1
|
Lalanza JF, Oyem JC, Huijgens PT, McCutcheon JE, Heijkoop R, Snoeren EMS. Behavioral and neural alterations of the ventral tegmental area by exposure to junk food in rats. Appetite 2025; 214:108146. [PMID: 40414303 DOI: 10.1016/j.appet.2025.108146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/21/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
The brain reward system is essential for regulating appetitive and consummatory behaviors in response to various incentive stimuli. Junk food, characterized by its high palatability, is particularly associated with the potential for excessive consumption. While prior studies indicate that excessive junk food intake can impact reward circuitry, the precise mechanisms underlying these effects remain elusive. Furthermore, it is unclear whether the functionality of this neural system is similarly altered in response to other natural rewards. In this study, we used fiber photometry combined with a behavioral reward test to investigate the effects of six weeks of excessive cafeteria (CAF) diet consumption on ventral tegmental area (VTA) neural activity and behavioral responses to food and sexual rewards in female rats. Our findings demonstrate that prolonged exposure to a CAF diet reduced the exploration and consumption of food rewards. These behavioral changes were accompanied by attenuated neural activity in the VTA. Similarly, reductions in VTA activity were observed in response to a sexual partner, although no significant behavioral differences were detected during sexual interactions. Moreover, a two-week reversal diet of standard chow proved insufficient to restore VTA neural activity in CAF-exposed animals, which continued to exhibit decreased VTA responses to both food rewards and sexual partners. Our results suggest that prolonged junk food exposure induces desensitization of the VTA, resulting in reduced responsiveness to natural rewards.
Collapse
Affiliation(s)
- Jaume F Lalanza
- Department of Psychology, UiT The Arctic University of Norway, Huginbakken 32, 9037, Tromsø, Norway
| | - John C Oyem
- Department of Psychology, UiT The Arctic University of Norway, Huginbakken 32, 9037, Tromsø, Norway
| | - Patty T Huijgens
- Department of Psychology, UiT The Arctic University of Norway, Huginbakken 32, 9037, Tromsø, Norway
| | - James E McCutcheon
- Department of Psychology, UiT The Arctic University of Norway, Huginbakken 32, 9037, Tromsø, Norway
| | - Roy Heijkoop
- Department of Psychology, UiT The Arctic University of Norway, Huginbakken 32, 9037, Tromsø, Norway
| | - Eelke M S Snoeren
- Department of Psychology, UiT The Arctic University of Norway, Huginbakken 32, 9037, Tromsø, Norway.
| |
Collapse
|
2
|
Shrivastava K, Athreya V, Lu Y, Luis-Islas J, Han A, Kowalski TF, Rossi MA. Energy state guides reward seeking via an extended amygdala to lateral hypothalamus pathway. Nat Commun 2025; 16:4474. [PMID: 40368884 PMCID: PMC12078644 DOI: 10.1038/s41467-025-59686-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 04/28/2025] [Indexed: 05/16/2025] Open
Abstract
Impaired regulation of food intake underlies numerous health problems, including obesity and type 2 diabetes, yet how brain systems controlling reward seeking become dysregulated to promote overeating is unknown. Glutamatergic neurons of the lateral hypothalamic area (LHA) are thought to act as a brake on feeding, which is dysregulated during diet-induced obesity. These neurons receive input from the extended amygdala, including the bed nucleus of the stria terminalis (BNST). However, the circuit mechanisms underlying the ability of this pathway to control feeding behavior and how they contribute to dysregulated eating are unclear. Here, we discover that BNST projections to LHA (BNST→LHA) promote reward seeking in an energy state-dependent manner by combining optogenetics, in vivo multiphoton calcium imaging, and electrophysiology in mice. Synaptic strength and neuronal function within the BNST→LHA pathway are dynamically regulated according to energy state to guide reward seeking. These findings suggest that hormonal factors modulate the function of the BNST→LHA pathway to align food seeking with current energy needs.
Collapse
Affiliation(s)
- Kuldeep Shrivastava
- Center for NeuroMetabolism, Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Vikshar Athreya
- Center for NeuroMetabolism, Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Yi Lu
- Center for NeuroMetabolism, Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Jorge Luis-Islas
- Center for NeuroMetabolism, Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Ashley Han
- Center for NeuroMetabolism, Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Tess F Kowalski
- Center for NeuroMetabolism, Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Mark A Rossi
- Center for NeuroMetabolism, Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA.
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA.
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA.
- Brain Health Institute, Rutgers University, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
3
|
Ammineni D, Park R. The compulsive eating paradigm: can psychedelics help in treating obesity? J Eat Disord 2025; 13:59. [PMID: 40197427 PMCID: PMC11978192 DOI: 10.1186/s40337-024-01186-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/20/2024] [Indexed: 04/10/2025] Open
Abstract
Obesity is a multifactorial disorder involving a behavioural aetiology in subsets of patients that traditional therapeutic approaches have failed to address. Drawing parallels with addiction, the rewarding aspects of a chronic energy-dense diet can compromise dopaminergic reward circuits, eventually causing individuals to become habitually responsive to food-related stimuli despite adverse health consequences. The maladaptive prediction of reward and motivational salience that becomes associated with food-related stimuli can exert top-down influence on perception and attention, promoting compulsive eating behaviour. Emerging research suggests that psychedelics, e.g., psilocybin and LSD, induce non-ordinary mental states where the influence of such behaviours could potentially be reduced and modified. Based on current evidence, mechanisms have been proposed which suggest that psychedelics might relax the top-down influence of high-level predictions encoded within neuronal hierarchies and sensitise them to bottom-up information flow. Additionally, psychedelics are thought to open a window of psychological flexibility, allowing people to potentially become open to new cognitive and behavioural strategies that can be offered via assisted psychotherapy. Therefore, psychedelics-assisted psychotherapy may encourage beneficial changes to eating behaviour, in those with maladaptive eating habits. While promising in theory, new research is needed to assess the potential efficacy of psychedelics-assisted psychotherapy in treating compulsive eating behaviour.
Collapse
|
4
|
Teixeira MR, Silva T, Felício RDFM, Bozza PT, Zembrzuski VM, de Mello Neto CB, da Fonseca ACP, Kohlrausch FB, Salum KCR. Exploring the genetic contribution in obesity: An overview of dopaminergic system genes. Behav Brain Res 2025; 480:115401. [PMID: 39689745 DOI: 10.1016/j.bbr.2024.115401] [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: 08/26/2024] [Revised: 12/13/2024] [Accepted: 12/13/2024] [Indexed: 12/19/2024]
Abstract
Obesity is a widespread global health concern that affects a significant portion of the population and is associated with reduced quality of life, morbidity, and mortality. It is considered a pandemic, with its prevalence constantly rising in Western countries. As a result, numerous studies have focused on understanding the elements that contribute to obesity. Researchers have focused on neurotransmitters in the brain to develop weight management drugs that regulate food intake. This review explores the literature on genetic influences on dopaminergic processes to determine whether genetic variation has an association with obesity in reward-responsive regions, including mesolimbic efferent and mesocortical areas. Various neurotransmitters play an essential role in regulating food intake, such as dopamine which controls through mesolimbic circuits in the brain that modulate food reward. Appetite stimulation, including primary reinforcers such as food, leads to an increase in dopamine release in the reward centers of the brain. This release is related to motivation and reinforcement, which determines the motivational weighting of the reinforcer. Changes in dopamine expression can lead to hedonic eating behaviors and contribute to the development of obesity. Genetic polymorphisms have been investigated due to their potential role in modulating the risk of obesity and eating behaviors. Therefore, it is crucial to assess the impact of genetic alterations that disrupt this pathway on the obesity phenotype.
Collapse
Affiliation(s)
- Myrela Ribeiro Teixeira
- Human Genetics Laboratory, Department of General Biology, Institute of Biology, Federal Fluminense University, Professor Marcos Waldemar de Freitas Reis Street, Niterói, RJ 24210-201, Brazil; Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), 4365 Brazil Avenue, Leônidas Deane Pavilion, Rio de Janeiro, RJ 21040-360, Brazil; Postgraduate Program in Science and Biotechnology, Department of General Biology, Institute of Biology, Federal Fluminense University, Professor Marcos Waldemar de Freitas Reis Street, Niterói, RJ 24210-201, Brazil
| | - Tamara Silva
- Genetics Laboratory, Grande Rio University/AFYA, Professor José de Souza Herdy Street, 1160 - Jardim Vinte e Cinco de Agosto, Duque de Caxias, RJ 25071-202, Brazil
| | - Rafaela de Freitas Martins Felício
- Congenital Malformation Epidemiology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), 4365 Brazil Avenue, Leônidas Deane Pavilion, Rio de Janeiro, RJ 21040-360, Brazil
| | - Patrícia Torres Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), 4365 Brazil Avenue, Rio de Janeiro, RJ 21040‑360, Brazil
| | - Verônica Marques Zembrzuski
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), 4365 Brazil Avenue, Leônidas Deane Pavilion, Rio de Janeiro, RJ 21040-360, Brazil
| | - Cicero Brasileiro de Mello Neto
- Human Genetics Laboratory, Department of General Biology, Institute of Biology, Federal Fluminense University, Professor Marcos Waldemar de Freitas Reis Street, Niterói, RJ 24210-201, Brazil; Postgraduate Program in Science and Biotechnology, Department of General Biology, Institute of Biology, Federal Fluminense University, Professor Marcos Waldemar de Freitas Reis Street, Niterói, RJ 24210-201, Brazil
| | - Ana Carolina Proença da Fonseca
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), 4365 Brazil Avenue, Leônidas Deane Pavilion, Rio de Janeiro, RJ 21040-360, Brazil; Genetics Laboratory, Grande Rio University/AFYA, Professor José de Souza Herdy Street, 1160 - Jardim Vinte e Cinco de Agosto, Duque de Caxias, RJ 25071-202, Brazil; Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), 4365 Brazil Avenue, Rio de Janeiro, RJ 21040‑360, Brazil; Postgraduate Program in Translational Biomedicine, Grande Rio University/AFYA, Professor José de Souza Herdy Street, 1160 - Jardim Vinte e Cinco de Agosto, Duque de Caxias, RJ 25071-202, Brazil
| | - Fabiana Barzotto Kohlrausch
- Human Genetics Laboratory, Department of General Biology, Institute of Biology, Federal Fluminense University, Professor Marcos Waldemar de Freitas Reis Street, Niterói, RJ 24210-201, Brazil
| | - Kaio Cezar Rodrigues Salum
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), 4365 Brazil Avenue, Leônidas Deane Pavilion, Rio de Janeiro, RJ 21040-360, Brazil; Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Professor Rodolpho Paulo Rocco Street, 255, University City, Rio de Janeiro, RJ 21941-617, Brazil.
| |
Collapse
|
5
|
Darcey VL, Guo J, Chi M, Chung ST, Courville AB, Gallagher I, Herscovitch P, Joseph PV, Howard R, La Noire M, Milley L, Schick A, Stagliano M, Turner S, Urbanski N, Yang S, Zhai N, Zhou MS, Hall KD. Brain dopamine responses to ultra-processed milkshakes are highly variable and not significantly related to adiposity in humans. Cell Metab 2025; 37:616-628.e5. [PMID: 40043691 DOI: 10.1016/j.cmet.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/03/2024] [Accepted: 02/04/2025] [Indexed: 03/12/2025]
Abstract
Ultra-processed foods high in fat and sugar have been theorized to be addictive due to their purported ability to induce an exaggerated post-ingestive brain dopamine response akin to drugs of abuse. Using [11C]raclopride positron emission tomography (PET) displacement methods used to measure brain dopamine responses to addictive drugs, we measured striatal dopamine responses beginning 30 min after ingesting an ultra-processed milkshake high in fat and sugar in 50 young, healthy adults over a wide body mass index (BMI) range (20-45 kg/m2). Surprisingly, milkshake consumption did not result in a significant post-ingestive dopamine response in the striatum (p = 0.62) nor in any striatal subregion (p > 0.33), and the highly variable interindividual responses were not significantly related to adiposity (BMI: r = 0.076, p = 0.51; % body fat: r = 0.16, p = 0.28). Thus, post-ingestive striatal dopamine responses to an ultra-processed milkshake were likely substantially smaller than for many addictive drugs and below the limits of detection using standard PET methods.
Collapse
Affiliation(s)
- Valerie L Darcey
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA; Center on Compulsive Behaviors, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Juen Guo
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Meible Chi
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stephanie T Chung
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Amber B Courville
- Human Energy and Body Weight Regulation Core, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Isabelle Gallagher
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter Herscovitch
- Positron Emission Tomography Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Paule V Joseph
- Section of Sensory Science and Metabolism, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA; National Institute of Nursing Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA; National Institute on Deafness and Other Communication Disorders, Smell and Taste Center, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Rebecca Howard
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Melissa La Noire
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lauren Milley
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alex Schick
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael Stagliano
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sara Turner
- Nutrition Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Nicholas Urbanski
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shanna Yang
- Nutrition Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Nan Zhai
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Megan S Zhou
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kevin D Hall
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
6
|
Mathes CM, Terrill SJ, Taborda-Bejarano JP, Chometton S, Witt MJ, Mendiratta G, Gilman EG, Hartswick DR, Anderson BM, Schier LA. Neurobehavioral plasticity in the rodent gustatory system induced by regular consumption of a low-calorie sweetener during adolescence. Sci Rep 2025; 15:2359. [PMID: 39824856 PMCID: PMC11742420 DOI: 10.1038/s41598-024-84391-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 12/23/2024] [Indexed: 01/20/2025] Open
Abstract
Habitual consumption of low-calorie sweeteners (LCS) during juvenile-adolescence can lead to greater sugar intake later in life. Here, we investigated if exposure to the LCS Acesulfame Potassium (Ace-K) during this critical period of development reprograms the taste system in a way that would alter hedonic responding for common dietary compounds. Results revealed that early-life LCS intake not only enhanced the avidity for a caloric sugar (fructose) when rats were in a state of caloric need, it increased acceptance of a bitterant (quinine) in Ace-K-exposed rats tested when middle-aged. These behavior shifts, which endured months after the end of Ace-K exposure, were accompanied by widespread changes in the peripheral taste system. The anterior tongue had fewer fungiform taste papillae, and the circumvallate papillae had a reduced anterior to posterior span and diminished expression of genes involved in sweet reception, sweet and bitter intracellular signaling, fructose transport, and cellular progeneration in the Ace-K-exposed rats. Ace-K exposure also led to a significant reduction in dopamine-producing cells of the ventral tegmental area. The collective findings reveal that LCS intake early in life alters the taste-brain axis and the behavioral responsiveness to both positive and negative tastants that are important determinants of dietary preferences.
Collapse
Affiliation(s)
- Clare M Mathes
- Department of Neuroscience, Baldwin Wallace University, Berea, OH, USA
| | - Sarah J Terrill
- Department of Neuroscience, Carthage College, Kenosha, WI, USA
| | | | - Sandrine Chometton
- Department of Biological Sciences, University of Southern California, 3616 Trousdale Parkway, AHF 252, Los Angeles, CA, 90089-0372, USA
- Centre for Taste and Feeding Behavior, CNRS, INRAE, Institut AgroDijon, University of Burgundy, Dijon, France
| | - Mallory J Witt
- Department of Neuroscience, Baldwin Wallace University, Berea, OH, USA
| | | | - Emily G Gilman
- Department of Neuroscience, Baldwin Wallace University, Berea, OH, USA
| | | | - Bo M Anderson
- Department of Neuroscience, Carthage College, Kenosha, WI, USA
| | - Lindsey A Schier
- Department of Biological Sciences, University of Southern California, 3616 Trousdale Parkway, AHF 252, Los Angeles, CA, 90089-0372, USA.
| |
Collapse
|
7
|
Ribeiro G, Fernandes AB, Oliveira FPM, Duarte JS, Oliveira M, Limbert C, Costa RM, Costa DC, Oliveira-Maia AJ. Postingestive reward acts through behavioral reinforcement and is conserved in obesity and after bariatric surgery. PLoS Biol 2024; 22:e3002936. [PMID: 39689052 DOI: 10.1371/journal.pbio.3002936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 11/13/2024] [Indexed: 12/19/2024] Open
Abstract
Postingestive nutrient stimulation conditions food preferences through striatal dopamine and may be associated with blunted brain responses in obesity. In a cross-sectional study, we tested flavor-nutrient conditioning (FNC) with maltodextrin-enriched yogurt, with maltodextrin previously optimized for concentration and dextrose equivalents (n = 57), and to mask texture cues (n = 102). After conditioning, healthy volunteers (n = 52) increased preference for maltodextrin-paired (+102 kcal, CS+), relative to control (+1.8 kcal, CS-) flavors, as assessed according to intake, but not pleasantness. In a clinical study (n = 61), behavioral conditioning without effects on pleasantness was confirmed across pre-bariatric candidates with obesity, weight-stable post-surgery patients, and healthy controls, without significant differences between groups. Striatal dopamine D2-like receptor (DD2lR) availability, assessed with [123I]IBZM SPECT, was reduced in the obesity group and strongly correlated with conditioning strength and a measure of restrained eating in patients with gastric bypass. These results show that postingestive nutrient stimulation influences human food choices through behavioral reinforcement, and is conserved in obesity and after bariatric surgery. Trial Registration: ISRCTN17965026: Dopaminergic neurotransmission in dietary learning and obesity.
Collapse
Affiliation(s)
- Gabriela Ribeiro
- Champalimaud Research & Clinical Centre, Champalimaud Foundation, Av. de Brasília, Doca de Pedrouços, Lisboa, Portugal
- Lisbon Academic Medical Centre PhD Program, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, Lisboa, Portugal
| | - Ana B Fernandes
- Champalimaud Research & Clinical Centre, Champalimaud Foundation, Av. de Brasília, Doca de Pedrouços, Lisboa, Portugal
- NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo Mártires da Pátria 130, Lisboa, Portugal
| | - Francisco P M Oliveira
- Champalimaud Research & Clinical Centre, Champalimaud Foundation, Av. de Brasília, Doca de Pedrouços, Lisboa, Portugal
| | - João S Duarte
- Department of Endocrinology, Centro Hospitalar de Lisboa Ocidental, Rua da Junqueira, Lisboa, Portugal
| | - Manuela Oliveira
- Department of Endocrinology, Centro Hospitalar de Lisboa Ocidental, Rua da Junqueira, Lisboa, Portugal
| | - Clotilde Limbert
- Department of Endocrinology, Centro Hospitalar de Lisboa Ocidental, Rua da Junqueira, Lisboa, Portugal
| | - Rui M Costa
- Department of Neuroscience and Neurology, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
- Allen Institute, Seattle, Washington State, United States of America
| | - Durval C Costa
- Champalimaud Research & Clinical Centre, Champalimaud Foundation, Av. de Brasília, Doca de Pedrouços, Lisboa, Portugal
| | - Albino J Oliveira-Maia
- Champalimaud Research & Clinical Centre, Champalimaud Foundation, Av. de Brasília, Doca de Pedrouços, Lisboa, Portugal
- NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo Mártires da Pátria 130, Lisboa, Portugal
| |
Collapse
|
8
|
Hagarty-Waite KA, Emmons HA, Fordahl SC, Erikson KM. The Influence of Strain and Sex on High Fat Diet-Associated Alterations of Dopamine Neurochemistry in Mice. Nutrients 2024; 16:3301. [PMID: 39408267 PMCID: PMC11479034 DOI: 10.3390/nu16193301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Objective: The objective of this study was to determine the influence of sex and strain on striatal and nucleus accumbens dopamine neurochemistry and dopamine-related behavior due to a high-saturated-fat diet (HFD). Methods: Male and female C57B6/J (B6J) and Balb/cJ (Balb/c) mice were randomly assigned to a control-fat diet (CFD) containing 10% kcal fat/g or a mineral-matched HFD containing 60% kcal fat/g for 12 weeks. Results: Intraperitoneal glucose tolerance testing (IPGTT) and elevated plus maze experiments (EPM) confirmed that an HFD produced marked blunting of glucose clearance and increased anxiety-like behavior, respectively, in male and female B6J mice. Electrically evoked dopamine release in the striatum and reuptake in the nucleus accumbens (NAc), as measured by ex vivo fast scan cyclic voltammetry, was reduced for HFD-fed B6J females. Impairment in glucose metabolism explained HFD-induced changes in dopamine neurochemistry for B6J males and, to a lesser extent, Balb/c males. The relative expressions of protein markers associated with the activation of microglia, ionized calcium binding adaptor molecule (Iba1) and cluster of differentiation molecule 11b (CD11b) in the striatum were increased due to an HFD for B6J males but were unchanged or decreased amongst HFD-fed Balb/c mice. Conclusions: Our findings demonstrate that strain and sex influence the insulin- and microglia-dependent mechanisms of alterations to dopamine neurochemistry and associated behavior due to an HFD.
Collapse
Affiliation(s)
| | | | | | - Keith M. Erikson
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC 27412, USA; (K.A.H.-W.); (H.A.E.); (S.C.F.)
| |
Collapse
|
9
|
Pescari D, Mihuta MS, Bena A, Stoian D. Comparative Analysis of Dietary Habits and Obesity Prediction: Body Mass Index versus Body Fat Percentage Classification Using Bioelectrical Impedance Analysis. Nutrients 2024; 16:3291. [PMID: 39408258 PMCID: PMC11479188 DOI: 10.3390/nu16193291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/13/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Obesity remains a widely debated issue, often criticized for the limitations in its identification and classification. This study aims to compare two distinct systems for classifying obesity: body mass index (BMI) and body fat percentage (BFP) as assessed by bioelectrical impedance analysis (BIA). By examining these measures, the study seeks to clarify how different metrics of body composition influence the identification of obesity-related risk factors. Methods: The study enrolled 1255 adults, comprising 471 males and 784 females, with a mean age of 36 ± 12 years. Participants exhibited varying degrees of weight status, including optimal weight, overweight, and obesity. Body composition analysis was conducted using the TANITA Body Composition Analyzer BC-418 MA III device (T5896, Tokyo, Japan), evaluating the following parameters: current weight, basal metabolic rate (BMR), adipose tissue (%), muscle mass (%), and hydration status (%). Results: Age and psychological factors like cravings, fatigue, stress, and compulsive eating were significant predictors of obesity in the BMI model but not in the BFP model. Additionally, having a family history of diabetes was protective in the BMI model (OR: 0.33, 0.11-0.87) but increased risk in the BFP model (OR: 1.66, 1.01-2.76). The BMI model demonstrates exceptional predictive ability (AUC = 0.998). In contrast, the BFP model, while still performing well, exhibits a lower AUC (0.975), indicating slightly reduced discriminative power compared to the BMI model. Conclusions: BMI classification demonstrates superior predictive accuracy, specificity, and sensitivity. This suggests that BMI remains a more reliable measure for identifying obesity-related risk factors compared to the BFP model.
Collapse
Affiliation(s)
- Denisa Pescari
- Department of Doctoral Studies, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
- Center for Molecular Research in Nephrology and Vascular Disease, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (A.B.); (D.S.)
| | - Monica Simina Mihuta
- Center for Molecular Research in Nephrology and Vascular Disease, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (A.B.); (D.S.)
| | - Andreea Bena
- Center for Molecular Research in Nephrology and Vascular Disease, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (A.B.); (D.S.)
- Discipline of Endocrinology, Second Department of Internal Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Dana Stoian
- Center for Molecular Research in Nephrology and Vascular Disease, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (A.B.); (D.S.)
- Discipline of Endocrinology, Second Department of Internal Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| |
Collapse
|
10
|
Luengo N, Goldfield GS, Obregón AM. Association between dopamine genes, adiposity, food addiction, and eating behavior in Chilean adult. Front Nutr 2024; 11:1466384. [PMID: 39385779 PMCID: PMC11463150 DOI: 10.3389/fnut.2024.1466384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 08/22/2024] [Indexed: 10/12/2024] Open
Abstract
Background A frequent consumption of high sugar/fat foods can affect dopamine signaling in the brain and cause sustained stimulation of the reward system. It has been hypothesized that a hypodopaminergic trait results in an individual overeating in order to increase brain DA. Genetic variants in this route have been connected with addiction and eating behaviors. Most studies focus on a specific SNP, and few studies have used multilocus genetic scores, which quantify genetic risk on a continuum. Aim To assess the relationship between multilocus genetic scores based on multiple gene variants in the dopaminergic pathway and measurements of anthropometry, eating behavior, food reinforcement, and food addiction (FA) in Chilean adults. Methods We recruited 221 Chilean adults for a cross-sectional study. A standard anthropometric measurement procedure was followed and eating behavior was examined using the Three Factor Eating questionnaire (TFEQ), Food Reinforcement Value Questionnaire (FRVQ), Yale Food Addiction Scale (YFAS) and 24-h diet recall. Multilocus genetic scores were calculated using TaqMan assays (rs1800497-rs1799732-rs6277-rs4680). Results No differences were found in the entire sample for anthropometric measurements, by MLGS. We found that participants with a score ≥ 2.0 in the MLGS showed higher food choices on the RVFQ and lower energy intake in protein, lipids, SAFA, MUFA, PUFA, dietary cholesterol, omega-3 and Omega-6 fatty acids in the 24-h recall (p < 0.05). Stratified by nutritional condition, the group with obesity had inferior scores on cognitive restriction, greater scores on uncontrolled eating, emotional eating, and responding to palatable food in the RVFQ. Also, in subjects with obesity, there was more food addiction in the group scoring "MLGS ≥2.0 or low dopamine signaling" (53%), compared to the group scored "MLGS <2.0 or high dopamine signaling" (23%) (p-value; 0.05). Emotional Eating scores correlated positively with MLGS in subjects with obesity. Conclusion In adults with obesity, the MLGS of the dopamine pathway, reflecting hypodopaminergic signaling, was associated with greater scores on food addiction and altered eating behavior traits.
Collapse
Affiliation(s)
- Nicole Luengo
- Escuela de Nutrición y Dietética, Facultad de Ciencias para el cuidado de la Salud, Universidad San Sebastián, Concepción, Chile
| | - Gary S. Goldfield
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Ana M. Obregón
- Escuela de Nutrición y Dietética, Facultad de Ciencias para el cuidado de la Salud, Universidad San Sebastián, Concepción, Chile
| |
Collapse
|
11
|
Steiner AM, Roscoe RF, Booze RM, Mactutus CF. Motivational dysregulation with melanocortin 4 receptor haploinsufficiency. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2024; 3:237-250. [PMID: 39741559 PMCID: PMC11683877 DOI: 10.1515/nipt-2024-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 10/18/2024] [Indexed: 01/03/2025]
Abstract
Obesity, by any standard, is a global health crisis. Both genetic and dietary contributions to the development and maintenance of obesity were integral factors of our experimental design. As mutations of the melanocortin 4 receptors (MC4R) are the leading monogenetic cause of obesity, MC4R haploinsufficient rats were fed a range of dietary fat (0-12 %) in a longitudinal design. Physiological and motivational assessments were performed using a locomotor task, a 5-choice sucrose preference task, an operant task with fixed and progressive ratios, as well as a distraction operant task. Dendritic spine morphology of medium spiny neurons (MSNs) of the nucleus accumbens (NAc), cells with ample D1 and D2 receptors, was also assessed. The percentage of lipid deposits in the liver of each rat was also analyzed using the Area Fraction Fractionator probe for stereological measurements. MC4R haploinsufficiency resulted in a phenotypic resemblance for adult-onset obesity that was exacerbated by the consumption of a high-fat diet. Results from the operant tasks indicate that motivational deficits due to MC4R haploinsufficiency were apparent prior to the onset of obesity and exacerbated by dietary fat consumption after obesity was well established. Moreover, MSN morphology shifted to longer spines with smaller head diameters for the MC4R+/- animals under the high-fat diet, suggesting a potential mechanism for the dysregulation of motivation to work for food. Increasing our knowledge of the neural circuitry/mechanisms responsible for the rewarding properties of food has significant implications for understanding energy balance and the development of obesity.
Collapse
Affiliation(s)
- Alex M. Steiner
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, University of South Carolina, Columbia, SC, USA
| | - Robert F. Roscoe
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, University of South Carolina, Columbia, SC, USA
| | - Rosemarie M. Booze
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, University of South Carolina, Columbia, SC, USA
| | - Charles F. Mactutus
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
12
|
Koceva A, Herman R, Janez A, Rakusa M, Jensterle M. Sex- and Gender-Related Differences in Obesity: From Pathophysiological Mechanisms to Clinical Implications. Int J Mol Sci 2024; 25:7342. [PMID: 39000449 PMCID: PMC11242171 DOI: 10.3390/ijms25137342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Obesity, primarily characterized by excessive fat accumulation, is a multifactorial chronic disease with an increasing global prevalence. Despite the well-documented epidemiology and significant advances in understanding its pathophysiology and clinical implications, the impact of sex is typically overlooked in obesity research. Worldwide, women have a higher likelihood to become obese compared to men. Although women are offered weight loss interventions more often and at earlier stages than men, they are more vulnerable to psychopathology. Men, on the other hand, are less likely to pursue weight loss intervention and are more susceptible to the metabolic implications of obesity. In this narrative review, we comprehensively explored sex- and gender-specific differences in the development of obesity, focusing on a variety of biological variables, such as body composition, fat distribution and energy partitioning, the impact of sex steroid hormones and gut microbiota diversity, chromosomal and genetic variables, and behavioural and sociocultural variables influencing obesity development in men and women. Sex differences in obesity-related comorbidities and varying effectiveness of different weight loss interventions are also extensively discussed.
Collapse
Affiliation(s)
- Andrijana Koceva
- Department of Endocrinology and Diabetology, University Medical Center Maribor, 2000 Maribor, Slovenia
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Rok Herman
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Andrej Janez
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Matej Rakusa
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Mojca Jensterle
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
13
|
Darcey VL, Guo J, Chi M, Chung ST, Courville AB, Gallagher I, Herscovitch P, Joseph PV, Howard R, LaNoire M, Milley L, Schick A, Stagliano M, Turner S, Urbanski N, Yang S, Zhai N, Zhou MS, Hall KD. Brain dopamine responses to ultra-processed milkshakes are highly variable and not significantly related to adiposity in humans. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.24.24309440. [PMID: 39108535 PMCID: PMC11302720 DOI: 10.1101/2024.06.24.24309440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
Ultra-processed foods high in fat and sugar may be addictive, in part, due to their purported ability to induce an exaggerated postingestive brain dopamine response akin to drugs of abuse. Using standard [11C]raclopride positron emission tomography (PET) displacement methods used to measure brain dopamine responses to addictive drugs, we measured postingestive striatal dopamine responses to an ultra-processed milkshake high in fat and sugar in 50 young, healthy adults over a wide body mass index range (BMI 20-45 kg/m2). Surprisingly, milkshake consumption did not result in significant postingestive dopamine response in the striatum (p=0.62) nor any striatal subregion (p>0.33) and the highly variable interindividual responses were not significantly related to adiposity (BMI: r=0.076, p=0.51; %body fat: r=0.16, p=0.28). Thus, postingestive striatal dopamine responses to an ultra-processed milkshake were likely substantially smaller than many addictive drugs and below the limits of detection using standard PET methods.
Collapse
Affiliation(s)
- Valerie L Darcey
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
- Center on Compulsive Behaviors, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Juen Guo
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Meible Chi
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stephanie T Chung
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Amber B Courville
- Human Energy and Body Weight Regulation Core, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Isabelle Gallagher
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter Herscovitch
- Positron Emission Tomography Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Paule V Joseph
- Section of Sensory Science and Metabolism, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, 20892, USA
- National Institute of Nursing Research, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, 20892, USA
- National Institute on Deafness and Other Communication Disorders, Smell and Taste Center, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, 20892, USA
| | - Rebecca Howard
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Melissa LaNoire
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lauren Milley
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alex Schick
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael Stagliano
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sara Turner
- Nutrition Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Nicholas Urbanski
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shanna Yang
- Nutrition Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Nan Zhai
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Megan S Zhou
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kevin D Hall
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
14
|
Zhou G, Lane G, Kahnt T, Zelano C. Structural Connectivity between Olfactory Tubercle and Ventrolateral Periaqueductal Gray Implicated in Human Feeding Behavior. J Neurosci 2024; 44:e2342232024. [PMID: 38755004 PMCID: PMC11209663 DOI: 10.1523/jneurosci.2342-23.2024] [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: 12/07/2023] [Revised: 02/16/2024] [Accepted: 03/19/2024] [Indexed: 05/18/2024] Open
Abstract
The olfactory tubercle (TUB), also called the tubular striatum, receives direct input from the olfactory bulb and, along with the nucleus accumbens, is one of the two principal components of the ventral striatum. As a key component of the reward system, the ventral striatum is involved in feeding behavior, but the vast majority of research on this structure has focused on the nucleus accumbens, leaving the TUB's role in feeding behavior understudied. Given the importance of olfaction in food seeking and consumption, olfactory input to the striatum should be an important contributor to motivated feeding behavior. Yet the TUB is vastly understudied in humans, with very little understanding of its structural organization and connectivity. In this study, we analyzed macrostructural variations between the TUB and the whole brain and explored the relationship between TUB structural pathways and feeding behavior, using body mass index (BMI) as a proxy in females and males. We identified a unique structural covariance between the TUB and the periaqueductal gray (PAG), which has recently been implicated in the suppression of feeding. We further show that the integrity of the white matter tract between the two regions is negatively correlated with BMI. Our findings highlight a potential role for the TUB-PAG pathway in the regulation of feeding behavior in humans.
Collapse
Affiliation(s)
- Guangyu Zhou
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Gregory Lane
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Thorsten Kahnt
- National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland 21224
| | - Christina Zelano
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| |
Collapse
|
15
|
Montalban E, Giralt A, Taing L, Nakamura Y, Pelosi A, Brown M, de Pins B, Valjent E, Martin M, Nairn AC, Greengard P, Flajolet M, Hervé D, Gambardella N, Roussarie JP, Girault JA. Operant Training for Highly Palatable Food Alters Translating Messenger RNA in Nucleus Accumbens D 2 Neurons and Reveals a Modulatory Role of Ncdn. Biol Psychiatry 2024; 95:926-937. [PMID: 37579933 PMCID: PMC11059129 DOI: 10.1016/j.biopsych.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND Highly palatable food triggers behavioral responses including strong motivation. These effects involve the reward system and dopamine neurons, which modulate neurons in the nucleus accumbens (NAc). The molecular mechanisms underlying the long-lasting effects of highly palatable food on feeding behavior are poorly understood. METHODS We studied the effects of 2-week operant conditioning of mice with standard or isocaloric highly palatable food. We investigated the behavioral responses and dendritic spine modifications in the NAc. We compared the translating messenger RNA in NAc neurons identified by the type of dopamine receptors they express, depending on the kind of food and training. We tested the consequences of invalidation of an abundant downregulated gene, Ncdn. RESULTS Operant conditioning for highly palatable food increased motivation for food even in well-fed mice. In wild-type mice, free choice between regular and highly palatable food increased weight compared with access to regular food only. Highly palatable food increased spine density in the NAc. In animals trained for highly palatable food, translating messenger RNAs were modified in NAc neurons expressing dopamine D2 receptors, mostly corresponding to striatal projection neurons, but not in neurons expressing D1 receptors. Knockout of Ncdn, an abundant downregulated gene, opposed the conditioning-induced changes in satiety-sensitive feeding behavior and apparent motivation for highly palatable food, suggesting that downregulation may be a compensatory mechanism. CONCLUSIONS Our results emphasize the importance of messenger RNA alterations in D2 striatal projection neurons in the NAc in the behavioral consequences of highly palatable food conditioning and suggest a modulatory contribution of Ncdn downregulation.
Collapse
Affiliation(s)
- Enrica Montalban
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France.
| | - Albert Giralt
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Lieng Taing
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Yuki Nakamura
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Assunta Pelosi
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Mallory Brown
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Benoit de Pins
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Emmanuel Valjent
- Institut de Génomique Fonctionnelle, University of Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Miquel Martin
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Reus, Spain; Instituto de investigaciones médicas Hospital del Mar, Barcelona, Spain
| | - Angus C Nairn
- Department of Psychiatry, Yale School of Medicine, Connecticut Mental Health Center, New Haven, Connecticut
| | - Paul Greengard
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Marc Flajolet
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Denis Hervé
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France
| | | | - Jean-Pierre Roussarie
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Jean-Antoine Girault
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France.
| |
Collapse
|
16
|
Senol E, Mohammad H. Current perspectives on brain circuits involved in food addiction-like behaviors. J Neural Transm (Vienna) 2024; 131:475-485. [PMID: 38216705 DOI: 10.1007/s00702-023-02732-4] [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: 08/28/2023] [Accepted: 12/17/2023] [Indexed: 01/14/2024]
Abstract
There is an emerging view that the increased availability of energy-dense foods in our society is contributing to excessive food consumption which could lead to food addiction-like behavior. Particularly, compulsive eating patterns are predominant in people suffering from eating disorders (binge-eating disorder, bulimia and anorexia nervosa) and obesity. Phenotypically, the behavioral pattern exhibits a close resemblance to individuals suffering from other forms of addiction (drug, sex, gambling). Growing body of evidence in neuroscience research is showing that excessive consumption of energy-dense foods alters the brain circuits implicated in reward, decision-making, control, habit formation, and emotions that are central to drug addiction. Here, we review the current understanding of the circuits of food addiction-like behaviors and highlight the future possibility of exploring those circuits to combat obesity and eating disorders.
Collapse
Affiliation(s)
- Esra Senol
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Hasan Mohammad
- Centre de Recherche en Biomédicine de Strasbourg (CRBS), L'Institut National de La Santé Et de La Recherche Médicale (Inserm) U1114, University of Strasbourg, Strasbourg, France.
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, 140306, India.
| |
Collapse
|
17
|
Kang A, Kwak MJ, Lee DJ, Lee JJ, Kim MK, Song M, Lee M, Yang J, Oh S, Kim Y. Dietary supplementation with probiotics promotes weight loss by reshaping the gut microbiome and energy metabolism in obese dogs. Microbiol Spectr 2024; 12:e0255223. [PMID: 38270436 PMCID: PMC10913549 DOI: 10.1128/spectrum.02552-23] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 12/08/2023] [Indexed: 01/26/2024] Open
Abstract
Obesity and overweight among companion animals are significant concerns, paralleling the issues observed in human populations. Recent research has highlighted the potential benefits of various probiotics in addressing weight-related changes, obesity, and associated pathologies. In this study, we delved into the beneficial probiotic mechanisms in high-fat-induced obese canines, revealing that Enterococcus faecium IDCC 2102 (IDCC 2102) and Bifidobacterium lactis IDCC 4301 (IDCC 4301) have the capacity to mitigate the increase in body weight and lipid accumulation in obese canines subjected to a high-fat diet and hyperlipidemic Caenorhabditis elegans (C. elegans) strain VS29. Both IDCC 2102 and IDCC 4301 demonstrated the ability to reduce systemic inflammation and hormonal disruptions induced by obesity. Notably, these probiotics induced modifications in the microbiota by promoting lactic acid bacteria, including Lactobacillaceae, Ruminococcaceae, and S24-7, with concomitant activation of pyruvate metabolism. IDCC 4301, through the generation of bacterial short-chain fatty acids and carboxylic acids, facilitated glycolysis and contributed to ATP synthesis. Meanwhile, IDCC 2102 produced bacterial metabolites such as acetic acid and butyric acid, exhibiting a particular ability to stimulate dopamine synthesis in a canine model. This stimulation led to the restoration of eating behavior and improvements in glucose and insulin tolerance. In summary, we propose novel probiotics for the treatment of obese animals based on the modifications induced by IDCC 2102 and IDCC 4301. These probiotics enhanced systemic energy utilization in response to high caloric intake, thereby preventing lipid accumulation and restoring stability to the fecal microbiota. Consequently, this intervention resulted in a reduction in systemic inflammation caused by the high-fat diet.IMPORTANCEProbiotic supplementation affected commensal bacterial proliferation, and administering probiotics increased glycolysis and activated pyruvate metabolism in the body, which is related to propanate metabolism as a result of pyruvate metabolism activation boosting bacterial fatty acid production via dopamine and carboxylic acid specialized pathways, hence contributing to increased ATP synthesis and energy metabolism activity.
Collapse
Affiliation(s)
- Anna Kang
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
| | - Min-Jin Kwak
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
| | - Daniel Junpyo Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
| | - Jeong Jae Lee
- Institute of Agricultural Science and Technology, Kyungpook National University, Daegu, South Korea
| | - Min Kyu Kim
- Division of Animal and Dairy Science, Chungnam National University, Daejeon, South Korea
| | - Minho Song
- Division of Animal and Dairy Science, Chungnam National University, Daejeon, South Korea
| | - Minjee Lee
- Ildong Bioscience, Pyeongtaek-si, Gyeonggi-do, South Korea
| | - Jungwoo Yang
- Ildong Bioscience, Pyeongtaek-si, Gyeonggi-do, South Korea
| | - Sangnam Oh
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, South Korea
| | - Younghoon Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
| |
Collapse
|
18
|
Zhang Y, Tsai TH, Ezrokhi M, Stoelzel C, Cincotta AH. Tyrosine Hydroxylase Knockdown at the Hypothalamic Supramammillary Nucleus Area Induces Obesity and Glucose Intolerance. Neuroendocrinology 2023; 114:483-510. [PMID: 38128505 PMCID: PMC11098027 DOI: 10.1159/000535944] [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: 09/23/2022] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
INTRODUCTION The supramammillary nucleus (SuMN) exerts influences on a wide range of brain functions including feeding and feeding-independent fuel metabolism. However, which specific neuronal type(s) within the SuMN manifest this influence has not been delineated. This study investigated the effect of SuMN tyrosine hydroxylase (TH) (rate-limiting enzyme in dopamine synthesis) knockdown (THx) on peripheral fuel metabolism. METHODS SuMN-THx was accomplished using a virus-mediated shRNA to locally knockdown TH gene expression at the SuMN. The impact of SuMN-THx was examined over 35-72 days in rats least prone to developing metabolic syndrome (MS) - female Sprague-Dawley rats resistant to the obesogenic effect of high fat diet (HFDr) and fed regular chow (RC) - upon body weight/fat, feeding, glucose tolerance, and insulin sensitivity. The influence of HFD, gender, and long-term response of SuMN-THx was subsequently investigated in female HFDr rats fed HFD, male HFDr rats fed RC, and female HFD-sensitive rats fed RC over 1 year, respectively. RESULTS SuMN-THx induced obesity and glucose intolerance, elevated plasma leptin and triglycerides, increased hepatic mRNA levels of gluconeogenic, lipogenic, and pro-inflammatory genes, reduced white adipose fatty acid oxidation rate, and altered plasma corticosterone level and hepatic circadian gene expression. Moreover, SuMN-THx increased feeding during the natural resting/fasting period and altered ghrelin feeding response suggesting ghrelin resistance. This MS-inducing effect was enhanced by HFD feeding, similarly observed in male rats and persisted over 1 year. DISCUSSION/CONCLUSION SuMN-THx induced long-term, gender-nonspecific, multiple pathophysiological changes leading to MS suggesting SuMN dopaminergic circuits communicating with other brain metabolism and behavior control centers modulate peripheral fuel metabolism.
Collapse
|
19
|
Parekh P, Begley P, Jessop M, Aplin M, Missir E, McMeekin H, Raczek G, Singh N, Dizdarevic S. Association between body mass index (BMI) and [ 123I]Ioflupane (DaTSCAN) availabilities in patients with parkinsonism using single-photon emission computed tomography-computed tomography (SPECT-CT). Eur J Hybrid Imaging 2023; 7:21. [PMID: 37981626 PMCID: PMC10657921 DOI: 10.1186/s41824-023-00181-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 09/14/2023] [Indexed: 11/21/2023] Open
Abstract
AIM [123I]Ioflupane (DaTSCAN) has a high binding affinity to the dopamine (DA) transporter (DaT) and tenfold less affinity to serotonin (5-HT) transporter (SERT). Both neurotransmitters are considered to contribute to body weight regulation. This study assesses the association between body mass index (BMI) and DaTSCAN availability in brain. METHOD Scans from 74 consecutive patients who had undergone DaTSCAN single-photon emission computed tomography-computed tomography (SPECT-CT) were used to obtain semi- and absolute quantitative data in several volumes of interest (VOIs). Relative semi-quantitative specific binding ratios (SBRs) from Chang attenuated SPECT were obtained from GE DaTQUANT. Absolute normalised concentration (NC) was calculated from attenuation/scatter corrected SPECT-CT images, using an adapted version of the EARL Ltd (European Association of Nuclear Medicine (EANM) Research 4 Life) template. Scans were subdivided into either degenerative parkinsonism (abnormal = 49), borderline (n = 14) or scan without evidence of dopaminergic deficit (SWEDD = 11) using visual assessment and SBR values by two nuclear medicine consultants. RESULTS SBRs did not correlate with BMI. However, NC values correlated negatively in the entire cohort, with the strongest correlation in the frontal (r = - 0.649. p = 0.000), occipital (r = - 0.555, p = 0.000) regions and pons (r = - 0.555, p = 0.000). In the abnormal (n = 49) and SWEDD group (n = 11), NC of the frontal region was the most correlated with BMI (r = - 0.570, p = 0.000; r = - 0.813, p = 0.002, respectively). In the borderline group (n = 14), the left posterior putamen displayed the strongest correlation (r = - 0.765, p = 0.001). CONCLUSION Absolute NC values demonstrate a strong inverse correlation with BMI, strongest in the extrastriatal regions. Due to the predominately non-overlapping distribution of DaT and SERT, this study suggests greater involvement of SERT in obesity with possible interplay with DA transmission.
Collapse
Affiliation(s)
- Puja Parekh
- Brighton and Sussex Medical School, Brighton, England
| | - Patrick Begley
- Nuclear Medicine Department, Royal Sussex County Hospital, University Hospitals Sussex NHS Foundation Trust, Brighton, England
| | - Maryam Jessop
- Nuclear Medicine Department, Royal Sussex County Hospital, University Hospitals Sussex NHS Foundation Trust, Brighton, England
| | - Mark Aplin
- Nuclear Medicine Department, Royal Sussex County Hospital, University Hospitals Sussex NHS Foundation Trust, Brighton, England
| | - Elena Missir
- Brighton and Sussex Medical School, Brighton, England
| | | | - Gosia Raczek
- Brighton and Sussex Medical School, Brighton, England
| | - Nitasha Singh
- Nuclear Medicine Department, Royal Sussex County Hospital, University Hospitals Sussex NHS Foundation Trust, Brighton, England
| | - Sabina Dizdarevic
- Clinical Imaging Science Centre, Neuroscience and Medicine, Brighton and Sussex Medical School, Brighton, England.
- Brighton and Sussex Medical School, Brighton, England.
- Nuclear Medicine Department, Royal Sussex County Hospital, University Hospitals Sussex NHS Foundation Trust, Brighton, England.
| |
Collapse
|
20
|
Slomp M, de Lange IGS, Mul JD, Schrantee A, la Fleur SE. Investigating Habenula Functional Connectivity and Reward-Related Activity in Obesity Using Human Connectome Project Data. Brain Connect 2023; 13:541-552. [PMID: 37578129 DOI: 10.1089/brain.2023.0034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023] Open
Abstract
Introduction: The habenula, a brain region involved in aversion, might negatively modulate caloric intake. Functional magnetic resonance imaging (fMRI) studies reported associations between weight loss and habenula functional connectivity. However, whether habenula resting-state functional connectivity (rsFC) and reward-related activity are altered in obesity is yet unknown. Methods: Using data from the Human Connectome Project, we included 300 subjects with various body mass indexes (BMIs) and a healthy long-term blood glucose (hemoglobin A1c [HbA1c]). In addition, we investigated a potential BMI × HbA1c interaction in a separate cohort including subjects with prediabetes (n = 72). Habenula rsFC was assessed using a region of interest (ROI)-to-ROI analysis. Furthermore, a separate analysis using gambling task fMRI data focused on reward-related habenula activity. Results: We did not find an association between BMI and habenula rsFC for any of the ROIs. For the exploratory analysis of the BMI × HbA1c effect, a significant interaction effect was found for the habenula-ventral tegmental area (VTA) connection, but this did not survive multiple comparisons correction. Monetary punishment compared with reward activated the bilateral habenula in the BMI sample, but this activity was not associated with BMI. Discussion: In conclusion, we did not find evidence for an association between BMI and habenula rsFC or reward-related activity. However, there might be an interaction between BMI and HbA1c for the habenula-VTA rsFC, suggestive of a role of the habenula in glucose regulation. Future studies should focus on metabolic parameters in their experimental design to confirm our findings and explore the precise role of the habenula in metabolism.
Collapse
Affiliation(s)
- Margo Slomp
- Endocrine Laboratory, Department of Laboratory Medicine, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology & Metabolism, Amsterdam, The Netherlands
- Metabolism and Reward Group, Royal Netherlands Academy of Arts and Sciences, Netherlands Institute of Neuroscience, Amsterdam, The Netherlands
| | - Ilke G S de Lange
- Endocrine Laboratory, Department of Laboratory Medicine, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
- Metabolism and Reward Group, Royal Netherlands Academy of Arts and Sciences, Netherlands Institute of Neuroscience, Amsterdam, The Netherlands
| | - Joram D Mul
- Amsterdam Neuroscience, Amsterdam, The Netherlands
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - Anouk Schrantee
- Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Susanne E la Fleur
- Endocrine Laboratory, Department of Laboratory Medicine, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology & Metabolism, Amsterdam, The Netherlands
- Metabolism and Reward Group, Royal Netherlands Academy of Arts and Sciences, Netherlands Institute of Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Carnell S, Thapaliya G, Jansen E, Chen L. Biobehavioral susceptibility for obesity in childhood: Behavioral, genetic and neuroimaging studies of appetite. Physiol Behav 2023; 271:114313. [PMID: 37544571 PMCID: PMC10591980 DOI: 10.1016/j.physbeh.2023.114313] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 06/06/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023]
Abstract
Modern food environments are conducive to overeating and weight gain, but not everyone develops obesity. One reason for this may be that individuals differ in appetitive characteristics, or traits, that manifest early in life and go on to influence their behavioral susceptibility to gain and maintain excess weight. Classic studies showing that eating behavior in children can be measured by behavioral paradigms such as tests of caloric compensation and eating in the absence of hunger inspired the development and validation of psychometric instruments to assess appetitive characteristics in children and infants. A large body of evidence now suggests that food approach traits increase obesity risk, while food avoidant traits, such as satiety responsiveness, decrease obesity risk. Twin studies and genetic association studies have demonstrated that appetitive characteristics are heritable, consistent with a biological etiology. However, family environment factors are also influential, with mounting evidence suggesting that genetic and environmental risk factors interact and correlate with consequences for child eating behavior and weight. Further, neuroimaging studies are revealing that individual differences in responses to visual food cues, as well as to small tastes and larger amounts of food, across a number of brain regions involved in reward/motivation, cognitive control and other functions, may contribute to individual variation in appetitive behavior. Growing evidence also suggests that variation on psychometric measures of appetite is associated with regional differences in brain structure, and differential patterns of resting state functional connectivity. Large prospective studies beginning in infancy promise to enrich our understanding of neural and other biological underpinnings of appetite and obesity development in early life, and how the interplay between genetic and environmental factors affects appetitive systems. The biobehavioral susceptibility model of obesity development and maintenance outlined in this narrative review has implications for prevention and treatment of obesity in childhood.
Collapse
Affiliation(s)
- Susan Carnell
- Division of Child and Adolescent Psychiatry, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore MD, USA.
| | - Gita Thapaliya
- Division of Child and Adolescent Psychiatry, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore MD, USA
| | - Elena Jansen
- Division of Child and Adolescent Psychiatry, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore MD, USA
| | - Liuyi Chen
- Division of Psychiatric Neuroimaging, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore MD, USA
| |
Collapse
|
22
|
Byrne ME, Tanofsky-Kraff M, Liuzzi L, Holroyd T, Parker MN, Bloomer BF, Nugent A, Brady SM, Yang SB, Turner SA, Pine DS, Yanovski JA. Neural underpinnings of threat bias in relation to loss-of-control eating behaviors among adolescent girls with high weight. Front Psychiatry 2023; 14:1276300. [PMID: 37965354 PMCID: PMC10642175 DOI: 10.3389/fpsyt.2023.1276300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/22/2023] [Indexed: 11/16/2023] Open
Abstract
Introduction Loss-of-control (LOC) eating, a key feature of binge-eating disorder, may relate attentional bias (AB) to highly salient interpersonal stimuli. The current pilot study used magnetoencephalography (MEG) to explore neural features of AB to socially threatening cues in adolescent girls with and without LOC-eating. Methods Girls (12-17 years old) with overweight or obesity (BMI >85th percentile) completed an AB measure on an affective dot-probe AB task during MEG and evoked neural responses to angry or happy (vs. neutral) face cues were captured. A laboratory test meal paradigm measured energy intake and macronutrient consumption patterns. Results Girls (N = 34; Mage = 15.5 ± 1.5 years; BMI-z = 1.7 ± 0.4) showed a blunted evoked response to the presentation of angry face compared with neutral face cues in the left dorsolateral prefrontal cortex, a neural region implicated in executive control and regulation processes, during attention deployment (p < 0.01). Compared with those without LOC-eating (N = 21), girls with LOC-eating (N = 13) demonstrated a stronger evoked response to angry faces in the visual cortex during attention deployment (p < 0.001). Visual and cognitive control ROIs had trends suggesting interaction with test meal intake patterns among girls with LOC-eating (ps = 0.01). Discussion These findings suggest that girls with overweight or obesity may fail to adaptively engage neural regions implicated in higher-order executive processes. This difficulty may relate to disinhibited eating patterns that could lead to excess weight gain.
Collapse
Affiliation(s)
- Meghan E. Byrne
- Section on Development and Affective Neuroscience, Emotion and Development Branch, National Institute of Mental Health (NIMH), Bethesda, MD, United States
- Division of Intramural Research, National Institutes of Health (NIH), Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| | - Marian Tanofsky-Kraff
- Division of Intramural Research, National Institutes of Health (NIH), Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
- Department of Medical and Clinical Psychology, USUHS, Bethesda, MD, United States
- Military Cardiovascular Outcomes Research (MiCOR) Program, Department of Medicine, Uniformed Services University of the Health Sciences (USUHS), Bethesda, MD, United States
| | - Lucrezia Liuzzi
- Section on Development and Affective Neuroscience, Emotion and Development Branch, National Institute of Mental Health (NIMH), Bethesda, MD, United States
| | - Tom Holroyd
- MEG Core Facility, National Institute of Mental Health (NIMH), Bethesda, MD, United States
| | - Megan N. Parker
- Division of Intramural Research, National Institutes of Health (NIH), Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
- Department of Medical and Clinical Psychology, USUHS, Bethesda, MD, United States
| | - Bess F. Bloomer
- Division of Intramural Research, National Institutes of Health (NIH), Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| | - Allison Nugent
- MEG Core Facility, National Institute of Mental Health (NIMH), Bethesda, MD, United States
| | - Sheila M. Brady
- Division of Intramural Research, National Institutes of Health (NIH), Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| | - Shanna B. Yang
- Nutrition Department, Clinical Center, NIH, Bethesda, MD, United States
| | - Sara A. Turner
- Nutrition Department, Clinical Center, NIH, Bethesda, MD, United States
| | - Daniel S. Pine
- Section on Development and Affective Neuroscience, Emotion and Development Branch, National Institute of Mental Health (NIMH), Bethesda, MD, United States
| | - Jack A. Yanovski
- Division of Intramural Research, National Institutes of Health (NIH), Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| |
Collapse
|
23
|
Yang R, Zhang Y, Deng Y, Yang Y, Zhong W, Zhu L. 2-Ethylhexyl Diphenyl Phosphate Causes Obesity in Zebrafish by Stimulating Overeating via Inhibition of Dopamine Receptor D2. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:14162-14172. [PMID: 37704188 DOI: 10.1021/acs.est.3c04070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Obesity is a popular public health problem worldwide and is mainly caused by overeating, but little is known about the impacts of synthetic chemicals on obesity. Herein, we evaluated the obesogenic effect caused by 2-ethylhexyl diphenyl phosphate (EHDPHP) on zebrafish. Adult zebrafish were exposed to 5, 35, and 245 μg/L of EHDPHP for 21 days. Results showed that EHDPHP exposure significantly promoted the feeding behavior of zebrafish, as evidenced by shorter reaction time, increased average food intake, feeding rate, and intake frequency (p < 0.05). Transcriptomic, real-time quantitative PCR, and neurotransmitter analyses revealed that the dopamine (DA) receptor D2 (DRD2) was inhibited, which interfered with the DA neural reward regulation system, thus stimulating food addiction to zebrafish. This was further verified by the restored DRD2 after 7 days of Halo (a DRD2 agonist) treatment. A strong interaction between EHDPHP and DRD2 was identified via molecular docking. As a consequence of the abnormal feeding behavior, the exposed fish exhibited significant obesity evidenced by increased body weight, body mass index, plasma total cholesterol, triglyceride, and body fat content. Additionally, the pathways linked to Parkinson's disease, alcoholism, and cocaine addiction were also disrupted, implying that EHDPHP might cause other neurological disorders via the disrupted DA system.
Collapse
Affiliation(s)
- Rongyan Yang
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Yuan Zhang
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Yun Deng
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Yi Yang
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Wenjue Zhong
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Lingyan Zhu
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| |
Collapse
|
24
|
Passeri A, Municchi D, Cavalieri G, Babicola L, Ventura R, Di Segni M. Linking drug and food addiction: an overview of the shared neural circuits and behavioral phenotype. Front Behav Neurosci 2023; 17:1240748. [PMID: 37767338 PMCID: PMC10520727 DOI: 10.3389/fnbeh.2023.1240748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Despite a lack of agreement on its definition and inclusion as a specific diagnosable disturbance, the food addiction construct is supported by several neurobiological and behavioral clinical and preclinical findings. Recognizing food addiction is critical to understanding how and why it manifests. In this overview, we focused on those as follows: 1. the hyperpalatable food effects in food addiction development; 2. specific brain regions involved in both food and drug addiction; and 3. animal models highlighting commonalities between substance use disorders and food addiction. Although results collected through animal studies emerged from protocols differing in several ways, they clearly highlight commonalities in behavioral manifestations and neurobiological alterations between substance use disorders and food addiction characteristics. To develop improved food addiction models, this heterogeneity should be acknowledged and embraced so that research can systematically investigate the role of specific variables in the development of the different behavioral features of addiction-like behavior in preclinical models.
Collapse
Affiliation(s)
- Alice Passeri
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
| | - Diana Municchi
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
| | - Giulia Cavalieri
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
| | | | - Rossella Ventura
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
- IRCCS San Raffaele, Rome, Italy
| | - Matteo Di Segni
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
| |
Collapse
|
25
|
Montalban E, Walle R, Castel J, Ansoult A, Hassouna R, Foppen E, Fang X, Hutelin Z, Mickus S, Perszyk E, Petitbon A, Berthelet J, Rodrigues-Lima F, Cebrian-Serrano A, Gangarossa G, Martin C, Trifilieff P, Bosch-Bouju C, Small DM, Luquet S. The Addiction-Susceptibility TaqIA/Ankk1 Controls Reward and Metabolism Through D 2 Receptor-Expressing Neurons. Biol Psychiatry 2023; 94:424-436. [PMID: 36805080 DOI: 10.1016/j.biopsych.2023.02.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/21/2023] [Accepted: 02/09/2023] [Indexed: 02/18/2023]
Abstract
BACKGROUND A large body of evidence highlights the importance of genetic variants in the development of psychiatric and metabolic conditions. Among these, the TaqIA polymorphism is one of the most commonly studied in psychiatry. TaqIA is located in the gene that codes for the ankyrin repeat and kinase domain containing 1 kinase (Ankk1) near the dopamine D2 receptor (D2R) gene. Homozygous expression of the A1 allele correlates with a 30% to 40% reduction of striatal D2R, a typical feature of addiction, overeating, and other psychiatric pathologies. The mechanisms by which the variant influences dopamine signaling and behavior are unknown. METHODS Here, we used transgenic and viral-mediated strategies to reveal the role of Ankk1 in the regulation of activity and functions of the striatum. RESULTS We found that Ankk1 is preferentially enriched in striatal D2R-expressing neurons and that Ankk1 loss of function in the dorsal and ventral striatum leads to alteration in learning, impulsivity, and flexibility resembling endophenotypes described in A1 carriers. We also observed an unsuspected role of Ankk1 in striatal D2R-expressing neurons of the ventral striatum in the regulation of energy homeostasis and documented differential nutrient partitioning in humans with or without the A1 allele. CONCLUSIONS Overall, our data demonstrate that the Ankk1 gene is necessary for the integrity of striatal functions and reveal a new role for Ankk1 in the regulation of body metabolism.
Collapse
Affiliation(s)
- Enrica Montalban
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France.
| | - Roman Walle
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - Julien Castel
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Anthony Ansoult
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Rim Hassouna
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Ewout Foppen
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Xi Fang
- Modern Diet and Physiology Research Center, New Haven, Connecticut; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Zach Hutelin
- Modern Diet and Physiology Research Center, New Haven, Connecticut; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Sophie Mickus
- Modern Diet and Physiology Research Center, New Haven, Connecticut; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Emily Perszyk
- Modern Diet and Physiology Research Center, New Haven, Connecticut; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Anna Petitbon
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - Jérémy Berthelet
- Université Paris Cité, CNRS, Unité Epigenetique et Destin Cellulaire, Paris, France
| | | | - Alberto Cebrian-Serrano
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany
| | - Giuseppe Gangarossa
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Claire Martin
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Pierre Trifilieff
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | | | - Dana M Small
- Modern Diet and Physiology Research Center, New Haven, Connecticut; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Serge Luquet
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France; Modern Diet and Physiology Research Center, New Haven, Connecticut.
| |
Collapse
|
26
|
Kang J, Park M, Oh CM, Kim T. High-fat diet-induced dopaminergic dysregulation induces REM sleep fragmentation and ADHD-like behaviors. Psychiatry Res 2023; 327:115412. [PMID: 37607442 DOI: 10.1016/j.psychres.2023.115412] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 07/27/2023] [Accepted: 08/10/2023] [Indexed: 08/24/2023]
Abstract
Consumption of a high-fat diet (HFD) has been associated with reduced wakefulness and various behavioral deficits, including anxiety, depression, and anhedonia. The dopaminergic system, which plays a crucial role in sleep and ADHD, is known to be vulnerable to chronic HFD. However, the association between HFD-induced behavioral and molecular changes remains unclear. Therefore, we investigated the effects of a HFD on the dopaminergic system and its association with behavioral deficits in male mice. The mice were divided into normal diet and HFD groups and were analyzed for sleep patterns, behavior tests, and transcription levels of dopamine-related genes in the brain. The HFD group showed decreased wakefulness, increased REM sleep with fragmented patterns, decreased time spent in the center zone of the open field test, shorter immobile time in the tail suspension test, impaired visuospatial memory, and reduced sucrose preference. Additionally, the HFD group had decreased mRNA levels of D1R, COMT, and DAT in the nucleus accumbens, which negatively correlated with REM sleep proportion and REM sleep bout count. The results suggest that HFD-induced behavioral deficits were resemblance to ADHD-like behavioral phenotypes and disturbs REM sleep by dysregulating the dopaminergic system.
Collapse
Affiliation(s)
- Jiseung Kang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Mincheol Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| | - Tae Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| |
Collapse
|
27
|
Barbosa DAN, Gattas S, Salgado JS, Kuijper FM, Wang AR, Huang Y, Kakusa B, Leuze C, Luczak A, Rapp P, Malenka RC, Hermes D, Miller KJ, Heifets BD, Bohon C, McNab JA, Halpern CH. An orexigenic subnetwork within the human hippocampus. Nature 2023; 621:381-388. [PMID: 37648849 PMCID: PMC10499606 DOI: 10.1038/s41586-023-06459-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 07/20/2023] [Indexed: 09/01/2023]
Abstract
Only recently have more specific circuit-probing techniques become available to inform previous reports implicating the rodent hippocampus in orexigenic appetitive processing1-4. This function has been reported to be mediated at least in part by lateral hypothalamic inputs, including those involving orexigenic lateral hypothalamic neuropeptides, such as melanin-concentrating hormone5,6. This circuit, however, remains elusive in humans. Here we combine tractography, intracranial electrophysiology, cortico-subcortical evoked potentials, and brain-clearing 3D histology to identify an orexigenic circuit involving the lateral hypothalamus and converging in a hippocampal subregion. We found that low-frequency power is modulated by sweet-fat food cues, and this modulation was specific to the dorsolateral hippocampus. Structural and functional analyses of this circuit in a human cohort exhibiting dysregulated eating behaviour revealed connectivity that was inversely related to body mass index. Collectively, this multimodal approach describes an orexigenic subnetwork within the human hippocampus implicated in obesity and related eating disorders.
Collapse
Affiliation(s)
- Daniel A N Barbosa
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sandra Gattas
- Department of Electrical Engineering and Computer Science, University of California, Irvine, Irvine, CA, USA
| | - Juliana S Salgado
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Fiene Marie Kuijper
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
- Université Paris Cité, Paris, France
- Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Allan R Wang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuhao Huang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Bina Kakusa
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Christoph Leuze
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Artur Luczak
- Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Paul Rapp
- Department of Military & Emergency Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Robert C Malenka
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Dora Hermes
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Kai J Miller
- Department of Neurosurgery, Mayo Clinic, Rochester, MN, USA
| | - Boris D Heifets
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Cara Bohon
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Jennifer A McNab
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Casey H Halpern
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Surgery, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA.
| |
Collapse
|
28
|
Arrue A, Olivas O, Erkoreka L, Alvarez FJ, Arnaiz A, Varela N, Bilbao A, Rodríguez JJ, Moreno-Calle MT, Gordo E, Marín E, Garcia-Cano J, Saez E, Gonzalez-Torres MÁ, Zumárraga M, Basterreche N. Multilocus Genetic Profile Reflecting Low Dopaminergic Signaling Is Directly Associated with Obesity and Cardiometabolic Disorders Due to Antipsychotic Treatment. Pharmaceutics 2023; 15:2134. [PMID: 37631349 PMCID: PMC10459305 DOI: 10.3390/pharmaceutics15082134] [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: 07/07/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Treatment with second-generation antipsychotics (SGAs) can cause obesity and other cardiometabolic disorders linked to D2 receptor (DRD2) and to genotypes affecting dopaminergic (DA) activity, within reward circuits. We explored the relationship of cardiometabolic alterations with single genetic polymorphisms DRD2 rs1799732 (NG_008841.1:g.4750dup -> C), DRD2 rs6277 (NG_008841.1:g.67543C>T), COMT rs4680 (NG_011526.1:g.27009G>A), and VNTR in both DRD4 NC_000011.10 (637269-640706) and DAT1 NC_000005.10 (1392794-1445440), as well as with a multilocus genetic profile score (MLGP). A total of 285 psychiatric patients treated with SGAs for at least three months were selected. Cardiometabolic parameters were classified according to ATP-III and WHO criteria. Blood samples were taken for routinely biochemical assays and PCR genotyping. Obesity (BMI, waist (W)), high diastolic blood pressure (DBP), and hypertriglyceridemia (HTG) were present in those genetic variants related to low dopaminergic activity: InsIns genotype in rs1799732 (BMI: OR: 2.91 [1.42-5.94]), DRD4-VNTR-L allele (W: OR: 1.73 [1.04-2.87]) and 9R9R variant in DAT1-VNTR (W: OR: 2.73 [1.16-6.40]; high DBP: OR: 3.33 [1.54-7.31]; HTG: OR: 4.38 [1.85-10.36]). A low MLGP score indicated a higher risk of suffering cardiometabolic disorders (BMI: OR: 1.23 [1.05-1.45]; W: OR: 1.18 [1.03-1.34]; high DBP: OR: 1.22 [1.06-1.41]; HTG: OR: 1.20 [1.04-1.39]). The MLGP score was more sensitive for detecting the risk of suffering these alterations. Low dopaminergic system function would contribute to increased obesity, BDP, and HTG following long-term SGA treatment.
Collapse
Affiliation(s)
- Aurora Arrue
- Mental Health Network Group, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (O.O.); (L.E.)
- Neurochemical Research Unit, Bizkaia Mental Health Network, Osakidetza Basque Health Service, 48903 Barakaldo, Spain
| | - Olga Olivas
- Mental Health Network Group, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (O.O.); (L.E.)
- Bizkaia Mental Health Network, Zaldibar Hospital, Osakidetza Basque Health Service, 48250 Zaldibar, Spain
| | - Leire Erkoreka
- Mental Health Network Group, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (O.O.); (L.E.)
- Department of Psychiatry, Galdakao-Usánsolo University Hospital, Osakidetza Basque Health Service, 48960 Galdakao, Spain
- Department of Neurosciences, Faculty of Medicine and Dentistry, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
| | - Francisco Jose Alvarez
- Research Unit, Cruces University Hospital, Osakidetza Basque Health Service, 48903 Barakaldo, Spain
| | - Ainara Arnaiz
- Mental Health Network Group, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (O.O.); (L.E.)
- Erandio Mental Health Center, Bizkaia Mental Health Network, Osakidetza Basque Health Service, 48950 Erandio, Spain
| | - Noemi Varela
- Mental Health Network Group, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (O.O.); (L.E.)
- Uribe Mental Health Center, Bizkaia Mental Health Network, Osakidetza Basque Health Service, 48990 Getxo, Spain
| | - Ainhoa Bilbao
- Animal Research Facility, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Jose-Julio Rodríguez
- Department of Neurosciences, Faculty of Medicine and Dentistry, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
- Functional Neuroanatomy, BioCruces Bizkaia Health Research Institute, Ikerbasque Basque Foundation for Science, 48903 Barakaldo, Spain
| | - María Teresa Moreno-Calle
- Mental Health Network Group, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (O.O.); (L.E.)
- Department of Psychiatry, Galdakao-Usánsolo University Hospital, Osakidetza Basque Health Service, 48960 Galdakao, Spain
| | - Estibaliz Gordo
- Bizkaia Mental Health Network, Zamudio Hospital, Osakidetza Basque Health Service, 48170 Zamudio, Spain
| | - Elena Marín
- Mental Health Network Group, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (O.O.); (L.E.)
- Bizkaia Mental Health Network, Bermeo Hospital, Osakidetza Basque Health Service, 48370 Bermeo, Spain
| | - Javier Garcia-Cano
- Alternatives to Hospitalization in Bilbao, Bizkaia Mental Health Network, Osakidetza Basque Health Service, 48903 Bilbao, Spain
| | - Estela Saez
- Mental Health Network Group, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (O.O.); (L.E.)
- Department of Psychiatry, Galdakao-Usánsolo University Hospital, Osakidetza Basque Health Service, 48960 Galdakao, Spain
| | - Miguel Ángel Gonzalez-Torres
- Department of Neurosciences, Faculty of Medicine and Dentistry, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
- Integrative Research Group in Mental Health, BioCruces Bizkaia Health Research Institute, 48013 Bilbao, Spain
- Department of Psychiatry, Basurto University Hospital, Osakidetza Basque Health Service, 48013 Bilbao, Spain
| | - Mercedes Zumárraga
- Mental Health Network Group, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (O.O.); (L.E.)
- Neurochemical Research Unit, Bizkaia Mental Health Network, Osakidetza Basque Health Service, 48903 Barakaldo, Spain
| | - Nieves Basterreche
- Functional Neuroanatomy, BioCruces Bizkaia Health Research Institute, Ikerbasque Basque Foundation for Science, 48903 Barakaldo, Spain
- Integrative Research Group in Mental Health, BioCruces Bizkaia Health Research Institute, 48013 Bilbao, Spain
| |
Collapse
|
29
|
Dunn JP, Lamichhane B, Smith GI, Garner A, Wallendorf M, Hershey T, Klein S. Dorsal striatal response to taste is modified by obesity and insulin resistance. Obesity (Silver Spring) 2023; 31:2065-2075. [PMID: 37475685 PMCID: PMC10767984 DOI: 10.1002/oby.23799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 07/22/2023]
Abstract
OBJECTIVE In preclinical models, insulin resistance in the dorsal striatum (DS) contributes to overeating. Although human studies support the concept of central insulin resistance, they have not investigated its effect on consummatory reward-induced brain activity. METHODS Taste-induced activation was assessed in the caudate and putamen of the DS with blood oxygen level-dependent (BOLD) functional magnetic resonance imaging. Three phenotypically distinct groups were studied: metabolically healthy lean, metabolically healthy obesity, and metabolically unhealthy obesity (MUO; presumed to have central insulin resistance). Participants with MUO also completed a weight loss intervention followed by a second functional magnetic resonance imaging session. RESULTS The three groups were significantly different at baseline consistent with the design. The metabolically healthy lean group had a primarily positive BOLD response, the MUO group had a primarily negative BOLD response, and the metabolically healthy obesity group had a response in between the two other groups. Food craving was predicted by taste-induced activation. After weight loss in the MUO group, taste-induced activation increased in the DS. CONCLUSIONS These data support the hypothesis that insulin resistance and obesity contribute to aberrant responses to taste in the DS, which is only partially attenuated by weight loss. Aberrant responses to food exposure may be a barrier to weight loss.
Collapse
Affiliation(s)
- Julia P. Dunn
- VA St. Louis Health Care System, St. Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Bidhan Lamichhane
- Department of Neurosurgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Gordon I. Smith
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Amy Garner
- VA St. Louis Health Care System, St. Louis, Missouri, USA
| | - Michael Wallendorf
- Division of Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Tamara Hershey
- Departments of Psychiatry and Radiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Samuel Klein
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
30
|
Becetti I, Bwenyi EL, de Araujo IE, Ard J, Cryan JF, Farooqi IS, Ferrario CR, Gluck ME, Holsen LM, Kenny PJ, Lawson EA, Lowell BB, Schur EA, Stanley TL, Tavakkoli A, Grinspoon SK, Singhal V. The Neurobiology of Eating Behavior in Obesity: Mechanisms and Therapeutic Targets: A Report from the 23rd Annual Harvard Nutrition Obesity Symposium. Am J Clin Nutr 2023; 118:314-328. [PMID: 37149092 PMCID: PMC10375463 DOI: 10.1016/j.ajcnut.2023.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/03/2023] [Accepted: 05/01/2023] [Indexed: 05/08/2023] Open
Abstract
Obesity is increasing at an alarming rate. The effectiveness of currently available strategies for the treatment of obesity (including pharmacologic, surgical, and behavioral interventions) is limited. Understanding the neurobiology of appetite and the important drivers of energy intake (EI) can lead to the development of more effective strategies for the prevention and treatment of obesity. Appetite regulation is complex and is influenced by genetic, social, and environmental factors. It is intricately regulated by a complex interplay of endocrine, gastrointestinal, and neural systems. Hormonal and neural signals generated in response to the energy state of the organism and the quality of food eaten are communicated by paracrine, endocrine, and gastrointestinal signals to the nervous system. The central nervous system integrates homeostatic and hedonic signals to regulate appetite. Although there has been an enormous amount of research over many decades regarding the regulation of EI and body weight, research is only now yielding potentially effective treatment strategies for obesity. The purpose of this article is to summarize the key findings presented in June 2022 at the 23rd annual Harvard Nutrition Obesity Symposium entitled "The Neurobiology of Eating Behavior in Obesity: Mechanisms and Therapeutic Targets." Findings presented at the symposium, sponsored by NIH P30 Nutrition Obesity Research Center at Harvard, enhance our current understanding of appetite biology, including innovative techniques used to assess and systematically manipulate critical hedonic processes, which will shape future research and the development of therapeutics for obesity prevention and treatment.
Collapse
Affiliation(s)
- Imen Becetti
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States.
| | - Esther L Bwenyi
- Metabolism Unit, Massachusetts General Hospital, Boston, MA, United States; Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Ivan E de Araujo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY, United States; Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Jamy Ard
- Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Bariatric and Weight Management Center, Wake Forest Baptist Health, Winston-Salem, NC, United States; Center on Diabetes, Obesity, and Metabolism, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Hypertension and Vascular Research Center, Cardiovascular Sciences Center, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Maya Angelou Center for Healthy Equity, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Ismaa Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom; Wellcome-Medical Research Council (MRC) Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom; Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, United Kingdom
| | - Carrie R Ferrario
- Department of Pharmacology, Psychology Department (Biopsychology Area), University of Michigan, Ann Arbor, MI, United States
| | - Marci E Gluck
- National Institutes of Health, Phoenix, AZ, United States; National Institute of Diabetes and Digestive and Kidney Disease, Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, Phoenix, AZ, United States
| | - Laura M Holsen
- Harvard Medical School, Boston, MA, United States; Division of Women's Health, Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States; Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, United States
| | - Paul J Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY, United States; Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Elizabeth A Lawson
- Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States; Neuroendocrine Unit, Massachusetts General Hospital, Boston, MA, United States
| | - Bradford B Lowell
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Ellen A Schur
- Division of General Internal Medicine, University of Washington, Seattle, WA, United States; Univeristy of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, United States; Univeristy of Washington Nutrition and Obesity Research Center, University of Washington, Seattle, WA, United States; Clinical and Translational Research Services Core, University of Washington, Seattle, WA, United States
| | - Takara L Stanley
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States; Metabolism Unit, Massachusetts General Hospital, Boston, MA, United States; Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Ali Tavakkoli
- Division of General and Gastrointestinal (GI) Surgery, Center for Weight Management and Wellness, Advanced Minimally Invasive Fellowship, Harvard Medical School, Boston, MA, United States
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital, Boston, MA, United States; Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Vibha Singhal
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States; Harvard Medical School, Boston, MA, United States; Pediatric Endocrinology and Obesity Medicine, Massachusetts General Hospital, Boston, MA, United States; Pediatric Program MGH Weight Center, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
31
|
Hidalgo Vira N, Oyarce K, Valladares Vega M, Goldfield GS, Guzmán-Gutiérrez E, Obregón AM. No association of the dopamine D2 receptor genetic bilocus score (rs1800497/rs1799732) on food addiction and food reinforcement in Chilean adults. Front Behav Neurosci 2023; 17:1067384. [PMID: 37064299 PMCID: PMC10102336 DOI: 10.3389/fnbeh.2023.1067384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 02/08/2023] [Indexed: 04/03/2023] Open
Abstract
Purpose: Different systems regulate food intake. In the reward system, dopamine (DA) is the main neurotransmitter, and a variety of genetic variants (rs1799732 and rs1800497) are associated with addiction. Addiction is a highly polygenic disease, where each allelic variant adds a small amount of vulnerability. Polymorphisms rs1799732 and rs1800497 are associated with eating behavior and hedonic hunger, but links to food addiction remain unclear.Aim: To evaluate the association between the bilocus profile (rs1799732-rs1800497) of the dopaminergic pathway with food reinforcement and food addiction in Chilean adults.Methods: A cross-sectional study recruited a convenience sample of 97 obese, 25 overweight, and 99 normal-weight adults (18–35 years). Anthropometric measurements were performed by standard procedures and eating behavior was assessed using the: Food Reinforcement Value Questionnaire (FRVQ) and Yale Food Addiction scale (YFAS). The DRD2 genotypes were determined by TaqMan assays (rs1800497 and rs1799732). A bilocus composite score was calculated.Results: In the normal weight group, individuals who were heterozygous for the rs1977932 variant (G/del) showed higher body weight (p-value 0.01) and abdominal circumference (p-value 0.01) compared to those who were homozygous (G/G). When analyzing rs1800497, a significant difference in BMI was observed for the normal weight group (p-value 0.02) where heterozygous showed higher BMI. In the obese group, homozygous A1/A1 showed higher BMI in comparison to A1/A2 and A2/A2 (p-value 0.03). Also, a significant difference in food reinforcement was observed in the rs1800497, where homozygous for the variant (A1A1) show less reinforcement (p-value 0.01).In relation to the bilocus score in the total sample, 11% showed “very low dopaminergic signaling”, 24.4% were “under”, 49.7% showed “intermediate signaling”, 12.7% showed “high” and 1.4% showed “very high”. No significant genotypic differences were observed in food reinforcement and food addiction by bilocus score.Conclusions: The results indicate that the genetic variants rs1799732 and rs1800497 (Taq1A) were associated with anthropometric measurements but not with food addiction or food reinforcement in Chilean university students. These results suggest that other genotypes, such as rs4680 and rs6277, which affect DA signaling capacity through a multilocus composite score, should be studied. Level V: Evidence obtained from a cross-sectional descriptive study.
Collapse
Affiliation(s)
- Nicole Hidalgo Vira
- Escuela de Tecnología Médica, Facultad de Medicina y Ciencia, Universidad San Sebastián, Concepción, Chile
- Escuela de Nutrición y Dietética, Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Concepción, Chile
| | - Karina Oyarce
- Escuela de Tecnología Médica, Facultad de Medicina y Ciencia, Universidad San Sebastián, Concepción, Chile
| | - Macarena Valladares Vega
- Escuela de Terapia Ocupacional, Facultad de Salud y Ciencias Sociales, Universidad de las Ámericas, Santiago, Chile
| | - Gary S. Goldfield
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Enrique Guzmán-Gutiérrez
- Pregnancy Diseases Laboratory, Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, Universidad de Concepción, Concepción, Chile
| | - Ana M. Obregón
- Escuela de Nutrición y Dietética, Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Concepción, Chile
- *Correspondence: Ana M. Obregón
| |
Collapse
|
32
|
Vourdoumpa A, Paltoglou G, Charmandari E. The Genetic Basis of Childhood Obesity: A Systematic Review. Nutrients 2023; 15:1416. [PMID: 36986146 PMCID: PMC10058966 DOI: 10.3390/nu15061416] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/05/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Overweight and obesity in childhood and adolescence represents one of the most challenging public health problems of our century owing to its epidemic proportions and the associated significant morbidity, mortality, and increase in public health costs. The pathogenesis of polygenic obesity is multifactorial and is due to the interaction among genetic, epigenetic, and environmental factors. More than 1100 independent genetic loci associated with obesity traits have been currently identified, and there is great interest in the decoding of their biological functions and the gene-environment interaction. The present study aimed to systematically review the scientific evidence and to explore the relation of single-nucleotide polymorphisms (SNPs) and copy number variants (CNVs) with changes in body mass index (BMI) and other measures of body composition in children and adolescents with obesity, as well as their response to lifestyle interventions. Twenty-seven studies were included in the qualitative synthesis, which consisted of 7928 overweight/obese children and adolescents at different stages of pubertal development who underwent multidisciplinary management. The effect of polymorphisms in 92 different genes was assessed and revealed SNPs in 24 genetic loci significantly associated with BMI and/or body composition change, which contribute to the complex metabolic imbalance of obesity, including the regulation of appetite and energy balance, the homeostasis of glucose, lipid, and adipose tissue, as well as their interactions. The decoding of the genetic and molecular/cellular pathophysiology of obesity and the gene-environment interactions, alongside with the individual genotype, will enable us to design targeted and personalized preventive and management interventions for obesity early in life.
Collapse
Affiliation(s)
- Aikaterini Vourdoumpa
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece
| | - George Paltoglou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece
| | - Evangelia Charmandari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece
- Division of Endocrinology and Metabolism, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
33
|
Montalban E, Giralt A, Taing L, Nakamura Y, Pelosi A, Brown M, de Pins B, Valjent E, Martin M, Nairn AC, Greengard P, Flajolet M, Herv D, Gambardella N, Roussarie JP, Girault JA. Operant training for highly palatable food alters translating mRNA in nucleus accumbens D2 neurons and reveals a modulatory role of Neurochondrin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531496. [PMID: 36945487 PMCID: PMC10028890 DOI: 10.1101/2023.03.07.531496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
BACKGROUND Highly palatable food triggers behavioral alterations reminiscent of those induced by addictive drugs. These effects involve the reward system and dopamine neurons, which modulate neurons in the nucleus accumbens (NAc). The molecular mechanisms underlying the effects of highly palatable food on feeding behavior are poorly understood. METHODS We studied the effects of 2-week operant conditioning of mice with standard or isocaloric highly palatable food. We investigated the behavioral effects and dendritic spine modifications in the NAc. We compared the translating mRNA in NAc neurons identified by the type of dopamine receptors they express, depending on the type of food and training. We tested the consequences of invalidation of an abundant downregulated gene, Ncdn (Neurochondrin). RESULTS Operant conditioning for highly palatable food increases motivation for food even in well-fed mice. In control mice, free access to regular or highly palatable food results in increased weight as compared to regular food only. Highly palatable food increases spine density in the NAc. In animals trained for highly palatable food, translating mRNAs are modified in NAc dopamine D2-receptor-expressing neurons, mostly corresponding to striatal projection neurons, but not in those expressing D1-receptors. Knock-out of Ncdn, an abundant down-regulated gene, opposes the conditioning-induced changes in satiety-sensitive feeding behavior and apparent motivation for highly palatable food, suggesting down-regulation may be a compensatory mechanism. CONCLUSIONS Our results emphasize the importance of mRNA alterations D2 striatal projection neurons in the NAc in the behavioral consequences of highly palatable food conditioning and suggest a modulatory contribution of Ncdn downregulation.
Collapse
|
34
|
Poghosyan V, Ioannou S, Al-Amri KM, Al-Mashhadi SA, Al-Mohammed F, Al-Otaibi T, Al-Saeed W. Spatiotemporal profile of altered neural reactivity to food images in obesity: Reward system is altered automatically and predicts efficacy of weight loss intervention. Front Neurosci 2023; 17:948063. [PMID: 36845430 PMCID: PMC9944082 DOI: 10.3389/fnins.2023.948063] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 01/24/2023] [Indexed: 02/10/2023] Open
Abstract
Introduction Obesity presents a significant public health problem. Brain plays a central role in etiology and maintenance of obesity. Prior neuroimaging studies have found that individuals with obesity exhibit altered neural responses to images of food within the brain reward system and related brain networks. However, little is known about the dynamics of these neural responses or their relationship to later weight change. In particular, it is unknown if in obesity, the altered reward response to food images emerges early and automatically, or later, in the controlled stage of processing. It also remains unclear if the pretreatment reward system reactivity to food images is predictive of subsequent weight loss intervention outcome. Methods In this study, we presented high-calorie and low-calorie food, and nonfood images to individuals with obesity, who were then prescribed lifestyle changes, and matched normal-weight controls, and examined neural reactivity using magnetoencephalography (MEG). We performed whole-brain analysis to explore and characterize large-scale dynamics of brain systems affected in obesity, and tested two specific hypotheses: (1) in obese individuals, the altered reward system reactivity to food images occurs early and automatically, and (2) pretreatment reward system reactivity predicts the outcome of lifestyle weight loss intervention, with reduced activity associated with successful weight loss. Results We identified a distributed set of brain regions and their precise temporal dynamics that showed altered response patterns in obesity. Specifically, we found reduced neural reactivity to food images in brain networks of reward and cognitive control, and elevated reactivity in regions of attentional control and visual processing. The hypoactivity in reward system emerged early, in the automatic stage of processing (< 150 ms post-stimulus). Reduced reward and attention responsivity, and elevated neural cognitive control were predictive of weight loss after six months in treatment. Discussion In summary, we have identified, for the first time with high temporal resolution, the large-scale dynamics of brain reactivity to food images in obese versus normal-weight individuals, and have confirmed both our hypotheses. These findings have important implications for our understanding of neurocognition and eating behavior in obesity, and can facilitate development of novel integrated treatment strategies, including tailored cognitive-behavioral and pharmacological therapies.
Collapse
Affiliation(s)
- Vahe Poghosyan
- Department of Neurophysiology, National Neuroscience Institute, King Fahad Medical City, Riyadh, Saudi Arabia,*Correspondence: Vahe Poghosyan,
| | - Stephanos Ioannou
- Department of Physiological Sciences, Alfaisal University, Riyadh, Saudi Arabia
| | - Khalid M. Al-Amri
- Obesity, Endocrinology and Metabolism Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Sufana A. Al-Mashhadi
- Research Unit, National Neuroscience Institute, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Fedaa Al-Mohammed
- Department of Neurophysiology, National Neuroscience Institute, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Tahani Al-Otaibi
- Department of Neurophysiology, National Neuroscience Institute, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Wjoud Al-Saeed
- Research Unit, National Neuroscience Institute, King Fahad Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
35
|
Pardo-Garcia TR, Gu K, Woerner RKR, Dus M. Food memory circuits regulate eating and energy balance. Curr Biol 2023; 33:215-227.e3. [PMID: 36528025 PMCID: PMC9877168 DOI: 10.1016/j.cub.2022.11.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 09/16/2022] [Accepted: 11/17/2022] [Indexed: 12/23/2022]
Abstract
In mammals, learning circuits play an essential role in energy balance by creating associations between sensory cues and the rewarding qualities of food. This process is altered by diet-induced obesity, but the causes and mechanisms are poorly understood. Here, we exploited the relative simplicity and wealth of knowledge about the D. melanogaster reinforcement learning network, the mushroom body, in order to study the relationship between the dietary environment, dopamine-induced plasticity, and food associations. We show flies that are fed a high-sugar diet cannot make associations between sensory cues and the rewarding properties of sugar. This deficit was caused by diet exposure, not fat accumulation, and specifically by lower dopamine-induced plasticity onto mushroom body output neurons (MBONs) during learning. Importantly, food memories dynamically tune the output of MBONs during eating, which instead remains fixed in sugar-diet animals. Interestingly, manipulating the activity of MBONs influenced eating and fat mass, depending on the diet. Altogether, this work advances our fundamental understanding of the mechanisms, causes, and consequences of the dietary environment on reinforcement learning and ingestive behavior.
Collapse
Affiliation(s)
- Thibaut R Pardo-Garcia
- The Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA; The Department of Molecular, Cellular, and Developmental Biology, College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kathleen Gu
- The Undergraduate Program in Neuroscience, College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Riley K R Woerner
- The Department of Molecular, Cellular, and Developmental Biology, College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Monica Dus
- The Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA; The Department of Molecular, Cellular, and Developmental Biology, College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA; The Undergraduate Program in Neuroscience, College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
36
|
Luo Y, Chen X, Zeng W, Xiao M, Liu Y, Gao X, Chen H. Associations of harsh, unpredictable environment, amygdala connectivity and overeating for children. Prog Neuropsychopharmacol Biol Psychiatry 2023; 120:110644. [PMID: 36167214 DOI: 10.1016/j.pnpbp.2022.110644] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 09/03/2022] [Accepted: 09/21/2022] [Indexed: 11/18/2022]
Abstract
OBJECTIVE In harsh and unpredictable environments, individuals tend to engage in activities that yield immediate rewards as delayed benefits can be unavailable. Substantial evidence suggests that a harsh and unpredictable childhood environment is associated with overeating. However, the neuromechanisms underlying this association remain unclear. This study aimed to investigate amygdala connectivity in relation to environmental harshness and unpredictability (EHU) from an evolutionary perspective and examine their relationship with overeating in children. METHODS Eighty-five children aged 8 to 12 years were scanned using a magnetic resonance imaging machine to assess resting-state functional connectivity (RSFC) of the two subregions of the amygdala (i.e., centromedial amygdala [CMA]; basolateral amygdala [BLA]). Self-reports of EHU and parental reports of overeating, including food responsiveness and enjoyment of food, were obtained cross-sectionally. Furthermore, findings indicated that children completed high- and low-calorie food portion choice tasks in the absence of hunger at 12 months of follow-up. RESULTS EHU was positively associated with parental reports of overeating, including food responsiveness and enjoyment, as well as children's selection of high-calorie food portion sizes. Moreover, static RSFC analyses revealed that EHU was negatively associated with bilateral BLA-left inferior frontal gyrus (IFG) connectivity, while dynamic RSFC analyses found that EHU was negatively associated with right CMA, left inferior parietal lobule, and right CMA-right precuneus connectivity. Particularly, the left BLA-left IFG connectivity mediated the association between EHU and parental reports of food responsiveness. CONCLUSION EHU was negatively associated with amygdala connectivity, which is implicated in the intrinsic processing of emotional regulation. Furthermore, deficits in emotional regulation resulted in increased energy intake. These insights provide a new perspective for understanding the developmental neuromechanisms underlying obesity.
Collapse
Affiliation(s)
- Yijun Luo
- Key Laboratory of Cognition and Personality (Ministry of Education), Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, China; School of Psychology, Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, China
| | - Ximei Chen
- Key Laboratory of Cognition and Personality (Ministry of Education), Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, China; School of Psychology, Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, China
| | - Weiyu Zeng
- Key Laboratory of Cognition and Personality (Ministry of Education), Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, China; School of Psychology, Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, China
| | - Mingyue Xiao
- Key Laboratory of Cognition and Personality (Ministry of Education), Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, China; School of Psychology, Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, China
| | - Yong Liu
- Key Laboratory of Cognition and Personality (Ministry of Education), Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, China; School of Psychology, Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, China
| | - Xiao Gao
- Key Laboratory of Cognition and Personality (Ministry of Education), Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, China; School of Psychology, Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, China
| | - Hong Chen
- Key Laboratory of Cognition and Personality (Ministry of Education), Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, China; School of Psychology, Southwest University, No. 2 Tiansheng Road, Beibei, Chongqing 400715, China.
| |
Collapse
|
37
|
Personalized Dietary Advice to Increase Protein Intake in Older Adults Does Not Affect the Gut Microbiota, Appetite or Central Processing of Food Stimuli in Community-Dwelling Older Adults: A Six-Month Randomized Controlled Trial. Nutrients 2023; 15:nu15020332. [PMID: 36678203 PMCID: PMC9862486 DOI: 10.3390/nu15020332] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
Expert groups argue to raise the recommended daily allowance for protein in older adults from 0.8 to 1.2 g/kg/day to prevent undernutrition. However, protein is thought to increase satiety, possibly through effects on gut microbiota and central appetite regulation. If true, raising daily protein intake may work counterproductively. In a randomized controlled trial, we evaluated the effects of dietary advice aimed at increasing protein intake to 1.2 g/kg adjusted body weight/day (g/kg aBW/day) on appetite and gut microbiota in 90 community-dwelling older adults with habitual protein intake <1.0 g/kg aBW/day (Nintervention = 47, Ncontrol = 43). Food intake was determined by 24-h dietary recalls and gut microbiota by 16S rRNA sequencing. Functional magnetic resonance imaging (fMRI) scans were performed in a subgroup of 48 participants to evaluate central nervous system responses to food-related stimuli. Both groups had mean baseline protein intake of 0.8 ± 0.2 g/kg aBW/day. At 6 months’ follow-up this increased to 1.2 ± 0.2 g/kg aBW/day for the intervention group and 0.9 ± 0.2 g/kg aBW/day for the control group. Microbiota composition was not affected, nor were appetite or brain activity in response to food-related stimuli. Increasing protein intake in older adults to 1.2 g/kg aBW/day does not negatively impact the gut microbiota or suppress appetite.
Collapse
|
38
|
Hanßen R, Schiweck C, Aichholzer M, Reif A, Edwin Thanarajah S. Food reward and its aberrations in obesity. Curr Opin Behav Sci 2022. [DOI: 10.1016/j.cobeha.2022.101224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
39
|
Duan Y, Zheng M, Wu J, Ma J, Xing X, Ma Z, Li S, Li Y, Xue X, Hua X, Xu J. Cerebral 18 F-fluorodeoxyglucose metabolism alteration of reward- and motivation-related regions in groups of different BMI classifications. Obesity (Silver Spring) 2022; 30:2213-2221. [PMID: 36321272 PMCID: PMC9828716 DOI: 10.1002/oby.23553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 07/05/2022] [Accepted: 07/08/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE This study explored the relationship between BMI and regional cerebral glucose metabolism and explicitly detected regions with significant differences in cerebral metabolism using positron emission tomography (PET)/magnetic resonance imaging in the resting state. METHODS Corresponding PET images acquired from 220 participants were sorted into four groups according to Asian BMI standards: underweight, normal weight, overweight, and obesity. Pearson correlation coefficient analysis was performed to assess the association between BMI and standard uptake value. The regional cerebral glucose metabolism was measured in the fasted state. The PET images were analyzed using statistical parameter maps. One-way ANOVA was used to explore differences in the standard uptake value as an indicator of regional cerebral glucose metabolism. RESULTS This study found that lower cerebral glucose metabolism in reward- and motivation-related regions was accompanied by more severe obesity and that regional cerebral glucose metabolism activities were negatively correlated with BMI. In addition, more severe obesity was accompanied by a larger range of areas with significant differences independent of current dietary status. CONCLUSIONS These findings suggest that the reward and motivation circuits may be a factor regulating energy balance and influencing the degree of obesity.
Collapse
Affiliation(s)
- Yu‐Jie Duan
- School of Rehabilitation ScienceShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Mou‐Xiong Zheng
- Department of Traumatology and Orthopedics, Yueyang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Jia‐Jia Wu
- Center of Rehabilitation Medicine, Yueyang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Jie Ma
- Center of Rehabilitation Medicine, Yueyang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xiang‐Xin Xing
- Center of Rehabilitation Medicine, Yueyang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Zhen‐Zhen Ma
- Department of Rehabilitation Medicine, Longhua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Si‐Si Li
- School of Rehabilitation ScienceShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Yu‐Lin Li
- School of Rehabilitation ScienceShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xin Xue
- School of Rehabilitation ScienceShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xu‐Yun Hua
- Department of Traumatology and Orthopedics, Yueyang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Jian‐Guang Xu
- School of Rehabilitation ScienceShanghai University of Traditional Chinese MedicineShanghaiChina
- Center of Rehabilitation Medicine, Yueyang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
- Engineering Research Center of Traditional Chinese Medicine Intelligent RehabilitationMinistry of EducationShanghaiChina
| |
Collapse
|
40
|
Devoto F, Coricelli C, Paulesu E, Zapparoli L. Neural circuits mediating food cue-reactivity: Toward a new model shaping the interplay of internal and external factors. Front Nutr 2022; 9:954523. [PMID: 36276811 PMCID: PMC9579536 DOI: 10.3389/fnut.2022.954523] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Francantonio Devoto
- Psychology Department and NeuroMi—Milan Centre for Neuroscience, University of Milano-Bicocca, Milan, Italy,*Correspondence: Francantonio Devoto
| | - Carol Coricelli
- Psychology Department, Western University, London, ON, Canada
| | - Eraldo Paulesu
- Psychology Department and NeuroMi—Milan Centre for Neuroscience, University of Milano-Bicocca, Milan, Italy,fMRI Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Galeazzi, Milan, Italy
| | - Laura Zapparoli
- Psychology Department and NeuroMi—Milan Centre for Neuroscience, University of Milano-Bicocca, Milan, Italy,fMRI Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Galeazzi, Milan, Italy,Laura Zapparoli
| |
Collapse
|
41
|
O'Connor RM, Kenny PJ. Utility of 'substance use disorder' as a heuristic for understanding overeating and obesity. Prog Neuropsychopharmacol Biol Psychiatry 2022; 118:110580. [PMID: 35636576 DOI: 10.1016/j.pnpbp.2022.110580] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 02/07/2023]
Abstract
Rates of obesity and obesity-associated diseases have increased dramatically in countries with developed economies. Substance use disorders (SUDs) are characterized by the persistent use of the substance despite negative consequences. It has been hypothesized that overconsumption of palatable energy dense food can elicit SUD-like maladaptive behaviors that contribute to persistent caloric intake beyond homeostatic need even in the face of negative consequences. Palatable food and drugs of abuse act on many of the same motivation-related circuits in the brain, and can induce, at least superficially, similar molecular, cellular, and physiological adaptations on these circuits. As such, applying knowledge about the neurobiological mechanisms of SUDs may serve as useful heuristic to better understand the persistent overconsumption of palatable food that contributes to obesity. However, many important differences exist between the actions of drugs of abuse and palatable food in the brain. This warrants caution when attributing weight gain and obesity to the manifestation of a putative SUD-related behavioral disorder. Here, we describe similarities and differences between compulsive drug use in SUDs and overconsumption in obesity and consider the merit of the concept of "food addiction".
Collapse
Affiliation(s)
- Richard M O'Connor
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States of America
| | - Paul J Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States of America.
| |
Collapse
|
42
|
Food Reward Alterations during Obesity Are Associated with Inflammation in the Striatum in Mice: Beneficial Effects of Akkermansia muciniphila. Cells 2022; 11:cells11162534. [PMID: 36010611 PMCID: PMC9406832 DOI: 10.3390/cells11162534] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
The reward system involved in hedonic food intake presents neuronal and behavioral dysregulations during obesity. Moreover, gut microbiota dysbiosis during obesity promotes low-grade inflammation in peripheral organs and in the brain contributing to metabolic alterations. The mechanisms underlying reward dysregulations during obesity remain unclear. We investigated if inflammation affects the striatum during obesity using a cohort of control-fed or diet-induced obese (DIO) male mice. We tested the potential effects of specific gut bacteria on the reward system during obesity by administrating Akkermansia muciniphila daily or a placebo to DIO male mice. We showed that dysregulations of the food reward are associated with inflammation and alterations in the blood–brain barrier in the striatum of obese mice. We identified Akkermansia muciniphila as a novel actor able to improve the dysregulated reward behaviors associated with obesity, potentially through a decreased activation of inflammatory pathways and lipid-sensing ability in the striatum. These results open a new field of research and suggest that gut microbes can be considered as an innovative therapeutic approach to attenuate reward alterations in obesity. This study provides substance for further investigations of Akkermansia muciniphila-mediated behavioral improvements in other inflammatory neuropsychiatric disorders.
Collapse
|
43
|
White O, Roeder N, Blum K, Eiden RD, Thanos PK. Prenatal Effects of Nicotine on Obesity Risks: A Narrative Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:9477. [PMID: 35954830 PMCID: PMC9368674 DOI: 10.3390/ijerph19159477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 11/16/2022]
Abstract
Nicotine usage by mothers throughout pregnancy has been observed to relate to numerous deleterious effects in children, especially relating to obesity. Children who have prenatally been exposed to nicotine tend to have lower birth weights, with an elevated risk of becoming overweight throughout development and into their adolescent and adult life. There are numerous theories as to how this occurs: catch-up growth theory, thrifty phenotype theory, neurotransmitter or endocrine imbalances theory, and a more recent examination on the genetic factors relating to obesity risk. In addition to the negative effect on bodyweight and BMI, individuals with obesity may also suffer from numerous comorbidities involving metabolic disease. These may include type 1 and 2 diabetes, high cholesterol levels, and liver disease. Predisposition for obesity with nicotine usage may also be associated with genetic risk alleles for obesity, such as the DRD2 A1 variant. This is important for prenatally nicotine-exposed individuals as an opportunity to provide early prevention and intervention of obesity-related risks.
Collapse
Affiliation(s)
- Olivia White
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA; (O.W.); (N.R.)
- Department of Psychology, University at Buffalo, Buffalo, NY 14203, USA
| | - Nicole Roeder
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA; (O.W.); (N.R.)
- Department of Psychology, University at Buffalo, Buffalo, NY 14203, USA
| | - Kenneth Blum
- Division of Addiction Research, Center for Psychiatry, Medicine & Primary Care (Office of Provost), Western University Health Sciences, Pomona, CA 91766, USA;
| | - Rina D. Eiden
- Department of Psychology, Social Science Research Institute, The Pennsylvania State University, University Park, PA 16801, USA;
| | - Panayotis K. Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA; (O.W.); (N.R.)
- Department of Psychology, University at Buffalo, Buffalo, NY 14203, USA
| |
Collapse
|
44
|
Abstract
The modern obesogenic environment contains an abundance of food cues (e.g., sight, smell of food) as well cues that are associated with food through learning and memory processes. Food cue exposure can lead to food seeking and excessive consumption in otherwise food-sated individuals, and a high level of food cue responsivity is a risk factor for overweight and obesity. Similar food cue responses are observed in experimental rodent models, and these models are therefore useful for mechanistically identifying the neural circuits mediating food cue responsivity. This review draws from both experimental rodent models and human data to characterize the behavioral and biological processes through which food-associated stimuli contribute to overeating and weight gain. Two rodent models are emphasized - cue-potentiated feeding and Pavlovian-instrumental transfer - that provide insight in the neural circuits and peptide systems underlying food cue responsivity. Data from humans are highlighted that reveal physiological, psychological, and neural mechanisms that connect food cue responsivity with overeating and weight gain. The collective literature identifies connections between heightened food cue responsivity and obesity in both rodents and humans, and identifies underlying brain regions (nucleus accumbens, amygdala, orbitofrontal cortex, hippocampus) and endocrine systems (ghrelin) that regulate food cue responsivity in both species. These species similarities are encouraging for the possibility of mechanistic rodent model research and further human research leading to novel treatments for excessive food cue responsivity in humans.
Collapse
Affiliation(s)
- Scott E Kanoski
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| | - Kerri N Boutelle
- Department of Pediatrics, Herbert Wertheim School of Public Health and Human Longevity Science, and Psychiatry, University of California San Diego, San Diego, CA, USA.
| |
Collapse
|
45
|
Kung PH, Soriano-Mas C, Steward T. The influence of the subcortex and brain stem on overeating: How advances in functional neuroimaging can be applied to expand neurobiological models to beyond the cortex. Rev Endocr Metab Disord 2022; 23:719-731. [PMID: 35380355 PMCID: PMC9307542 DOI: 10.1007/s11154-022-09720-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/21/2022] [Indexed: 12/13/2022]
Abstract
Functional neuroimaging has become a widely used tool in obesity and eating disorder research to explore the alterations in neurobiology that underlie overeating and binge eating behaviors. Current and traditional neurobiological models underscore the importance of impairments in brain systems supporting reward, cognitive control, attention, and emotion regulation as primary drivers for overeating. Due to the technical limitations of standard field strength functional magnetic resonance imaging (fMRI) scanners, human neuroimaging research to date has focused largely on cortical and basal ganglia effects on appetitive behaviors. The present review draws on animal and human research to highlight how neural signaling encoding energy regulation, reward-learning, and habit formation converge on hypothalamic, brainstem, thalamic, and striatal regions to contribute to overeating in humans. We also consider the role of regions such as the mediodorsal thalamus, ventral striatum, lateral hypothalamus and locus coeruleus in supporting habit formation, inhibitory control of food craving, and attentional biases. Through these discussions, we present proposals on how the neurobiology underlying these processes could be examined using functional neuroimaging and highlight how ultra-high field 7-Tesla (7 T) fMRI may be leveraged to elucidate the potential functional alterations in subcortical networks. Focus is given to how interactions of these regions with peripheral endocannabinoids and neuropeptides, such as orexin, could be explored. Technical and methodological aspects regarding the use of ultra-high field 7 T fMRI to study eating behaviors are also reviewed.
Collapse
Affiliation(s)
- Po-Han Kung
- Melbourne School of Psychological Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Victoria, Australia
| | - Carles Soriano-Mas
- Psychiatry and Mental Health Group, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Neuroscience Program, L'Hospitalet de Llobregat, Spain
- CIBERSAM, Carlos III Health Institute, Madrid, Spain
- Department of Social Psychology and Quantitative Psychology, University of Barcelona, Barcelona, Spain
| | - Trevor Steward
- Melbourne School of Psychological Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia.
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Victoria, Australia.
| |
Collapse
|
46
|
Fang X, Davis X, Flack KD, Duncan C, Li F, White M, Grilo C, Small DM. Dietary adaptation for weight loss maintenance at Yale (DAWLY): Protocol and predictions for a randomized controlled trial. Front Nutr 2022; 9:940064. [PMID: 35967820 PMCID: PMC9369668 DOI: 10.3389/fnut.2022.940064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background Current therapies for obesity treatment are effective at producing short-term weight loss, but weight loss maintenance remains a significant challenge. Here we investigate the impact of pre-intervention dietary fat intake on the efficacy of a dietary supplement to support weight loss maintenance. Preclinical work demonstrates that a vagal afferent pathway critical for sensing dietary lipids is blunted by a high-fat diet (HFD), resulting in a reduced preference for a low-fat emulsion and severe blunting of the dopamine (DA) response to the gastric infusion of lipids. Infusion of the gut lipid messenger oleoylethanolamide (OEA), which is also depleted by HFD, immediately reverses this DA blunting and restores preference for the low-fat emulsion. Studies of OEA supplementation for weight loss in humans have had limited success. Given the strong effect of HFD on this pathway, we designed a study to test whether the efficacy of OEA as a weight loss treatment is related to pre-intervention habitual intake of dietary fat. Methods/Design We employed a randomized, double-blind, placebo-controlled trial in which 100 adults with overweight/obesity (OW/OB) were randomized to receive either OEA or placebo daily for 16 months. Following a baseline evaluation of diet, metabolic health, adiposity, and brain response to a palatable an energy dense food, participants in both groups underwent a 4-month behavioral weight loss intervention (LEARN®) followed by a 1-year maintenance period. The study aims are to (1) determine if pre-intervention dietary fat intake moderates the ability of OEA to improve weight loss and weight loss maintenance after a gold standard behavioral weight loss treatment; (2) identify biomarkers that predict outcome and optimize a stratification strategy; and (3) test a model underlying OEA's effectiveness. Discussion Focusing on interventions that target the gut-brain axis is supported by mounting evidence for the role of gut-brain signaling in food choice and the modulation of this circuit by diet. If successful, this work will provide support for targeting the gut-brain pathway for weight loss maintenance using a precision medicine approach that is easy and inexpensive to implement. Clinical Trial Registration [www.ClinicalTrials.gov], identifier [NCT04614233].
Collapse
Affiliation(s)
- Xi Fang
- Modern Diet and Physiology Research Center, New Haven, CT, United States
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Xue Davis
- Modern Diet and Physiology Research Center, New Haven, CT, United States
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Kyle D. Flack
- Department of Dietetics and Human Nutrition, College of Agriculture, Foods, and Environment, University of Kentucky, Lexington, KY, United States
| | - Chavonn Duncan
- Modern Diet and Physiology Research Center, New Haven, CT, United States
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Fangyong Li
- Yale Center for Analytical Sciences, Yale School of Public Health, New Haven, CT, United States
| | - Marney White
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- Department of Social and Behavioral Sciences, Yale School of Public Health, New Haven, CT, United States
| | - Carlos Grilo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Dana M. Small
- Modern Diet and Physiology Research Center, New Haven, CT, United States
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
47
|
de Wouters d’Oplinter A, Huwart SJP, Cani PD, Everard A. Gut microbes and food reward: From the gut to the brain. Front Neurosci 2022; 16:947240. [PMID: 35958993 PMCID: PMC9358980 DOI: 10.3389/fnins.2022.947240] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Inappropriate food intake behavior is one of the main drivers for fat mass development leading to obesity. Importantly the gut microbiota-mediated signals have emerged as key actors regulating food intake acting mainly on the hypothalamus, and thereby controlling hunger or satiety/satiation feelings. However, food intake is also controlled by the hedonic and reward systems leading to food intake based on pleasure (i.e., non-homeostatic control of food intake). This review focus on both the homeostatic and the non-homeostatic controls of food intake and the implication of the gut microbiota on the control of these systems. The gut-brain axis is involved in the communications between the gut microbes and the brain to modulate host food intake behaviors through systemic and nervous pathways. Therefore, here we describe several mediators of the gut-brain axis including gastrointestinal hormones, neurotransmitters, bioactive lipids as well as bacterial metabolites and compounds. The modulation of gut-brain axis by gut microbes is deeply addressed in the context of host food intake with a specific focus on hedonic feeding. Finally, we also discuss possible gut microbiota-based therapeutic approaches that could lead to potential clinical applications to restore food reward alterations. Therapeutic applications to tackle these dysregulations is of utmost importance since most of the available solutions to treat obesity present low success rate.
Collapse
|
48
|
Alterations in Functional and Structural Connectivity of Basal Ganglia Network in Patients with Obesity. Brain Topogr 2022; 35:453-463. [PMID: 35780276 DOI: 10.1007/s10548-022-00906-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 06/15/2022] [Indexed: 11/02/2022]
Abstract
Obesity is related to overconsumption of high-calorie (HiCal) food, which is modulated by brain reward and inhibitory control circuitries. The basal ganglia (BG) are a key set of nuclei within the reward circuitry, but obesity-associated functional and structural abnormalities of BG have not been well studied. Resting-state functional MRI with independent component analysis (ICA) and probabilistic tractography were employed to investigate differences in BG-related functional-(FC) and structural connectivity (SC) between 32 patients with obesity (OB) and 35 normal-weight (NW) participants. Compared to NW, OB showed significantly lower FC strength in the caudate nucleus within the BG network, and seed-based FC analysis showed lower FC between caudate and dorsolateral prefrontal cortex (DLPFC), which was negatively correlated with craving for HiCal food cues. Further SC analysis revealed that OB showed lower SC than NW between left caudate and left DLPFC as measured with fractional anisotropy (FA). Alterations in FC and SC between caudate and DLPFC in obese patients, which highlights the role of BG network in modulating the balance between reward and inhibitory-control.
Collapse
|
49
|
Kanarik M, Grimm O, Mota NR, Reif A, Harro J. ADHD co-morbidities: A review of implication of gene × environment effects with dopamine-related genes. Neurosci Biobehav Rev 2022; 139:104757. [PMID: 35777579 DOI: 10.1016/j.neubiorev.2022.104757] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 06/25/2022] [Accepted: 06/26/2022] [Indexed: 02/07/2023]
Abstract
ADHD is a major burden in adulthood, where co-morbid conditions such as depression, substance use disorder and obesity often dominate the clinical picture. ADHD has substantial shared heritability with other mental disorders, contributing to comorbidity. However, environmental risk factors exist but their interaction with genetic makeup, especially in relation to comorbid disorders, remains elusive. This review for the first time summarizes present knowledge on gene x environment (GxE) interactions regarding the dopamine system. Hitherto, mainly candidate (GxE) studies were performed, focusing on the genes DRD4, DAT1 and MAOA. Some evidence suggest that the variable number tandem repeats in DRD4 and MAOA may mediate GxE interactions in ADHD generally, and comorbid conditions specifically. Nevertheless, even for these genes, common variants are bound to suggest risk only in the context of gender and specific environments. For other polymorphisms, evidence is contradictory and less convincing. Particularly lacking are longitudinal studies testing the interaction of well-defined environmental with polygenic risk scores reflecting the dopamine system in its entirety.
Collapse
Affiliation(s)
- Margus Kanarik
- Chair of Neuropsychopharmacology, Institute of Chemistry, University of Tartu, Ravila 14A Chemicum, 50411 Tartu, Estonia
| | - Oliver Grimm
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Nina Roth Mota
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Jaanus Harro
- Chair of Neuropsychopharmacology, Institute of Chemistry, University of Tartu, Ravila 14A Chemicum, 50411 Tartu, Estonia; Psychiatry Clinic, North Estonia Medical Centre, Paldiski Road 52, 10614 Tallinn, Estonia.
| |
Collapse
|
50
|
Morales I. Brain regulation of hunger and motivation: The case for integrating homeostatic and hedonic concepts and its implications for obesity and addiction. Appetite 2022; 177:106146. [PMID: 35753443 DOI: 10.1016/j.appet.2022.106146] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/16/2022] [Accepted: 06/21/2022] [Indexed: 11/19/2022]
Abstract
Obesity and other eating disorders are marked by dysregulations to brain metabolic, hedonic, motivational, and sensory systems that control food intake. Classic approaches in hunger research have distinguished between hedonic and homeostatic processes, and have mostly treated these systems as independent. Hindbrain structures and a complex network of interconnected hypothalamic nuclei control metabolic processes, energy expenditure, and food intake while mesocorticolimbic structures are though to control hedonic and motivational processes associated with food reward. However, it is becoming increasingly clear that hedonic and homeostatic brain systems do not function in isolation, but rather interact as part of a larger network that regulates food intake. Incentive theories of motivation provide a useful route to explore these interactions. Adapting incentive theories of motivation can enable researchers to better how motivational systems dysfunction during disease. Obesity and addiction are associated with profound alterations to both hedonic and homeostatic brain systems that result in maladaptive patterns of consumption. A subset of individuals with obesity may experience pathological cravings for food due to incentive sensitization of brain systems that generate excessive 'wanting' to eat. Further progress in understanding how the brain regulates hunger and appetite may depend on merging traditional hedonic and homeostatic concepts of food reward and motivation.
Collapse
Affiliation(s)
- Ileana Morales
- Department of Psychology, University of Michigan, 530 Church Street, Ann Arbor, MI, 48109-1043, USA.
| |
Collapse
|