preferences, but more slowly than the changes in meal patterns, and ate proportionally more energy from complex carbohydrates and protein and proportionally less fat. Overall, the pattern of results suggests that after RYGB rats quickly learn to adjust their size, eating rate, and distribution of meals without altering meal number and to shift their macronutrient intake away from fat; these changes appear to be more related to postingestive events than to a fundamental decline in the palatability of food choices.
Collapse
Affiliation(s)
- Ginger D. Blonde
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA;
| | - Ruth K. Price
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK;
| | - Carel W. le Roux
- Diabetes Complications Research Centre, Conway Institute, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland;
| | - Alan C. Spector
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA;
| |
Collapse
|
19
|
Modulation of Adipocyte Metabolism by Microbial Short-Chain Fatty Acids. Nutrients 2021; 13:nu13103666. [PMID: 34684670 PMCID: PMC8538331 DOI: 10.3390/nu13103666] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/17/2021] [Accepted: 10/17/2021] [Indexed: 12/13/2022] Open
Abstract
Obesity and its complications—including type 2 diabetes, cardiovascular disease, and certain cancers—constitute a rising global epidemic that has imposed a substantial burden on health and healthcare systems over the years. It is becoming increasingly clear that there is a link between obesity and the gut microbiota. Gut dysbiosis, characterized as microbial imbalance, has been consistently associated with obesity in both humans and animal models, and can be reversed with weight loss. Emerging evidence has shown that microbial-derived metabolites such as short-chain fatty acids (SCFAs)—including acetate, propionate, and butyrate—provide benefits to the host by impacting organs beyond the gut, including adipose tissue. In this review, we summarize what is currently known regarding the specific mechanisms that link gut-microbial-derived SCFAs with adipose tissue metabolism, such as adipogenesis, lipolysis, and inflammation. In addition, we explore indirect mechanisms by which SCFAs can modulate adipose tissue metabolism, such as via perturbation of gut hormones, as well as signaling to the brain and the liver. Understanding how the modulation of gut microbial metabolites such as SCFAs can impact adipose tissue function could lead to novel therapeutic strategies for the prevention and treatment of obesity.
Collapse
|
20
|
The Function of Gastrointestinal Hormones in Obesity-Implications for the Regulation of Energy Intake. Nutrients 2021; 13:nu13061839. [PMID: 34072172 PMCID: PMC8226753 DOI: 10.3390/nu13061839] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023] Open
Abstract
The global burden of obesity and the challenges of prevention prompted researchers to investigate the mechanisms that control food intake. Food ingestion triggers several physiological responses in the digestive system, including the release of gastrointestinal hormones from enteroendocrine cells that are involved in appetite signalling. Disturbed regulation of gut hormone release may affect energy homeostasis and contribute to obesity. In this review, we summarize the changes that occur in the gut hormone balance during the pre- and postprandial state in obesity and the alterations in the diurnal dynamics of their plasma levels. We further discuss how obesity may affect nutrient sensors on enteroendocrine cells that sense the luminal content and provoke alterations in their secretory profile. Gastric bypass surgery elicits one of the most favorable metabolic outcomes in obese patients. We summarize the effect of different strategies to induce weight loss on gut enteroendocrine function. Although the mechanisms underlying obesity are not fully understood, restoring the gut hormone balance in obesity by targeting nutrient sensors or by combination therapy with gut peptide mimetics represents a novel strategy to ameliorate obesity.
Collapse
|
21
|
Effect of Obesity on the Expression of Nutrient Receptors and Satiety Hormones in the Human Colon. Nutrients 2021; 13:nu13041271. [PMID: 33924402 PMCID: PMC8070384 DOI: 10.3390/nu13041271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/07/2021] [Accepted: 04/09/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Receptors located on enteroendocrine cells (EECs) of the colon can detect nutrients in the lumen. These receptors regulate appetite through a variety of mechanisms, including hormonal and neuronal signals. We assessed the effect of obesity on the expression of these G-protein coupled receptors (GPCRs) and hormones at both mRNA and protein level. Methods: qPCR and immunohistochemistry were used to examine colonic tissue from cohorts of patients from the Netherlands (proximal and sigmoid tissue) and the United Kingdom (tissue from across the colon) and patients were grouped by body mass index (BMI) value (BMI < 25 and BMI ≥ 25). Results: The mRNA expression of the hormones/signaling molecules serotonin, glucagon, peptide YY (PYY), CCK and somatostatin were not significantly different between BMI groups. GPR40 mRNA expression was significantly increased in sigmoid colon samples in the BMI ≥ 25 group, but not proximal colon. GPR41, GPR109a, GPR43, GPR120, GPRC6A, and CaSR mRNA expression were unaltered between low and high BMI. At the protein level, serotonin and PYY containing cell numbers were similar in high and low BMI groups. Enterochromaffin cells (EC) showed high degree of co-expression with amino acid sensing receptor, CaSR while co-expression with PYY containing L-cells was limited, regardless of BMI. Conclusions: While expression of medium/long chain fatty acid receptor GPR40 was increased in the sigmoid colon of the high BMI group, expression of other nutrient sensing GPCRs, and expression profiles of EECs involved in peripheral mechanisms of appetite regulation were unchanged. Collectively, these data suggest that in human colonic tissue, EEC and nutrient-sensing receptor expression profiles are not affected despite changes to BMI.
Collapse
|
22
|
Castorina S, Barresi V, Luca T, Privitera G, De Geronimo V, Lezoche G, Cosentini I, Di Vincenzo A, Barbatelli G, Giordano A, Taus M, Nicolai A, Condorelli DF, Cinti S. Gastric ghrelin cells in obese patients are hyperactive. Int J Obes (Lond) 2020; 45:184-194. [PMID: 33230309 DOI: 10.1038/s41366-020-00711-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 09/07/2020] [Accepted: 11/02/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND/OBJECTIVES Distribution and activity of ghrelin cells in the stomach of obese subjects are controversial. SUBJECTS/METHODS We examined samples from stomachs removed by sleeve gastrectomy in 49 obese subjects (normoglycemic, hyperglycemic and diabetic) and quantified the density of ghrelin/chromogranin endocrine cells by immunohistochemistry. Data were compared with those from 13 lean subjects evaluated by gastroscopy. In 44 cases (11 controls and 33 obese patients) a gene expression analysis of ghrelin and its activating enzyme ghrelin O-acyl transferase (GOAT) was performed. In 21 cases (4 controls and 17 obese patients) the protein levels of unacylated and acylated-ghrelin were measured by ELISA tests. In 18 cases (4 controls and 14 obese patients) the morphology of ghrelin-producing cells was evaluated by electron microscopy. RESULTS The obese group, either considered as total population or divided into subgroups, did not show any significant difference in ghrelin cell density when compared with control subjects. Inter-glandular smooth muscle fibres were increased in obese patients. In line with a positive trend of the desacylated form found by ELISA, Ghrelin and GOAT mRNA expression in obese patients was significantly increased. The unique ghrelin cell ultrastructure was maintained in all obese groups. In the hyperglycemic obese patients, the higher ghrelin expression matched with ultrastructural signs of endocrine hyperactivity, including expanded rough endoplasmic reticulum and reduced density, size and electron-density of endocrine granules. A positive correlation between ghrelin gene expression and glycemic values, body mass index and GOAT was also found. All obese patients with type 2 diabetes recovered from diabetes at follow-up after 5 months with a 16.5% of weight loss. CONCLUSIONS Given the known inhibitory role on insulin secretion of ghrelin, these results suggest a possible role for gastric ghrelin overproduction in the complex architecture that takes part in the pathogenesis of type 2 diabetes.
Collapse
Affiliation(s)
- Sergio Castorina
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy.,Mediterranean Foundation "G.B. Morgagni", Catania, Italy
| | - Vincenza Barresi
- Department of Biomedical and Biotechnological Sciences, Section of Medical Biochemistry, University of Catania, Catania, Italy
| | - Tonia Luca
- Mediterranean Foundation "G.B. Morgagni", Catania, Italy
| | | | | | - Giovanni Lezoche
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Ancona, Italy
| | - Ilaria Cosentini
- Department of Biomedical and Biotechnological Sciences, Section of Medical Biochemistry, University of Catania, Catania, Italy
| | - Angelica Di Vincenzo
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Ancona, Italy
| | - Giorgio Barbatelli
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Ancona, Italy
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Ancona, Italy
| | - Marina Taus
- Dietetic Unit and Clinical Nutrition, United Hospitals of Ancona, Ancona, Italy
| | - Albano Nicolai
- Dietetic Unit and Clinical Nutrition, United Hospitals of Ancona, Ancona, Italy
| | - Daniele Filippo Condorelli
- Department of Biomedical and Biotechnological Sciences, Section of Medical Biochemistry, University of Catania, Catania, Italy
| | - Saverio Cinti
- Mediterranean Foundation "G.B. Morgagni", Catania, Italy. .,Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Ancona, Italy. .,Dietetic Unit and Clinical Nutrition, United Hospitals of Ancona, Ancona, Italy.
| |
Collapse
|
23
|
Shackley M, Ma Y, Tate EW, Brown AJH, Frost G, Hanyaloglu AC. Short Chain Fatty Acids Enhance Expression and Activity of the Umami Taste Receptor in Enteroendocrine Cells via a Gα i/o Pathway. Front Nutr 2020; 7:568991. [PMID: 33195366 PMCID: PMC7658341 DOI: 10.3389/fnut.2020.568991] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/29/2020] [Indexed: 12/20/2022] Open
Abstract
The short chain fatty acids (SCFAs) acetate, butyrate and propionate, are produced by fermentation of non-digestible carbohydrates by the gut microbiota and regulate appetite, adiposity, metabolism, glycemic control, and immunity. SCFAs act at two distinct G protein coupled receptors (GPCRs), FFAR2 and FFAR3 and are expressed in intestinal enteroendocrine cells (EECs), where they mediate anorectic gut hormone release. EECs also express other GPCRs that act as nutrient sensors, thus SCFAs may elicit some of their health-promoting effects by altering GPCR expression in EECs and enhance gut sensitivity to dietary molecules. Here, we identify that exposure of the murine EEC STC-1 cell line or intestinal organoids to physiological concentrations of SCFAs enhances mRNA levels of the umami taste receptors TASR1 and TASR3, without altering levels of the SCFA GPCRs, FFAR2 and FFAR3. Treatment of EECs with propionate or butyrate, but not acetate, increased levels of umami receptor transcripts, while propionate also reduced CCK expression. This was reversed by inhibiting Gαi/o signaling with pertussis toxin, suggesting that SCFAs act through FFAR2/3 to alter gene expression. Surprisingly, neither a FFAR3 nor a FFAR2 selective ligand could increase TASR1/TASR3 mRNA levels. We assessed the functional impact of increased TASR1/TASR3 expression using unique pharmacological properties of the umami taste receptor; namely, the potentiation of signaling by inosine monophosphate. Activation of umami taste receptor induced inositol-1-phosphate and calcium signaling, and butyrate pretreatment significantly enhanced such signaling. Our study reveals that SCFAs may contribute to EEC adaptation and alter EEC sensitivity to bioactive nutrients.
Collapse
Affiliation(s)
- Matilda Shackley
- Section of Nutrition Research, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, United Kingdom.,Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, United Kingdom
| | - Yue Ma
- Section of Nutrition Research, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, United Kingdom
| | - Edward W Tate
- Department of Chemistry, Imperial College London, London, United Kingdom
| | | | - Gary Frost
- Section of Nutrition Research, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, United Kingdom
| | - Aylin C Hanyaloglu
- Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, United Kingdom
| |
Collapse
|
24
|
Luscombe VB, Lucy D, Bataille CJR, Russell AJ, Greaves DR. 20 Years an Orphan: Is GPR84 a Plausible Medium-Chain Fatty Acid-Sensing Receptor? DNA Cell Biol 2020; 39:1926-1937. [PMID: 33001759 DOI: 10.1089/dna.2020.5846] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
GPR84 is an inflammation-induced receptor highly expressed on immune cells, yet its endogenous ligand is still unknown. This makes any interpretation of its physiological activity in vivo difficult. However, experiments with potent synthetic agonists have highlighted what the receptor can do, namely, enhance proinflammatory signaling and macrophage effector functions such as phagocytosis. Developing drugs to block these effects has attracted interest from the scientific community with the aim of decreasing disease activity in inflammatory disorders or enhancing inflammation resolution. In this review, we critically reassess the widely held belief that the major role of GPR84 is that of being a medium-chain fatty acid (MCFA) receptor. While MCFAs have been shown to activate GPR84, it remains to be demonstrated that they are present in relevant tissues at appropriate concentrations. In contrast to four other "full-time" free fatty acid receptor subtypes, GPR84 is not expressed by enteroendocrine cells and has limited expression in the gastrointestinal tract. Across multiple tissues and cell types, the highest expression levels of GPR84 are observed hours after exposure to an inflammatory stimulus. These factors obscure the relationship between ligand and receptor in the human body and do not support the exclusive physiological pairing of MCFAs with GPR84. To maximize the chances of developing efficacious drugs for inflammatory diseases, we must advance our understanding of GPR84 and what it does in vivo.
Collapse
Affiliation(s)
- Vincent B Luscombe
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Daniel Lucy
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom.,Department of Chemistry and University of Oxford, Oxford, United Kingdom
| | | | - Angela J Russell
- Department of Chemistry and University of Oxford, Oxford, United Kingdom.,Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - David R Greaves
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
25
|
Free Fatty Acid Receptors 2 and 3 as Microbial Metabolite Sensors to Shape Host Health: Pharmacophysiological View. Biomedicines 2020; 8:biomedicines8060154. [PMID: 32521775 PMCID: PMC7344995 DOI: 10.3390/biomedicines8060154] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/30/2020] [Accepted: 06/01/2020] [Indexed: 12/13/2022] Open
Abstract
The role of the gut microbiome in human health is becoming apparent. The major functional impact of the gut microbiome is transmitted through the microbial metabolites that are produced in the gut and interact with host cells either in the local gut environment or are absorbed into circulation to impact distant cells/organs. Short-chain fatty acids (SCFAs) are the major microbial metabolites that are produced in the gut through the fermentation of non-digestible fibers. SCFAs are known to function through various mechanisms, however, their signaling through free fatty acid receptors 2 and 3 (FFAR2/3; type of G-coupled protein receptors) is a new therapeutic approach. FFAR2/3 are widely expressed in diverse cell types in human and mice, and function as sensors of SCFAs to change several physiological and cellular functions. FFAR2/3 modulate neurological signaling, energy metabolism, intestinal cellular homeostasis, immune response, and hormone synthesis. FFAR2/3 function through Gi and/or Gq signaling, that is mediated through specific structural features of SCFAs-FFAR2/3 bindings and modulating specific signaling pathway. In this review, we discuss the wide-spread expression and structural homologies between human and mice FFAR2/3, and their role in different human health conditions. This information can unlock opportunities to weigh the potential of FFAR2/3 as a drug target to prevent human diseases.
Collapse
|
26
|
Abstract
Reviewed here are multiple mouse models of vertical sleeve gastrectomy (VSG) and Roux-en Y gastric bypass (RYGB) that have emerged over the past decade. These models use diverse approaches to both operative and perioperative procedures. Scrutinizing the benefits and pitfalls of each surgical model and what to expect in terms of post-operative outcomes will enhance our assessment of studies using mouse models, as well as advance our understanding of their translational potential. Two mouse models of bariatric surgery, VSG-lembert and RYGB-small pouch, demonstrate low mortality and most closely recapitulate the human forms of surgery. The use of liquid diets can be minimized, and in mice, RYGB demonstrates more reliable and longer lasting effects on weight loss compared to that of VSG.
Collapse
|
27
|
Laessle C, Jin K, Seifert GJ, Timme-Bronsert S, Fichtner-Feigl S, Marjanovic G, Fink JM. Putting the Hindgut Hypothesis to the Test in a Diabetic Zucker Rat Model. Obes Surg 2020; 29:4000-4007. [PMID: 31367988 DOI: 10.1007/s11695-019-04079-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND The hindgut theory hypothesizes a key role of differential hindgut stimulation following metabolic procedures in ameliorating diabetes mellitus. We used two strategies to remove the hindgut from intestinal continuity in order to analyze its impact on diabetes mellitus. METHODS Loop duodeno-jejunostomy (DJOS) with exclusion of one-third of total intestinal length was performed in 3 groups of 9-week-old Zucker diabetic fatty rats. In group 1, no further alteration of the intestinal tract was made. Group 2 received additional ileal exclusion (IE). Group 3 underwent additional resection of 50% of the ileum with side-to-side ileocecal anastomosis (IR). One, 2, and 4 months after surgery, fasting blood glucose levels, oral glucose tolerance tests (OGTT), and glucose-stimulated hormone analyses were conducted, and bile acid blood levels were compared. Body weight was documented weekly. RESULTS In relation to DJOS, glucose control was not impaired in IR or IE. On the contrary, only IR could maintain preOP glucose values until 4 months. There were no significant weight differences between the groups. Confirming effective ileal diversion, bile acid blood levels were significantly higher in the DJOS group compared with both IR and IE (p = 0.0025 and p = 0.0047). Operative interventions had no impact on GLP-1 levels at any time point (ANOVA p > 0.05 for all). Insulin secretion was preserved in all groups. CONCLUSION This data supports the hypothesis that the mechanisms driving amelioration of diabetes mellitus are complex and cannot be reduced to the ileum.
Collapse
Affiliation(s)
- Claudia Laessle
- Department of General and Visceral Surgery, Faculty of Medicine, Medical Center - University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany.
| | - Ke Jin
- Department of General and Visceral Surgery, Faculty of Medicine, Medical Center - University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - Gabriel J Seifert
- Department of General and Visceral Surgery, Faculty of Medicine, Medical Center - University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - Sylvia Timme-Bronsert
- Faculty of Medicine, Institute of Pathology, Medical Center - University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - Stefan Fichtner-Feigl
- Department of General and Visceral Surgery, Faculty of Medicine, Medical Center - University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - Goran Marjanovic
- Department of General and Visceral Surgery, Faculty of Medicine, Medical Center - University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - Jodok Matthias Fink
- Department of General and Visceral Surgery, Faculty of Medicine, Medical Center - University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| |
Collapse
|
28
|
Ginés I, Gil-Cardoso K, D’Addario C, Falconi A, Bellia F, Blay MT, Terra X, Ardévol A, Pinent M, Beltrán-Debón R. Long-Lasting Effects of GSPE on Ileal GLP-1R Gene Expression Are Associated with a Hypomethylation of the GLP-1R Promoter in Female Wistar Rats. Biomolecules 2019; 9:biom9120865. [PMID: 31842341 PMCID: PMC6995503 DOI: 10.3390/biom9120865] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/03/2019] [Accepted: 12/10/2019] [Indexed: 12/14/2022] Open
Abstract
Flavonoids have been shown to modulate GLP-1 in obesity. GLP-1 induces some of its effects through the intestinal GLP-1 receptor (GLP-1R), though no data exist on how flavonoids affect this receptor. Here, we examine how a dose of grape seed proanthocyanidin extract (GSPE) with anti-obesity activity affects intestinal GLP-1R and analyze whether epigenetics play a role in the long-lasting effects of GSPE. We found that 10-day GSPE administration prior to the cafeteria diet upregulated GLP-1R mRNA in the ileum 17 weeks after the GSPE treatment. This was associated with a hypomethylation of the GLP-1R promoter near the region where the SP1 transcription factor binds. In the colon, the cafeteria diet upregulated GLP-1R without showing any GSPE effect. In conclusion, we have identified long-lasting GSPE effects on GLP-1R gene expression in the ileum that are partly mediated by hypomethylation at the gene promoter and may affect the SP1 binding factor.
Collapse
Affiliation(s)
- Iris Ginés
- MoBioFood Research Group, Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, c/Marcel·lí Domingo 1, 43007 Tarragona, Spain; (I.G.); (K.G.-C.); (M.T.B.); (X.T.); (A.A.); (R.B.-D.)
| | - Katherine Gil-Cardoso
- MoBioFood Research Group, Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, c/Marcel·lí Domingo 1, 43007 Tarragona, Spain; (I.G.); (K.G.-C.); (M.T.B.); (X.T.); (A.A.); (R.B.-D.)
| | - Claudio D’Addario
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via Renato Balzarini 1, 64100 Teramo, Italy; (C.D.); (A.F.); (F.B.)
| | - Anastasia Falconi
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via Renato Balzarini 1, 64100 Teramo, Italy; (C.D.); (A.F.); (F.B.)
| | - Fabio Bellia
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via Renato Balzarini 1, 64100 Teramo, Italy; (C.D.); (A.F.); (F.B.)
| | - M Teresa Blay
- MoBioFood Research Group, Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, c/Marcel·lí Domingo 1, 43007 Tarragona, Spain; (I.G.); (K.G.-C.); (M.T.B.); (X.T.); (A.A.); (R.B.-D.)
| | - Ximena Terra
- MoBioFood Research Group, Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, c/Marcel·lí Domingo 1, 43007 Tarragona, Spain; (I.G.); (K.G.-C.); (M.T.B.); (X.T.); (A.A.); (R.B.-D.)
| | - Anna Ardévol
- MoBioFood Research Group, Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, c/Marcel·lí Domingo 1, 43007 Tarragona, Spain; (I.G.); (K.G.-C.); (M.T.B.); (X.T.); (A.A.); (R.B.-D.)
| | - Montserrat Pinent
- MoBioFood Research Group, Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, c/Marcel·lí Domingo 1, 43007 Tarragona, Spain; (I.G.); (K.G.-C.); (M.T.B.); (X.T.); (A.A.); (R.B.-D.)
- Correspondence: ; Tel.: +34-977-55-9566; Fax: +34-977-55-8232
| | - Raúl Beltrán-Debón
- MoBioFood Research Group, Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, c/Marcel·lí Domingo 1, 43007 Tarragona, Spain; (I.G.); (K.G.-C.); (M.T.B.); (X.T.); (A.A.); (R.B.-D.)
| |
Collapse
|
29
|
Kimura I, Ichimura A, Ohue-Kitano R, Igarashi M. Free Fatty Acid Receptors in Health and Disease. Physiol Rev 2019; 100:171-210. [PMID: 31487233 DOI: 10.1152/physrev.00041.2018] [Citation(s) in RCA: 488] [Impact Index Per Article: 97.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fatty acids are metabolized and synthesized as energy substrates during biological responses. Long- and medium-chain fatty acids derived mainly from dietary triglycerides, and short-chain fatty acids (SCFAs) produced by gut microbial fermentation of the otherwise indigestible dietary fiber, constitute the major sources of free fatty acids (FFAs) in the metabolic network. Recently, increasing evidence indicates that FFAs serve not only as energy sources but also as natural ligands for a group of orphan G protein-coupled receptors (GPCRs) termed free fatty acid receptors (FFARs), essentially intertwining metabolism and immunity in multiple ways, such as via inflammation regulation and secretion of peptide hormones. To date, several FFARs that are activated by the FFAs of various chain lengths have been identified and characterized. In particular, FFAR1 (GPR40) and FFAR4 (GPR120) are activated by long-chain saturated and unsaturated fatty acids, while FFAR3 (GPR41) and FFAR2 (GPR43) are activated by SCFAs, mainly acetate, butyrate, and propionate. In this review, we discuss the recent reports on the key physiological functions of the FFAR-mediated signaling transduction pathways in the regulation of metabolism and immune responses. We also attempt to reveal future research opportunities for developing therapeutics for metabolic and immune disorders.
Collapse
Affiliation(s)
- Ikuo Kimura
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| | - Atsuhiko Ichimura
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| | - Ryuji Ohue-Kitano
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| | - Miki Igarashi
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| |
Collapse
|
30
|
Neuro-hormonal mechanisms underlying changes in reward related behaviors following weight loss surgery: Potential pharmacological targets. Biochem Pharmacol 2019; 164:106-114. [DOI: 10.1016/j.bcp.2019.04.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/03/2019] [Indexed: 12/11/2022]
|