1
|
Kim N, Kim S, Park S, Kim EK. Adenosine transmission from hypothalamic tanycytes to AGRP/NPY neurons regulates energy homeostasis. Exp Mol Med 2025:10.1038/s12276-025-01449-6. [PMID: 40316705 DOI: 10.1038/s12276-025-01449-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/14/2025] [Accepted: 03/03/2025] [Indexed: 05/04/2025] Open
Abstract
Tanycytes are a pivotal component of the hypothalamic network that controls energy homeostasis. Despite their importance, the regulatory mechanisms governing tanycyte-neuron interactions in response to metabolic signals remain unexplored. Here we report that adenosine signaling between tanycytes and AGRP/NPY neurons is crucial for tanycytic metabolic regulation mediated by translocator protein 18 kDa (TSPO). Tanycyte-specific Tspo-knockout mice displayed reduced food consumption and weight loss associated with the downregulation of Agrp and Npy expression under high-fat diet feeding. Tspo-deficient tanycytes had elevated levels of intracellular ATP, which was released via connexin 43 hemichannels and extracellularly converted into adenosine by tanycytic ectonucleotidases. The adenosine signal was perceived by adenosine A1 receptors on adjacent AGRP/NPY neurons, reducing ERK phosphorylation, which in turn downregulated Agrp and Npy expression. Our findings underscore the anorexic role of adenosine as a gliotransmitter in the intricate communication between tanycytes and neurons for regulating appetite and body weight.
Collapse
Affiliation(s)
- Nayoun Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Seolsong Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Seokjae Park
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
- Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Eun-Kyoung Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea.
- Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea.
| |
Collapse
|
2
|
Toledo M, Martínez-Martínez S, Van Hul M, Laudo B, Eyre E, Pelicaen R, Puel A, Altirriba J, Gómez-Valadés AG, Inderhees J, Moreno-Indias I, Pozo M, Chivite I, Milà-Guasch M, Haddad-Tóvolli R, Obri A, Fos-Domènech J, Tahiri I, Llana SR, Ramírez S, Monelli E, Schwaninger M, Cani PD, Nogueiras R, Claret M. Rapid modulation of gut microbiota composition by hypothalamic circuits in mice. Nat Metab 2025:10.1038/s42255-025-01280-3. [PMID: 40263603 DOI: 10.1038/s42255-025-01280-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 03/17/2025] [Indexed: 04/24/2025]
Abstract
In recent years, the gut microbiota and derived metabolites have emerged as relevant players in modulating several brain functions, including energy balance control1-3. This form of distant communication mirrors that of metabolic hormones (for example, leptin, ghrelin), which convey information about the organism's energy status by exerting effects on diverse brain regions, including the master homeostatic centre, the hypothalamus4. However, whether the hypothalamus is also able to influence gut microbiota composition remains enigmatic. Here we present a study designed to unravel this challenging question. To this aim, we used chemogenetics5 (to selectively activate or inhibit hypothalamic pro-opiomelanocortin or agouti-related peptide neurons) or centrally administered leptin or ghrelin to male mice. Subsequently, we conducted microbiota composition analysis throughout the gut using 16S rRNA gene sequencing. Our results showed that these brain interventions significantly changed the gut microbiota in an anatomical and short-term (2-4 h) fashion. Transcriptomic analysis indicated that these changes were associated with the reconfiguration of neuronal and synaptic pathways in the duodenum concomitant with increased sympathetic tone. Interestingly, diet-induced obesity attenuated the brain-mediated changes triggered by leptin in gut microbiota communities and sympathetic activation. Our findings reveal a previously unanticipated brain-gut axis that acutely attunes microbiota composition on fast timescales, with potential implications for meal-to-meal adjustments and systemic energy balance control.
Collapse
Affiliation(s)
- Míriam Toledo
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sara Martínez-Martínez
- Department of Physiology (CIMUS), School of Medicine-Instituto de Investigaciones Sanitarias (IDIS), Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - Matthias Van Hul
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
| | - Berta Laudo
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Elena Eyre
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Rudy Pelicaen
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Anthony Puel
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
| | - Jordi Altirriba
- Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Alicia G Gómez-Valadés
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Julica Inderhees
- Bioanalytic Core Facility, Center for Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
- German Research Centre for Cardiovascular Research (DZHK), Lübeck, Germany
| | - Isabel Moreno-Indias
- Department of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, Málaga, Spain
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Macarena Pozo
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Iñigo Chivite
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maria Milà-Guasch
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Roberta Haddad-Tóvolli
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Arnaud Obri
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Júlia Fos-Domènech
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Iasim Tahiri
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sergio R Llana
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sara Ramírez
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Erika Monelli
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Markus Schwaninger
- German Research Centre for Cardiovascular Research (DZHK), Lübeck, Germany
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Patrice D Cani
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université Catholique de Louvain, Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium.
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université Catholique de Louvain, Brussels, Belgium.
| | - Rubén Nogueiras
- Department of Physiology (CIMUS), School of Medicine-Instituto de Investigaciones Sanitarias (IDIS), Universidad de Santiago de Compostela, Santiago de Compostela, Spain.
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain.
- Galicia Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain.
| | - Marc Claret
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain.
- School of Medicine, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
3
|
Anjom-Shoae J, Hajishafiee M, Fitzgerald PC, Coleman R, Martin AM, Poppitt SD, Lee M, Higgs S, Rehfeld JF, Holst JJ, Veedfald S, Horowitz M, Feinle-Bisset C. Acute decrease in the plasma tryptophan-to-large-neutral-amino-acids ratio attenuates the effects of L-tryptophan on gut hormones and energy intake in healthy males: a randomized, cross-over, exploratory trial. Am J Clin Nutr 2025; 121:816-825. [PMID: 39978467 DOI: 10.1016/j.ajcnut.2025.02.016] [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: 09/25/2024] [Revised: 02/09/2025] [Accepted: 02/13/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND L-tryptophan ("Trp") and L-leucine ("Leu"), when administered intraduodenally, increase plasma cholecystokinin (CCK) and glucagon-like peptide 1 (GLP-1) and stimulate pyloric pressures, which all slow gastric emptying and suppress subsequent energy intake. The circulating Trp-to-large-neutral-amino-acids ("Trp/LNAAs") ratio is also inversely related to energy intake. OBJECTIVES This exploratory study characterized the impact of standardized changes in the plasma Trp/LNAAs ratio, achieved by combining a fixed-load intraduodenal infusion of Trp with increasing loads of Leu, on the appetite-inhibitory effects of enteral Trp. METHODS Twelve males of normal weight [mean ± standard deviation; age: 23 ± 2 y; body mass index (in kg/m2: 23±1)], received on 4 separate occasions, 90-min iso-osmotic intraduodenal infusions of 1) isotonic 0.9% saline ("control"), 2) Trp (0.15 kcal/min; "Trp"), 3) Trp + Leu (0.22 kcal/min; "Trp+Leu-0.22"), or 4) Trp + Leu (0.45 kcal/min; "Trp+Leu-0.45"), in a randomized, double-blind, cross-over fashion. Immediately postinfusion ad-libitum energy intake was quantified. Plasma CCK, GLP-1, amino acid concentrations, and antropyloroduodenal pressures were measured throughout. RESULTS Although there was a transient stimulation of CCK and GLP-1 by Trp + Leu - 0.45 (at t = 15 min), only Trp led to a sustained increase in plasma CCK (P = 0.04) and GLP-1 (P = 0.009) from t = 60-90 min, and stimulated pyloric pressures (P = 0.01), compared with control. Only Trp reduced energy intake [kcal (mean ± standard error of the mean); control: 1085 ± 49, Trp: 881 ± 75, Trp + Leu - 0.22: 963 ± 57, Trp + Leu - 0.45: 932 ± 60] compared with control (P = 0.008). The Trp/LNAAs ratio was dose-dependently decreased by Trp + Leu - 0.22 and Trp + Leu - 0.45, compared with Trp (all P = 0.001), and energy intake correlated inversely with the Trp/LNAAs ratio (R = -0.38; P = 0.02). CONCLUSIONS Acute reduction in the Trp/LNAAs ratio appears to be associated with a diminished capacity of Trp to stimulate CCK and GLP-1 and suppress energy intake. Although these observations should be interpreted with caution given the exploratory nature of the study, they attest to the complexity of the relationships between pre and postabsorptive mechanisms underlying Trp's appetite-inhibitory effect. This trial was registered at the Australian New Zealand clinical trial registry as ACTRN12620001275954.
Collapse
Affiliation(s)
- Javad Anjom-Shoae
- Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Maryam Hajishafiee
- Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Penelope Ce Fitzgerald
- Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Rosie Coleman
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Alyce M Martin
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Sally D Poppitt
- Department of Medicine, Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Michelle Lee
- School of Psychology, Faculty of Medicine, Health and Life Science, Swansea University, Swansea, United Kingdom
| | - Suzanne Higgs
- School of Psychology, University of Birmingham, Birmingham, United Kingdom
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Simon Veedfald
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Horowitz
- Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide Medical School, University of Adelaide, Adelaide, Australia; Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, Australia
| | - Christine Feinle-Bisset
- Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide Medical School, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
4
|
Wang Y, Qiu W, Kernodle S, Parker C, Padilla MA, Su J, Tomlinson AJ, Oldham S, Field J, Bernard E, Hornigold D, Rhodes CJ, Olson DP, Seeley RJ, Myers MG. Roles for Prlhr/GPR10 and Npffr2/GPR74 in feeding responses to PrRP. Mol Metab 2025; 92:102093. [PMID: 39755369 PMCID: PMC11773474 DOI: 10.1016/j.molmet.2024.102093] [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: 10/31/2024] [Revised: 12/06/2024] [Accepted: 12/28/2024] [Indexed: 01/06/2025] Open
Abstract
OBJECTIVE Several groups of neurons in the NTS suppress food intake, including Prlh-expressing neurons (NTSPrlh cells). Not only does the artificial activation of NTSPrlh cells decrease feeding, but also the expression of Prlh (which encodes the neuropeptide PrRP) and neurotransmission by NTSPrlh neurons contributes to the restraint of food intake and body weight, especially in animals fed a high fat diet (HFD). We set out to determine roles for putative PrRP receptors in the response to NTS PrRP and exogenous PrRP-related peptides. METHODS We used animals lacking PrRP receptors GPR10 and/or GPR74 (encoded by Prlhr and Npffr2, respectively) to determine roles for each in the restraint of food intake and body weight by the increased expression of Prlh in NTSPrlh neurons (NTSPrlhOX mice) and in response to the anorectic PrRP analog, p52. RESULTS Although Prlhr played a crucial role in the restraint of food intake and body weight in HFD-fed control animals, the combined absence of Prlhr and Npffr2 was required to abrogate the restraint of food intake in NTSPrlhOX mice. p52 suppressed feeding independently of both receptors, however. CONCLUSIONS Hence, each receptor can participate in the NTSPrlh-mediated suppression of food intake and body weight gain, while PrRP analog treatment can mediate its effects via distinct systems. While Prlhr plays a crucial role in the physiologic restraint of weight gain, the action of either receptor is capable of ameliorating obesity in response to enhanced NTSPrlh signaling.
Collapse
Affiliation(s)
- Yi Wang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, the Second Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Weiwei Qiu
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Stace Kernodle
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Carly Parker
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Jiaao Su
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Stephanie Oldham
- Early Cardiovascular Renal and Metabolism, BioPharmaceuticals, R&D, AstraZeneca, Cambridge, UK
| | - Joss Field
- Early Cardiovascular Renal and Metabolism, BioPharmaceuticals, R&D, AstraZeneca, Cambridge, UK
| | - Elise Bernard
- Hit Discovery, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - David Hornigold
- Early Cardiovascular Renal and Metabolism, BioPharmaceuticals, R&D, AstraZeneca, Cambridge, UK
| | - Christopher J Rhodes
- Early Cardiovascular Renal and Metabolism, BioPharmaceuticals, R&D, AstraZeneca, Cambridge, UK
| | - David P Olson
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Martin G Myers
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
5
|
Soengas JL, Comesaña S, Blanco AM, Conde-Sieira M. Feed Intake Regulation in Fish: Implications for Aquaculture. REVIEWS IN FISHERIES SCIENCE & AQUACULTURE 2025; 33:8-60. [DOI: 10.1080/23308249.2024.2374259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- José L. Soengas
- Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Laboratorio de Fisioloxía Animal, Centro de Investigación Mariña, Universidade de Vigo, Vigo, Spain
| | - Sara Comesaña
- Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Laboratorio de Fisioloxía Animal, Centro de Investigación Mariña, Universidade de Vigo, Vigo, Spain
| | - Ayelén M. Blanco
- Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Laboratorio de Fisioloxía Animal, Centro de Investigación Mariña, Universidade de Vigo, Vigo, Spain
| | - Marta Conde-Sieira
- Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Laboratorio de Fisioloxía Animal, Centro de Investigación Mariña, Universidade de Vigo, Vigo, Spain
| |
Collapse
|
6
|
Guerin S, Henry G, Le Normand L, Cahu A, Hiolle M, Baniel A, Dupont D, Boudry G. Micellar casein and sodium caseinate supramolecular structure differently impacts subsequent food intake in pigs. Food Res Int 2025; 200:115465. [PMID: 39779120 DOI: 10.1016/j.foodres.2024.115465] [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: 09/10/2024] [Revised: 11/15/2024] [Accepted: 11/26/2024] [Indexed: 01/04/2025]
Abstract
Dietary protein reduces energy intake in following meals by signaling directly or indirectly to the brain. We recently observed differences in plasma amino acid kinetics and intra-gastric behavior between micellar casein (MC) and sodium caseinate (SC) in pigs, two factors that impact food intake. Our objective was to clarify whether the supramolecular structure of casein, given as a preload to pigs, impacts on subsequent food intake. Overnight fasted pigs were allowed to consume casein drinks differing in casein macromolecular structure (SC vs MC) within 5 min in a cross-over study. Ad libitum intake of their regular feed was assessed during 1 h, either 1 or 4 h after casein drink ingestion. To evaluate the potential mechanisms at play, gastric emptying of the casein drinks radiolabeled with 99Tc-colloïd was followed using gamma-scintigraphy while plasma kinetics of ghrelin, GLP-1, insulin and free amino acids were evaluated. The amount of feed consumed 1, but not 4 h, after SC ingestion was lower than the amount of feed consumed after MC ingestion (P = 0.03). Gastric emptying parameters, plasma ghrelin, GLP-1 and insulin kinetics after both types of casein ingestion were not significantly different (P > 0.05). However, plasma free amino acid concentrations, known to reduce food intake, increased after both SC and MC ingestion but was greater after SC than MC ingestion from 60 to 120 min (P = 0.009). In conclusion, casein supramolecular structure in a preload drink impacts differently subsequent energy intake, likely due to difference in amino acid bioavailability. Micellar casein exhibits less anorectic effect than sodium caseinate, a property that could benefit population with high protein need but low appetite such as elderly.
Collapse
Affiliation(s)
- Sylvie Guerin
- Institut Numecan, INSERM, INRAE, Univ Rennes, Rennes, France
| | | | | | - Armelle Cahu
- Institut Numecan, INSERM, INRAE, Univ Rennes, Rennes, France
| | | | | | | | - Gaëlle Boudry
- Institut Numecan, INSERM, INRAE, Univ Rennes, Rennes, France.
| |
Collapse
|
7
|
Nagao T, Braga JD, Chen S, Thongngam M, Chartkul M, Yanaka N, Kumrungsee T. Synergistic effects of peripheral GABA and GABA-transaminase inhibitory drugs on food intake control and weight loss in high-fat diet-induced obese mice. Front Pharmacol 2024; 15:1487585. [PMID: 39415835 PMCID: PMC11480068 DOI: 10.3389/fphar.2024.1487585] [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: 08/28/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Background Developing anti-obesity interventions targeting appetite or food intake, the primary driver of obesity, remains challenging. Here, we demonstrated that dietary γ-aminobutyric acid (GABA) with GABA-degradation inhibitory drugs could be an anti-obesity intervention possessing strong food intake-suppressive and weight-loss effects. Methods High-fat (HF)-diet-induced obese mice were divided into six groups receiving either the HF diet or the 2% GABA-HF diet with daily administration of PBS or the GABA-degradation inhibitory drugs, vigabatrin and ethanolamine-O-sulfate (EOS). In 24-h fast-induced refeeding, lean mice with a basal diet were used, and food intake was measured from 0.5 to 24 h after refeeding. Results Coadministration of the 2% GABA-HF diet with vigabatrin or EOS significantly decreased food intake (-53%, -35%) and body weight (-22%, -16%) within 11 days in obese mice, along with a marked increase in plasma GABA levels. Mice receiving dietary GABA alone or the drugs alone exhibited no such effects. Hypothalamic GABA levels increased in drug-treated mice, regardless of diet. At 0.5 h after refeeding, food intake was similar in all groups. However, at 0.5 h, plasma GABA levels were markedly increased only in mice receiving coadministration of dietary GABA and the drugs, and their food intake was completely inhibited for over 6 h, while mice in other groups gradually increased their food intake. Conclusion Combining dietary GABA with GABA-degradation inhibitory drugs effectively suppresses food intake and promotes weight loss in obese mice, primarily through increased plasma GABA availability. These findings may advance the development of food intake-controlling strategies for obesity management.
Collapse
Affiliation(s)
- Tomoka Nagao
- Program of Food and AgriLife Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Jason D. Braga
- Program of Food and AgriLife Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
- Institute of Food Science and Technology, College of Agriculture, Food, Environment and Natural Resources, Cavite State University, Indang, Philippines
| | - Siyi Chen
- Program of Food and AgriLife Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Masubon Thongngam
- Department of Food Science and Technology, Faculty of Agro-Industry, Kasetsart University, Bangkok, Thailand
| | - Maesaya Chartkul
- Weight Care Clinic, Health Promotion Center, Bangkok Chanthaburi Hospital, Chanthaburi, Thailand
| | - Noriyuki Yanaka
- Program of Food and AgriLife Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Thanutchaporn Kumrungsee
- Program of Food and AgriLife Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
- Smart Agriculture, Graduate School of Innovation and Practice for Smart Society, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
8
|
Worth AA, Feetham CH, Morrissey NA, Luckman SM. Paraventricular oxytocin neurons impact energy intake and expenditure: projections to the bed nucleus of the stria terminalis reduce sucrose consumption. Front Endocrinol (Lausanne) 2024; 15:1449326. [PMID: 39286269 PMCID: PMC11402739 DOI: 10.3389/fendo.2024.1449326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Background The part played by oxytocin and oxytocin neurons in the regulation of food intake is controversial. There is much pharmacological data to support a role for oxytocin notably in regulating sugar consumption, however, several recent experiments have questioned the importance of oxytocin neurons themselves. Methods Here we use a combination of histological and chemogenetic techniques to investigate the selective activation or inhibition of oxytocin neurons in the hypothalamic paraventricular nucleus (OxtPVH). We then identify a pathway from OxtPVH neurons to the bed nucleus of the stria terminalis using the cell-selective expression of channel rhodopsin. Results OxtPVH neurons increase their expression of cFos after both physiological (fast-induced re-feeding or oral lipid) and pharmacological (systemic administration of cholecystokinin or lithium chloride) anorectic signals. Chemogenetic activation of OxtPVH neurons is sufficient to decrease free-feeding in Oxt Cre:hM3Dq mice, while inhibition in Oxt Cre:hM4Di mice attenuates the response to administration of cholecystokinin. Activation of OxtPVH neurons also increases energy expenditure and core-body temperature, without a significant effect on locomotor activity. Finally, the selective, optogenetic stimulation of a pathway from OxtPVH neurons to the bed nucleus of the stria terminalis reduces the consumption of sucrose. Conclusion Our results support a role for oxytocin neurons in the regulation of whole-body metabolism, including a modulatory action on food intake and energy expenditure. Furthermore, we demonstrate that the pathway from OxtPVH neurons to the bed nucleus of the stria terminalis can regulate sugar consumption.
Collapse
Affiliation(s)
| | | | | | - Simon M. Luckman
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
9
|
Comesaña S, Antomagesh F, Soengas JL, Blanco AM, Vijayan MM. Valine administration in the hypothalamus alters the brain and plasma metabolome in rainbow trout. Am J Physiol Regul Integr Comp Physiol 2024; 327:R261-R273. [PMID: 38881412 DOI: 10.1152/ajpregu.00056.2024] [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: 03/03/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/18/2024]
Abstract
Central administration of valine has been shown to cause hyperphagia in fish. Although mechanistic target of rapamycin (mTOR) is involved in this response, the contributions to feed intake of central and peripheral metabolite changes due to excess valine are unknown. Here, we investigated whether intracerebroventricular injection of valine modulates central and peripheral metabolite profiles and may provide insights into feeding response in fish. Juvenile rainbow trout (Oncorhynchus mykiss) were administered an intracerebroventricular injection of valine (10 µg·µL-1 at 1 μL·100·g-1 body wt), and the metabolite profile in plasma, hypothalamus, and rest of the brain (composing of telencephalon, optic tectum, cerebellum, and medulla oblongata) was carried out by liquid chromatography-mass spectrometry (LC/MS)-based metabolomics. Valine administration led to a spatially distinct metabolite profile at 1 h postinjection in the brain: enrichment of amino acid metabolism and energy production pathways in the rest of the brain but not in hypothalamus. This suggests a role for extrahypothalamic input in the regulation of feed intake. Also, there was enrichment of several amino acids, including tyrosine, proline, valine, phenylalanine, and methionine, in plasma in response to valine. Changes in liver transcript abundance and protein expression reflect an increased metabolic capacity, including energy production from glucose and fatty acids, and a lower protein kinase B (Akt) phosphorylation in the valine group. Altogether, valine intracerebroventricular administration affects central and peripheral metabolism in rainbow trout, and we propose a role for the altered metabolite profile in modulating the feeding response to this branched-chain amino acid.NEW & NOTEWORTHY Valine causes hyperphagia in fish when it is centrally administered; however, the exact mechanisms are far from clear. We tested how intracerebroventricular injection of valine in rainbow trout affected the brain and plasma metabolome. The metabolite changes in response to valine were more evident in the rest of the brain compared with the hypothalamus. Furthermore, we demonstrated for the first time that central valine administration affects peripheral metabolism in rainbow trout.
Collapse
Affiliation(s)
- Sara Comesaña
- Centro de Investigación Mariña, Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo, Spain
| | | | - José L Soengas
- Centro de Investigación Mariña, Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo, Spain
| | - Ayelén M Blanco
- Centro de Investigación Mariña, Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo, Spain
| | | |
Collapse
|
10
|
Kanata MC, Yanni AE, Koliaki C, Pateras I, Anastasiou IA, Kokkinos A, Karathanos VT. Effects of Wheat Biscuits Enriched with Plant Proteins Incorporated into an Energy-Restricted Dietary Plan on Postprandial Metabolic Responses of Women with Overweight/Obesity. Nutrients 2024; 16:1229. [PMID: 38674919 PMCID: PMC11053654 DOI: 10.3390/nu16081229] [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: 03/13/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
This study investigates the effect of daily consumption of wheat biscuits enriched with plant proteins in postprandial metabolic responses of women with overweight/obesity who follow an energy-restricted diet. Thirty apparently healthy women participated in a 12-week randomized controlled trial and were assigned either to a control (CB) or an intervention (PB) group. Participants consumed daily either a conventional (CB) or an isocaloric wheat biscuit enriched with plant proteins (PB) containing high amounts of amino acids with appetite-regulating properties, i.e., BCAAs and L-arg. At baseline and the end of the intervention, a mixed meal tolerance test was performed. The responses of glucose, insulin, ghrelin, GLP-1, and glicentin were evaluated over 180 min. After 12 weeks, both groups experienced significant decreases in body weight, fat mass, and waist circumference. In the PB group, a trend towards higher weight loss was observed, accompanied by lower carbohydrate, fat, and energy intakes (p < 0.05 compared to baseline and CB group), while decreases in fasting insulin and the HOMA-IR index were also observed (p < 0.05 compared to baseline). In both groups, similar postprandial glucose, ghrelin, and GLP-1 responses were detected, while iAUC for insulin was lower (p < 0.05). Interestingly, the iAUC of glicentin was greater in the PB group (p < 0.05 compared to baseline). Subjective appetite ratings were beneficially affected in both groups (p < 0.05). Consumption of wheat biscuits enriched in plant proteins contributed to greater weight loss, lower energy intake, and insulin resistance and had a positive impact on postprandial glicentin response, a peptide that can potentially predict long-term weight loss and decreased food intake.
Collapse
Affiliation(s)
- Maria-Christina Kanata
- Laboratory of Chemistry-Biochemistry-Physical Chemistry of Foods, Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece; (M.-C.K.); (V.T.K.)
| | - Amalia E. Yanni
- Laboratory of Chemistry-Biochemistry-Physical Chemistry of Foods, Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece; (M.-C.K.); (V.T.K.)
| | - Chrysi Koliaki
- First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 15772 Athens, Greece; (C.K.); (I.A.A.); (A.K.)
| | - Irene Pateras
- ELBISCO S.A., Industrial and Commercial Food Company, 21st Km Marathonos Avenue, 19009 Pikermi, Greece;
| | - Ioanna A. Anastasiou
- First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 15772 Athens, Greece; (C.K.); (I.A.A.); (A.K.)
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Alexander Kokkinos
- First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 15772 Athens, Greece; (C.K.); (I.A.A.); (A.K.)
| | - Vaios T. Karathanos
- Laboratory of Chemistry-Biochemistry-Physical Chemistry of Foods, Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece; (M.-C.K.); (V.T.K.)
| |
Collapse
|
11
|
Blanco AM, Antomagesh F, Comesaña S, Soengas JL, Vijayan MM. Chronic cortisol stimulation enhances hypothalamus-specific enrichment of metabolites in the rainbow trout brain. Am J Physiol Endocrinol Metab 2024; 326:E382-E397. [PMID: 38294699 DOI: 10.1152/ajpendo.00410.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/08/2024] [Accepted: 01/24/2024] [Indexed: 02/01/2024]
Abstract
The hypothalamus is a key integrating center that is involved in the initiation of the corticosteroid stress response, and in regulating nutrient homeostasis. Although cortisol, the principal glucocorticoid in humans and teleosts, plays a central role in feeding regulation, the mechanisms are far from clear. We tested the hypothesis that the metabolic changes to cortisol exposure signal an energy excess in the hypothalamus, leading to feeding suppression during stress in fish. Rainbow trout (Oncorhynchus mykiss) were administered a slow-release cortisol implant for 3 days, and the metabolite profiles in the plasma, hypothalamus, and the rest of the brain were assessed. Also, U-13C-glucose was injected into the hypothalamus by intracerebroventricular (ICV) route, and the metabolic fate of this energy substrate was followed in the brain regions by metabolomics. Chronic cortisol treatment reduced feed intake, and this corresponded with a downregulation of the orexigenic gene agrp, and an upregulation of the anorexigenic gene cart in the hypothalamus. The U-13C-glucose-mediated metabolite profiling indicated an enhancement of glycolytic flux and tricarboxylic acid intermediates in the rest of the brain compared with the hypothalamus. There was no effect of cortisol treatment on the phosphorylation status of AMPK or mechanistic target of rapamycin in the brain, whereas several endogenous metabolites, including leucine, citrate, and lactate were enriched in the hypothalamus, suggesting a tissue-specific metabolic shift in response to cortisol stimulation. Altogether, our results suggest that the hypothalamus-specific enrichment of leucine and the metabolic fate of this amino acid, including the generation of lipid intermediates, contribute to cortisol-mediated feeding suppression in fish.NEW & NOTEWORTHY Elevated cortisol levels during stress suppress feed intake in animals. We tested whether the feed suppression is associated with cortisol-mediated alteration in hypothalamus metabolism. The brain metabolome revealed a hypothalamus-specific metabolite profile suggesting nutrient excess. Specifically, we noted the enrichment of leucine and citrate in the hypothalamus, and the upregulation of pathways involved in leucine metabolism and fatty acid synthesis. This cortisol-mediated energy substrate repartitioning may modulate the feeding/satiety centers leading to the feeding suppression.
Collapse
Affiliation(s)
- Ayelén M Blanco
- Centro de Investigación Mariña, Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo, Spain
| | | | - Sara Comesaña
- Centro de Investigación Mariña, Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo, Spain
| | - José L Soengas
- Centro de Investigación Mariña, Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo, Spain
| | | |
Collapse
|
12
|
Müller M, Van Liefferinge E, Tilbrook A, van Barneveld R, Roura E. Excess dietary Lys reduces feed intake, stimulates jejunal CCK secretion and alters essential and non-essential blood AA profile in pigs. J Anim Sci Biotechnol 2024; 15:24. [PMID: 38369505 PMCID: PMC10874532 DOI: 10.1186/s40104-023-00971-9] [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: 07/29/2023] [Accepted: 12/06/2023] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND Commercial diets are frequently formulated to meet or exceed nutrient levels including those of limiting essential amino acids (AA) covering potential individual variations within the herd. However, the provision of dietary excess of AA, such as Lys, may lead to reduced appetite and growth in pigs. The mechanisms modulating these responses have not been extensively investigated. This study evaluated the effect of Lys dietary excesses on performance and satiety biomarkers in post weaning pigs. METHODS Twenty-four pigs aged 21 d and weighing 6.81 ± 0.12 kg (mean ± SEM) were individually housed and offered 1 of 4 dietary treatments for 3 weeks: a diet containing a standardized ileal digestible Lys reaching 100% (T0), 120% (T1), 150% (T2) or 200% (T3) of the NRC (2012) requirements. At the end of the experiment, blood samples from the cephalic vein of the T0 and T3 groups were obtained for AA analysis. In addition, primary intestinal cultures from T0 pigs were used, following their humane killing, to evaluate the effect of Lys on gut hormone secretion and AA sensors gene expression under ex vivo conditions. RESULTS Feed intake was linearly reduced (P < 0.001) and the weight gain to feed ratio reduced (P < 0.10) with increased dietary levels of Lys during the third- and first-week post weaning, respectively. Cholecystokinin concentration (P < 0.05) and the metabotropic glutamate receptor 1 and the solute carrier family 7 member 2 (P < 0.10) gene expression was enhanced in proximal jejunum tissues incubated with Lys at 20 mmol/L when compared to the control (Lys 0 mmol/L). Plasma Lys and Glu (P < 0.05) concentration increased in the T3 compared to T0 pigs. In contrast, plasma levels of His, Val, Thr, Leu (P < 0.05) and Gln (P < 0.10) were lower in T3 than T0 pigs. CONCLUSION The present results confirm that excess dietary Lys inhibits hunger in pigs. Moreover, the results provide evidence of pre- and post-absorptive mechanisms modulating these responses. Lys dietary excesses should be narrowed, when possible, to avoid negative effects of the AA on appetite in pigs.
Collapse
Affiliation(s)
- Maximiliano Müller
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Elout Van Liefferinge
- Laboratory of Animal Nutrition and Animal Product Quality (LANUPRO), Department of Animal Sciences and Aquatic Ecology, Ghent University, 339000, Ghent, Flanders, Belgium
| | - Alan Tilbrook
- Centre for Animal Science, Queensland Alliance for Agriculture and Food Innovation and the School of Veterinary Science, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | | | - Eugeni Roura
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, Queensland, 4072, Australia.
| |
Collapse
|
13
|
Yu Z, Han J, Li L, Zhang Q, Chen A, Chen J, Wang K, Jin J, Li H, Chen G. Chronic triclosan exposure induce impaired glucose tolerance by altering the gut microbiota. Food Chem Toxicol 2024; 183:114305. [PMID: 38052405 DOI: 10.1016/j.fct.2023.114305] [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] [Received: 09/20/2023] [Revised: 11/10/2023] [Accepted: 11/27/2023] [Indexed: 12/07/2023]
Abstract
Triclosan (TCS) is an antimicrobial compound incorporated into more than 2000 consumer products. This compound is frequently detected in the human body and causes ubiquitous contamination in the environment, thereby raising concerns about its impact on human health and environmental pollution. Here, we demonstrated that 20 weeks' exposure of TCS drove the development of glucose intolerance by inducing compositional and functional alterations in intestinal microbiota in rats. Fecal-transplantation experiments corroborated the involvement of gut microbiota in TCS-induced glucose-tolerance impairment. 16S rRNA gene-sequencing analysis of cecal contents showed that TCS disrupted the gut microbiota composition in rats and increased the ratio of Firmicutes to Bacteroidetes. Cecal metabolomic analyses detected that TCS altered host metabolic pathways that are linked to host glucose and amino acid metabolism, particularly branched-chain amino acid (BCAA) biosynthesis. BCAA measurement confirmed the increase in serum BCAAs in rats exposed to TCS. Western blot and immunostaining results further confirmed that elevated BCAAs stimulated mTOR, a nutrient-sensing complex, and following IRS-1 serine phosphorylation, resulted in insulin resistance and glucose intolerance. These results suggested that TCS may induce glucose metabolism imbalance by regulating BCAA concentration by remodeling the gut microbiota.
Collapse
Affiliation(s)
- Zhen Yu
- Fujian Provincial Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou, 350001, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, China
| | - Junyong Han
- Fujian Provincial Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou, 350001, China
| | - Lisha Li
- Fujian Provincial Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou, 350001, China
| | - Qiufeng Zhang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Ayun Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Jinyan Chen
- Fujian Provincial Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou, 350001, China
| | - Kun Wang
- Fujian Provincial Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou, 350001, China
| | - Jingjun Jin
- Fujian Provincial Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou, 350001, China
| | - Huangyuan Li
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, China.
| | - Gang Chen
- Department of Endocrinology, Fujian Provincial Hospital, Fuzhou, 350001, China.
| |
Collapse
|
14
|
Huang C, Luo Y, Zeng B, Chen Y, Liu Y, Chen W, Liao X, Liu Y, Wang Y, Wang X. Branched-chain amino acids prevent obesity by inhibiting the cell cycle in an NADPH-FTO-m 6A coordinated manner. J Nutr Biochem 2023; 122:109437. [PMID: 37666478 DOI: 10.1016/j.jnutbio.2023.109437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 08/15/2023] [Accepted: 08/30/2023] [Indexed: 09/06/2023]
Abstract
Obesity has become a major health crisis in the past decades. Branched-chain amino acids (BCAA), a class of essential amino acids, exerted beneficial health effects with regard to obesity and its related metabolic dysfunction, although the underlying reason is unknown. Here, we show that BCAA supplementation alleviates high-fat diet (HFD)-induced obesity and insulin resistance in mice and inhibits adipogenesis in 3T3-L1 cells. Further, we find that BCAA prevent the mitotic clonal expansion (MCE) of preadipocytes by reducing cyclin A2 (CCNA2) and cyclin-dependent kinase 2 (CDK2) expression. Mechanistically, BCAA decrease the concentration of nicotinamide adenine dinucleotide phosphate (NADPH) in adipose tissue and 3T3-L1 cells by reducing glucose-6-phosphate dehydrogenase (G6PD) expression. The reduced NADPH attenuates the expression of fat mass and obesity-associated (FTO) protein, a well-known m6A demethylase, to increase the N6-methyladenosine (m6A) levels of Ccna2 and Cdk2 mRNA. Meanwhile, the high m6A levels of Ccna2 and Cdk2 mRNA are recognized by YTH N6-methyladenosine RNA binding protein 2 (YTHDF2), which results in mRNA decay and reduction of their protein expressions. Overall, our data demonstrate that BCAA inhibit obesity and adipogenesis by reducing CDK2 and CCNA2 expression via an NADPH-FTO-m6A coordinated manner in vivo and in vitro, which raises a new perspective on the role of m6A in the BCAA regulation of obesity and adipogenesis.
Collapse
Affiliation(s)
- Chaoqun Huang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Yaojun Luo
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Botao Zeng
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Yushi Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Youhua Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Wei Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Xing Liao
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Yuxi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Yizhen Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Xinxia Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang province, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
15
|
Peleg-Raibstein D, Viskaitis P, Burdakov D. Eat, seek, rest? An orexin/hypocretin perspective. J Neuroendocrinol 2023; 35:e13259. [PMID: 36994677 DOI: 10.1111/jne.13259] [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: 12/21/2022] [Revised: 03/06/2023] [Accepted: 03/10/2023] [Indexed: 03/19/2023]
Abstract
Seeking and ingesting nutrients is an essential cycle of life in all species. In classical neuropsychology these two behaviours are viewed as fundamentally distinct from each other, and known as appetitive and consummatory, respectively. Appetitive behaviour is highly flexible and diverse, but typically involves increased locomotion and spatial exploration. Consummatory behaviour, in contrast, typically requires reduced locomotion. Another long-standing concept is "rest and digest", a hypolocomotive response to calorie intake, thought to facilitate digestion and storage of energy after eating. Here, we note that the classical seek➔ingest➔rest behavioural sequence is not evolutionarily advantageous for all ingested nutrients. Our limited stomach capacity should be invested wisely, rather than spent on the first available nutrient. This is because nutrients are not simply calories: some nutrients are more essential for survival than others. Thus, a key choice that needs to be made soon after ingestion: to eat more and rest, or to terminate eating and search for better food. We offer a perspective on recent work suggesting how nutrient-specific neural responses shape this choice. Specifically, the hypothalamic hypocretin/orexin neurons (HONs) - cells that promote hyperlocomotive explorative behaviours - are rapidly and differentially modulated by different ingested macronutrients. Dietary non-essential (but not essential) amino acids activate HONs, while glucose depresses HONs. This nutrient-specific HON modulation engages distinct reflex arcs, seek➔ingest➔seek and seek➔ingest➔rest, respectively. We propose that these nutri-neural reflexes evolved to facilitate optimal nutrition despite the limitations of our body.
Collapse
Affiliation(s)
- Daria Peleg-Raibstein
- ETH Zürich, Department of Health Sciences and Technology, Schorenstrasse 16, 8603 Schwerzenbach, Switzerland
| | - Paulius Viskaitis
- ETH Zürich, Department of Health Sciences and Technology, Schorenstrasse 16, 8603 Schwerzenbach, Switzerland
| | - Denis Burdakov
- ETH Zürich, Department of Health Sciences and Technology, Schorenstrasse 16, 8603 Schwerzenbach, Switzerland
| |
Collapse
|
16
|
Agostini D, Gervasi M, Ferrini F, Bartolacci A, Stranieri A, Piccoli G, Barbieri E, Sestili P, Patti A, Stocchi V, Donati Zeppa S. An Integrated Approach to Skeletal Muscle Health in Aging. Nutrients 2023; 15:nu15081802. [PMID: 37111021 PMCID: PMC10141535 DOI: 10.3390/nu15081802] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
A decline in muscle mass and function represents one of the most problematic changes associated with aging, and has dramatic effects on autonomy and quality of life. Several factors contribute to the inexorable process of sarcopenia, such as mitochondrial and autophagy dysfunction, and the lack of regeneration capacity of satellite cells. The physiologic decline in muscle mass and in motoneuron functionality associated with aging is exacerbated by the sedentary lifestyle that accompanies elderly people. Regular physical activity is beneficial to most people, but the elderly need well-designed and carefully administered training programs that improve muscle mass and, consequently, both functional ability and quality of life. Aging also causes alteration in the gut microbiota composition associated with sarcopenia, and some advances in research have elucidated that interventions via the gut microbiota-muscle axis have the potential to ameliorate the sarcopenic phenotype. Several mechanisms are involved in vitamin D muscle atrophy protection, as demonstrated by the decreased muscular function related to vitamin D deficiency. Malnutrition, chronic inflammation, vitamin deficiencies, and an imbalance in the muscle-gut axis are just a few of the factors that can lead to sarcopenia. Supplementing the diet with antioxidants, polyunsaturated fatty acids, vitamins, probiotics, prebiotics, proteins, kefir, and short-chain fatty acids could be potential nutritional therapies against sarcopenia. Finally, a personalized integrated strategy to counteract sarcopenia and maintain the health of skeletal muscles is suggested in this review.
Collapse
Affiliation(s)
- Deborah Agostini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Marco Gervasi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Fabio Ferrini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Alessia Bartolacci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Alessandro Stranieri
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Giovanni Piccoli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Elena Barbieri
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Piero Sestili
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Antonino Patti
- Sport and Exercise Sciences Research Unit, Department of Psychology, Educational Science and Human Movement, University of Palermo, 90128 Palermo, Italy
| | - Vilberto Stocchi
- Department of Human Science for Promotion of Quality of Life, Università Telematica San Raffaele, 00166 Rome, Italy
| | - Sabrina Donati Zeppa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| |
Collapse
|
17
|
Yuan F, Wu S, Zhou Z, Jiao F, Yin H, Niu Y, Jiang H, Chen S, Guo F. Leucine deprivation results in antidepressant effects via GCN2 in AgRP neurons. LIFE METABOLISM 2023; 2:load004. [PMID: 39872511 PMCID: PMC11748975 DOI: 10.1093/lifemeta/load004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/20/2023] [Accepted: 02/03/2023] [Indexed: 01/30/2025]
Abstract
Essential amino acids (EAAs) are crucial nutrients, whose levels change in rodents and patients with depression. However, how the levels of a single EAA affects depressive behaviors remains elusive. Here, we demonstrate that although deprivation of the EAA leucine has no effect in unstressed mice, it remarkably reverses the depression-like behaviors induced by chronic restraint stress (CRS). This beneficial effect is independent of feeding and is applicable to the dietary deficiency of other EAAs. Furthermore, the effect of leucine deprivation is suppressed by central injection of leucine or mimicked by central injection of leucinol. Moreover, hypothalamic agouti-related peptide (AgRP) neural activity changes during CRS and leucine deprivation, and chemogenetically inhibiting AgRP neurons eliminates the antidepressant effects of leucine deprivation. Finally, the leucine deprivation-regulated behavioral effects are mediated by amino acid sensor general control non-derepressible 2 (GCN2) in AgRP neurons. Taken together, our results suggest a new drug target and/or dietary intervention for the reduction of depressive symptoms.
Collapse
Affiliation(s)
- Feixiang Yuan
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Shangming Wu
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ziheng Zhou
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Fuxin Jiao
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hanrui Yin
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuguo Niu
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Haizhou Jiang
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shanghai Chen
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Feifan Guo
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
18
|
Lueders B, Kanney BC, Krone MJ, Gannon NP, Vaughan RA. Effect of branched-chain amino acids on food intake and indicators of hunger and satiety- a narrative summary. HUMAN NUTRITION & METABOLISM 2022; 30:200168. [DOI: 10.1016/j.hnm.2022.200168] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
19
|
Park S, Oh S, Kim EK. Glucagon-like peptide-1 analog liraglutide leads to multiple metabolic alterations in diet-induced obese mice. J Biol Chem 2022; 298:102682. [PMID: 36356900 PMCID: PMC9730228 DOI: 10.1016/j.jbc.2022.102682] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/27/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022] Open
Abstract
Liraglutide, a glucagon-like peptide-1 analog, has beneficial metabolic effects in patients with type 2 diabetes and obesity. Although the high efficacy of liraglutide as an anti-diabetic and anti-obesity drug is well known, liraglutide-induced metabolic alterations in diverse tissues remain largely unexplored. Here, we report the changes in metabolic profiles induced by a 2-week subcutaneous injection of liraglutide in diet-induced obese mice fed a high-fat diet for 8 weeks. Our comprehensive metabolomic analyses of the hypothalamus, plasma, liver, and skeletal muscle showed that liraglutide intervention led to various metabolic alterations in comparison with diet-induced obese or nonobese mice. We found that liraglutide remarkably coordinated not only fatty acid metabolism in the hypothalamus and skeletal muscle but also amino acid and carbohydrate metabolism in plasma and liver. Comparative analyses of metabolite dynamics revealed that liraglutide rewired intertissue metabolic correlations. Our study points to a previously unappreciated metabolic alteration by liraglutide in several tissues, which may underlie its therapeutic effects within and across the tissues.
Collapse
Affiliation(s)
- Seokjae Park
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea,Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Sungjoon Oh
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea,Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Eun-Kyoung Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea,Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea,For correspondence: Eun-Kyoung Kim
| |
Collapse
|
20
|
Yanni AE, Kokkinos A, Binou P, Papaioannou V, Halabalaki M, Konstantopoulos P, Simati S, Karathanos VT. Postprandial Glucose and Gastrointestinal Hormone Responses of Healthy Subjects to Wheat Biscuits Enriched with L-Arginine or Branched-Chain Amino Acids of Plant Origin. Nutrients 2022; 14:nu14204381. [PMID: 36297065 PMCID: PMC9611898 DOI: 10.3390/nu14204381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
The study investigates the effects of wheat biscuits supplemented with plant flours originating from legumes/seeds enriched either in L-arginine (L-arg) or branched-chain amino acids (BCAAs) on postprandial glucose response of healthy subjects. Gastrointestinal hormone and amino acid responses as well as subjective appetite sensations are also evaluated. Subjects consumed wheat-based biscuits, enriched either in L-arg (ArgB) or BCAAs (BCAAsB) or a conventional wheat biscuit (CB) or a glucose solution (GS) in an acute randomized crossover design. Responses of glucose, insulin, ghrelin, glucagon-like peptide-1 (GLP-1), peptide YY (PYY) and glicentin, as well as those of L-arginine, L-leucine, L-isoleucine and L-valine, were evaluated over 180 min. Consumption of ArgB and BCAAsB elicited lower glucose iAUC compared to GS (p < 0.05). A lower iAUC for insulin was observed after consumption of BCAAsB (p < 0.05 compared to CB and ArgB), while ArgB elicited higher iAUC for GLP-1 accompanied by higher glicentin response (p < 0.05 compared to CB). BCAAsB and ArgB increased postprandial amino acid concentrations and caused stronger satiety effects compared to CB. Increasing protein content of wheat biscuits with supplementation of plant flours originating from legumes/seeds decreases postprandial glycemia and provides with healthier snack alternatives which can easily be incorporated into diet.
Collapse
Affiliation(s)
- Amalia E. Yanni
- Laboratory of Chemistry-Biochemistry-Physical Chemistry of Foods, Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece
- Correspondence: ; Tel.: +30-210-9549174
| | - Alexander Kokkinos
- 1st Department of Propaedeutic and Internal Medicine, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Panagiota Binou
- Laboratory of Chemistry-Biochemistry-Physical Chemistry of Foods, Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece
| | - Varvara Papaioannou
- Division of Pharmacognosy and Natural Products Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Maria Halabalaki
- Division of Pharmacognosy and Natural Products Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Panagiotis Konstantopoulos
- Laboratory of Experimental Surgery and Surgery Research, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Stamatia Simati
- 1st Department of Propaedeutic and Internal Medicine, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Vaios T. Karathanos
- Laboratory of Chemistry-Biochemistry-Physical Chemistry of Foods, Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece
| |
Collapse
|
21
|
Zhu Y, Bailey D, Childress A, Dawson JA, Binks M, Dhurandhar NV. Greater protein quality of an egg breakfast may be inadequate to induce satiety during weight loss, compared with a cereal breakfast of equal protein quantity. Int J Food Sci Nutr 2022; 73:1096-1103. [PMID: 36237122 DOI: 10.1080/09637486.2022.2133097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
We compared the effects of consuming egg-breakfast of superior protein quality to cereal-breakfast of similar energy density and protein quantity, but lower protein quality. Two, two-week randomised crossover clinical trials included 30 otherwise healthy women with overweight or obesity. Subjects received counselling to follow a reduced-calorie diet. Under supervision, participants consumed either breakfast for one-week then crossed over to the opposite breakfast. Experiment-1 outcome variables included post-breakfast appetite hormones, glucose and insulin, subjective markers of satiety and energy intake at lunch and dinner. In Experiment-2, an appealing food (brownies) was included in lunch. Following the breakfasts, Experiment-1 showed no significant differences in outcome variables. In Experiment-2, the egg-breakfast increased fullness (p = 0.038), but lunch-time energy intake was not different. If these findings apply to other breakfasts, it suggests that in comparing two breakfasts with similar protein quantity, the greater protein quality of a breakfast may not be adequate to induce satiety.
Collapse
Affiliation(s)
- Yaqiong Zhu
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Dylan Bailey
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Allison Childress
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - John A Dawson
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Martin Binks
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | | |
Collapse
|
22
|
Olszewski PK, Noble EE, Paiva L, Ueta Y, Blevins JE. Oxytocin as a potential pharmacological tool to combat obesity. J Neuroendocrinol 2022; 34:e13106. [PMID: 35192207 PMCID: PMC9372234 DOI: 10.1111/jne.13106] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/06/2022] [Accepted: 02/08/2022] [Indexed: 11/30/2022]
Abstract
The neuropeptide oxytocin (OT) has emerged as an important anorexigen in the regulation of food intake and energy balance. It has been shown that the release of OT and activation of hypothalamic OT neurons coincide with food ingestion. Its effects on feeding have largely been attributed to limiting meal size through interactions in key regulatory brain regions governing the homeostatic control of food intake such as the hypothalamus and hindbrain in addition to key feeding reward areas such as the nucleus accumbens and ventral tegmental area. Furthermore, the magnitude of an anorexigenic response to OT and feeding-related activation of the brain OT circuit are modified by the composition and flavor of a diet, as well as by a social context in which a meal is consumed. OT is particularly effective in reducing consumption of carbohydrates and sweet tastants. Pharmacologic, genetic, and pair-feeding studies indicate that OT-elicited weight loss cannot be fully explained by reductions of food intake and that the overall impact of OT on energy balance is also partly a result of OT-elicited changes in lipolysis, energy expenditure, and glucose regulation. Peripheral administration of OT mimics many of its effects when it is given into the central nervous system, raising the questions of whether and to what extent circulating OT acts through peripheral OT receptors to regulate energy balance. Although OT has been found to elicit weight loss in female mice, recent studies have indicated that sex and estrous cycle may impact oxytocinergic modulation of food intake. Despite the overall promising basic research data, attempts to use OT in the clinical setting to combat obesity and overeating have generated somewhat mixed results. The focus of this mini-review is to briefly summarize the role of OT in feeding and metabolism, address gaps and inconsistencies in our knowledge, and discuss some of the limitations to the potential use of chronic OT that should help guide future research on OT as a tailor-made anti-obesity therapeutic.
Collapse
Affiliation(s)
- Pawel K Olszewski
- Faculty of Science and Engineering, University of Waikato, Waikato, New Zealand
- Department of Food Science and Nutrition, College of Food, Agricultural and Natural Resource Sciences, University of Minnesota, St Paul, Minnesota, USA
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Emily E Noble
- Department of Nutritional Sciences, University of Georgia, Athens, Georgia, USA
| | - Luis Paiva
- Instituto de Ciencia Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - James E Blevins
- Department of Veterans Affairs Medical Center, VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Seattle, Washington, USA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
23
|
Stier C, Koschker AC, Kim M, Stier R, Chiappetta S, Stein J. Fast-track rescue weight reduction therapy to achieve rapid technical operability for emergency bariatric surgery in patients with life-threatening inoperable severe obesity - A proof of concept study. Clin Nutr ESPEN 2022; 50:238-246. [PMID: 35871930 DOI: 10.1016/j.clnesp.2022.05.009] [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: 10/28/2021] [Revised: 05/14/2022] [Accepted: 05/17/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND AND AIMS Severe obesity (BMI ≥60 kg/m2) in multimorbid patients can be acutely life-threatening. While emergency weight-loss surgery is urgently needed to preserve life, most patients are in an inoperable state. Pre-surgical bridging therapy is required to achieve technical operability through weight reduction. Standard bridging using an intragastric balloon (IB) can achieve operability in 6 months but is unsuitable for some patients in a critical condition. A non-invasive fast-track rescue therapy to achieve very rapid operability is urgently needed. We investigated whether a rescue weight reduction therapy (RWR) consisting of liraglutide, a glucagon-like peptide-1 (GLP-1) receptor agonist, a leucine-rich amino acid infusion and a hypocaloric diet, can accelerate readiness for emergency surgery in patients with acutely life-threatening severe obesity. METHODS In this proof-of-concept study, prospective data from patients treated with RWR (intervention group 1, n = 26) were mathematically matched with retrospective biometric data of 26 patients with severe obesity (historic control group 2) who underwent standard 6-month bridging with IB. A rating scale was developed to identify patients needing urgent fast-track bridging. RESULTS Rapid weight loss was observed in all patients on the RWR therapy. All achieved operability after a mean RWR bridging duration of 20.7 ± 6.9 days. Baseline weight was 236.3 ± 35.8 kg in group 1 compared with 230.1 ± 32.7 kg in group 2. Mean body weight loss during RWR was 27.5 ± 14.1 kg, compared with 20.9 ± 10.5 kg in group 2 (P = 0.0629). CONCLUSIONS Pre-operative bridging using liraglutide in combination with a leucine-rich amino acid infusion and hypocaloric diet was effective in all cases of acutely life-threatening severe obesity, achieving technical operability within only ca. 2-4 weeks. This therapy has potential as a life-saving rescue therapy for multimorbid patients with severe obesity who were previously untreatable. This study is registered at ClinicalTrials.gov (identifier: NCT02616003).
Collapse
Affiliation(s)
- Christine Stier
- Department of Obesity and Metabolic Surgery, Sana Klinikum Offenbach, Offenbach am Main, Germany; Sana Obesity Center North Rhine Westphalia, Hürth, Germany; Department of General, Visceral and Transplant Surgery, University of Aachen, Germany; Division of Endocrinology and Diabetes at the Department of Internal Medicine, University of Würzburg, Germany.
| | - Ann-Cathrin Koschker
- Division of Endocrinology and Diabetes at the Department of Internal Medicine, University of Würzburg, Germany
| | - Mia Kim
- Clinic and Policlinic for General, Visceral, Transplantation, Vascular and Pediatric Surgery, University of Würzburg, Germany; Münchner Klinik Harlaching, Clinic for General and Visceral Surgery, Munich, Germany
| | - Raphael Stier
- Department of General, Visceral, Tumor and Transplant Surgery, University of Cologne, Germany
| | - Sonja Chiappetta
- Bariatric and Metabolic Surgery Unit, Ospedale Evangelico Betania, Naples, Italy
| | - Jürgen Stein
- Department of Gastroenterology and Clinical Nutrition, DGD Clinics Sachsenhausen, Frankfurt am Main, Germany
| |
Collapse
|
24
|
Binou P, Yanni AE, Kartsioti K, Barmpagianni A, Konstantopoulos P, Karathanos VT, Kokkinos A. Wheat Biscuits Enriched with Plant-Based Protein Contribute to Weight Loss and Beneficial Metabolic Effects in Subjects with Overweight/Obesity. Nutrients 2022; 14:nu14122516. [PMID: 35745249 PMCID: PMC9231350 DOI: 10.3390/nu14122516] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/08/2022] [Accepted: 06/14/2022] [Indexed: 12/23/2022] Open
Abstract
The present study aimed to assess the impact of daily consumption of a snack fortified with plant proteins with high content in amino acids with appetite regulating properties (BCAAs and L-arginine), as part of a dietary intervention, on weight loss. Seventy adults without diabetes (26 male, 44 female) and with overweight/obesity participated in a 12-week restricted dietary intervention and were randomized to either a control or an intervention group, consuming daily 70 g of conventional wheat biscuits (CB) or an isocaloric amount of wheat biscuits enriched with plant proteins (PB) originating from legumes and seeds, respectively. Anthropometric characteristics were measured and venous blood samples were collected at baseline and at the end of the intervention. Decreases in body weight, body fat mass and waist circumference were observed in both groups. Participants in the intervention group experienced greater weight loss (7.6 ± 2.7 vs. 6.2 ± 2.7%, p = 0.025) and marginally significant larger decrease in body fat mass (4.9 ± 2.2 vs. 3.9 ± 2.4 kg, p = 0.059). A moderate reduction in IL-1β levels (p = 0.081), a significantly higher decrease in TNF-α levels (p < 0.001) and a marginally significant greater leptin decrease (p = 0.066) in subjects of the PB group were noticed. Greater reductions in caloric and carbohydrate intake and a trend towards a higher decrease in fat intake were also observed in participants of this group. Incorporation of plant-based proteins with high content in amino acids with appetite-regulating properties in wheat biscuits may contribute to greater weight loss and improvement of metabolic parameters in subjects who are overweight or obese. Protein enrichment of snacks offers a beneficial qualitative manipulation that could be successfully incorporated in a diet plan.
Collapse
Affiliation(s)
- Panagiota Binou
- Laboratory of Chemistry-Biochemistry-Physical Chemistry of Foods, Department of Nutrition and Dietetics, Harokopio University of Athens, 17671 Athens, Greece; (P.B.); (K.K.); (V.T.K.)
| | - Amalia E. Yanni
- Laboratory of Chemistry-Biochemistry-Physical Chemistry of Foods, Department of Nutrition and Dietetics, Harokopio University of Athens, 17671 Athens, Greece; (P.B.); (K.K.); (V.T.K.)
- Correspondence: ; Tel.: +30-2109549174
| | - Klio Kartsioti
- Laboratory of Chemistry-Biochemistry-Physical Chemistry of Foods, Department of Nutrition and Dietetics, Harokopio University of Athens, 17671 Athens, Greece; (P.B.); (K.K.); (V.T.K.)
| | - Aikaterini Barmpagianni
- 1st Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.B.); (A.K.)
| | - Panagiotis Konstantopoulos
- Laboratory of Experimental Surgery and Surgery Research, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Vaios T. Karathanos
- Laboratory of Chemistry-Biochemistry-Physical Chemistry of Foods, Department of Nutrition and Dietetics, Harokopio University of Athens, 17671 Athens, Greece; (P.B.); (K.K.); (V.T.K.)
| | - Alexander Kokkinos
- 1st Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.B.); (A.K.)
| |
Collapse
|
25
|
Viskaitis P, Arnold M, Garau C, Jensen LT, Fugger L, Peleg-Raibstein D, Burdakov D. Ingested non-essential amino acids recruit brain orexin cells to suppress eating in mice. Curr Biol 2022; 32:1812-1821.e4. [PMID: 35316652 DOI: 10.1016/j.cub.2022.02.067] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 01/10/2022] [Accepted: 02/23/2022] [Indexed: 12/22/2022]
Abstract
Ingested nutrients are proposed to control mammalian behavior by modulating the activity of hypothalamic orexin/hypocretin neurons (HONs). Previous in vitro studies showed that nutrients ubiquitous in mammalian diets, such as non-essential amino acids (AAs) and glucose, modulate HONs in distinct ways. Glucose inhibits HONs, whereas non-essential (but not essential) AAs activate HONs. The latter effect is of particular interest because its purpose is unknown. Here, we show that ingestion of a dietary-relevant mix of non-essential AAs activates HONs and shifts behavior from eating to exploration. These effects persisted despite ablation of a key neural gut → brain communication pathway, the cholecystokinin-sensitive vagal afferents. The behavioral shift induced by the ingested non-essential AAs was recapitulated by targeted HON optostimulation and abolished in mice lacking HONs. Furthermore, lick microstructure analysis indicated that intragastric non-essential AAs and HON optostimulation each reduce the size, but not the frequency, of consumption bouts, thus implicating food palatability modulation as a mechanism for the eating suppression. Collectively, these results suggest that a key purpose of HON activation by ingested, non-essential AAs is to suppress eating and re-initiate food seeking. We propose and discuss possible evolutionary advantages of this, such as optimizing the limited stomach capacity for ingestion of essential nutrients.
Collapse
Affiliation(s)
- Paulius Viskaitis
- ETH Zürich, Department of Health Sciences and Technology, Schorenstrasse, Schwerzenbach 8603, Switzerland
| | - Myrtha Arnold
- ETH Zürich, Department of Health Sciences and Technology, Schorenstrasse, Schwerzenbach 8603, Switzerland
| | - Celia Garau
- University of Leicester, Department of Neuroscience, Psychology & Behaviour, University Road, Leicester LE1 9HN, UK
| | - Lise T Jensen
- Aarhus University, Department of Clinical Medicine - Department of Clinical Immunology, Palle Juul-Jensens Boulevard, Aarhus 8200, Denmark
| | - Lars Fugger
- Aarhus University, Department of Clinical Medicine - Department of Clinical Immunology, Palle Juul-Jensens Boulevard, Aarhus 8200, Denmark; University of Oxford, Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, Division of Clinical Neurology, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Daria Peleg-Raibstein
- ETH Zürich, Department of Health Sciences and Technology, Schorenstrasse, Schwerzenbach 8603, Switzerland
| | - Denis Burdakov
- ETH Zürich, Department of Health Sciences and Technology, Schorenstrasse, Schwerzenbach 8603, Switzerland.
| |
Collapse
|
26
|
Abstract
BACKGROUND Obesity develops due to an imbalance in energy homeostasis, wherein energy intake exceeds energy expenditure. Accumulating evidence shows that manipulations of dietary protein and their component amino acids affect the energy balance, resulting in changes in fat mass and body weight. Amino acids are not only the building blocks of proteins but also serve as signals regulating multiple biological pathways. SCOPE OF REVIEW We present the currently available evidence regarding the effects of dietary alterations of a single essential amino acid (EAA) on energy balance and relevant signaling mechanisms at both central and peripheral levels. We summarize the association between EAAs and obesity in humans and the clinical use of modifying the dietary EAA composition for therapeutic intervention in obesity. Finally, similar mechanisms underlying diets varying in protein levels and diets altered of a single EAA are described. The current review would expand our understanding of the contribution of protein and amino acids to energy balance control, thus helping discover novel therapeutic approaches for obesity and related diseases. MAJOR CONCLUSIONS Changes in circulating EAA levels, particularly increased branched-chain amino acids (BCAAs), have been reported in obese human and animal models. Alterations in dietary EAA intake result in improvements in fat and weight loss in rodents, and each has its distinct mechanism. For example, leucine deprivation increases energy expenditure, reduces food intake and fat mass, primarily through regulation of the general control nonderepressible 2 (GCN2) and mammalian target of rapamycin (mTOR) signaling. Methionine restriction by 80% decreases fat mass and body weight while developing hyperphagia, primarily through fibroblast growth factor 21 (FGF-21) signaling. Some effects of diets with different protein levels on energy homeostasis are mediated by similar mechanisms. However, reports on the effects and underlying mechanisms of dietary EAA imbalances on human body weight are few, and more investigations are needed in future.
Collapse
Affiliation(s)
- Fei Xiao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China
| | - Feifan Guo
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China.
| |
Collapse
|
27
|
Wu CT, Chaffin AT, Ryan KK. Fibroblast Growth Factor 21 Facilitates the Homeostatic Control of Feeding Behavior. J Clin Med 2022; 11:580. [PMID: 35160033 PMCID: PMC8836936 DOI: 10.3390/jcm11030580] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/01/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) is a stress hormone that is released from the liver in response to nutritional and metabolic challenges. In addition to its well-described effects on systemic metabolism, a growing body of literature now supports the notion that FGF21 also acts via the central nervous system to control feeding behavior. Here we review the current understanding of FGF21 as a hormone regulating feeding behavior in rodents, non-human primates, and humans. First, we examine the nutritional contexts that induce FGF21 secretion. Initial reports describing FGF21 as a 'starvation hormone' have now been further refined. FGF21 is now better understood as an endocrine mediator of the intracellular stress response to various nutritional manipulations, including excess sugars and alcohol, caloric deficits, a ketogenic diet, and amino acid restriction. We discuss FGF21's effects on energy intake and macronutrient choice, together with our current understanding of the underlying neural mechanisms. We argue that the behavioral effects of FGF21 function primarily to maintain systemic macronutrient homeostasis, and in particular to maintain an adequate supply of protein and amino acids for use by the cells.
Collapse
Affiliation(s)
| | | | - Karen K. Ryan
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA 95616, USA; (C.-T.W.); (A.T.C.)
| |
Collapse
|
28
|
Recommended egg intake in children: past, present, and future. NUTR HOSP 2022; 39:44-51. [DOI: 10.20960/nh.04311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
29
|
Khan MS, Spann RA, Münzberg H, Yu S, Albaugh VL, He Y, Berthoud HR, Morrison CD. Protein Appetite at the Interface between Nutrient Sensing and Physiological Homeostasis. Nutrients 2021; 13:4103. [PMID: 34836357 PMCID: PMC8620426 DOI: 10.3390/nu13114103] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/04/2021] [Accepted: 11/11/2021] [Indexed: 12/19/2022] Open
Abstract
Feeding behavior is guided by multiple competing physiological needs, as animals must sense their internal nutritional state and then identify and consume foods that meet nutritional needs. Dietary protein intake is necessary to provide essential amino acids and represents a specific, distinct nutritional need. Consistent with this importance, there is a relatively strong body of literature indicating that protein intake is defended, such that animals sense the restriction of protein and adaptively alter feeding behavior to increase protein intake. Here, we argue that this matching of food consumption with physiological need requires at least two concurrent mechanisms: the first being the detection of internal nutritional need (a protein need state) and the second being the discrimination between foods with differing nutritional compositions. In this review, we outline various mechanisms that could mediate the sensing of need state and the discrimination between protein-rich and protein-poor foods. Finally, we briefly describe how the interaction of these mechanisms might allow an animal to self-select between a complex array of foods to meet nutritional needs and adaptively respond to changes in either the external environment or internal physiological state.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Christopher D. Morrison
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA; (M.S.K.); (R.A.S.); (H.M.); (S.Y.); (V.L.A.); (Y.H.); (H.-R.B.)
| |
Collapse
|
30
|
Worth AA, Luckman SM. Do oxytocin neurones affect feeding? J Neuroendocrinol 2021; 33:e13035. [PMID: 34495565 PMCID: PMC11475321 DOI: 10.1111/jne.13035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/30/2021] [Accepted: 08/19/2021] [Indexed: 12/24/2022]
Abstract
There has been a long history of research on the effects of oxytocin on feeding behaviour. The classic-held view is that the neurohormone is anorexigenic at least in rodents, although the data for humans are not so clear cut. Likewise, a physiological role for oxytocin is disputed. Thus, although pharmacological, anatomical and physiological data suggest oxytocin may have a function in satiety signalling, this view is not supported by the latest research using the genetic recording and manipulation of oxytocin neurones. Here, we avoid a discussion of the pharmacological effects of oxytocin and examine evidence, from both sides of the argument, concerning whether the endogenous oxytocin system has a role in the regulation of normal feeding.
Collapse
Affiliation(s)
- Amy A. Worth
- Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Simon M. Luckman
- Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| |
Collapse
|
31
|
Niu J, Tong J, Blevins JE. Oxytocin as an Anti-obesity Treatment. Front Neurosci 2021; 15:743546. [PMID: 34720864 PMCID: PMC8549820 DOI: 10.3389/fnins.2021.743546] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 09/16/2021] [Indexed: 12/19/2022] Open
Abstract
Obesity is a growing health concern, as it increases risk for heart disease, hypertension, type 2 diabetes, cancer, COVID-19 related hospitalizations and mortality. However, current weight loss therapies are often associated with psychiatric or cardiovascular side effects or poor tolerability that limit their long-term use. The hypothalamic neuropeptide, oxytocin (OT), mediates a wide range of physiologic actions, which include reproductive behavior, formation of prosocial behaviors and control of body weight. We and others have shown that OT circumvents leptin resistance and elicits weight loss in diet-induced obese rodents and non-human primates by reducing both food intake and increasing energy expenditure (EE). Chronic intranasal OT also elicits promising effects on weight loss in obese humans. This review evaluates the potential use of OT as a therapeutic strategy to treat obesity in rodents, non-human primates, and humans, and identifies potential mechanisms that mediate this effect.
Collapse
Affiliation(s)
- JingJing Niu
- VA Puget Sound Health Care System, Office of Research and Development, Medical Research Service, Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| | - Jenny Tong
- VA Puget Sound Health Care System, Office of Research and Development, Medical Research Service, Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| | - James E. Blevins
- VA Puget Sound Health Care System, Office of Research and Development, Medical Research Service, Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
32
|
Tran PV, Tamura Y, Pham CV, Elhussiny MZ, Han G, Chowdhury VS, Furuse M. Neuropeptide Y modifies a part of diencephalic catecholamine but not indolamine metabolism in chicks depending on feeding status. Neuropeptides 2021; 89:102169. [PMID: 34229214 DOI: 10.1016/j.npep.2021.102169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 11/16/2022]
Abstract
The role of the monoaminergic system in the feeding behavior of neonatal chicks has been reported, but the functional relationship between the metabolism of monoamines and appetite-related neuropeptides is still unclear. This study aimed to investigate the changes in catecholamine and indolamine metabolism in response to the central action of neuropeptide Y (NPY) in different feeding statuses and the underlying mechanisms. In Experiment 1, the diencephalic concentrations of amino acids and monoamines following the intracerebroventricular (ICV) injection of NPY (375 pmol/10 μl/chick), saline solution under ad libitum, and fasting conditions for 30 min were determined. Central NPY significantly decreased L-tyrosine concentration, the precursor of catecholamines under feeding condition, but not under fasting condition. Central NPY significantly increased dopamine metabolites, including 3,4-dihydroxyphenylacetic acid and homovanillic acid (HVA). The concentration of 3-methoxy-4-hydroxyphenylglycol was significantly reduced under feeding condition, but did not change under fasting condition by NPY. However, no effects of NPY on indolamine metabolism were found in either feeding status. Therefore, the mechanism of action of catecholamines with central NPY under feeding condition was elucidated in Experiment 2. Central NPY significantly attenuated diencephalic gene expression of catecholaminergic synthetic enzymes, such as tyrosine hydroxylase, L-aromatic amino acid decarboxylase, and GTP cyclohydrolase I after 30 min of feeding. In Experiment 3, co-injection of α-methyl-L-tyrosine, an inhibitor of tyrosine hydroxylase with NPY, moderately attenuated the orexigenic effect of NPY, accompanied by a significant positive correlation between food intake and HVA levels. In Experiment 4, there was a significant interaction between NPY and clorgyline, an inhibitor of monoamine oxidase A with ICV co-injection which implies that co-existence of NPY and clorgyline enhances the orexigenic effect of NPY. In conclusion, central NPY modifies a part of catecholamine metabolism, which is illustrated by the involvement of dopamine transmission and metabolism under feeding but not fasting conditions.
Collapse
Affiliation(s)
- Phuong V Tran
- Laboratory of Regulation in Metabolism and Behavior, Graduate School of Bioresource and Bioenvironmental Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Yui Tamura
- Laboratory of Regulation in Metabolism and Behavior, Graduate School of Bioresource and Bioenvironmental Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Cuong V Pham
- Laboratory of Regulation in Metabolism and Behavior, Graduate School of Bioresource and Bioenvironmental Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Mohamed Z Elhussiny
- Laboratory of Regulation in Metabolism and Behavior, Graduate School of Bioresource and Bioenvironmental Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Guofeng Han
- Laboratory of Regulation in Metabolism and Behavior, Graduate School of Bioresource and Bioenvironmental Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Vishwajit S Chowdhury
- Laboratory of Stress Physiology and Metabolism, Division of Experimental Natural Science, Faculty of Arts and Science, Graduate School of Bioresource and Bioenvironmental Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Mitsuhiro Furuse
- Laboratory of Regulation in Metabolism and Behavior, Graduate School of Bioresource and Bioenvironmental Science, Kyushu University, Fukuoka 819-0395, Japan.
| |
Collapse
|
33
|
Supruniuk E, Żebrowska E, Chabowski A. Branched chain amino acids-friend or foe in the control of energy substrate turnover and insulin sensitivity? Crit Rev Food Sci Nutr 2021; 63:2559-2597. [PMID: 34542351 DOI: 10.1080/10408398.2021.1977910] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Branched chain amino acids (BCAA) and their derivatives are bioactive molecules with pleiotropic functions in the human body. Elevated fasting blood BCAA concentrations are considered as a metabolic hallmark of obesity, insulin resistance, dyslipidaemia, nonalcoholic fatty liver disease, type 2 diabetes and cardiovascular disease. However, since increased BCAA amount is observed both in metabolically healthy and obese subjects, a question whether BCAA are mechanistic drivers of insulin resistance and its morbidities or only markers of metabolic dysregulation, still remains open. The beneficial effects of BCAA on body weight and composition, aerobic capacity, insulin secretion and sensitivity demand high catabolic potential toward amino acids and/or adequate BCAA intake. On the opposite, BCAA-related inhibition of lipogenesis and lipolysis enhancement may preclude impairment in insulin sensitivity. Thereby, the following review addresses various strategies pertaining to the modulation of BCAA catabolism and the possible roles of BCAA in energy homeostasis. We also aim to elucidate mechanisms behind the heterogeneity of ramifications associated with BCAA modulation.
Collapse
Affiliation(s)
- Elżbieta Supruniuk
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Ewa Żebrowska
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
34
|
Kerem L, Lawson EA. The Effects of Oxytocin on Appetite Regulation, Food Intake and Metabolism in Humans. Int J Mol Sci 2021; 22:7737. [PMID: 34299356 PMCID: PMC8306733 DOI: 10.3390/ijms22147737] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/18/2022] Open
Abstract
The hypothalamic peptide oxytocin and its receptor are involved in a range of physiological processes, including parturition, lactation, cell growth, wound healing, and social behavior. More recently, increasing evidence has established the effects of oxytocin on food intake, energy expenditure, and peripheral metabolism. In this review, we provide a comprehensive description of the central oxytocinergic system in which oxytocin acts to shape eating behavior and metabolism. Next, we discuss the peripheral beneficial effects oxytocin exerts on key metabolic organs, including suppression of visceral adipose tissue inflammation, skeletal muscle regeneration, and bone tissue mineralization. A brief summary of oxytocin actions learned from animal models is presented, showing that weight loss induced by chronic oxytocin treatment is related not only to its anorexigenic effects, but also to the resulting increase in energy expenditure and lipolysis. Following an in-depth discussion on the technical challenges related to endogenous oxytocin measurements in humans, we synthesize data related to the association between endogenous oxytocin levels, weight status, metabolic syndrome, and bone health. We then review clinical trials showing that in humans, acute oxytocin administration reduces food intake, attenuates fMRI activation of food motivation brain areas, and increases activation of self-control brain regions. Further strengthening the role of oxytocin in appetite regulation, we review conditions of hypothalamic insult and certain genetic pathologies associated with oxytocin depletion that present with hyperphagia, extreme weight gain, and poor metabolic profile. Intranasal oxytocin is currently being evaluated in human clinical trials to learn whether oxytocin-based therapeutics can be used to treat obesity and its associated sequela. At the end of this review, we address the fundamental challenges that remain in translating this line of research to clinical care.
Collapse
Affiliation(s)
- Liya Kerem
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA;
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children, Boston, MA 02114, USA
| | - Elizabeth A. Lawson
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA;
| |
Collapse
|
35
|
Quante M, Iske J, Heinbokel T, Desai BN, Cetina Biefer HR, Nian Y, Krenzien F, Matsunaga T, Uehara H, Maenosono R, Azuma H, Pratschke J, Falk CS, Lo T, Sheu E, Tavakkoli A, Abdi R, Perkins D, Alegre ML, Banks AS, Zhou H, Elkhal A, Tullius SG. Restored TDCA and valine levels imitate the effects of bariatric surgery. eLife 2021; 10:e62928. [PMID: 34155969 PMCID: PMC8257250 DOI: 10.7554/elife.62928] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 05/20/2021] [Indexed: 12/12/2022] Open
Abstract
Background Obesity is widespread and linked to various co-morbidities. Bariatric surgery has been identified as the only effective treatment, promoting sustained weight loss and the remission of co-morbidities. Methods Metabolic profiling was performed on diet-induced obese (DIO) mice, lean mice, and DIO mice that underwent sleeve gastrectomies (SGx). In addition, mice were subjected to intraperitoneal (i.p.) injections with taurodeoxycholic acid (TDCA) and valine. Indirect calorimetry was performed to assess food intake and energy expenditure. Expression of appetite-regulating hormones was assessed through quantification of isolated RNA from dissected hypothalamus tissue. Subsequently, i.p. injections with a melanin-concentrating hormone (MCH) antagonist and intrathecal administration of MCH were performed and weight loss was monitored. Results Mass spectrometric metabolomic profiling revealed significantly reduced systemic levels of TDCA and L-valine in DIO mice. TDCA and L-valine levels were restored after SGx in both human and mice to levels comparable with lean controls. Systemic treatment with TDCA and valine induced a profound weight loss analogous to effects observed after SGx. Utilizing indirect calorimetry, we confirmed reduced food intake as causal for TDCA/valine-mediated weight loss via a central inhibition of the MCH. Conclusions In summary, we identified restored TDCA/valine levels as an underlying mechanism of SGx-derived effects on weight loss. Of translational relevance, TDCA and L-valine are presented as novel agents promoting weight loss while reversing obesity-associated metabolic disorders. Funding This work has been supported in part by a grant from NIH (UO-1 A1 132898 to S.G.T., DP and MA). M.Q. was supported by the IFB Integrated Research and Treatment Centre Adiposity Diseases (Leipzig, Germany) and the German Research Foundation (QU 420/1-1). J.I. was supported by the Biomedical Education Program (BMEP) of the German Academic Exchange Service (DAAD). T.H. (HE 7457/1-1) and F.K. (KR 4362/1-1) were supported by the German Research Foundation (DFG). H.R.C.B. was supported the Swiss Society of Cardiac Surgery. Y.N. was supported by the Chinese Scholarship Council (201606370196) and Central South University. H.U., T.M. and R.M. were supported by the Osaka Medical Foundation. C.S.F. was supported by the German Research Foundation (DFG, SFB738, B3).
Collapse
Affiliation(s)
- Markus Quante
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
- Department of General, Visceral and Transplant Surgery, University Hospital TübingenTübingenGermany
| | - Jasper Iske
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
- Institute of Transplant Immunology, Hannover Medical SchoolHannover, Lower SaxonyGermany
| | - Timm Heinbokel
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
- Department of Pathology, Charité Universitätsmedizin BerlinBerlinGermany
| | - Bhavna N Desai
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical CenterBostonUnited States
| | - Hector Rodriguez Cetina Biefer
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
- Department of Cardiovascular Surgery, Charité Universitätsmedizin BerlinBerlinGermany
| | - Yeqi Nian
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
- Department of Urology, The Second Xiangya Hospital, Central South UniversityChangshaChina
| | - Felix Krenzien
- Department of Visceral, Abdominal and Transplantation Surgery, Charité Universitätsmedizin BerlinBerlinGermany
| | - Tomohisa Matsunaga
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
- Department of Urology, Faculty of Medicine, Osaka Medical and Pharmaceutical UniversityOsakaJapan
| | - Hirofumi Uehara
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
- Department of Urology, Faculty of Medicine, Osaka Medical and Pharmaceutical UniversityOsakaJapan
| | - Ryoichi Maenosono
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
- Department of Urology, Faculty of Medicine, Osaka Medical and Pharmaceutical UniversityOsakaJapan
| | - Haruhito Azuma
- Department of Urology, Faculty of Medicine, Osaka Medical and Pharmaceutical UniversityOsakaJapan
| | - Johann Pratschke
- Department of Visceral, Abdominal and Transplantation Surgery, Charité Universitätsmedizin BerlinBerlinGermany
| | - Christine S Falk
- Institute of Transplant Immunology, Hannover Medical SchoolHannover, Lower SaxonyGermany
| | - Tammy Lo
- Division of Gastrointestinal and General Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Eric Sheu
- Division of Gastrointestinal and General Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Ali Tavakkoli
- Division of Gastrointestinal and General Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Reza Abdi
- Renal Division, Brigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - David Perkins
- Division of Nephrology, Department of Medicine, University of Illinois at ChicagoChicagoUnited States
| | - Maria-Luisa Alegre
- Department of Medicine, Section of Rheumatology, The University of ChicagoChicagoUnited States
| | - Alexander S Banks
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical CenterBostonUnited States
| | - Hao Zhou
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
| | - Abdallah Elkhal
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
| | - Stefan G Tullius
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
36
|
Dale N. Biological insights from the direct measurement of purine release. Biochem Pharmacol 2021; 187:114416. [PMID: 33444569 DOI: 10.1016/j.bcp.2021.114416] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 12/23/2022]
Abstract
Although purinergic signalling has been a well-established and accepted mechanism of chemical communication for many years, it remains important to measure the extracellular concentration of ATP and adenosine in real time. In this review I summarize the reasons why such measurements are still needed, how they provide additional mechanistic insight and give an overview of the techniques currently available to make spatially localised measurements of ATP and adenosine in real time. To illustrate the impact of direct real-time measurements, I explore CO2 and nutrient sensing in the medulla oblongata and hypothalamus. In both of these examples, the sensing involves hemichannel mediated ATP release from glial cells. For CO2 the hemichannels involved, connexin26, are directly CO2-sensitive. This mechanism contributes to the chemosensory control of breathing. In the hypothamalus, specialised glial cells, tanycytes, directly contact the cerebrospinal fluid in the 3rd ventricle and sense nutrients via sweet and umami taste receptors. Nutrient sensing by tanycytes is likely to contribute to the control of body weight as their selective stimulation alters food intake. To illustrate the importance of direct adenosine measurements, I consider the complex and multiple mechanisms of activity-dependent adenosine release in different brain regions. This activity dependent release of adenosine is likely to mediate important feedback regulation and may also be involved in controlling the sleep-wake state. I finish by briefly considering the potential of whole blood purine measurements in clinical practice.
Collapse
Affiliation(s)
- Nicholas Dale
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK.
| |
Collapse
|
37
|
Li RJW, Batchuluun B, Zhang SY, Abraham MA, Wang B, Lim YM, Yue JTY, Lam TKT. Nutrient infusion in the dorsal vagal complex controls hepatic lipid and glucose metabolism in rats. iScience 2021; 24:102366. [PMID: 33870148 PMCID: PMC8044434 DOI: 10.1016/j.isci.2021.102366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 02/08/2021] [Accepted: 03/24/2021] [Indexed: 11/22/2022] Open
Abstract
Hypothalamic regulation of lipid and glucose homeostasis is emerging, but whether the dorsal vagal complex (DVC) senses nutrients and regulates hepatic nutrient metabolism remains unclear. Here, we found in rats DVC oleic acid infusion suppressed hepatic secretion of triglyceride-rich very-low-density lipoprotein (VLDL-TG), which was disrupted by inhibiting DVC long-chain fatty acyl-CoA synthetase that in parallel disturbed lipid homeostasis during intravenous lipid infusion. DVC glucose infusion elevated local glucose levels similarly as intravenous glucose infusion and suppressed hepatic glucose production. This was independent of lactate metabolism as inhibiting lactate dehydrogenase failed to disrupt glucose sensing and neither could DVC lactate infusion recapitulate glucose effect. DVC oleic acid and glucose infusion failed to lower VLDL-TG secretion and glucose production in high-fat fed rats, while inhibiting DVC farnesoid X receptor enhanced oleic acid but not glucose sensing. Thus, an impairment of DVC nutrient sensing may lead to the disruption of lipid and glucose homeostasis in metabolic syndrome. DVC oleic acid infusion lowers hepatic secretion of VLDL-TG in chow but not HF rats Inhibition of ACSL in the DVC negates lipid sensing DVC glucose infusion lowers hepatic glucose production in chow but not HF rats Inhibition of FXR in the DVC enhances oleic acid but not glucose sensing in HF rats
Collapse
Affiliation(s)
- Rosa J W Li
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada.,Toronto General Hospital Research Institute, UHN, MaRS Center, TMDT 101 College Street, 10-705, Toronto, ON M5G 1L7, Canada
| | - Battsetseg Batchuluun
- Toronto General Hospital Research Institute, UHN, MaRS Center, TMDT 101 College Street, 10-705, Toronto, ON M5G 1L7, Canada
| | - Song-Yang Zhang
- Toronto General Hospital Research Institute, UHN, MaRS Center, TMDT 101 College Street, 10-705, Toronto, ON M5G 1L7, Canada
| | - Mona A Abraham
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada.,Toronto General Hospital Research Institute, UHN, MaRS Center, TMDT 101 College Street, 10-705, Toronto, ON M5G 1L7, Canada
| | - Beini Wang
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada.,Toronto General Hospital Research Institute, UHN, MaRS Center, TMDT 101 College Street, 10-705, Toronto, ON M5G 1L7, Canada
| | - Yu-Mi Lim
- Toronto General Hospital Research Institute, UHN, MaRS Center, TMDT 101 College Street, 10-705, Toronto, ON M5G 1L7, Canada.,Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Republic of Korea
| | - Jessica T Y Yue
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Tony K T Lam
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada.,Toronto General Hospital Research Institute, UHN, MaRS Center, TMDT 101 College Street, 10-705, Toronto, ON M5G 1L7, Canada.,Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada.,Banting and Best Diabetes Centre, University of Toronto, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
38
|
Edwards MM, Nguyen HK, Herbertson AJ, Dodson AD, Wietecha T, Wolden-Hanson T, Graham JL, O'Brien KD, Havel PJ, Blevins JE. Chronic hindbrain administration of oxytocin elicits weight loss in male diet-induced obese mice. Am J Physiol Regul Integr Comp Physiol 2021; 320:R471-R487. [PMID: 33470901 PMCID: PMC8238148 DOI: 10.1152/ajpregu.00294.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/15/2021] [Accepted: 01/16/2021] [Indexed: 02/08/2023]
Abstract
Previous studies indicate that oxytocin (OT) administration reduces body weight in high-fat diet (HFD)-induced obese (DIO) rodents through both reductions in food intake and increases in energy expenditure. We recently demonstrated that chronic hindbrain [fourth ventricular (4V)] infusions of OT evoke weight loss in DIO rats. Based on these findings, we hypothesized that chronic 4V OT would elicit weight loss in DIO mice. We assessed the effects of 4V infusions of OT (16 nmol/day) or vehicle over 28 days on body weight, food intake, and body composition. OT reduced body weight by approximately 4.5% ± 1.4% in DIO mice relative to OT pretreatment body weight (P < 0.05). These effects were associated with reduced adiposity and adipocyte size [inguinal white adipose tissue (IWAT)] (P < 0.05) and attributed, in part, to reduced energy intake (P < 0.05) at a dose that did not increase kaolin intake (P = NS). OT tended to increase uncoupling protein-1 expression in IWAT (0.05 < P < 0.1) suggesting that OT stimulates browning of WAT. To assess OT-elicited changes in brown adipose tissue (BAT) thermogenesis, we examined the effects of 4V OT on interscapular BAT temperature (TIBAT). 4V OT (1 µg) elevated TIBAT at 0.75 (P = 0.08), 1, and 1.25 h (P < 0.05) postinjection; a higher dose (5 µg) elevated TIBAT at 0.75-, 1-, 1.25-, 1.5-, 1.75- (P < 0.05), and 2-h (0.05 < P < 0.1) postinjection. Together, these findings support the hypothesis that chronic hindbrain OT treatment evokes sustained weight loss in DIO mice by reducing energy intake and increasing BAT thermogenesis at a dose that is not associated with evidence of visceral illness.
Collapse
MESH Headings
- Adipocytes, Brown/drug effects
- Adipocytes, Brown/metabolism
- Adipocytes, Brown/pathology
- Adipocytes, White/drug effects
- Adipocytes, White/metabolism
- Adipocytes, White/pathology
- Adiposity/drug effects
- Animals
- Anti-Obesity Agents/administration & dosage
- Diet, High-Fat
- Disease Models, Animal
- Eating/drug effects
- Energy Intake/drug effects
- Infusions, Intraventricular
- Leptin/blood
- Male
- Mice, Inbred C57BL
- Obesity/drug therapy
- Obesity/metabolism
- Obesity/pathology
- Obesity/physiopathology
- Oxytocin/administration & dosage
- Rhombencephalon/drug effects
- Rhombencephalon/physiopathology
- Thermogenesis/drug effects
- Uncoupling Protein 1/metabolism
- Weight Loss/drug effects
- Mice
Collapse
Affiliation(s)
- Melise M Edwards
- Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Veteran Affairs Puget Sound Health Care System, Seattle, Washington
| | - Ha K Nguyen
- Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Veteran Affairs Puget Sound Health Care System, Seattle, Washington
| | - Adam J Herbertson
- Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Veteran Affairs Puget Sound Health Care System, Seattle, Washington
| | - Andrew D Dodson
- Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Veteran Affairs Puget Sound Health Care System, Seattle, Washington
| | - Tomasz Wietecha
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, Washington
| | - Tami Wolden-Hanson
- Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Veteran Affairs Puget Sound Health Care System, Seattle, Washington
| | - James L Graham
- Department of Nutrition, University of California, Davis, California
| | - Kevin D O'Brien
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, Washington
- Division of Cardiology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Peter J Havel
- Department of Nutrition, University of California, Davis, California
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, California
| | - James E Blevins
- Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Veteran Affairs Puget Sound Health Care System, Seattle, Washington
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
39
|
Adjustment of Whey:Casein Ratio from 20:80 to 60:40 in Milk Formulation Affects Food Intake and Brainstem and Hypothalamic Neuronal Activation and Gene Expression in Laboratory Mice. Foods 2021; 10:foods10030658. [PMID: 33808819 PMCID: PMC8003661 DOI: 10.3390/foods10030658] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 02/06/2023] Open
Abstract
Adjustment of protein content in milk formulations modifies protein and energy levels, ensures amino acid intake and affects satiety. The shift from the natural whey:casein ratio of ~20:80 in animal milk is oftentimes done to reflect the 60:40 ratio of human milk. Studies show that 20:80 versus 60:40 whey:casein milks differently affect glucose metabolism and hormone release; these data parallel animal model findings. It is unknown whether the adjustment from the 20:80 to 60:40 ratio affects appetite and brain processes related to food intake. In this set of studies, we focused on the impact of the 20:80 vs. 60:40 whey:casein content in milk on food intake and feeding-related brain processes in the adult organism. By utilising laboratory mice, we found that the 20:80 whey:casein milk formulation was consumed less avidly and was less preferred than the 60:40 formulation in short-term choice and no-choice feeding paradigms. The relative PCR analyses in the hypothalamus and brain stem revealed that the 20:80 whey:casein milk intake upregulated genes involved in early termination of feeding and in an interplay between reward and satiety, such as melanocortin 3 receptor (MC3R), oxytocin (OXT), proopiomelanocortin (POMC) and glucagon-like peptide-1 receptor (GLP1R). The 20:80 versus 60:40 whey:casein formulation intake differently affected brain neuronal activation (assessed through c-Fos, an immediate-early gene product) in the nucleus of the solitary tract, area postrema, ventromedial hypothalamic nucleus and supraoptic nucleus. We conclude that the shift from the 20:80 to 60:40 whey:casein ratio in milk affects short-term feeding and relevant brain processes.
Collapse
|
40
|
da Silva CD, de Oliveira DR, Perrone ÍT, Fonseca CH, Garcia ES. Low-fat, lactose-free and leucine-enriched chocolate cow milk prototype: A preliminary study on sensorial acceptability and gastrointestinal complaints following exhaustive exercise. J Int Soc Sports Nutr 2021; 18:14. [PMID: 33568169 PMCID: PMC7874447 DOI: 10.1186/s12970-020-00406-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 12/26/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chocolate milk has gained recent scientific support as a recovery drink. However, it is known that high exercise-demand triggers gastrointestinal discomfort which continues post-exercise, thereby hindering this nutritional strategy. In addition, those who are lactose intolerant cannot benefit from a milk-based beverage. Thus, the aim of this preliminary study was to develop a low-fat, lactose-free, and leucine-enriched chocolate cow milk prototype (CML) representing nutrition-related recommendations for football players, as well as assess athletes' individual subjective outcomes for gastrointestinal complaints and sensorial acceptability in a field-based setting following strenuous team-sport physical demands. METHODS This study followed a single group and repeated-measured design with 10 football players (23 ± 2 yrs., 74 ± 14 kg, 174 ± 5 cm) who consumed CML following a 90-min football match simulation protocol (FMP). The total CML intake to achieve 0.150 g leucine·kg [BW]·h- 1 occurred in aliquots of 50, 30 and 20% at 0-, 45- and 75-min post-FMP, respectively. Athletes were evaluated by the prevalence, the type and severity (bloating, nausea, flatulence, and gastric reflux) of gastrointestinal complaints and sensorial acceptability (overall perception, appearance, consistency, and flavour) after drinking each aliquot in a 4-h recovery period. RESULTS The CML showed higher scores for "Product Acceptability Index" (88%) and sensorial acceptability (~ 8 in 9-point hedonic scale). Kendall's W with bootstrapped resample (95%CI) revealed agreement among respondents as "moderate" (overall perception, flavour) to "strong" (appearance, consistency) and with no significant agreement differences between rater response in the timeline analysis (0.57 up to 0.87; p > 0.05). Agresti-Caffo add-4 analysis (95% confidence interval, [95%CI]) revealed no differences in each time-point analysis versus baseline for athletes classified as having severe gastrointestinal symptoms, but confirmed concern with bloating (three athletes showed a transient response at 2-h and only one continued until 3-h; p = 0.051). CONCLUSIONS These preliminary findings suggest that CML presents good taste and high acceptability by the sampled athletes. Thus, CML may be an alternative sport drink for immediate post-workout supplementation to overcome the energy deficit, offer co-ingested leucine, maintain palatability and adherence including lactose intolerance following a team sport-specific fatigue. TRIAL REGISTRATION RBR-2vmpz9 , 10/12/2019, retrospectively registered.
Collapse
Affiliation(s)
- Cristiano D da Silva
- Department of Physical Education, Institute of Life Sciences, Federal University of Juiz de Fora, Campus: Governador Valadares, Rua Manoel Byrro, 241 - Vila Bretas, Governador Valadares, MG, 35010-260, Brazil. .,School of Physical Education, Physiotherapy and Occupational Therapy, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Dirce R de Oliveira
- Department of Basic Life Sciences Institute of Life Sciences , Federal University of Juiz de Fora , Campus Governador Valadares, MG, Governador Valadares, Brazil
| | - Ítalo T Perrone
- Department of Food Engineering, Center of Exact Sciences, Federal University of Viçosa, Viçosa, MG, Brazil.,Pharmaceutical Department, Faculty of Pharmacy, Federal University of Juiz de Fora, Campus Juiz de Fora, MG, Brazil
| | - Carlos H Fonseca
- Pharmaceutical Department, Institute of Life Sciences, Federal University of Juiz de Fora, Campus Governador Valadares, MG, Brazil
| | - Emerson S Garcia
- Department of Physical Education, Federal University of Maranhão, São Luís, Maranhão, Brazil
| |
Collapse
|
41
|
Abstract
Sensing and responding to changes in nutrient levels, including those of glucose, lipids, and amino acids, by the body is necessary for survival. Accordingly, perturbations in nutrient sensing are tightly linked with human pathologies, particularly metabolic diseases such as obesity, type 2 diabetes mellitus, and other complications of metabolic syndromes. The conventional view is that amino acids are fundamental elements for protein and peptide synthesis, while recent studies have revealed that amino acids are also important bioactive molecules that play key roles in signaling pathways and metabolic regulation. Different pathways that sense intracellular and extracellular levels of amino acids are integrated and coordinated at the organismal level, and, together, these pathways maintain whole metabolic homeostasis. In this review, we discuss the studies describing how important sensing signals respond to amino acid availability and how these sensing mechanisms modulate metabolic processes, including energy, glucose, and lipid metabolism. We further discuss whether dysregulation of amino acid sensing signals can be targeted to promote metabolic disorders, and discuss how to translate these mechanisms to treat human diseases. This review will help to enhance our overall understanding of the correlation between amino acid sensing and metabolic homeostasis, which have important implications for human health.
Collapse
Affiliation(s)
- Xiaoming Hu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Feifan Guo
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
42
|
Althammer F, Eliava M, Grinevich V. Central and peripheral release of oxytocin: Relevance of neuroendocrine and neurotransmitter actions for physiology and behavior. HANDBOOK OF CLINICAL NEUROLOGY 2021; 180:25-44. [PMID: 34225933 DOI: 10.1016/b978-0-12-820107-7.00003-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The hypothalamic neuropeptide oxytocin (OT) is critically involved in the modulation of socio-emotional behavior, sexual competence, and pain perception and anticipation. While intracellular signaling of OT and its receptor (OTR), as well as the functional connectivity of hypothalamic and extra-hypothalamic OT projections, have been recently explored, it remains elusive how one single molecule has pleotropic effects from cell proliferation all the way to modulation of complex cognitive processes. Moreover, there are astonishing species-dependent differences in the way OT regulates various sensory modalities such as touch, olfaction, and vision, which can be explained by differences in OTR expression in brain regions processing sensory information. Recent research highlights a small subpopulation of OT-synthesizing cells, namely, parvocellular cells, which merely constitute 1% of the total number of OT cells but act as "master cells' that regulate the activity of the entire OT system. In this chapter, we summarize the latest advances in the field of OT research with a particular focus on differences between rodents, monkeys and humans and highlight the main differences between OT and its "sister" peptide arginine-vasopressin, which often exerts opposite effects on physiology and behavior.
Collapse
Affiliation(s)
- Ferdinand Althammer
- Neuroscience Department, Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, United States
| | - Marina Eliava
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
43
|
Dalvi P, Loganathan N, Mcilwraith EK, Tran A, Belsham DD. Hypothalamic Cell Models. CELLULAR ENDOCRINOLOGY IN HEALTH AND DISEASE 2021:27-77. [DOI: 10.1016/b978-0-12-819801-8.00002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
44
|
Wang J, Liang XF, He S, Zhang YP, Li J, Huang K, Shi LJ, Ren P. Valine acts as a nutritional signal in brain to activate TORC1 and attenuate postprandial ammonia-N excretion in Chinese perch (Siniperca chuatsi). FISH PHYSIOLOGY AND BIOCHEMISTRY 2020; 46:2015-2025. [PMID: 32749664 DOI: 10.1007/s10695-020-00767-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 01/13/2020] [Indexed: 06/11/2023]
Abstract
An emerging concept is that the hypothalamic nutrient sensor can regulate peripheral energy metabolism via a brain-liver circuit. Valine is an essential branched-chain amino acid (BCAA) that drives intracellular signaling cascades by the activation of target of rapamycin complex 1 (TORC1) which is critical to protein metabolism in mammals. However, in teleost fish, it remains scarce in this area especially about how the intraventricular (ICV) injection of valine can mediate the protein metabolism in peripheral organs. This study would tentatively explore the effects of ICV injection of valine on protein metabolism in peripheral organs through evaluating the postprandial ammonia-N excretion rate in Chinese perch. The control group was injected with 5-μL PBS, and the Val group was injected with 20-μg L valine dissolved into 5-μL PBS. The ammonia-N excretion rate of Val group was lower than control group at 4-, 12-, and 24-h postinjection, while the concertation of plasma glucose was increased sharply at 0.5-, 4-, 12-, and 24-h postinjection. We further checked both mRNA level and the enzyme activity of glutamate dehydrogenase (GDH) in the liver and adenosine monophosphate deaminase (AMPD) in muscle, and we found that they were obviously decreased in Val group at 4-, 12-, and 24-h postinjection. The phosphorylation level of ribosomal protein S6, a downstream target protein of TORC1, was markedly enhanced in the liver of Val group at 4-, 12-, and 24-h postinjection. Collectively, these results illustrated that ICV injection of valine can attenuate protein degradation in peripheral organs by depressing the GDH and AMPD enzyme activity; on the other hand, the injected valine can trigger the activation of TORC1 in the liver via a brain-liver circuit and then interdict proteolysis.
Collapse
Affiliation(s)
- Jie Wang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
| | - Xu-Fang Liang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China.
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China.
| | - Shan He
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
| | - Yan-Peng Zhang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
| | - Jiao Li
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
| | - Kang Huang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
| | - Lin-Jie Shi
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
| | - Ping Ren
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
| |
Collapse
|
45
|
Tsang AH, Nuzzaci D, Darwish T, Samudrala H, Blouet C. Nutrient sensing in the nucleus of the solitary tract mediates non-aversive suppression of feeding via inhibition of AgRP neurons. Mol Metab 2020; 42:101070. [PMID: 32898712 PMCID: PMC7549147 DOI: 10.1016/j.molmet.2020.101070] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/26/2020] [Accepted: 09/02/2020] [Indexed: 01/03/2023] Open
Abstract
The nucleus of the solitary tract (NTS) is emerging as a major site of action for the appetite-suppressive effects of leading pharmacotherapies currently investigated to treat obesity. However, our understanding of how NTS neurons regulate appetite remains incomplete. OBJECTIVES In this study, we used NTS nutrient sensing as an entry point to characterize stimulus-defined neuronal ensembles engaged by the NTS to produce physiological satiety. METHODS We combined histological analysis, neuroanatomical assessment using inducible viral tracing tools, and functional tests to characterize hindbrain-forebrain circuits engaged by NTS leucine sensing to suppress hunger. RESULTS We found that NTS detection of leucine engages NTS prolactin-releasing peptide (PrRP) neurons to inhibit AgRP neurons via a population of leptin receptor-expressing neurons in the dorsomedial hypothalamus. This circuit is necessary for the anorectic response to NTS leucine, the appetite-suppressive effect of high-protein diets, and the long-term control of energy balance. CONCLUSIONS These results extend the integrative capability of AgRP neurons to include brainstem nutrient sensing inputs.
Collapse
Affiliation(s)
- Anthony H Tsang
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Danae Nuzzaci
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Tamana Darwish
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Havish Samudrala
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Clémence Blouet
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
46
|
Nutritional modulation of leptin expression and leptin action in obesity and obesity-associated complications. J Nutr Biochem 2020; 89:108561. [PMID: 33249183 DOI: 10.1016/j.jnutbio.2020.108561] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 09/11/2020] [Accepted: 11/21/2020] [Indexed: 12/12/2022]
Abstract
In obesity, an elevated accumulation and dysregulation of adipose tissue, due to an imbalance between energy intake and energy expenditure, usually coexists with the loss of responsiveness to leptin in central nervous system, and subsequently with hyperleptinemia. Leptin, a peptide hormone mainly produced by white adipose tissue, regulates energy homeostasis by stimulating energy expenditure and inhibiting food intake. Human obesity is characterized by increased plasma leptin levels, which have been related with different obesity-associated complications, such as chronic inflammatory state (risk factor for diabetes, cardiovascular and autoimmune diseases), as well as infertility and different types of cancer. Besides, leptin is also produced by placenta, and high leptin levels during pregnancy may be related with some pathological conditions such as gestational diabetes. This review focuses on the current insights and emerging concepts on potentially valuable nutrients and food components that may modulate leptin metabolism. Notably, several dietary food components, such as phenols, peptides, and vitamins, are able to decrease inflammation and improve leptin sensitivity by up- or down-regulation of leptin signaling molecules. On the other hand, some food components, such as saturated fatty acids may worsen chronic inflammation increasing the risk for pathological complications. Future research into nutritional mechanisms that restore leptin metabolism and signals of energy homeostasis may inspire new treatment options for obesity-related disorders.
Collapse
|
47
|
Wald HS, Chandra A, Kalluri A, Ong ZY, Hayes MR, Grill HJ. NTS and VTA oxytocin reduces food motivation and food seeking. Am J Physiol Regul Integr Comp Physiol 2020; 319:R673-R683. [PMID: 33026822 DOI: 10.1152/ajpregu.00201.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Oxytocin (OT) is a neuropeptide whose central receptor-mediated actions include reducing food intake. One mechanism of its behavioral action is the amplification of the feeding inhibitory effects of gastrointestinal (GI) satiation signals processed by hindbrain neurons. OT treatment also reduces carbohydrate intake in humans and rodents, and correspondingly, deficits in central OT receptor (OT-R) signaling increase sucrose self-administration. This suggests that additional processes contribute to central OT effects on feeding. This study investigated the hypothesis that central OT reduces food intake by decreasing food seeking and food motivation. As central OT-Rs are expressed widely, a related focus was to assess the role of one or more OT-R-expressing nuclei in food motivation and food-seeking behavior. OT was delivered to the lateral ventricle (LV), nucleus tractus solitarius (NTS), or ventral tegmental area (VTA), and a progressive ratio (PR) schedule of operant reinforcement and an operant reinstatement paradigm were used to measure motivated feeding behavior and food-seeking behavior, respectively. OT delivered to the LV, NTS, or VTA reduced 1) motivation to work for food and 2) reinstatement of food-seeking behavior. Results provide a novel and additional interpretation for central OT-driven food intake inhibition to include the reduction of food motivation and food seeking.
Collapse
Affiliation(s)
- Hallie S Wald
- Department of Psychology, Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ananya Chandra
- Department of Psychology, Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anita Kalluri
- Department of Psychology, Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Zhi Yi Ong
- School of Psychology, University of New South Wales, Sydney, Australia
| | - Matthew R Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Harvey J Grill
- Department of Psychology, Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
48
|
Impact of Protein Intake in Older Adults with Sarcopenia and Obesity: A Gut Microbiota Perspective. Nutrients 2020; 12:nu12082285. [PMID: 32751533 PMCID: PMC7468805 DOI: 10.3390/nu12082285] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
The continuous population increase of older adults with metabolic diseases may contribute to increased prevalence of sarcopenia and obesity and requires advocacy of optimal nutrition treatments to combat their deleterious outcomes. Sarcopenic obesity, characterized by age-induced skeletal-muscle atrophy and increased adiposity, may accelerate functional decline and increase the risk of disability and mortality. In this review, we explore the influence of dietary protein on the gut microbiome and its impact on sarcopenia and obesity. Given the associations between red meat proteins and altered gut microbiota, a combination of plant and animal-based proteins are deemed favorable for gut microbiota eubiosis and muscle-protein synthesis. Additionally, high-protein diets with elevated essential amino-acid concentrations, alongside increased dietary fiber intake, may promote gut microbiota eubiosis, given the metabolic effects derived from short-chain fatty-acid and branched-chain fatty-acid production. In conclusion, a greater abundance of specific gut bacteria associated with increased satiation, protein synthesis, and overall metabolic health may be driven by protein and fiber consumption. This could counteract the development of sarcopenia and obesity and, therefore, represent a novel approach for dietary recommendations based on the gut microbiota profile. However, more human trials utilizing advanced metabolomic techniques to investigate the microbiome and its relationship with macronutrient intake, especially protein, are warranted.
Collapse
|
49
|
Interaction of glucose sensing and leptin action in the brain. Mol Metab 2020; 39:101011. [PMID: 32416314 PMCID: PMC7267726 DOI: 10.1016/j.molmet.2020.101011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/27/2020] [Accepted: 05/05/2020] [Indexed: 01/10/2023] Open
Abstract
Background In response to energy abundant or deprived conditions, nutrients and hormones activate hypothalamic pathways to maintain energy and glucose homeostasis. The underlying CNS mechanisms, however, remain elusive in rodents and humans. Scope of review Here, we first discuss brain glucose sensing mechanisms in the presence of a rise or fall of plasma glucose levels, and highlight defects in hypothalamic glucose sensing disrupt in vivo glucose homeostasis in high-fat fed, obese, and/or diabetic conditions. Second, we discuss brain leptin signalling pathways that impact glucose homeostasis in glucose-deprived and excessed conditions, and propose that leptin enhances hypothalamic glucose sensing and restores glucose homeostasis in short-term high-fat fed and/or uncontrolled diabetic conditions. Major conclusions In conclusion, we believe basic studies that investigate the interaction of glucose sensing and leptin action in the brain will address the translational impact of hypothalamic glucose sensing in diabetes and obesity.
Collapse
|
50
|
An Allostatic Theory of Oxytocin. Trends Cogn Sci 2020; 24:515-528. [PMID: 32360118 DOI: 10.1016/j.tics.2020.03.008] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 12/31/2022]
Abstract
Oxytocin has garnered considerable interest for its role in social behavior, as well as for the potential of intranasal administration to treat social difficulties. However, current theoretical models for the role of oxytocin in social behavior pay little consideration to its evolutionary and developmental history. This article aims to broaden our understanding of the role of oxytocin in social behavior by adopting an ethological approach through the lens of Nikolaas Tinbergen's 'four questions' - how does oxytocin work; how does the role of oxytocin change during development; how does oxytocin enhance survival; and how did the oxytocin system evolve? We argue that oxytocin is most accurately described as an allostatic hormone that modulates both social and non-social behavior by maintaining stability through changing environments.
Collapse
|