1
|
Cruz-Trujillo R, Díaz-Urbina D, Díaz-Gandarilla JA, Vidal-López DG, Escartín-Pérez RE, Mancilla-Diaz JM, Florán B, Tejas-Juárez JG. Stimulation of Dopamine D4 Receptors in the Nucleus Accumbens Shell Increases Palatable Food Intake in Satiated Male Rats: Modulation by NMDA and AMPA Receptors. Brain Sci 2024; 14:1103. [PMID: 39595865 PMCID: PMC11592207 DOI: 10.3390/brainsci14111103] [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: 10/06/2024] [Revised: 10/26/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Palatability significantly influences food consumption, often leading to overeating and obesity by activating the brain's reward systems. The nucleus accumbens (NAc) plays a central role in this process, modulating reward mechanisms primarily via dopamine through D2-like receptors (D2R, D3R, D4R). While the involvement of D2 receptors in feeding is well-documented, the role of D4 receptors (D4Rs) is less clear. METHODS Male Wistar rats received intra-NAc shell microinjections of the D4R agonist PD-168077 and the antagonist L-745870. This study also examined the modulation between D4R and glutamatergic transmission by administration of NMDA, NMDA receptor antagonist AP-5, AMPA, and AMPA receptor antagonist CNQX. RESULTS PD-168077 increased sweet solution intake by 46%, an effect that was reversed by L-745870. Pre-treatment with NMDA prevented the stimulatory effect of PD-168077, whereas the NMDA receptor antagonist AP-5 had no such effect. Additionally, AMPA administration reduced sweet solution intake by 63%, counteracting the effect of PD-168077, while the AMPA receptor antagonist CNQX, on its own, increased intake by 40%. CONCLUSIONS These findings suggest that D4Rs promote hedonic feeding by modulating glutamatergic transmission in the NAc shell, highlighting the complexity of D4R involvement in food intake regulation. This study underscores the potential of targeting D4Rs for therapeutic interventions in eating disorders and obesity, though further research is essential to clarify the precise mechanisms through which D4R modulates AMPA and NMDA receptor activity in feeding behavior.
Collapse
Affiliation(s)
- Refugio Cruz-Trujillo
- Escuela de Ciencias Químicas, Universidad Autónoma de Chiapas (UNACH), Carretera Panamericana Ocozocoautla-Cintalapa Km. 2.5, Ocozocoautla de Espinosa 29140, Mexico;
- Departamento de Químicos Farmacobiólogos, Universidad Pablo Guardado Chávez (UPGCH), Libramiento Norte Oriente No. 3450, Tuxtla Gutiérrez 29040, Mexico
| | - Daniel Díaz-Urbina
- Laboratory on Neurobiology of Compulsive Behaviors, NIMH, National Institutes of Health, Bethesda, MD 20892, USA;
- Laboratorio de Neurobiología de la Alimentación, Grupo de Investigación en Nutrición, División de Investigación y Posgrado de la FES Iztacala, UNAM, Ciudad de México 04510, Mexico; (R.E.E.-P.); (J.M.M.-D.)
| | - José Alfredo Díaz-Gandarilla
- División Académica Multidisciplinaria de Comalcalco, Universidad Juárez Autónoma de Tabasco (UJAT), Comalcalco 86658, Mexico;
| | | | - Rodrigo Erick Escartín-Pérez
- Laboratorio de Neurobiología de la Alimentación, Grupo de Investigación en Nutrición, División de Investigación y Posgrado de la FES Iztacala, UNAM, Ciudad de México 04510, Mexico; (R.E.E.-P.); (J.M.M.-D.)
| | - Juan Manuel Mancilla-Diaz
- Laboratorio de Neurobiología de la Alimentación, Grupo de Investigación en Nutrición, División de Investigación y Posgrado de la FES Iztacala, UNAM, Ciudad de México 04510, Mexico; (R.E.E.-P.); (J.M.M.-D.)
| | - Benjamín Florán
- Centro de Investigación y de Estudios Avanzados del IPN, Departamento de Fisiología, Biofísica y Neurociencias, Ciudad de México 07360, Mexico;
| | - Juan Gabriel Tejas-Juárez
- División Académica Multidisciplinaria de Comalcalco, Universidad Juárez Autónoma de Tabasco (UJAT), Comalcalco 86658, Mexico;
| |
Collapse
|
2
|
D'Aquila PS. Dopamine, activation of ingestion and evaluation of response efficacy: a focus on the within-session time-course of licking burst number. Psychopharmacology (Berl) 2024; 241:1111-1124. [PMID: 38702473 PMCID: PMC11106101 DOI: 10.1007/s00213-024-06600-1] [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: 02/07/2024] [Accepted: 04/24/2024] [Indexed: 05/06/2024]
Abstract
RATIONALE Evidence on the effect of dopamine D1-like and D2-like receptor antagonists on licking microstructure and the forced swimming response led us to suggest that (i) dopamine on D1-like receptors plays a role in activating reward-directed responses and (ii) the level of response activation is reboosted based on a process of evaluation of response efficacy requiring dopamine on D2-like receptors. A main piece of evidence in support of this hypothesis is the observation that the dopamine D2-like receptor antagonist raclopride induces a within-session decrement of burst number occurring after the contact with the reward. The few published studies with a detailed analysis of the time-course of this measure were conducted in our laboratory. OBJECTIVES The aim of this review is to recapitulate and discuss the evidence in support of the analysis of the within-session burst number as a behavioural substrate for the study of the mechanisms governing ingestion, behavioural activation and the related evaluation processes, and its relevance in the analysis of drug effects on ingestion. CONCLUSIONS The evidence gathered so far suggests that the analysis of the within-session time-course of burst number provides an important behavioural substrate for the study of the mechanisms governing ingestion, behavioural activation and the related evaluation processes, and might provide decisive evidence in the analysis of the effects of drugs on ingestion. However, further evidence from independent sources is necessary to validate the use and the proposed interpretation of this measure.
Collapse
Affiliation(s)
- Paolo S D'Aquila
- Dipartimento di Scienze Biomediche, Università di Sassari, Viale S. Pietro 43/b, Sassari, 07100, Italy.
| |
Collapse
|
3
|
Fadahunsi N, Petersen J, Metz S, Jakobsen A, Vad Mathiesen C, Silke Buch-Rasmussen A, Kurgan N, Kjærgaard Larsen J, Andersen RC, Topilko T, Svendsen C, Apuschkin M, Skovbjerg G, Hendrik Schmidt J, Houser G, Elgaard Jager S, Bach A, Deshmukh AS, Kilpeläinen TO, Strømgaard K, Madsen KL, Clemmensen C. Targeting postsynaptic glutamate receptor scaffolding proteins PSD-95 and PICK1 for obesity treatment. SCIENCE ADVANCES 2024; 10:eadg2636. [PMID: 38427737 PMCID: PMC10906926 DOI: 10.1126/sciadv.adg2636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/29/2024] [Indexed: 03/03/2024]
Abstract
Human genome-wide association studies (GWAS) suggest a functional role for central glutamate receptor signaling and plasticity in body weight regulation. Here, we use UK Biobank GWAS summary statistics of body mass index (BMI) and body fat percentage (BF%) to identify genes encoding proteins known to interact with postsynaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-d-aspartate (NMDA) receptors. Loci in/near discs large homolog 4 (DLG4) and protein interacting with C kinase 1 (PICK1) reached genome-wide significance (P < 5 × 10-8) for BF% and/or BMI. To further evaluate the functional role of postsynaptic density protein-95 (PSD-95; gene name: DLG4) and PICK1 in energy homeostasis, we used dimeric PSD-95/disc large/ZO-1 (PDZ) domain-targeting peptides of PSD-95 and PICK1 to demonstrate that pharmacological inhibition of PSD-95 and PICK1 induces prolonged weight-lowering effects in obese mice. Collectively, these data demonstrate that the glutamate receptor scaffolding proteins, PICK1 and PSD-95, are genetically linked to obesity and that pharmacological targeting of their PDZ domains represents a promising therapeutic avenue for sustained weight loss.
Collapse
Affiliation(s)
- Nicole Fadahunsi
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jonas Petersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Sophia Metz
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Alexander Jakobsen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cecilie Vad Mathiesen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Alberte Silke Buch-Rasmussen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Nigel Kurgan
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jeppe Kjærgaard Larsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Rita C. Andersen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Charlotte Svendsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Mia Apuschkin
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Grethe Skovbjerg
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Gubra, Hørsholm, Denmark
| | - Jan Hendrik Schmidt
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Grace Houser
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sara Elgaard Jager
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders Bach
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Atul S. Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Tuomas O. Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Strømgaard
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Kenneth L. Madsen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Berger G, Corris JD, Fields SE, Hao L, Scarpa LL, Bello NT. Systematic Review of Binge Eating Rodent Models for Developing Novel or Repurposing Existing Pharmacotherapies. Biomolecules 2023; 13:742. [PMID: 37238615 PMCID: PMC10216509 DOI: 10.3390/biom13050742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/18/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Recent advances in developing and screening candidate pharmacotherapies for psychiatric disorders have depended on rodent models. Eating disorders are a set of psychiatric disorders that have traditionally relied on behavioral therapies for effective long-term treatment. However, the clinical use of Lisdexamfatamine for binge eating disorder (BED) has furthered the notion of using pharmacotherapies for treating binge eating pathologies. While there are several binge eating rodent models, there is not a consensus on how to define pharmacological effectiveness within these models. Our purpose is to provide an overview of the potential pharmacotherapies or compounds tested in established rodent models of binge eating behavior. These findings will help provide guidance for determining pharmacological effectiveness for potential novel or repurposed pharmacotherapies.
Collapse
Affiliation(s)
- Gregory Berger
- Endocrinology and Animal Biosciences Graduate Program, Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Joshua D. Corris
- Endocrinology and Animal Biosciences Graduate Program, Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Spencer E. Fields
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Nutritional Sciences Graduate Program, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Lihong Hao
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Lori L. Scarpa
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Nicholas T. Bello
- Endocrinology and Animal Biosciences Graduate Program, Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Rutgers Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| |
Collapse
|
5
|
Zakariaª EM, Abdel-Ghanyª RH, Elgharbawyª AS, Alsemehᵇ AE, Metwallyª SS. A novel approach to repositioning memantine for metabolic syndrome-induced steatohepatitis: Modulation of hepatic autophagy, inflammation, and fibrosis. Life Sci 2023; 319:121509. [PMID: 36822316 DOI: 10.1016/j.lfs.2023.121509] [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: 11/23/2022] [Revised: 02/07/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023]
Abstract
AIMS This study investigated the possible hepatoprotective effects of memantine, compared to pioglitazone, in rat steatohepatitis, emphasizing its role in modulating hepatic autophagy. MAIN METHODS Metabolic syndrome (MetS) was provoked in adult male Wistar rats by a high fructose/fat/salt regimen for eight weeks. Then, rats were administered either memantine or pioglitazone daily for 10 weeks (both at 20 mg/kg, orally). An oral glucose tolerance test (OGTT) was done at the end of the study, and serum liver enzymes, lipids, and fasting blood glucose were measured. Also, hepatic contents of inflammatory, oxidative, and autophagy markers were quantified. Additionally, histopathological examinations of general hepatic structure and glycogen content were performed. KEY FINDINGS Compared to the MetS rats, memantine normalized fasting serum insulin, Homeostatic Model Assessment (HOMA-IR), serum lipids, and liver enzymes (ALT and AST). Memantine also markedly reduced hepatic inflammatory markers; NF-κB and TNF-α. In addition, hepatic NRF2 and GSH were augmented, while hepatic MDA was reduced by memantine. Interestingly, livers of the memantine group showed elevated Beclin1 and LC3 and reduced p62 contents compared to the MetS group indicating that memantine preserved hepatic autophagy. Histopathological examination revealed that memantine ameliorated hepatic steatosis and inflammation. Pioglitazone also mitigated most of the steatohepatitis-related changes, however, memantine was more effective in most of the studied parameters. SIGNIFICANCE The hepatoprotective effect of memantine against steatohepatitis is mediated, at least partly, through conserving hepatic autophagy along with anti-inflammatory, antioxidant, and anti-fibrotic effects.
Collapse
Affiliation(s)
- Esraa M Zakariaª
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; Department of Anatomy, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt.
| | - Rasha H Abdel-Ghanyª
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; Department of Anatomy, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Atef S Elgharbawyª
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; Department of Anatomy, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Amira Ebrahim Alsemehᵇ
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; Department of Anatomy, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Sami S Metwallyª
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; Department of Anatomy, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
6
|
Eating Compulsivity in Inpatients with Severe Obesity and the General Population: The Italian Version of the Measure of Eating Compulsivity (MEC10-IT). Nutrients 2023; 15:nu15061378. [PMID: 36986106 PMCID: PMC10052425 DOI: 10.3390/nu15061378] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
This study aimed to validate and investigate the psychometric properties of the Italian version of the Measure of Eating Compulsivity-10 (MEC10-IT) in a sample of inpatients with severe obesity (Study 1), and to test the measurement invariance of the measure across non-clinical and clinical samples (Study 2). In the first study, a confirmatory factorial analysis (CFA) was conducted among 452 patients in order to confirm the factorial structure of the MEC10-IT. In the second study, the psychometric properties of the MEC10-IT were tested on 453 inpatients with severe obesity and a community sample of 311 participants. The CFA confirmed the factorial structure of the MEC10-IT among an Italian sample of adult inpatients with severe obesity (Study 1). The MEC10-IT was also demonstrated to be invariant between the clinical and the community sample and to possess good psychometric properties, as well as excellent screening abilities for classifying individuals with problematic eating behaviors (Study 2). In conclusion, the MEC10-IT could be considered as a valid and reliable tool for the assessment of compulsive eating in both non-clinical and clinical samples and represents a psychometrically sound measure for clinical and research purposes.
Collapse
|
7
|
Aljuwaiser M, Alayadhi N, Ozidu V, Shafik Zakhari SA, Rushdy R, Naguy A. Clinical Indications of Memantine in Psychiatry-Science or Art? PSYCHOPHARMACOLOGY BULLETIN 2023; 53:30-38. [PMID: 36873917 PMCID: PMC9981340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Background Memenatine is USFDA approved for dementia of Alzheimer's disease. Apart from this indication, trend of its use in psychiatry is on the rise addressing a multitude of disorders. Study question Memantine remains one of only few psychotropic drugs with antiglutamate activity. This might impart it a therapeutic potential in treatment-resistant major psychiatric disorders characterized by neuroprogression. We reviewed memantine basic pharmacology and its diversifying clinical indications while examining the extant evidence. Methods EMBASE, Ovid MEDLINE, PubMed, Scopus, Web of Science, and Cochrane Database of Systemic Reviews were searched for all relevant studies up to date of November, 2022. Results Sound evidence supports use of memantine for major neuro-cognitive disorder due to Alzheimer's disease and severe vascular dementia, obsessive-compulsive disorder, treatment-resistant schizophrenia, and, ADHD. Modicum evidence supports use of memantine for PTSD, GAD and pathological gambling. Less compelling evidence is present for use in catatonia. No evidence supports use for core symptoms of autism spectrum disorder. Conclusions Memantine is an important addition to the psychopharmacological armamentarium. Level of evidence supporting the use of memantine in these off-label indications is highly variable, and hence, sound clinical judgment is necessary for its proper use and placement in real-life psychiatric practice and psychopharmacotherapy algorithms.
Collapse
Affiliation(s)
| | - Nadyah Alayadhi
- Alayadhi, BSc (Pharma, UK), MSc, PhD (UK), Head, Pharmacy Department, KCMH, Kuwait
| | - Victoria Ozidu
- Ozidu, MBBch, MSc, MRCPsych (UK), General Adult Psychiatrist, Greater Manchester Mental Health Trust, UK
| | | | - Reda Rushdy
- Rushdy, MD, PhD, Professor, and Chairperson, Psychiatric Departments, Azhar University, Egypt
| | - Ahmed Naguy
- Naguy, MBBch, MSc, Child/Adolescent Psychiatrist, Al-Manara CAP Centre, Kuwait Centre for Mental Health (KCMH), Jamal Abdul-Nassir St, Shuwaikh, State of Kuwait
| |
Collapse
|
8
|
Sketriene D, Battista D, Lalert L, Kraiwattanapirom N, Thai HN, Leeboonngam T, Knackstedt LA, Nithianantharajah J, Sumithran P, Lawrence AJ, Brown RM. Compulsive-like eating of high-fat high-sugar food is associated with 'addiction-like' glutamatergic dysfunction in obesity prone rats. Addict Biol 2022; 27:e13206. [PMID: 36001420 PMCID: PMC9541933 DOI: 10.1111/adb.13206] [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: 11/19/2021] [Revised: 04/13/2022] [Accepted: 06/22/2022] [Indexed: 11/29/2022]
Abstract
Chronic overeating is a core feature of diet-induced obesity. There is increasing evidence that in vulnerable individuals, such overeating could become compulsive, resembling an addictive disorder. The transition to compulsive substance use has been linked with changes at glutamatergic synapses in the nucleus accumbens. In this study, we investigated a potential link between such glutamatergic dysregulation and compulsive-like eating using a rat model of diet-induced obesity. A conditioned suppression task demonstrated that diet-induced obese rats display eating despite negative consequences, as their consumption was insensitive to an aversive cue. Moreover, nucleus accumbens expression of GluA1 and xCT proteins was upregulated in diet-induced obese animals. Lastly, both a computed 'addiction score' (based on performance across three criteria) and weight gain were positively correlated with changes in GluA1 and xCT expression in the nucleus accumbens. These data demonstrate that the propensity for diet-induced obesity is associated with compulsive-like eating of highly palatable food and is accompanied by 'addiction-like' glutamatergic dysregulation in the nucleus accumbens, thus providing neurobiological evidence of addiction-like pathology in this model of obesity.
Collapse
Affiliation(s)
- Diana Sketriene
- Mental Health Research ThemeThe Florey Institute of Neuroscience and Mental HealthParkvilleMelbourneAustralia
- The Florey Department of Neuroscience and Mental HealthUniversity of MelbourneParkvilleMelbourneAustralia
| | - Damien Battista
- Mental Health Research ThemeThe Florey Institute of Neuroscience and Mental HealthParkvilleMelbourneAustralia
| | - Laddawan Lalert
- Mental Health Research ThemeThe Florey Institute of Neuroscience and Mental HealthParkvilleMelbourneAustralia
- School of MedicineWalailak UniversityNakhon Si ThammaratThailand
| | - Natcharee Kraiwattanapirom
- Mental Health Research ThemeThe Florey Institute of Neuroscience and Mental HealthParkvilleMelbourneAustralia
- Institute of Molecular BiosciencesMahidol UniversityNakhon PathomThailand
| | - Han Ngoc Thai
- Mental Health Research ThemeThe Florey Institute of Neuroscience and Mental HealthParkvilleMelbourneAustralia
| | - Tanawan Leeboonngam
- Mental Health Research ThemeThe Florey Institute of Neuroscience and Mental HealthParkvilleMelbourneAustralia
- The Florey Department of Neuroscience and Mental HealthUniversity of MelbourneParkvilleMelbourneAustralia
| | | | - Jess Nithianantharajah
- Mental Health Research ThemeThe Florey Institute of Neuroscience and Mental HealthParkvilleMelbourneAustralia
| | - Priya Sumithran
- Department of Medicine (St Vincent's)University of MelbourneMelbourneAustralia
- Department of EndocrinologyAustin HealthMelbourneAustralia
| | - Andrew J. Lawrence
- Mental Health Research ThemeThe Florey Institute of Neuroscience and Mental HealthParkvilleMelbourneAustralia
| | - Robyn M. Brown
- Mental Health Research ThemeThe Florey Institute of Neuroscience and Mental HealthParkvilleMelbourneAustralia
- The Florey Department of Neuroscience and Mental HealthUniversity of MelbourneParkvilleMelbourneAustralia
- Department of Biochemistry and PharmacologyUniversity of MelbourneParkvilleMelbourneAustralia
| |
Collapse
|
9
|
The role of the nucleus accumbens and ventral pallidum in feeding and obesity. Prog Neuropsychopharmacol Biol Psychiatry 2021; 111:110394. [PMID: 34242717 DOI: 10.1016/j.pnpbp.2021.110394] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 05/31/2021] [Accepted: 06/29/2021] [Indexed: 02/04/2023]
Abstract
Obesity is a growing global epidemic that stems from the increasing availability of highly-palatable foods and the consequent enhanced calorie consumption. Extensive research has shown that brain regions that are central to reward seeking modulate feeding and evidence linking obesity to pathology in such regions have recently started to accumulate. In this review we focus on the contribution of two major interconnected structures central to reward processing, the nucleus accumbens and the ventral pallidum, to obesity. We first review the known literature linking these structures to feeding behavior, then discuss recent advances connecting pathology in the nucleus accumbens and ventral pallidum to obesity, and finally examine the similarities and differences between drug addiction and obesity in the context of these two structures. The understanding of how pathology in brain regions involved in reward seeking and consumption may drive obesity and how mechanistically similar obesity and addiction are, is only now starting to be revealed. We hope that future research will advance knowledge in the field and open new avenues to studying and treating obesity.
Collapse
|
10
|
Rehn S, Raymond JS, Boakes RA, Leenaars CHC. A systematic review and meta-analysis of animal models of binge eating - Part 1: Definitions and food/drink intake outcomes. Neurosci Biobehav Rev 2021; 132:1137-1156. [PMID: 34742923 DOI: 10.1016/j.neubiorev.2021.10.036] [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: 09/13/2021] [Revised: 10/19/2021] [Accepted: 10/28/2021] [Indexed: 12/09/2022]
Abstract
Binge eating involves consuming excessive amounts of food within a discrete period of time and is associated with significant impairments in binge-eating disorder and bulimia nervosa. While research on clinical binge eating has provided valuable aetiological insights, animal models allow for closer examination of environmental, biological, and developmental risk factors. Numerous animal models of binge eating exist and differ widely in operational definitions of bingeing, animal characteristics and methodological parameters. The current review aimed to synthesise the available published evidence on these models. A systematic review of binge definitions in 170 articles found most studies displayed good face validity. Meta-analyses on 150 articles confirmed that the amount of food or drink consumed by animals under binge conditions was larger than that of non-binge conditions across many protocols. The meta-regression revealed species, strain, and sex moderated binge effect size, with the largest effect observed in studies with female animals and mice. Risk of bias assessment identified that improved reporting of allocation, baseline characteristics and outcome assessment is required in future studies.
Collapse
Affiliation(s)
- Simone Rehn
- The University of Sydney, Faculty of Science, School of Psychology, Sydney, 2006 NSW, Australia.
| | - Joel S Raymond
- The University of Sydney, Faculty of Science, School of Psychology, Sydney, 2006 NSW, Australia; The University of Sydney, Brain and Mind Centre, 94 Mallett Street, Camperdown, Sydney, NSW 2050, Australia
| | - Robert A Boakes
- The University of Sydney, Faculty of Science, School of Psychology, Sydney, 2006 NSW, Australia
| | - Cathalijn H C Leenaars
- Institute for Laboratory Animal Science, Hannover Medical School, 30625, Hannover, Germany; Department for Health Evidence, Radboud University Medical Centre, 6600, Nijmegen, The Netherlands
| |
Collapse
|
11
|
Sahraei M, Sahraei H, Rahimi M, Khosravi M, Ganjkhani M, Meftahi GH. Anxiogenic and anxiolytic effects of memantine injected into the ventral hippocampus in male stressed mice. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2021; 19:581-589. [PMID: 34533006 DOI: 10.1515/jcim-2021-0159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 08/24/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES The effects of intra-ventral hippocampal memantine administration in male NMRI stressed mice were studied. METHODS Two stainless steel gauge 23 guide cannulas were placed in the middle part of the mice ventral hippocampus using stereotaxic coordination. Seven days later, the animals were undergone to the stress protocol as follows: They experience four consecutive electro-foot shock stress sessions lasting for 10 min. Five or 30 min before each stress session, the animals received intra-ventral hippocampal (0.1, 1 and, 5 µg/mouse) or intraperitoneal (1, 5, and 10 mg/kg) memantine respectively. Eight days after stress termination, the animals were tested either for the maintenance of either anxiety (elevated plus maze) or depression (forced swimming test). RESULTS Animals show anxiety eight days after stress termination. Intra-ventral hippocampal infusion of memantine (5 µg/mouse) 5 min before stress inhibited the anxiety-like behaviors. However, other doses of the drug exacerbate the stress effect. The drug, when injected peripherally exacerbated the stress effect in all doses. The drug by itself had no effect. In addition, animals also show depression nine days after stress termination and memantine (0.1, 1, and 5 µg/mouse) reduced the stress effect. The drug (0.1 µg/mouse) by itself induced depression in the animals. However, the drug when injected peripherally reduced the stress effect in all doses. CONCLUSIONS It could be concluded that NMDA glutamate receptors in the ventral hippocampus may play a pivotal role in the mediation of maintenance of anxiety and depression induced by stress in the mice.
Collapse
Affiliation(s)
- Mohammad Sahraei
- School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hedayat Sahraei
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Masoomeh Rahimi
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Khosravi
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mahin Ganjkhani
- Department of Physiology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | |
Collapse
|
12
|
Converging vulnerability factors for compulsive food and drug use. Neuropharmacology 2021; 196:108556. [PMID: 33862029 DOI: 10.1016/j.neuropharm.2021.108556] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/29/2021] [Accepted: 04/03/2021] [Indexed: 12/12/2022]
Abstract
Highly palatable foods and substance of abuse have intersecting neurobiological, metabolic and behavioral effects relevant for understanding vulnerability to conditions related to food (e.g., obesity, binge eating disorder) and drug (e.g., substance use disorder) misuse. Here, we review data from animal models, clinical populations and epidemiological evidence in behavioral, genetic, pathophysiologic and therapeutic domains. Results suggest that consumption of highly palatable food and drugs of abuse both impact and conversely are regulated by metabolic hormones and metabolic status. Palatable foods high in fat and/or sugar can elicit adaptation in brain reward and withdrawal circuitry akin to substances of abuse. Intake of or withdrawal from palatable food can impact behavioral sensitivity to drugs of abuse and vice versa. A robust literature suggests common substrates and roles for negative reinforcement, negative affect, negative urgency, and impulse control deficits, with both highly palatable foods and substances of abuse. Candidate genetic risk loci shared by obesity and alcohol use disorders have been identified in molecules classically associated with both metabolic and motivational functions. Finally, certain drugs may have overlapping therapeutic potential to treat obesity, diabetes, binge-related eating disorders and substance use disorders. Taken together, data are consistent with the hypotheses that compulsive food and substance use share overlapping, interacting substrates at neurobiological and metabolic levels and that motivated behavior associated with feeding or substance use might constitute vulnerability factors for one another. This article is part of the special issue on 'Vulnerabilities to Substance Abuse'.
Collapse
|
13
|
Adermark L, Gutierrez S, Lagström O, Hammarlund M, Licheri V, Johansson ME. Weight gain and neuroadaptations elicited by high fat diet depend on fatty acid composition. Psychoneuroendocrinology 2021; 126:105143. [PMID: 33493754 DOI: 10.1016/j.psyneuen.2021.105143] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 11/28/2022]
Abstract
Overconsumption of food is a major health concern in the western world. Palatable food has been shown to alter the activity of neural circuits, and obesity has been linked to alterations in the connectivity between the hypothalamus and cortical regions involved in decision-making and reward processing, putatively modulating the incentive value of food. Outlining neurophysiological adaptations induced by dietary intake of high fat diets (HFD) is thus valuable to establish how the diet by itself may promote overeating. To this end, C57BL/6 mice were fed HFD rich in either saturated fatty acids (HFD-S) or polyunsaturated fatty acids (HFD-P), or a low-fat control diet (LFD) for four weeks. Food and energy intake were monitored and ex vivo electrophysiology was employed to assess neuroadaptations in lateral hypothalamus (LH) and corticostriatal circuits, previously associated with food intake. In addition, the effects of dietary saturated and polyunsaturated fatty acids on the gene expression of NMDA, AMPA and GABAA receptor subunits in the hypothalamus were investigated. Our data shows that mice fed HFD-P had increased daily food and energy intake compared with mice fed HFD-S or LFD. However, this increase in energy intake had no obesogenic effects. Electrophysiological recordings demonstrated that HFD-P had a selective effect on glutamatergic neurotransmission in the LH, which was concomitant with a change in mRNA expression of AMPA receptor subtypes Gria1, Gria3 and Gria4, with no effect on the mRNA expression of NMDA receptor subtypes or GABAA receptor subtypes. Furthermore, while synaptic output from corticostriatal subregions was not significantly modulated by diet, synaptic plasticity in the form of long-term depression (LTD) was impaired in the dorsomedial striatum of mice fed HFD-S. In conclusion, this study suggests that the composition of fatty acids in the diet not only affects weight gain, but may also modulate neuronal function and plasticity in brain regions involved in food intake.
Collapse
Affiliation(s)
- Louise Adermark
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden; Deparment of Pharmacology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden.
| | - Saray Gutierrez
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Oona Lagström
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Maria Hammarlund
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Valentina Licheri
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Maria E Johansson
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| |
Collapse
|
14
|
Huang XT, Yang JX, Wang Z, Zhang CY, Luo ZQ, Liu W, Tang SY. Activation of N-methyl-D-aspartate receptor regulates insulin sensitivity and lipid metabolism. Theranostics 2021; 11:2247-2262. [PMID: 33500723 PMCID: PMC7797674 DOI: 10.7150/thno.51666] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/19/2020] [Indexed: 12/13/2022] Open
Abstract
RATIONALE Although significant progress has been made in understanding the mechanisms of steatosis and insulin resistance, the physiological functions of regulators in these processes remain largely elusive. Evidence has suggested that the glutamate/N-methyl-D-aspartic acid receptor (NMDAR) axis contributes to acute lung injury, pulmonary arterial hypertension, and diabetes, but the specific metabolic contribution of the glutamate/NMDAR axis is not clear. Here we provide data at the animal, cellular, and molecular levels to support the role of the glutamate/NMDAR axis as a therapeutic target for metabolic syndrome in obesity. Methods: We examined the glutamate level in the obese mouse induced by a high-fat diet (HFD) for 12 weeks. To assess the role of NMDAR in insulin sensitivity and lipid metabolism, we tested the effects of Memantine (an NMDAR antagonist) and NMDA (an NMDAR agonist) on mice fed with HFD or standard chow diet. The in vitros NMDAR roles were analyzed in hepatocytes and potential mechanisms involved in regulating lipid metabolism were investigated. Results: Glutamate was increased in the serum of HFD-treated mice. The NMDAR blockade by Memantine decreased the susceptibility to insulin resistance and hepatic steatosis in obese mice. NMDA treatment for 6 months induced obesity in mice, characterized by hyperglycemia, hyperlipidemia, insulin resistance, and pathological changes in the liver. We provided in vitro evidence demonstrating that NMDAR activation facilitated metabolic syndrome in obesity through promoting lipid accumulation. NMDAR inhibition attenuated lipid accumulation induced by palmitic acid. Mechanistically, NMDAR activation impaired fatty acid oxidation by reducing PPARα phosphorylation and activity. The PPARα activity reduction induced by NMDAR activation was reversibly mediated by ERK1/2 signaling. Conclusion: These findings revealed that targeting NMDAR might be a promising therapeutic strategy for metabolic syndrome in obesity.
Collapse
Affiliation(s)
- Xiao-Ting Huang
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Jun-Xiao Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zun Wang
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Zi-Qiang Luo
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Wei Liu
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Si-Yuan Tang
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| |
Collapse
|
15
|
Boswell RG, Potenza MN, Grilo CM. The Neurobiology of Binge-eating Disorder Compared with Obesity: Implications for Differential Therapeutics. Clin Ther 2021; 43:50-69. [PMID: 33257092 PMCID: PMC7902428 DOI: 10.1016/j.clinthera.2020.10.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 10/30/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE Emerging work indicates divergence in the neurobiologies of binge-eating disorder (BED) and obesity despite their frequent co-occurrence. This review highlights specific distinguishing aspects of BED, including elevated impulsivity and compulsivity possibly involving the mesocorticolimbic dopamine system, and discusses implications for differential therapeutics for BED. METHODS This narrative review describes epidemiologic, clinical, genetic, and preclinical differences between BED and obesity. Subsequently, this review discusses human neuroimaging work reporting differences in executive functioning, reward processing, and emotion reactivity in BED compared with obesity. Finally, on the basis of the neurobiology of BED, this review identifies existing and new therapeutic agents that may be most promising given their specific targets based on putative mechanisms of action relevant specifically to BED. FINDINGS BED is characterized by elevated impulsivity and compulsivity compared with obesity, which is reflected in divergent neurobiological characteristics and effective pharmacotherapies. Therapeutic agents that influence both reward and executive function systems may be especially effective for BED. IMPLICATIONS Greater attention to impulsivity/compulsivity-related, reward-related, and emotion reactivity-related processes may enhance conceptualization and treatment approaches for patients with BED. Consideration of these distinguishing characteristics and processes could have implications for more targeted pharmacologic treatment research and interventions.
Collapse
Affiliation(s)
- Rebecca G Boswell
- Yale School of Medicine, Department of Psychiatry, New Haven, CT, USA.
| | - Marc N Potenza
- Yale School of Medicine, Department of Psychiatry, New Haven, CT, USA; Connecticut Mental Health Center, New Haven, CT, USA; Connecticut Council on Problem Gambling, Wethersfield, CT, USA; Yale School of Medicine, Child Study Center, New Haven, CT, USA; Yale University, Department of Neuroscience, New Haven, CT, USA
| | - Carlos M Grilo
- Yale School of Medicine, Department of Psychiatry, New Haven, CT, USA; Yale University, Department of Psychology, New Haven, CT, USA
| |
Collapse
|
16
|
Galistu A, D'Aquila PS. Memantine effects on ingestion microstructure and the effect of administration time: A within-subject study. PLoS One 2020; 15:e0239270. [PMID: 32936829 PMCID: PMC7494107 DOI: 10.1371/journal.pone.0239270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/03/2020] [Indexed: 01/22/2023] Open
Abstract
In a between-subject comparison of two memantine administration schedules we observed that treatment with the NMDA receptor antagonist memantine before testing sessions reduced ingestion of a 10% sucrose solution in rats, due to reduced licking burst size, thus suggesting a blunted hedonic response. Conversely, daily post-session administration reduced burst number, indicating a reduced level of behavioural activation, likely due to the development of conditioned taste aversion (CTA). In this study, the effect of pre-session and post-session memantine administration was investigated within-subjects. Memantine was administered in daily intraperitoneal injections for 13 days, on alternate days, either 1-h before–“before testing” sessions—or immediately after a 30-min session–“after testing” sessions. The effects on the microstructure of licking for a 10% sucrose solution were examined in the course of treatment and for 21 days after treatment discontinuation. The results show reduced burst size in the “before testing” sessions, without effects on the intra-burst lick rate, an index of motoric effects. Moreover, burst number was reduced since the third session of both administration conditions until the end of treatment. Interestingly, the effect of memantine of reducing the activation of ingestive behaviour was less pronounced in this study with respect to that observed with the previous study post-session administration schedule, in spite of the longer treatment. This apparent paradox might be explained if one considers these effects as instances of a memory-related effect, such as the development of CTA. In the framework of this hypothesis, the “before testing” sessions, not being followed by memantine administration, can be considered as extinction sessions performed every other day. Moreover, the animals treated with memantine at the highest dose failed to recover to pre-treatment ingestion levels 21 days after treatment discontinuation, while the animals treated after testing sessions in the previously published study showed a complete recovery well before the 15th day test. Within the same interpretative framework, this might depend by the reduced number and frequency of the extinction trials—i.e. the number of the sessions run after treatment discontinuation—in the present study. These results provide further support to the conclusion that memantine administration before sessions reduce burst size, an effect which is likely due to blockade of NMDA receptors occurring during behavioural testing. The observation that this effect can be obtained even in absence of a reduced intra-burst lick rate, which rules out the involvement of motor impairment, provides an important piece of evidence in support to the interpretation of this effect as a blunted hedonic response. Moreover, these results provide further evidence that burst number reduction is due to a memory-related effect induced by memantine administration after sessions.
Collapse
Affiliation(s)
- Adriana Galistu
- Dipartimento di Scienze Biomediche, Università di Sassari, Sassari, Italy
| | - Paolo S D'Aquila
- Dipartimento di Scienze Biomediche, Università di Sassari, Sassari, Italy
| |
Collapse
|
17
|
Lee HS, Giunti E, Sabino V, Cottone P. Consummatory, Feeding Microstructural, and Metabolic Effects Induced by Limiting Access to Either a High-Sucrose or a High-Fat Diet. Nutrients 2020; 12:nu12061610. [PMID: 32486207 PMCID: PMC7352440 DOI: 10.3390/nu12061610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/24/2020] [Accepted: 05/28/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Binge eating disorder (BED) is characterized by recurrent binge eating episodes consisting of rapid consumption of excessive amounts of highly palatable, energy-dense food within discrete periods of time. The aim of this study was to test the consummatory, food microstructural, and metabolic effects of a one hour limited access to either a high-sucrose diet (HSD) or a high-fat diet (HFD) in an operant rat model of binge-like eating. Methods: Female rats were subject to a binge-like eating procedure in which a HSD, a HFD, or a standard chow diet were provided in a fixed ratio 1 (FR1) operant schedule of reinforcement. Results: Limiting access to either a HSD or a HFD promoted binge-like eating as compared to the control chow diet. However, binge-like eating of HSD, but not HFD, was based on a true increase in the amount of food consumed, an increased eating rate, and a decrease in the intake of the home-cage standard chow, altogether suggesting an increase in palatability. Moreover, while HSD rats consumed overall less energy than HFD rats, the former were more energy efficient and gained more body weight than the latter. Conclusions: These results provide information on how the quality of food can deeply influence the behavioral and metabolic outcomes of binge-like eating.
Collapse
|
18
|
Hicks C, Sabino V, Cottone P. The Alpha-1 Adrenergic Receptor Antagonist Prazosin Reduces Binge-Like Eating in Rats. Nutrients 2020; 12:nu12061569. [PMID: 32481494 PMCID: PMC7352795 DOI: 10.3390/nu12061569] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/18/2020] [Accepted: 05/26/2020] [Indexed: 01/19/2023] Open
Abstract
Background: Binge-eating disorder is a pervasive addiction-like disorder that is defined by excessive and uncontrollable consumption of food within brief periods of time. The aim of the current study was to examine the role of the brain noradrenergic system in binge-like eating through the use of the alpha-1 adrenergic receptor antagonist prazosin. Methods: For this purpose, we employed a limited access model whereby male Wistar rats were allowed to nosepoke for either chow (Chow rats) or a sugary, highly palatable food (Palatable rats) for 1 h/day. The effects of prazosin (0, 0.5, 1 and 2 mg/kg, i.p.) were tested in a fixed ratio 1 (FR1) and progressive ratio (PR) schedule of reinforcement. Results: The results show that prazosin preferentially reduced the responses for palatable food in a FR1 reinforcement schedule; when tested in a PR schedule of reinforcement, prazosin increased breakpoint in both Chow and Palatable rats, but more potently and more efficaciously in the latter. Our results suggest that prazosin treatment preferentially increased the motivational properties of the palatable diet. Conclusions: The current findings provide the characterization of the effects of prazosin on binge-like eating and offer support to the existing literature showing the important role of the noradrenergic system in addiction-like behavior.
Collapse
|
19
|
Ferragud A, Velázquez-Sánchez C, Abdullatif AA, Sabino V, Cottone P. Withdrawal from Extended, Intermittent Access to A Highly Palatable Diet Impairs Hippocampal Memory Function and Neurogenesis: Effects of Memantine. Nutrients 2020; 12:nu12051520. [PMID: 32456193 PMCID: PMC7284648 DOI: 10.3390/nu12051520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/14/2020] [Accepted: 05/21/2020] [Indexed: 12/21/2022] Open
Abstract
Background: Compulsive eating can be promoted by intermittent access to palatable food and is often accompanied by cognitive deficits and reduction in hippocampal plasticity. Here, we investigated the effects of intermittent access to palatable food on hippocampal function and neurogenesis. Methods: Male Wistar rats were either fed chow for 7 days/week (Chow/Chow group), or fed chow intermittently for 5 days/week followed by a palatable diet for 2 days/week (Chow/Palatable group). Hippocampal function and neurogenesis were assessed either during withdrawal or following renewed access to palatable food. Furthermore, the ability of the uncompetitive N-methyl-d-aspartate receptor (NMDAR) antagonist memantine to prevent the diet-induced memory deficits and block the maladaptive feeding was tested. Results: Palatable food withdrawn Chow/Palatable rats showed both a weakened ability for contextual spatial processing and a bias in their preference for a “novel cue” over a “novel place,” compared to controls. They also showed reduced expression of immature neurons in the dentate gyrus of the hippocampus as well as a withdrawal-dependent decrease of proliferating cells. Memantine treatment was able both to reverse the memory deficits and to reduce the excessive intake of palatable diet and the withdrawal-induced hypophagia in food cycling rats. Conclusions: In summary, our results provide evidence that withdrawal from highly palatable food produces NMDAR-dependent deficits in hippocampal function and a reduction in hippocampal neurogenesis.
Collapse
|
20
|
Moore CF, Leonard MZ, Micovic NM, Miczek KA, Sabino V, Cottone P. Reward sensitivity deficits in a rat model of compulsive eating behavior. Neuropsychopharmacology 2020; 45:589-596. [PMID: 31622973 PMCID: PMC7021808 DOI: 10.1038/s41386-019-0550-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/07/2019] [Accepted: 10/08/2019] [Indexed: 01/31/2023]
Abstract
Compulsive eating behavior is hypothesized to be driven in part by reward deficits likely due to neuroadaptations to the mesolimbic dopamine (DA) system. Therefore, the aim of this study was to assess deficits in reward system functioning and mesolimbic DA after alternating a standard chow with palatable diet, a model of compulsive eating. In this model, rats in the control group (Chow/Chow) are provided a standard chow diet 7 days a week, while the experimental group (Chow/Palatable) is provided chow for 5 days a week ("C Phase"), followed by 2 days of access to a highly palatable sucrose diet ("P Phase"). We first tested the sensitivity to d-Amphetamine's stimulatory, reward-enhancing, and primary rewarding effects using a locomotor activity assay, an intracranial self-stimulation (ICSS) procedure, and a conditioned place preference test, respectively. We then quantified DA release in the nucleus accumbens (NAc) shell after treatment with d-Amphetamine using in vivo microdialysis, quantified levels of tyrosine hydroxylase (TH) and dopamine transporter (DAT) mRNA using quantitative polymerase chain reaction (qPCR), and lastly, quantified baseline extracellular DA and function of DAT in vivo using quantitative "no-net-flux" microdialysis. Chow/Palatable rats displayed blunted d-Amphetamine-induced locomotor activity, insensitivity to d-Amphetamine potentiation of ICSS threshold, and decreased place preference for d-Amphetamine during the P Phase. We found that Chow/Palatable rats had blunted DA efflux following d-Amphetamine treatment. Furthermore, DAT mRNA was increased in Chow/Palatable rats during the P Phase. Finally, quantitative "no-net-flux" microdialysis revealed reduced extracellular baseline DA and DAT function in Chow/Palatable rats. Altogether, these results provide evidence of reduced reward system functioning and related neuroadaptations in the DA and DAT systems in this model of compulsive eating. Reward deficits, resulting from repeated overeating, may in turn contribute to the perpetuation of compulsive eating behavior.
Collapse
Affiliation(s)
- Catherine F. Moore
- 0000 0004 0367 5222grid.475010.7Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA USA ,0000 0004 0367 5222grid.475010.7Graduate Program for Neuroscience, Boston University School of Medicine, Boston, MA USA
| | - Michael Z. Leonard
- 0000 0004 1936 7531grid.429997.8Department of Psychology, Tufts University, Medford, MA USA
| | - Nicholas M. Micovic
- 0000 0004 0367 5222grid.475010.7Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA USA
| | - Klaus A. Miczek
- 0000 0004 1936 7531grid.429997.8Department of Psychology, Tufts University, Medford, MA USA ,0000 0004 1936 7531grid.429997.8Departments of Neuroscience, Psychiatry and Pharmacology, Tufts University, Boston, MA USA
| | - Valentina Sabino
- 0000 0004 0367 5222grid.475010.7Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA USA
| | - Pietro Cottone
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
21
|
Lisdexamfetamine suppresses instrumental and consummatory behaviors supported by foods with varying degrees of palatability: Exploration of a binge-like eating model. Pharmacol Biochem Behav 2020; 189:172851. [DOI: 10.1016/j.pbb.2020.172851] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/26/2019] [Accepted: 01/09/2020] [Indexed: 01/05/2023]
|
22
|
Daily memantine treatment blunts hedonic response to sucrose in rats. Psychopharmacology (Berl) 2020; 237:103-114. [PMID: 31414153 DOI: 10.1007/s00213-019-05348-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 08/09/2019] [Indexed: 02/07/2023]
Abstract
RATIONALE Preclinical and clinical studies suggest the potential use of memantine in the treatment of binge eating disorder. The aim of this study was to further investigate the mechanisms by which memantine influences the motivational aspects of ingestion through the analysis of licking microstructure. To interpret treatment effects in relation to drug action at specific functionally relevant times, we compared the effect of two different administration schedules. METHODS Memantine was administered daily for a week, either 1 h before or immediately after a 30-min daily session. The effects on the microstructure of licking for a 10% sucrose solution in rats were examined in the course of treatment and for 15 days after treatment discontinuation. RESULTS Treatment before testing reduced ingestion due to reduced burst size and increased latency in the first session. However, a progressive increase in burst number across sessions led to a full recovery of ingestion levels by the end of treatment. Daily post-session administration induced a dramatic decrease of activation of licking behaviour, indicated by reduced burst number, accompanied to reduced burst size. A slow recovery of ingestion took place after treatment discontinuation. CONCLUSION These results suggest a reduced hedonic/reward evaluation response, an effect likely due to NMDA receptor blockade occurring during the testing time and support the hypothesis that memantine interferes with the hedonic/non-homeostatic mechanisms regulating food intake and food-seeking. The effect of post-session administration might be explained by the development of conditioned taste aversion.
Collapse
|
23
|
Moore CF, Panciera JI, Sabino V, Cottone P. Neuropharmacology of compulsive eating. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0024. [PMID: 29352024 DOI: 10.1098/rstb.2017.0024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2017] [Indexed: 12/22/2022] Open
Abstract
Compulsive eating behaviour is a transdiagnostic construct observed in certain forms of obesity and eating disorders, as well as in the proposed construct of 'food addiction'. Compulsive eating can be conceptualized as comprising three elements: (i) habitual overeating, (ii) overeating to relieve a negative emotional state, and (iii) overeating despite adverse consequences. Neurobiological processes that include maladaptive habit formation, the emergence of a negative affect, and dysfunctions in inhibitory control are thought to drive the development and persistence of compulsive eating behaviour. These complex psychobehavioural processes are under the control of various neuropharmacological systems. Here, we describe the current evidence implicating these systems in compulsive eating behaviour, and contextualize them within the three elements. A better understanding of the neuropharmacological substrates of compulsive eating behaviour has the potential to significantly advance the pharmacotherapy for feeding-related pathologies.This article is part of a discussion meeting issue 'Of mice and mental health: facilitating dialogue between basic and clinical neuroscientists'.
Collapse
Affiliation(s)
- Catherine F Moore
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, 72 E. Concord Street, R-618, Boston, MA 02118, USA.,Graduate Program for Neuroscience, Boston University School of Medicine, 72 E. Concord Street, R-618, Boston, MA 02118, USA
| | - Julia I Panciera
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, 72 E. Concord Street, R-618, Boston, MA 02118, USA.,MS in Medical Sciences Program, Graduate Medical Sciences, Boston University School of Medicine, 72 E. Concord Street, R-618, Boston, MA 02118, USA.,Master of Public Health Program, Department of Health Policy and Management, Boston University School of Public Health, 715 Albany Street, Boston, MA, USA
| | - Valentina Sabino
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, 72 E. Concord Street, R-618, Boston, MA 02118, USA
| | - Pietro Cottone
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, 72 E. Concord Street, R-618, Boston, MA 02118, USA
| |
Collapse
|
24
|
Deng SN, Yan YH, Zhu TL, Ma BK, Fan HR, Liu YM, Li WG, Li F. Long-Term NMDAR Antagonism Correlates Weight Loss With Less Eating. Front Psychiatry 2019; 10:15. [PMID: 30800078 PMCID: PMC6375831 DOI: 10.3389/fpsyt.2019.00015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/10/2019] [Indexed: 12/12/2022] Open
Abstract
Memantine hydrochloride is an uncompetitive N-methyl-D-aspartate (NMDA) antagonist for treatment of moderate-to-severe Alzheimer's disease. Several studies have shown that memantine can significantly correct the binge-like eating behavior in human and animal models. People with overeating behavior are more likely to be obese. Therefore, we suppose that memantine would be a good candidate for the treatment of obesity. In this study, memantine was shown to increase weight loss in obese mice induced by high fat diet. Memantine was shown to decrease food intake without inducing abdominal discomfort and anxiety, suggesting that this compound would be a good candidate drug for obesity control.
Collapse
Affiliation(s)
- Shi-Ning Deng
- Developmental and Behavioral Pediatric Department and Child Primary Care Department, Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Shanghai Institute for Pediatric Research, Xinhua Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Hua Yan
- Key Laboratory of Brain Functional Genomics, Ministry of Education, East China Normal University, Shanghai, China
| | - Tai-Lin Zhu
- Key Laboratory of Brain Functional Genomics, Ministry of Education, East China Normal University, Shanghai, China
| | - Bing-Ke Ma
- Key Laboratory of Brain Functional Genomics, Ministry of Education, East China Normal University, Shanghai, China
| | - Hui-Ran Fan
- Developmental and Behavioral Pediatric Department and Child Primary Care Department, Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Shanghai Institute for Pediatric Research, Xinhua Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan-Mei Liu
- Developmental and Behavioral Pediatric Department and Child Primary Care Department, Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Shanghai Institute for Pediatric Research, Xinhua Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Guang Li
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Li
- Developmental and Behavioral Pediatric Department and Child Primary Care Department, Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Shanghai Institute for Pediatric Research, Xinhua Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
25
|
Abstract
Binge eating disorder is an addiction-like disorder characterized by recurrent, excessive food consumption within discrete periods of time, and it has been linked to increased trait impulsivity. Within impulsivity components, while impulsive action was shown to predict binge-like and addictive-like eating, the role of impulsive choice is instead unknown. The goal of this study was to determine if impulsive choice predicted, or was altered by binge-like eating of a sugary, highly palatable diet. We utilized a modified adjusting delay task procedure in free-fed rats to assess impulsive choice behavior, that is. the tendency to respond for a larger, delayed reward over a lesser, immediate reward. We found that baseline impulsive choice was not a predictor of binge-like eating in 1-h sessions of palatable diet operant self-administration. Furthermore, binge-like eating of the same palatable diet had no effect on later impulsive choice behavior. Thus, our data suggest that, unlike impulsive action, impulsive choice behavior does not predict binge-like eating in rats.
Collapse
|
26
|
Abstract
Eating disorders and some forms of obesity are characterized by addictive-like, compulsive eating behavior which contains numerous similarities with compulsive drug use. Food intake is in part mediated by reward and reinforcement processes that can become dysregulated in these disorders. Additionally, impairments in inhibitory control regulation of reward-related responding can cause or further exacerbate binge and compulsive eating. Dysfunctions in two neurotransmitter systems in the mesocorticolimbic pathway, dopamine and glutamate, are thought to contribute to maladaptive eating behaviors. The trace amine associated receptor 1 (TAAR1) system is a promising therapeutic target for compulsive eating behavior due to the role of TAAR1 in synaptic transmission and in the modulation of dopaminergic and glutamatergic signaling. In support of this notion, the TAAR1 agonist RO5256390 was found to decrease the reinforcing effects of palatable food-cues and to reduce binge-like and compulsive-like eating of palatable food. Additionally, prolonged, intermittent access to palatable food was shown to downregulate TAAR1 in the prefrontal cortex, suggesting a potential role for TAAR1 signaling in inhibitory control processes. Research into the role of TAAR1 in addiction, including TAAR1’s ability to modulate psychostimulant reward and reinforcement, bolsters support for TAAR1 agonism as a pharmacological treatment for compulsive eating and other addictive behaviors. This review summarizes the evidence for TAAR1 agonism as a promising therapeutic for compulsive eating behavior as well as the hypothesized mechanism responsible for these effects.
Collapse
Affiliation(s)
- Catherine F Moore
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, United States.,The Graduate Program for Neuroscience, Boston University School of Medicine, Boston, MA, United States
| | - Valentina Sabino
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, United States
| | - Pietro Cottone
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
27
|
A High-fat, High-sugar 'Western' Diet Alters Dorsal Striatal Glutamate, Opioid, and Dopamine Transmission in Mice. Neuroscience 2017; 372:1-15. [PMID: 29289718 DOI: 10.1016/j.neuroscience.2017.12.036] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/28/2017] [Accepted: 12/20/2017] [Indexed: 12/13/2022]
Abstract
Understanding neuroadaptations involved in obesity is critical for developing new approaches to treatment. Diet-induced neuroadaptations within the dorsal striatum have the capacity to drive excessive food seeking and consumption. Five-week-old C57BL/6J mice consumed a high-fat, high-sugar 'western diet' (WD) or a control 'standard diet' (SD) for 16 weeks. Weight gain, glucose tolerance, and insulin tolerance were measured to confirm an obese-like state. Following these 16 weeks, electrophysiological recordings were made from medium spiny neurons (MSNs) in the medial (DMS) and lateral (DLS) portions of dorsal striatum to evaluate diet effects on neuronal excitability and synaptic plasticity. In addition, fast-scan cyclic voltammetry evaluated dopamine transmission in these areas. WD mice gained significantly more weight and consumed more calories than SD mice and demonstrated impaired glucose tolerance. Electrophysiology data revealed that MSNs from WD mice demonstrated increased AMPA-to-NMDA receptor current ratio and prolonged spontaneous glutamate-mediated currents, specifically in the DLS. Evoked dopamine release was also significantly greater and reuptake slower in both subregions of WD striatum. Finally, dorsal striatal MSNs from WD mice were significantly less likely to demonstrate mu-opioid receptor-mediated synaptic plasticity. Neuronal excitability and GABAergic transmission were unaffected by diet in either striatal subregion. Our results demonstrate that a high-fat, high-sugar diet alters facets of glutamate, dopamine, and opioid signaling within the dorsal striatum, with some subregion specificity. These alterations within a brain area known to play a role in food motivation/consumption and habitual behavior are highly relevant for the clinical condition of obesity and its treatment.
Collapse
|
28
|
Moore CF, Schlain GS, Mancino S, Sabino V, Cottone P. A behavioral and pharmacological characterization of palatable diet alternation in mice. Pharmacol Biochem Behav 2017; 163:1-8. [PMID: 29097161 PMCID: PMC5911178 DOI: 10.1016/j.pbb.2017.10.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 10/09/2017] [Accepted: 10/29/2017] [Indexed: 12/28/2022]
Abstract
Obesity and eating disorders are widespread in Western societies. Both the increased availability of highly palatable foods and dieting are major risk factors contributing to the epidemic of disorders of feeding. The purpose of this study was to characterize an animal model of maladaptive feeding induced by intermittent access to a palatable diet alternation in mice. In this study, mice were either continuously provided with standard chow food (Chow/Chow), or provided with standard chow for 2days and a high-sucrose, palatable food for 1day (Chow/Palatable). Following stability of intake within the cycling paradigm, we then investigated the effects of several pharmacological treatments on excessive eating of palatable food: naltrexone, an opioid receptor antagonist, SR141716A, a cannabinoid-1 receptor antagonist/inverse agonist, and BD-1063, a sigma-1 receptor antagonist. Over successive cycles, Chow/Palatable mice showed an escalation of palatable food intake within the first hour of renewed access to palatable diet and displayed hypophagia upon its removal. Naltrexone, SR141716A, and BD-1063 all reduced overconsumption of palatable food during this first hour. Here we provide evidence of strong face and convergent validity in a palatable diet alternation model in mice, confirming multiple shared underlying mechanisms of pathological eating across species, and thus making it a useful therapeutic development tool.
Collapse
Affiliation(s)
- Catherine F Moore
- Laboratory of Addictive Disorders, Departments of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, USA; Graduate Program for Neuroscience, Boston University School of Medicine, Boston, MA, USA
| | - Gabrielle S Schlain
- Laboratory of Addictive Disorders, Departments of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Samantha Mancino
- Laboratory of Addictive Disorders, Departments of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Valentina Sabino
- Laboratory of Addictive Disorders, Departments of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Pietro Cottone
- Laboratory of Addictive Disorders, Departments of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
29
|
Moore CF, Sabino V, Koob GF, Cottone P. Neuroscience of Compulsive Eating Behavior. Front Neurosci 2017; 11:469. [PMID: 28883784 PMCID: PMC5573809 DOI: 10.3389/fnins.2017.00469] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/08/2017] [Indexed: 01/14/2023] Open
Abstract
A systematic characterization of compulsivity in pathological forms of eating has been proposed in the context of three functional domains: (1) habitual overeating; (2) overeating to relieve a negative emotional state; and (3) overeating despite aversive consequences. In this review, we provide evidence supporting this hypothesis and we differentiate the nascent field of neurocircuits and neurochemical mediators of compulsive eating through their underlying neuropsychobiological processes. A better understanding of the neurobiological mechanisms that lead to compulsive eating behavior can improve behavioral and pharmacological intervention for disorders of pathological eating.
Collapse
Affiliation(s)
- Catherine F Moore
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of MedicineBoston, MA, United States.,Graduate Program for Neuroscience, Boston University School of MedicineBoston, MA, United States
| | - Valentina Sabino
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of MedicineBoston, MA, United States
| | - George F Koob
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthBethesda, MD, United States
| | - Pietro Cottone
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of MedicineBoston, MA, United States
| |
Collapse
|
30
|
Abstract
Binge-eating disorder (BED) is the most prevalent eating disorder with estimates of 2-5% of the general adult population. Nonetheless, its pathophysiology is poorly understood. Furthermore, there exist few therapeutic options for its effective treatment. Here we review the current state of binge-eating neurobiology and pharmacology, drawing from clinical therapeutic, neuroimaging, cognitive, human genetic and animal model studies. These studies, which are still in their infancy, indicate that while there are many gaps in our knowledge, several key neural substrates appear to underpin binge-eating and may be conserved between human and animals. This observation suggests that behavioral intermediate phenotypes or endophenotypes relevant to BED may be modeled in animals, facilitating the identification and testing of novel pharmacological targets. The development of novel, safe and effective pharmacological therapies for the treatment of BED will enhance the ability of clinicians to provide optimal care for people with BED.
Collapse
Affiliation(s)
- Peter H Hutson
- Department of Neurobiology, CNS Discovery, Teva Pharmaceuticals, West Chester, PA, USA.
| | - Iris M Balodis
- Peter Boris Centre for Addiction Research, Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Marc N Potenza
- Department of Psychiatry, Child Study Center, Yale University School of Medicine, New Haven, CT, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA; National Center on Addiction and Substance Abuse, USA; Connecticut Mental Health Center, New Haven, CT, USA
| |
Collapse
|
31
|
Shariff M, Klenowski P, Morgan M, Patkar O, Mu E, Bellingham M, Belmer A, Bartlett SE. Binge-like sucrose consumption reduces the dendritic length and complexity of principal neurons in the adolescent rat basolateral amygdala. PLoS One 2017; 12:e0183063. [PMID: 28813474 PMCID: PMC5558950 DOI: 10.1371/journal.pone.0183063] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 07/28/2017] [Indexed: 11/18/2022] Open
Abstract
A compelling body of evidence suggests that the worldwide obesity epidemic is underpinned by excessive sugar consumption, typified by the modern western diet. Furthermore, evidence is beginning to emerge of maladaptive changes in the mesolimbic reward pathway of the brain in relation to excess sugar consumption that highlights the importance of examining this neural circuitry in an attempt to understand and subsequently mitigate the associated morbidities with obesity. While the basolateral amygdala (BLA) has been shown to mediate the reinforcing properties of drugs of abuse, it has also been shown to play an important role in affective and motivated behaviours and has been shown to undergo maladaptive changes in response to drugs of abuse and stress. Given the overlap in neural circuitry affected by drugs of abuse and sucrose, we sought to examine the effect of short- and long-term binge-like sucrose consumption on the morphology of the BLA principal neurons using an intermittent-access two-bottle choice paradigm. We used Golgi-Cox staining to impregnate principal neurons from the BLA of short- (4 week) and long-term (12 week) sucrose consuming adolescent rats and compared these to age-matched water controls. Our results indicate possibly maladaptive changes to the dendritic architecture of BLA principal neurons, particularly on apical dendrites following long-term sucrose consumption. Specifically, our results show reduced total dendritic arbor length of BLA principal neurons following short- and long-term sucrose consumption. Additionally, we found that long-term binge-like sucrose consumption caused a significant reduction in the length and complexity of apical dendrites. Taken together, our results highlight the differences between short- and long-term binge-like sucrose consumption on BLA principal neuron morphology and are suggestive of a perturbation in the diverse synaptic inputs to these neurons.
Collapse
Affiliation(s)
- Masroor Shariff
- Institute of Health and Biomedical Innovation at Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Paul Klenowski
- Institute of Health and Biomedical Innovation at Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Michael Morgan
- Institute of Health and Biomedical Innovation at Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Omkar Patkar
- Institute of Health and Biomedical Innovation at Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Erica Mu
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Mark Bellingham
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Arnauld Belmer
- Institute of Health and Biomedical Innovation at Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Selena E. Bartlett
- Institute of Health and Biomedical Innovation at Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
32
|
Cuesto G, Everaerts C, León LG, Acebes A. Molecular bases of anorexia nervosa, bulimia nervosa and binge eating disorder: shedding light on the darkness. J Neurogenet 2017; 31:266-287. [PMID: 28762842 DOI: 10.1080/01677063.2017.1353092] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Eating-disorders (EDs) consequences to human health are devastating, involving social, mental, emotional, physical and life-threatening aspects, concluding on impairment and death in cases of extreme anorexia nervosa. It also implies that people suffering an ED need to find psychiatric and psychological help as soon as possible to achieve a fully physical and emotional recovery. Unfortunately, to date, there is a crucial lack of efficient clinical treatment to these disorders. In this review, we present an overview concerning the actual pharmacological and psychological treatments, the knowledge of cells, circuits, neuropeptides, neuromodulators and hormones in the human brain- and other organs- underlying these disorders, the studies in animal models and, finally, the genetic approaches devoted to face this challenge. We will also discuss the need for new perspectives, avenues and strategies to be developed in order to pave the way to novel and more efficient therapeutics.
Collapse
Affiliation(s)
- Germán Cuesto
- a Centre for Biomedical Research of the Canary Islands , Institute of Biomedical Technologies, University of La Laguna , Tenerife , Spain
| | - Claude Everaerts
- b Centre des Sciences du Goût et de l'Alimentation , UMR 6265 CNRS, UMR 1324 INRA, Université de Bourgogne Franche-Comté , Dijon , France
| | - Leticia G León
- c Cancer Pharmacology Lab , AIRC Start Up Unit, University of Pisa , Pisa , Italy
| | - Angel Acebes
- a Centre for Biomedical Research of the Canary Islands , Institute of Biomedical Technologies, University of La Laguna , Tenerife , Spain
| |
Collapse
|
33
|
Camacho A, Montalvo-Martinez L, Cardenas-Perez RE, Fuentes-Mera L, Garza-Ocañas L. Obesogenic diet intake during pregnancy programs aberrant synaptic plasticity and addiction-like behavior to a palatable food in offspring. Behav Brain Res 2017; 330:46-55. [DOI: 10.1016/j.bbr.2017.05.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/30/2017] [Accepted: 05/04/2017] [Indexed: 02/07/2023]
|
34
|
Pathological Overeating: Emerging Evidence for a Compulsivity Construct. Neuropsychopharmacology 2017; 42:1375-1389. [PMID: 27922596 PMCID: PMC5436113 DOI: 10.1038/npp.2016.269] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 11/18/2016] [Accepted: 11/22/2016] [Indexed: 12/11/2022]
Abstract
Compulsive eating behavior is a transdiagnostic construct that is characteristic of medical and psychiatric conditions such as forms of obesity and eating disorders. Although feeding research is moving toward a better understanding of the proposed addictive properties of food, the components and the mechanisms contributing to compulsive eating are not yet clearly defined or understood. Current understanding highlights three elements of compulsive behavior as it applies to pathological overeating: (1) habitual overeating; (2) overeating to relieve a negative emotional state; and (3) overeating despite aversive consequences. These elements emerge through mechanisms involving pathological habit formation through an aberrant learning process, the emergence of a negative emotional state, and dysfunctions in behavioral control. Dysfunctions in systems within neurocircuitries that comprise the basal ganglia, the extended amygdala, and the prefrontal cortex result in compulsive eating behaviors. Here, we present evidence to relate compulsive eating behavior and addiction and to characterize their underlying neurobiological mechanisms. A major need to improve understanding of compulsive eating through the integration of complex motivational, emotional, and cognitive constructs is warranted.
Collapse
|
35
|
The Trace Amine-Associated Receptor 1 Agonist RO5256390 Blocks Compulsive, Binge-like Eating in Rats. Neuropsychopharmacology 2017; 42:1458-1470. [PMID: 27711047 PMCID: PMC5436108 DOI: 10.1038/npp.2016.233] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/28/2016] [Accepted: 09/30/2016] [Indexed: 01/20/2023]
Abstract
Compulsive, binge eating of highly palatable food constitutes a core feature of some forms of obesity and eating disorders, as well as of the recently proposed disorder of food addiction. Trace amine-associated receptor 1 (TAAR1) is a highly conserved G-protein-coupled receptor bound by endogenous trace amines. TAAR1 agonists have been shown to reduce multiple behavioral effects of drugs of abuse through their actions on the mesocorticolimbic system. In this study, we hypothesized that TAAR1 may have a role in compulsive, binge-like eating; we tested this hypothesis by assessing the effects of a TAAR1 agonist, RO5256390, in multiple excessive feeding-related behaviors induced by limiting access to a highly palatable diet in rats. Our results show that RO5256390 blocked binge-like eating in rats responding 1 h per day for a highly palatable sugary diet. Consistent with a palatability-selective effect, drug treatment selectively reduced the rate and regularity of palatable food responding, but it did not affect either baseline intake or food restriction-induced overeating of the standard chow diet. Furthermore, RO5256390 fully blocked compulsive-like eating when the palatable diet was offered in an aversive compartment of a light/dark conflict box, and blocked the conditioned rewarding properties of palatable food, as well as palatable food-seeking behavior in a second-order schedule of reinforcement. Drug treatment had no effect on either anxiety-like or depressive-like behavior, and it did not affect control performance in any of the tests. Importantly, rats exposed to palatable food showed decreased TAAR1 levels in the medial prefrontal cortex (mPFC), and RO5256390 microinfused into the infralimbic, but not prelimbic, subregion of the mPFC-reduced binge-like eating. Altogether, these results provide evidence for TAAR1 agonism as a novel pharmacological treatment for compulsive, binge eating.
Collapse
|
36
|
Kirkpatrick SL, Goldberg LR, Yazdani N, Babbs RK, Wu J, Reed ER, Jenkins DF, Bolgioni A, Landaverde KI, Luttik KP, Mitchell KS, Kumar V, Johnson WE, Mulligan MK, Cottone P, Bryant CD. Cytoplasmic FMR1-Interacting Protein 2 Is a Major Genetic Factor Underlying Binge Eating. Biol Psychiatry 2017; 81:757-769. [PMID: 27914629 PMCID: PMC5386810 DOI: 10.1016/j.biopsych.2016.10.021] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 09/14/2016] [Accepted: 10/04/2016] [Indexed: 01/11/2023]
Abstract
BACKGROUND Eating disorders are lethal and heritable; however, the underlying genetic factors are unknown. Binge eating is a highly heritable trait associated with eating disorders that is comorbid with mood and substance use disorders. Therefore, understanding its genetic basis will inform therapeutic development that could improve several comorbid neuropsychiatric conditions. METHODS We assessed binge eating in closely related C57BL/6 mouse substrains and in an F2 cross to identify quantitative trait loci associated with binge eating. We used gene targeting to validate candidate genetic factors. Finally, we used transcriptome analysis of the striatum via messenger RNA sequencing to identify the premorbid transcriptome and the binge-induced transcriptome to inform molecular mechanisms mediating binge eating susceptibility and establishment. RESULTS C57BL/6NJ but not C57BL/6J mice showed rapid and robust escalation in palatable food consumption. We mapped a single genome-wide significant quantitative trait locus on chromosome 11 (logarithm of the odds = 7.4) to a missense mutation in cytoplasmic FMR1-interacting protein 2 (Cyfip2). We validated Cyfip2 as a major genetic factor underlying binge eating in heterozygous knockout mice on a C57BL/6N background that showed reduced binge eating toward a wild-type C57BL/6J-like level. Transcriptome analysis of premorbid genetic risk identified the enrichment terms morphine addiction and retrograde endocannabinoid signaling, whereas binge eating resulted in the downregulation of a gene set enriched for decreased myelination, oligodendrocyte differentiation, and expression. CONCLUSIONS We identified Cyfip2 as a major significant genetic factor underlying binge eating and provide a behavioral paradigm for future genome-wide association studies in populations with increased genetic complexity.
Collapse
Affiliation(s)
- Stacey L. Kirkpatrick
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Department of Psychiatry, Boston University School of Medicine, Boston, MA USA
| | - Lisa R. Goldberg
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Department of Psychiatry, Boston University School of Medicine, Boston, MA USA,Graduate Program in Biomolecular Pharmacology, Boston University School of Medicine, Boston, MA USA
| | - Neema Yazdani
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Department of Psychiatry, Boston University School of Medicine, Boston, MA USA,Graduate Program in Biomolecular Pharmacology, Boston University School of Medicine, Boston, MA USA,Transformative Training Program in Addiction Science, Boston University
| | - R. Keith Babbs
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Department of Psychiatry, Boston University School of Medicine, Boston, MA USA
| | - Jiayi Wu
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Department of Psychiatry, Boston University School of Medicine, Boston, MA USA,Transformative Training Program in Addiction Science, Boston University,Ph.D. Program in Biomedical Sciences, Graduate Program in Genetics and Genomics, Boston University School of Medicine
| | - Eric R. Reed
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Department of Psychiatry, Boston University School of Medicine, Boston, MA USA,Ph.D. Program in Bioinformatics, Boston University, Boston, MA USA
| | - David F. Jenkins
- Ph.D. Program in Bioinformatics, Boston University, Boston, MA USA,Computational Biomedicine, Boston University School of Medicine, Boston, MA USA
| | - Amanda Bolgioni
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Department of Psychiatry, Boston University School of Medicine, Boston, MA USA,Graduate Program in Biomolecular Pharmacology, Boston University School of Medicine, Boston, MA USA
| | - Kelsey I. Landaverde
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Department of Psychiatry, Boston University School of Medicine, Boston, MA USA
| | - Kimberly P. Luttik
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Department of Psychiatry, Boston University School of Medicine, Boston, MA USA
| | - Karen S. Mitchell
- Department of Psychiatry, Boston University School of Medicine, Boston, MA USA
| | | | - W. Evan Johnson
- Computational Biomedicine, Boston University School of Medicine, Boston, MA USA
| | - Megan K. Mulligan
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN USA
| | - Pietro Cottone
- Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics and Department of Psychiatry, Boston University School of Medicine, Boston, MA USA
| | - Camron D. Bryant
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Department of Psychiatry, Boston University School of Medicine, Boston, MA USA,*Corresponding Author Camron D. Bryant, Ph.D., Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Department of Psychiatry, 72 E. Concord St., L-606C, Boston, MA 02118 USA, P: (617) 638-4489 F: (617) 638-4329
| |
Collapse
|
37
|
Yates JR, Gunkel BT, Rogers KK, Hughes MN, Prior NA. Effects of N-methyl-D-aspartate receptor ligands on sensitivity to reinforcer magnitude and delayed reinforcement in a delay-discounting procedure. Psychopharmacology (Berl) 2017; 234:461-473. [PMID: 27837332 PMCID: PMC5226882 DOI: 10.1007/s00213-016-4469-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 11/02/2016] [Indexed: 11/26/2022]
Abstract
RATIONALE The N-methyl-D-aspartate (NMDA) receptor has been recently identified as an important mediator of impulsive choice, as assessed in delay discounting. Although discounting is independently influenced by sensitivity to reinforcer magnitude and delayed reinforcement, few studies have examined how NMDA receptor ligands differentially affect these parameters. OBJECTIVES The current study examined the effects of various NMDA receptor ligands on sensitivity to reinforcer magnitude and delayed reinforcement in a delay-discounting procedure. METHODS Following behavioral training, rats received treatments of the following NMDA receptor ligands: the uncompetitive antagonists ketamine (0, 1.0, 5.0, or 10.0 mg/kg; i.p.), MK-801 (0, 0.003, 0.01, or 0.03 mg/kg; s.c.), and memantine (0, 2.5, 5.0, or 10.0 mg/kg; i.p.), the competitive antagonist CGS 19755 (0, 5.0, 10.0, or 20.0 mg/kg; s.c.), the non-competitive NR2B subunit-selective antagonist ifenprodil (0, 1.0, 3.0, or 10.0 mg/kg; i.p), and the partial agonist D-cycloserine (0, 3.25, 15.0, or 30.0 mg/kg; s.c.). RESULTS When an exponential model was used to describe discounting, CGS 19755 (5.0 mg/kg) increased impulsive choice without altering sensitivity to reinforcer magnitude. Conversely, ketamine (10.0 mg/kg), memantine (5.0 mg/kg), and ifenprodil (10.0 mg/kg) decreased sensitivity to reinforcer magnitude without altering impulsive choice. MK-801 and D-cycloserine did not alter delay-discounting performance, although two-way ANOVA analyses indicated D-cycloserine (15.0 mg/kg) decreased impulsive choice. CONCLUSIONS The behavioral changes observed in delay discounting following administration of NMDA receptor antagonists do not always reflect an alteration in impulsive choice. These results emphasize the utility in employing quantitative methods to assess drug effects in delay discounting.
Collapse
Affiliation(s)
- Justin R Yates
- Department of Psychological Science, Northern Kentucky University, 1 Nunn Drive, Highland Heights, KY, 41099, USA.
| | - Benjamin T Gunkel
- Department of Psychological Science, Northern Kentucky University, 1 Nunn Drive, Highland Heights, KY, 41099, USA
| | - Katherine K Rogers
- Department of Psychological Science, Northern Kentucky University, 1 Nunn Drive, Highland Heights, KY, 41099, USA
| | - Mallory N Hughes
- Department of Psychological Science, Northern Kentucky University, 1 Nunn Drive, Highland Heights, KY, 41099, USA
| | - Nicholas A Prior
- Department of Psychological Science, Northern Kentucky University, 1 Nunn Drive, Highland Heights, KY, 41099, USA
| |
Collapse
|
38
|
Kraft TT, Huang D, LaMagna S, Warshaw D, Natanova E, Sclafani A, Bodnar RJ. Acquisition and expression of fat-conditioned flavor preferences are differentially affected by NMDA receptor antagonism in BALB/c and SWR mice. Eur J Pharmacol 2017; 799:26-32. [PMID: 28132914 DOI: 10.1016/j.ejphar.2017.01.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 01/24/2017] [Accepted: 01/25/2017] [Indexed: 10/20/2022]
Abstract
Conditioned flavor preferences are elicited by fat (Intralipid) in inbred mouse strains with BALB/c and SWR mice displaying among the most robust preferences. Dopamine D1 and opioid receptor antagonism differentially reduces the acquisition (learning) and expression (maintenance) of fat-conditioned flavor preferences in these two strains. Because noncompetitive NMDA receptor antagonism with MK-801 differentially altered sugar-conditioned flavor preferences in these strains, and because NMDA receptors are involved in fat intake, the present study examined whether MK-801 differentially altered expression and acquisition of fat (Intralipid)-conditioned flavor preferences in BALB/c and SWR mice. In expression studies, food-restricted male mice alternately consumed a flavored (CS+, e.g., cherry, 5 sessions) 5% Intralipid solution and a differently-flavored (CS-, e.g., grape, 5 sessions) 0.5% Intralipid solution. Two-bottle CS choice tests occurred following vehicle or MK-801 (100, 200µg/kg). MK-801 blocked expression of Intralipid-CFP at both doses in BALB/c mice, but only at the 100µg/kg dose in SWR mice. In acquisition studies, groups of BALB/c (0, 100µg/kg) and SWR (0, 100µg/kg) male mice were treated prior to the ten acquisition training sessions followed by six 2-bottle CS choice tests without injections. MK-801 eliminated acquisition of Intralipid-conditioned flavor preferences in BALB/c mice, and actually changed the preference to an avoidance response in SWR mice. Thus, NMDA receptor signaling appears essential especially for the learning of fat-conditioned flavor preferences in both mouse strains.
Collapse
Affiliation(s)
- Tamar T Kraft
- CUNY Neuroscience Collaborative, CUNY Graduate Center, New York, NY, USA
| | - Donald Huang
- Department of Psychology, Queens College, CUNY, New York, NY, USA
| | - Sam LaMagna
- Department of Psychology, Queens College, CUNY, New York, NY, USA
| | - Deena Warshaw
- Department of Psychology, Queens College, CUNY, New York, NY, USA
| | - Elona Natanova
- Department of Psychology, Queens College, CUNY, New York, NY, USA
| | - Anthony Sclafani
- CUNY Neuroscience Collaborative, CUNY Graduate Center, New York, NY, USA; Department of Psychology, Brooklyn College, CUNY, New York, NY, USA
| | - Richard J Bodnar
- CUNY Neuroscience Collaborative, CUNY Graduate Center, New York, NY, USA; Department of Psychology, Queens College, CUNY, New York, NY, USA.
| |
Collapse
|
39
|
Diepenbroek C, Rijnsburger M, Eggels L, van Megen K, Ackermans M, Fliers E, Kalsbeek A, Serlie M, la Fleur S. Infusion of fluoxetine, a serotonin reuptake inhibitor, in the shell region of the nucleus accumbens increases blood glucose concentrations in rats. Neurosci Lett 2017; 637:85-90. [DOI: 10.1016/j.neulet.2016.11.045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/16/2016] [Accepted: 11/22/2016] [Indexed: 12/15/2022]
|
40
|
Hopf FW. Do specific NMDA receptor subunits act as gateways for addictive behaviors? GENES BRAIN AND BEHAVIOR 2016; 16:118-138. [PMID: 27706932 DOI: 10.1111/gbb.12348] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/27/2016] [Accepted: 10/03/2016] [Indexed: 12/19/2022]
Abstract
Addiction to alcohol and drugs is a major social and economic problem, and there is considerable interest in understanding the molecular mechanisms that promote addictive drives. A number of proteins have been identified that contribute to expression of addictive behaviors. NMDA receptors (NMDARs), a subclass of ionotropic glutamate receptors, have been of particular interest because their physiological properties make them an attractive candidate for gating induction of synaptic plasticity, a molecular change thought to mediate learning and memory. NMDARs are generally inactive at the hyperpolarized resting potentials of many neurons. However, given sufficient depolarization, NMDARs are activated and exhibit long-lasting currents with significant calcium permeability. Also, in addition to stimulating neurons by direct depolarization, NMDARs and their calcium signaling can allow strong and/or synchronized inputs to produce long-term changes in other molecules (such as AMPA-type glutamate receptors) which can last from days to years, binding internal and external stimuli in a long-term memory trace. Such memories could allow salient drug-related stimuli to exert strong control over future behaviors and thus promote addictive drives. Finally, NMDARs may themselves undergo plasticity, which can alter subsequent neuronal stimulation and/or the ability to induce plasticity. This review will address recent and past findings suggesting that NMDAR activity promotes drug- and alcohol-related behaviors, with a particular focus on GluN2B subunits as possible central regulators of many addictive behaviors, as well as newer studies examining the importance of non-canonical NMDAR subunits and endogenous NMDAR cofactors.
Collapse
Affiliation(s)
- F W Hopf
- Alcohol and Addiction Research Group, Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| |
Collapse
|
41
|
|
42
|
Chen ZZ, Yang DD, Zhao Z, Yan H, Ji J, Sun XL. Memantine mediates neuroprotection via regulating neurovascular unit in a mouse model of focal cerebral ischemia. Life Sci 2016; 150:8-14. [PMID: 26920629 DOI: 10.1016/j.lfs.2016.02.081] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 02/12/2016] [Accepted: 02/22/2016] [Indexed: 01/15/2023]
Abstract
AIMS Memantine is a low-moderate affinity and uncompetitive N-methyl-d-aspartate receptor (NMDAR) antagonist, which is also a potential neuroprotectant in acute ischemic stroke for its particular action profiles. The present study was to reveal the mechanisms involved in the neuroprotection of memantine. MAIN METHODS We used a mouse model of permanent focal cerebral ischemia via middle cerebral artery occlusion to verify our hypothesis. 2,3,5-Triphenyltetrazolium chloride staining was used to compare infarct size. The amount of astrocytes and the somal volume of the microglia cell body were analyzed by immunohistochemistry and stereological estimates. Western blotting was used to determine the protein expressions. KEY FINDINGS Memantine prevented cerebral ischemia-induced brain infarct and neuronal injury, and reduced oxygen-glucose deprivation-induced cortical neuronal apoptosis. Moreover, memantine reduced the amount of the damaged astrocytes and over activated microglia after 24h of ischemia. In the early phase of ischemia, higher production of MMP-9 was observed, and thereby collagen IV was dramatically disrupted. Meanwhile, the post-synaptic density protein 95(PSD-95) was also severely cleavaged. Memantine decreased MMP-9 secretion, prevented the degradation of collagen IV in mouse brain. PSD-95 cleavage was also inhibited by memantine. SIGNIFICANCE These results suggested that memantine exerted neuroprotection effects in acute ischemic brain damage, partially via improving the functions of neurovascular unit. Taking all these findings together, we consider that memantine might be a promising protective agent against ischemic stroke.
Collapse
Affiliation(s)
- Zheng-Zhen Chen
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, PR China
| | - Dan-Dan Yang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, PR China
| | - Zhan Zhao
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, PR China
| | - Hui Yan
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, PR China
| | - Juan Ji
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, PR China
| | - Xiu-Lan Sun
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, PR China.
| |
Collapse
|
43
|
Abstract
Binge eating disorder is characterized by excessive, uncontrollable consumption of palatable food within brief periods of time. Excessive intake of palatable food is thought to be driven by hedonic, rather than energy homeostatic, mechanisms. However, reward processing does not only comprise consummatory actions; a key component is represented by the anticipatory phase directed at procuring the reward. This phase is highly influenced by environmental food-associated stimuli, which can robustly enhance the desire to eat even in the absence of physiological needs. The opioid system (endogenous peptides and their receptors) has been strongly linked to the rewarding aspects of palatable food intake, and perhaps represents the key system involved in hedonic overeating. Here we review evidence suggesting that the opioid system can also be regarded as one of the systems that regulates the anticipatory incentive processes preceding binge eating hedonic episodes.
Collapse
|
44
|
Pitman KA, Borgland SL. Changes in mu-opioid receptor expression and function in the mesolimbic system after long-term access to a palatable diet. Pharmacol Ther 2015. [DOI: 10.1016/j.pharmthera.2015.07.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
45
|
Lardeux S, Kim JJ, Nicola SM. Intermittent-access binge consumption of sweet high-fat liquid does not require opioid or dopamine receptors in the nucleus accumbens. Behav Brain Res 2015; 292:194-208. [PMID: 26097003 DOI: 10.1016/j.bbr.2015.06.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 06/04/2015] [Accepted: 06/07/2015] [Indexed: 12/31/2022]
Abstract
Binge eating disorders are characterized by episodes of intense consumption of high-calorie food. In recently developed animal models of binge eating, rats given intermittent access to such food escalate their consumption over time. Consumption of calorie-dense food is associated with neurochemical changes in the nucleus accumbens, including dopamine release and alterations in dopamine and opioid receptor expression. Therefore, we hypothesized that binge-like consumption on intermittent access schedules is dependent on opioid and/or dopamine neurotransmission in the accumbens. To test this hypothesis, we asked whether injection of dopamine and opioid receptor antagonists into the core and shell of the accumbens reduced consumption of a sweet high-fat liquid in rats with and without a history of intermittent binge access to the liquid. Although injection of a μ opioid agonist increased consumption, none of the antagonists (including μ opioid, δ opioid, κ opioid, D1 dopamine and D2 dopamine receptor antagonists, as well as the broad-spectrum opioid receptor antagonist naltrexone) reduced consumption, and this was the case whether or not the animals had a prior history of intermittent access. These results suggest that consumption of sweet, fatty food does not require opioid or dopamine receptor activation in the accumbens even under intermittent access conditions that resemble human binge episodes.
Collapse
Affiliation(s)
- Sylvie Lardeux
- Department of Psychiatry, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - James J Kim
- Department of Psychiatry, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Saleem M Nicola
- Department of Psychiatry, Albert Einstein College of Medicine, Bronx, NY 10461, United States; Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, United States.
| |
Collapse
|