Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Psychol Med ; 54(6): 1184-1195, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37920985

ABSTRACT

BACKGROUND: Some preliminary research suggests higher rates of gastrointestinal disease in individuals with eating disorders (EDs). However, research is limited, and it remains unknown what etiologic factors account for observed associations. This was the first study to examine how EDs and dimensional ED symptoms (e.g. body dissatisfaction, binge eating) are phenotypically and etiologically associated with gastrointestinal disease in a large, population-based twin sample. METHODS: Adult female (N = 2980) and male (N = 2903) twins from the Michigan State University Twin Registry reported whether they had a lifetime ED (anorexia nervosa, bulimia nervosa, or binge-eating disorder) and completed a measure of dimensional ED symptoms. We coded the presence/absence of lifetime gastrointestinal disease (e.g. inflammatory bowel disease) based on responses to questions regarding chronic illnesses and medications. We first examined whether twins with gastrointestinal disease had higher rates of EDs and ED symptoms, then used correlated factors twin models to investigate genetic and environmental contributions to the overlap between disorders. RESULTS: Twins with gastrointestinal disease had significantly greater dimensional ED symptoms (ß = 0.21, p < 0.001) and odds of a lifetime ED (OR 2.90, p = 0.001), regardless of sex. Shared genetic factors fully accounted for the overlap between disorders, with no significant sex differences in etiologic associations. CONCLUSIONS: Comorbidity between EDs and gastrointestinal disease may be explained by overlap in genetic influences, potentially including inflammatory genes implicated in both types of disorders. Screening for gastrointestinal disease in people with EDs, and EDs in those with gastrointestinal disease, is warranted.


Subject(s)
Anorexia Nervosa , Binge-Eating Disorder , Bulimia Nervosa , Feeding and Eating Disorders , Gastrointestinal Diseases , Adult , Humans , Female , Male , Feeding and Eating Disorders/epidemiology , Feeding and Eating Disorders/genetics , Bulimia Nervosa/epidemiology , Bulimia Nervosa/genetics , Bulimia Nervosa/diagnosis , Anorexia Nervosa/genetics , Binge-Eating Disorder/genetics , Gastrointestinal Diseases/epidemiology , Gastrointestinal Diseases/genetics
2.
Psychoneuroendocrinology ; 158: 106384, 2023 12.
Article in English | MEDLINE | ID: mdl-37708824

ABSTRACT

The heritability of eating disorder (ED) symptoms increases dramatically across gonadarche in girls. Past studies suggest these developmental differences could be due to pubertal activation of estrogen, but findings have been limited to only one ED symptom (i.e., binge eating). The current study examined whether estrogen contributes to gonadarcheal differences in genetic influences on overall levels of ED symptoms as well as key cognitive symptoms (i.e., weight/shape concerns) that are present across all EDs and are early risk factors for eating pathology. Given that binge eating frequently co-occurs with all of these symptoms, analyses also examined whether estrogen effects exist for overall levels of ED symptoms and body weight/shape concerns after accounting for the known effects of estrogen on genetic risk for binge eating. Participants included 964 female twins (ages 8-16) from the Michigan State University Twin Registry. Overall levels of ED symptoms were assessed with the Minnesota Eating Behavior Survey (MEBS) total score. Weight/shape concerns were assessed with a latent factor modeled using subscales from the MEBS and the Eating Disorder Examination Questionnaire. Estradiol levels were assessed with saliva samples. Twin moderation models were used to examine whether genetic influences on overall levels of ED symptoms and weight/shape concerns differed significantly across estradiol levels. Although initial models suggested modest differences in genetic influences on overall levels of ED symptoms across estradiol levels, these effects were eliminated when binge eating was accounted for in the models. In addition, weight/shape concerns did not show significant moderation of genetic influences by estradiol in models with or without binge eating. Taken together, results are significant in suggesting that individual differences in estradiol levels during gonadarche have a unique and specific impact on genetic risk for binge eating, while other etiologic factors must contribute to increased heritability of cognitive ED symptoms during this key developmental period in girls.


Subject(s)
Binge-Eating Disorder , Feeding and Eating Disorders , Female , Humans , Binge-Eating Disorder/genetics , Estradiol , Estrogens , Feeding Behavior , Child , Adolescent
4.
Nat Genet ; 55(9): 1462-1470, 2023 09.
Article in English | MEDLINE | ID: mdl-37550530

ABSTRACT

Binge eating disorder (BED) is the most common eating disorder, yet its genetic architecture remains largely unknown. Studying BED is challenging because it is often comorbid with obesity, a common and highly polygenic trait, and it is underdiagnosed in biobank data sets. To address this limitation, we apply a supervised machine-learning approach (using 822 cases of individuals diagnosed with BED) to estimate the probability of each individual having BED based on electronic medical records from the Million Veteran Program. We perform a genome-wide association study of individuals of African (n = 77,574) and European (n = 285,138) ancestry while controlling for body mass index to identify three independent loci near the HFE, MCHR2 and LRP11 genes and suggest APOE as a risk gene for BED. We identify shared heritability between BED and several neuropsychiatric traits, and implicate iron metabolism in the pathophysiology of BED. Overall, our findings provide insights into the genetics underlying BED and suggest directions for future translational research.


Subject(s)
Binge-Eating Disorder , Humans , Binge-Eating Disorder/genetics , Binge-Eating Disorder/psychology , Genome-Wide Association Study , Obesity/genetics , Phenotype , Iron
5.
Int J Mol Sci ; 23(23)2022 Dec 03.
Article in English | MEDLINE | ID: mdl-36499556

ABSTRACT

Recurrent Binge Eating (BE) episodes characterize several eating disorders. Here, we attempted to reassemble a condition closer to BE disorder, and we analyzed whether recurrent episodes might evoke molecular alterations in the hypothalamus of rats. The hypothalamus is a brain region which is sensitive to stress and relevant in motivated behaviors, such as food intake. A well-characterized animal model of BE, in which a history of intermittent food restriction and stress induce binge-like palatable food consumption, was used to analyze the transcriptional regulation of the endocannabinoid system (ECS). We detected, in rats showing the BE behavior, an up-regulated gene expression of cannabinoid type-1 receptor (CB1), sn-1-specific diacylglycerol lipase, as well as fatty acid amide hydrolase (Faah) and monoacylglycerol lipase. A selective reduction in DNA methylation was also observed at the promoter of Faah, which is consistent with the changes in the gene expression. Moreover, BE behavior in rats was associated with an increase in anandamide (AEA) levels. Our findings support the relevant role of the ECS in the regulation of food intake in rats subjected to repeated BE episodes, and, in particular, on AEA signaling, acting via CB1 and FAAH modulation. Notably, the epigenetic regulation of the Faah gene might suggest this enzyme as a possible target for developing new therapeutical approaches.


Subject(s)
Binge-Eating Disorder , Rats , Female , Animals , Binge-Eating Disorder/genetics , Epigenesis, Genetic , Endocannabinoids/metabolism , Amidohydrolases/genetics , Amidohydrolases/metabolism , Monoacylglycerol Lipases/genetics , Monoacylglycerol Lipases/metabolism , Receptors, Cannabinoid/metabolism , Hypothalamus/metabolism , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Eating
6.
J Prev Med Hyg ; 63(2 Suppl 3): E297-E305, 2022 Jun.
Article in English | MEDLINE | ID: mdl-36479493

ABSTRACT

Eating disorders such as anorexia nervosa, bulimia nervosa and binge-eating disorder, have a deep social impact, concluding with death in cases of severe disease. Eating disorders affect up to 5% of the population in the industrialized countries, but probably the phenomenon is under-detection and under-diagnosis. Eating disorders are multifactorial disorders, resulting from the interaction between environmental triggers, psychological factors, but there is also a strong genetic component. In fact, genetic factors predispose for approximately 33-84% to anorexia nervosa, 28-83% to bulimia nervosa, and 41-57% to binge eating disorder. Twins and family studies have provided an unassailable proof on the heritability of these disorders. Other types of genetic studies, including genome-wide association studies, whole genome sequencing and linkage analysis, allowed to identify the genes and their variants associated with eating disorders and moreover global collaborative efforts have led to delineate the etiology of these disorders. Next Generation Sequencing technologies can be considered as an ideal diagnostic approach to identify not only the common variants, such as single nucleotide polymorphism, but also rare variants. Here we summarize the present knowledge on the molecular etiology and genetic determinants of eating disorders including serotonergic genes, dopaminergic genes, opioid genes, appetite regulation genes, endocannabinoid genes and vitamin D3.


Subject(s)
Anorexia Nervosa , Binge-Eating Disorder , Bulimia Nervosa , Humans , Binge-Eating Disorder/genetics , Developed Countries , Genome-Wide Association Study , Anorexia Nervosa/genetics , Bulimia Nervosa/genetics
7.
Clin Epigenetics ; 14(1): 108, 2022 08 30.
Article in English | MEDLINE | ID: mdl-36042529

ABSTRACT

BACKGROUND: The neuropeptide oxytocin (OXT) plays a role in the regulation of eating behavior and metabolism. OXT functioning is altered in patients with eating and weight disorders, and a variant of the oxytocin receptor gene (OXTR) has been associated with impulsive eating behavior as it is seen in patients with binge eating disorder (BED). Gene × environment interactions could play a role in BED. One mechanism mediating this interaction is the epigenetic alteration of gene expression. We therefore investigated if DNA methylation of the OXTR differs between individuals with obesity depending on a comorbid BED. We analyzed DNA methylation of the OXTR in peripheral blood of 227 individuals on the obesity spectrum (mean age: 40.3 ± 13.1 yrs; mean BMI: 38.6 ± 7.3 kg/m2), 130 of which were diagnosed with BED. RESULTS: There were no overall differences in OXTR methylation between participants with and those without BED (p > 0.05), while both subgroups were comparable regarding age and body mass index (BMI), but significantly differed in sex distribution (p = 0.035). We found no relationship between mean DNA methylation and BMI or self-reported eating disorder (ED) pathology. Analyzing potential sex differences revealed a significantly lower OXTR DNA methylation in male participants with BED as compared to those without BED (p = 0.017). No such difference was found in the female subsample (p > 0.05). CONCLUSIONS: Clinically significant binge eating pathology might be associated with lower OXTR DNA methylation exclusively in males. The differential DNA methylation of OXTR in males with BED supports the view that BED represents a phenotype within the obesity spectrum that is characterized by specific vulnerability factors. A better understanding of the epigenetic underpinnings of the OXT system might contribute to the refinement of OXT administration approaches as potential interventions in eating and weight disorders.


Subject(s)
Binge-Eating Disorder , Receptors, Oxytocin , Binge-Eating Disorder/genetics , DNA Methylation , Female , Humans , Male , Obesity/genetics , Oxytocin , Receptors, Oxytocin/genetics , Receptors, Oxytocin/metabolism
8.
Int J Eat Disord ; 55(8): 1031-1041, 2022 08.
Article in English | MEDLINE | ID: mdl-35502471

ABSTRACT

OBJECTIVE: Using preliminary data from the Binge-Eating Genetics Initiative (BEGIN), we evaluated the feasibility of delivering an eating disorder digital app, Recovery Record, through smartphone and wearable technology for individuals with binge-type eating disorders. METHODS: Participants (n = 170; 96% female) between 18 and 45 years old with lived experience of binge-eating disorder or bulimia nervosa and current binge-eating episodes were recruited through the Recovery Record app. They were randomized into a Watch (first-generation Apple Watch + iPhone) or iPhone group; they engaged with the app over 30 days and completed baseline and endpoint surveys. Retention, engagement, and associations between severity of illness and engagement were evaluated. RESULTS: Significantly more participants in the Watch group completed the study (p = .045); this group had greater engagement than the iPhone group (p's < .05; pseudo-R2 McFadden effect size = .01-.34). Overall, binge-eating episodes, reported for the previous 28 days, were significantly reduced from baseline (mean = 12.3) to endpoint (mean = 6.4): most participants in the Watch (60%) and iPhone (66%) groups reported reduced binge-eating episodes from baseline to endpoint. There were no significant group differences across measures of binge eating. In the Watch group, participants with fewer episodes of binge eating at baseline were more engaged (p's < .05; pseudo-R2 McFadden  = .01-.02). Engagement did not significantly predict binge eating at endpoint nor change in binge-eating episodes from baseline to endpoint for both the Watch and iPhone groups. DISCUSSION: Using wearable technology alongside iPhones to deliver an eating disorder app may improve study completion and app engagement compared with using iPhones alone.


Subject(s)
Binge-Eating Disorder , Bulimia Nervosa , Adolescent , Adult , Binge-Eating Disorder/diagnosis , Binge-Eating Disorder/genetics , Feasibility Studies , Female , Humans , Male , Middle Aged , Smartphone , Surveys and Questionnaires , Young Adult
9.
Eat Weight Disord ; 27(4): 1505-1512, 2022 May.
Article in English | MEDLINE | ID: mdl-34478125

ABSTRACT

AIM: The aim of this study was to analyze the association and susceptibility of Single Nucleotide Polymorphisms (SNPs) in the DRD2 and BDNF genes with BED in patients with weight regain in the postoperative period of bariatric surgery. METHODS: One hundred and seventy-seven individuals who underwent bariatric surgery with weight regain were evaluated and divided into two groups according to the BED diagnostic. The individuals were submitted to an anthropometric evaluation, analysis of the presence of BED using a validated questionnaire, and blood collection for genotyping of the polymorphisms rs6265 (BDNF) and rs1800497 (DRD2) by real-time polymerase chain reaction (RT-PCR). RESULTS: The presence of wild-type alleles for rs1800497 (CC) and rs6265 (GG) was more frequent in patients without BED. Nevertheless, the presence of one or two variant alleles for rs1800497 (CT + TT) and rs6265 (GA + AA) was more frequent in patients with BED. The combination of the two studied SNPs prevailed in patients with BED. CONCLUSIONS: The presence of allele frequency of rs1800497 SNP in the DRD2 gene and rs6265 SNP in the BDNF gene, isolated and/or combined, indicated an additional risk for the development of BED in patients with obesity, especially in the context of weight regain. LEVEL OF EVIDENCE: III (evidence obtained from the case-control analytic study).


Subject(s)
Bariatric Surgery , Binge-Eating Disorder , Binge-Eating Disorder/genetics , Brain-Derived Neurotrophic Factor/genetics , Humans , Polymorphism, Single Nucleotide , Receptors, Dopamine D2/genetics , Weight Gain/genetics
10.
Nutrients ; 13(3)2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33807560

ABSTRACT

The genetic polymorphisms involved in the physiopathology of binge eating disorder (BED) are currently unclear. This systematic review aims to highlight and summarize the research on polymorphisms that is conducted in the BED. We looked for observational studies where there was a genetic comparison between adults with BED, in some cases also with obesity or overweight, and healthy controls or obesity/overweight without BED. Our protocol was written using PRISMA. It is registered at PROSPERO (identification: CRD42020198645). To identify potentially relevant documents, the following bibliographic databases were searched without a time limit, but until September 2020: PubMed, PsycINFO, Scopus, and Web of Science. In total, 21 articles were included in the qualitative analysis of the systematic review, as they met the eligibility criteria. Within the selected studies, 41 polymorphisms of 17 genes were assessed. Overall, this systematic review provides a list of potentially useful genetic polymorphisms involved in BED: 5-HTTLPR (5-HTT), Taq1A (ANKK1/DRD2), A118G (OPRM1), C957T (DRD2), rs2283265 (DRD2), Val158Met (COMT), rs6198 (GR), Val103Ile (MC4R), Ile251Leu (MC4R), rs6265 (BNDF), and Leu72Met (GHRL). It is important to emphasize that Taq1A is the polymorphism that showed, in two different research groups, the most significant association with BED. The remaining polymorphisms need further evidence to be confirmed.


Subject(s)
Binge-Eating Disorder/genetics , Genetic Predisposition to Disease/genetics , Obesity/genetics , Overweight/genetics , Polymorphism, Genetic , Adolescent , Adult , Female , Humans , Male , Observational Studies as Topic , Young Adult
11.
Nutrients ; 13(5)2021 Apr 22.
Article in English | MEDLINE | ID: mdl-33922358

ABSTRACT

Binge-eating disorder, recently accepted as a diagnostic category, is differentiated from bulimia nervosa in that the former shows the presence of binge-eating episodes and the absence of compensatory behavior. Epigenetics is a conjunct of mechanisms (like DNA methylation) that regulate gene expression, which are dependent on environmental changes. Analysis of DNA methylation in eating disorders shows that it is reduced. The present study aimed to analyze the genome-wide DNA methylation differences between individuals diagnosed with BED and BN. A total of 46 individuals were analyzed using the Infinium Methylation EPIC array. We found 11 differentially methylated sites between BED- and BN-diagnosed individuals, with genome-wide significance. Most of the associations were found in genes related to metabolic processes (ST3GAL4, PRKAG2, and FRK), which are hypomethylated genes in BED. Cg04781532, located in the body of the PRKAG2 gene (protein kinase AMP-activated non-catalytic subunit gamma 2), was hypomethylated in individuals with BED. Agonists of PRKAG2, which is the subunit of AMPK (AMP-activated protein kinase), are proposed to treat obesity, BED, and BN. The present study contributes important insights into the effect that BED could have on PRKAG2 activation.


Subject(s)
Binge-Eating Disorder/diagnosis , Binge-Eating Disorder/genetics , DNA Methylation/genetics , Metabolism/genetics , Adolescent , Anorexia Nervosa/diagnosis , Anorexia Nervosa/genetics , Female , Humans , Male , Pilot Projects
12.
Neuropharmacology ; 196: 108556, 2021 09 15.
Article in English | MEDLINE | ID: mdl-33862029

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'.


Subject(s)
Binge-Eating Disorder/physiopathology , Brain/physiopathology , Food Addiction/physiopathology , Obesity/physiopathology , Substance-Related Disorders/physiopathology , Animals , Binge-Eating Disorder/genetics , Binge-Eating Disorder/metabolism , Brain/metabolism , Food Addiction/genetics , Food Addiction/metabolism , Genetic Predisposition to Disease , Humans , Obesity/genetics , Obesity/metabolism , Reinforcement, Psychology , Reward , Risk Factors , Substance-Related Disorders/genetics , Substance-Related Disorders/metabolism
13.
Int J Eat Disord ; 54(5): 785-793, 2021 05.
Article in English | MEDLINE | ID: mdl-33644868

ABSTRACT

OBJECTIVE: Genome-wide association studies have identified multiple genomic regions associated with anorexia nervosa. No genome-wide studies of other eating disorders, such as bulimia nervosa and binge-eating disorder, have been performed, despite their substantial heritability. Exploratively, we aimed to identify traits that are genetically associated with binge-type eating disorders. METHOD: We calculated genome-wide polygenic scores for 269 trait and disease outcomes using PRSice v2.2 and their association with anorexia nervosa, bulimia nervosa, and binge-eating disorder in up to 640 cases and 17,050 controls from the UK Biobank. Significant associations were tested for replication in the Avon Longitudinal Study of Parents and Children (up to 217 cases and 3,018 controls). RESULTS: Individuals with binge-type eating disorders had higher polygenic scores than controls for other psychiatric disorders, including depression, schizophrenia, and attention deficit hyperactivity disorder, and higher polygenic scores for body mass index. DISCUSSION: Our findings replicate some of the known comorbidities of eating disorders on a genomic level and motivate a deeper investigation of shared and unique genomic factors across the three primary eating disorders.


Subject(s)
Anorexia Nervosa , Binge-Eating Disorder , Bulimia Nervosa , Feeding and Eating Disorders , Anorexia Nervosa/diagnosis , Anorexia Nervosa/genetics , Binge-Eating Disorder/diagnosis , Binge-Eating Disorder/genetics , Bulimia Nervosa/diagnosis , Bulimia Nervosa/genetics , Child , Genome-Wide Association Study , Genomics , Humans , Longitudinal Studies
14.
Gene ; 781: 145538, 2021 May 20.
Article in English | MEDLINE | ID: mdl-33631245

ABSTRACT

BACKGROUND: The genetics of binge-eating disorder (BED) is an emerging topic, with dopaminergic genes being implicated in its etiology due to the role that dopamine (DA) plays in food reward sensitivity and self-regulation of eating behavior. However, no study to date has examined if DA genes influence response to behavioral treatment of BED. OBJECTIVE: The primary objective of this study was to examine the ability of DA-associated polymorphisms to predict BED treatment response measured using binge frequency over 12 months. As secondary objectives, this study examined cross-sectional relationships between these polymorphisms and anthropometrics in women living with and without BED and obesity. METHODS: Women aged 18-64 years old were genotyped for the DA-related SNPs DRD2/ANKK1 Taq1A (rs1800497) and COMT (rs4680), as well as the DA-related uVNTRs DAT-1 (SLC6A3) and MAO-A. A multi-locus DA composite score was formed from these 4 polymorphisms using genotypes known to have a functional impact resulting in modified DA signaling. Binge frequency (Eating Disorder Examination - Interview) and body composition (Tanita BC-418) were assessed in a pre-post analysis to examine genetic predictors of treatment response in women living with obesity and BED. Secondary data analysis was conducted on a cross-sectional comparison of three groups of women enrolled in trial group treatment for BED: women living with obesity and BED (n = 72), obesity without BED (n = 27), and normal-weight without BED (n = 45). RESULTS: There were no significant genotype × time interactions related to anthropometrics or binge frequency for any individual DA genotypes, or to the composite score reflecting DA availability. At baseline, there were no significant between-group differences in frequencies of DA-related alleles, nor were there associations between genotypes and anthropometrics. CONCLUSIONS: Our study found no evidence to suggest that the DRD2/ANKK1 Taq1A, COMT, MAO-A, or DAT-1 polymorphisms are associated with response to behavioral intervention for BED as measured by changes in binge frequency. Future studies should examine a greater variety of dopaminergic polymorphisms, other candidate genes that target other neurotransmitter systems, as well as examine their impact on both behavioral and pharmacological-based treatment for BED.


Subject(s)
Binge-Eating Disorder/genetics , Dopamine/genetics , Polymorphism, Genetic , Adolescent , Adult , Binge-Eating Disorder/metabolism , Catechol O-Methyltransferase/genetics , Cross-Sectional Studies , Female , Genotype , Humans , Middle Aged , Monoamine Oxidase/genetics , Protein Serine-Threonine Kinases/genetics , Receptors, Dopamine D2/genetics , Young Adult
15.
Nutr Res ; 83: 86-93, 2020 11.
Article in English | MEDLINE | ID: mdl-33038759

ABSTRACT

Binge eating disorder (BED) is known as the most common eating disorder with both psychosocial and biological factors involved. In this regard, there is a need to recognize probable disturbances in substances involved in food intake regulation in BED. In this study, we hypothesized that the levels of endocannabinoids, fatty acid amid hydrolase (FAAH) gene polymorphisms, and appetite regulatory substances are different in overweight and obese women with and without BED. A Binge Eating Scale was used to estimate the prevalence of BED in 180 women classified as overweight or obese. The levels of anandamide (AEA), 2-arachidonoylglycerol (2-AG), leptin, insulin, and orexin-A were measured by enzyme-linked immunosorbent assay kits. The subjects were genotyped for polymorphisms of FAAH gene using amplification refractory mutation system-polymerase chain reaction. Data were analyzed using SPSS software. About 41.6% (n = 75) of the subjects were diagnosed with BED. Women with BED exhibited significantly higher levels of AEA, 2-AG, leptin, and insulin compared to non-BED women (P < .05). Binary logistic regression analysis also showed that AEA, leptin, and insulin were the predictors of having BED after adjusting for body mass index (P < .05). In addition, the frequency of A allele of FAAH gene was higher in women with BED compared to women without BED; however, there were no significant differences between these 2 groups (P = .08). These results supported our hypothesis in the cases of AEA, 2-AG, leptin, and insulin but not orexin and FAAH gene polymorphisms. The findings of the current study provide further evidence concerning the role of these substances in BED.


Subject(s)
Amidohydrolases/genetics , Binge-Eating Disorder/genetics , Binge-Eating Disorder/metabolism , Endocannabinoids/blood , Adult , Arachidonic Acids/blood , Body Mass Index , Cross-Sectional Studies , Female , Genotype , Glycerides/blood , Humans , Insulin/blood , Leptin/blood , Obesity/genetics , Obesity/metabolism , Orexins/blood , Overweight/genetics , Overweight/metabolism , Polymorphism, Genetic , Polyunsaturated Alkamides/blood
16.
BMC Psychiatry ; 20(1): 307, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32546136

ABSTRACT

BACKGROUND: The Binge Eating Genetics Initiative (BEGIN) is a multipronged investigation examining the interplay of genomic, gut microbiota, and behavioral factors in bulimia nervosa and binge-eating disorder. METHODS: 1000 individuals who meet current diagnostic criteria for bulimia nervosa or binge-eating disorder are being recruited to collect saliva samples for genotyping, fecal sampling for microbiota characterization, and recording of 30 days of passive data and behavioral phenotyping related to eating disorders using the app Recovery Record adapted for the Apple Watch. DISCUSSION: BEGIN examines the interplay of genomic, gut microbiota, and behavioral factors to explore etiology and develop predictors of risk, course of illness, and response to treatment in bulimia nervosa and binge-eating disorder. We will optimize the richness and longitudinal structure of deep passive and active phenotypic data to lay the foundation for a personalized precision medicine approach enabling just-in-time interventions that will allow individuals to disrupt eating disorder behaviors in real time before they occur. TRIAL REGISTRATION: The ClinicalTrials.gov identifier is NCT04162574. November 14, 2019, Retrospectively Registered.


Subject(s)
Binge-Eating Disorder , Bulimia Nervosa , Bulimia , Feeding and Eating Disorders , Binge-Eating Disorder/genetics , Bulimia/genetics , Bulimia Nervosa/genetics , Feeding Behavior , Humans
17.
Appetite ; 150: 104678, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32209386

ABSTRACT

Binge eating is a heritable symptom of eating disorders with an unknown genetic etiology. Rodent models for binge-like eating (BLE) of palatable food permit the study of genetic and biological mechanisms. We previously genetically mapped a coding mutation in Cyfip2 associated with increased BLE of sweetened palatable food in the C57BL/6NJ versus C57BL/6J substrain. The increase in BLE in C57BL/6NJ mice was associated with a decrease in transcription of genes enriched for myelination in the striatum. Here, we tested the hypothesis that decreasing myelin levels with the demyelinating agent cuprizone would enhance BLE. Mice were treated with a 0.3% cuprizone home cage diet for two weeks. Cuprizone induced similar weight loss in both substrains and sexes that recovered within 48 h after removal of cuprizone. Following a three-week recovery period, mice were trained for BLE in an intermittent, limited access procedure. Surprisingly, cuprizone significantly reduced BLE in male but not female C57BL/6NJ mice while having no effect in C57BL/6J mice. Cuprizone also reduced myelin basic protein (MBP) at seven weeks post-cuprizone removal while having no effect on myelin-associated glycoprotein at this time point. C57BL/6NJ mice also showed less MBP than C57BL/6J mice. There were no statistical interactions of Treatment with Sex on MBP levels, indicating that differences in MBP reduction are unlikely to account for sex differences in BLE. To summarize, cuprizone induced an unexpected, significant reduction in BLE in C57BL/6NJ males, which could indicate genotype-dependent sex differences in the biological mechanisms of BLE.


Subject(s)
Binge-Eating Disorder/drug therapy , Cuprizone/pharmacology , Myelin Sheath/drug effects , Nerve Tissue Proteins/pharmacology , Sex Characteristics , Animals , Binge-Eating Disorder/genetics , Corpus Striatum/metabolism , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL
18.
Sci Rep ; 9(1): 15709, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31673099

ABSTRACT

Identifying genetic variants that regulate binge eating (BE) is critical for understanding the factors that control this behavior and for the development of pharmacological treatment strategies. Although several studies have revealed specific genes capable of affecting BE behavior, less is known about how genetic variation modulates BE. Thus, through a paradigm that promoted binge-like food intake through intermittent access to high calorie diet (HCD), we quantified food-intake in four inbred mouse strains: C57Bl/6J (B6), NOD/LtJ (NOD), 129S1/SvlmJ (S1), and A/J (AJ). We report that genetic variation likely influences the chronic regulation of food intake and the binge-like consumption of a palatable HCD. AJ mice consumed more of both standard chow and HCD than the other three strains tested when both diets were available ad libitum, while S1 mice consumed significantly less HCD than other strains during intermittent HCD access. Behavioral differences were also associated with differential changes in c-Fos immunohistochemistry in brain regions traditionally associated with appetite regulation. Our results identify 129S1/SvlmJ as a strain that exhibits low levels of binge feeding behavior and suggests that this strain could be useful in the investigation of the influence of genetic variation in the control of binge food intake.


Subject(s)
Feeding Behavior , Genetic Variation , Animals , Binge-Eating Disorder/genetics , Disease Models, Animal , Female , Mice , Mice, Inbred Strains
19.
Expert Rev Neurother ; 19(9): 867-879, 2019 09.
Article in English | MEDLINE | ID: mdl-31269819

ABSTRACT

Introduction: Clinical and genetic study of psychiatric conditions has underscored the co-occurrence of complex phenotypes and the need to refine them. Bipolar Disorder (BD) and Binge Eating (BE) behavior are common psychiatric conditions that have high heritability and high co-occurrence, such that at least one quarter of BD patients have BE (BD + BE). Genetic studies of BD alone and of BE alone suggest complex polygenic risk models, with many genetic risk loci yet to be identified. Areas covered: We review studies of the epidemiology of BD+BE, its clinical features (cognitive traits, psychiatric comorbidity, and role of obesity), genomic studies (of BD, eating disorders (ED) defined by BE, and BD + BE), and therapeutic implications of BD + BE. Expert opinion: Subphenotyping of complex psychiatric disorders reduces heterogeneity and increases statistical power and effect size; thus, it enhances our capacity to find missing genetic (and other) risk factors. BD + BE has a severe clinical picture and genetic studies suggests a distinct genetic architecture. Differential therapeutic interventions may be needed for patients with BD + BE compared with BD patients without BE. Recognizing the BD + BE subphenotype is an example of moving towards more precise clinical and genetic entities.


Subject(s)
Binge-Eating Disorder , Bipolar Disorder , Comorbidity , Binge-Eating Disorder/epidemiology , Binge-Eating Disorder/genetics , Binge-Eating Disorder/physiopathology , Bipolar Disorder/epidemiology , Bipolar Disorder/genetics , Bipolar Disorder/physiopathology , Humans
20.
J Psychopharmacol ; 33(12): 1550-1561, 2019 12.
Article in English | MEDLINE | ID: mdl-31161847

ABSTRACT

BACKGROUND: Pharmacological treatment approaches for eating disorders, such as binge eating disorder and bulimia nervosa, are currently limited. METHODS AND AIMS: Using a well-characterized animal model of binge eating, we investigated the epigenetic regulation of the A2A Adenosine Receptor (A2AAR) and dopaminergic D2 receptor (D2R) genes. RESULTS: Gene expression analysis revealed a selective increase of both receptor mRNAs in the amygdaloid complex of stressed and restricted rats, which exhibited binge-like eating, when compared to non-stressed and non-restricted rats. Consistently, pyrosequencing analysis revealed a significant reduction of the percentage of DNA methylation but only at the A2AAR promoter region in rats showing binge-like behaviour compared to the control animals. Focusing thus on A2AAR agonist (VT 7) administration (which inhibited the episode of binge systemically at 0.1 mg/kg or intra-central amygdala (CeA) injection at 900 ng/side) induced a significant increase of A2AAR mRNA levels in restricted and stressed rats when compared to the control group. In addition, we observed a significant decrease in A2AAR mRNA levels in rats treated with the A2AAR antagonist (ANR 94) at 1 mg/kg. Consistent changes in the DNA methylation status of the A2AAR promoter were found in restricted and stressed rats after administration of VT 7 or ANR 94. CONCLUSION: We confirm the role of A2AAR in binge eating behaviours, and we underline the importance of epigenetic regulation of the A2AAR gene, possibly due to a compensatory mechanism to counteract the effect of binge eating. We suggest that A2AAR activation, inducing receptor gene up-regulation, could be relevant to reduction of food consumption.


Subject(s)
Binge-Eating Disorder/genetics , Bulimia/genetics , Receptor, Adenosine A2A/genetics , Receptors, Dopamine D2/genetics , Adenine/analogs & derivatives , Adenine/pharmacology , Amygdala/metabolism , Animals , Binge-Eating Disorder/physiopathology , DNA Methylation/genetics , Disease Models, Animal , Epigenesis, Genetic , Female , Gene Expression Regulation , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...