Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Psychiatry ; 27(2): 947-955, 2022 02.
Article in English | MEDLINE | ID: mdl-34719691

ABSTRACT

Neurological and psychiatric disorders, including substance use disorders, share a range of symptoms, which could be the result of shared genetic background. Many genetic loci have been identified for these disorders using genome-wide association studies, but conclusive evidence about cell types wherein these loci are active is lacking. We aimed to uncover implicated brain cell types in neuropsychiatric traits and to assess consistency in results across RNA datasets and methods. We therefore comprehensively employed cell type enrichment methods by integrating single-cell transcriptomic data from mouse brain regions with an unprecedented dataset of 42 human genome-wide association study results of neuropsychiatric, substance use and behavioral/quantitative brain-related traits (n = 12,544,007 individuals). Single-cell transcriptomic datasets from the Karolinska Institute and 10x Genomics were used. Cell type enrichment was determined using Linkage Disequilibrium Score Regression, Multi-marker Analysis of GenoMic Annotation, and Data-driven Expression Prioritized Integration for Complex Traits. We found the largest degree of consistency across methods for implication of pyramidal cells in schizophrenia and cognitive performance. For other phenotypes, such as bipolar disorder, two methods implicated the same cell types, i.e., medium spiny neurons and pyramidal cells. For autism spectrum disorders and anorexia nervosa, no consistency in implicated cell types was observed across methods. We found no evidence for astrocytes being consistently implicated in neuropsychiatric traits. In conclusion, we provide comprehensive evidence for a subset of neuronal cell types being consistently implicated in several, but not all psychiatric disorders, while non-neuronal cell types seem less implicated.


Subject(s)
Bipolar Disorder , Schizophrenia , Animals , Bipolar Disorder/genetics , Genome-Wide Association Study , Humans , Mice , Neurons , RNA-Seq , Schizophrenia/genetics
2.
Tijdschr Psychiatr ; 64(5): 301-305, 2022.
Article in Dutch | MEDLINE | ID: mdl-35735041

ABSTRACT

BACKGROUND: Translational (genetic) research focuses on the translation of preclinical research into practice. While many genetic studies have been conducted in recent years, the results do not simply translate to the clinic.
AIM: To visualize the steps through which translational genetic research contributes to the unraveling of the biological backgrounds of psychiatric disorders, in particular of eating disorders.
METHOD: Literature review.
RESULTS: Genetic studies have unraveled a mechanism underlying the hunger and satiety system. There is hope that genome-wide studies of eating disorders will lead to identification of neural circuits in which associated genes cluster. New techniques, such as opto- and chemogenetics, provide the opportunity to define the precise role of these circuits in eating disorders.
CONCLUSION: New techniques in molecular neuroscience allow the unravelling of the complexity of how the brain works and some of those techniques (such as chemogenetics) are being further developed for application in humans. However, it will be years before we can definitively translate this into the treatment of psychiatric disorders.
.


Subject(s)
Feeding and Eating Disorders , Motivation , Brain , Feeding and Eating Disorders/genetics , Feeding and Eating Disorders/therapy , Humans
3.
Int J Obes (Lond) ; 42(4): 655-661, 2018 04.
Article in English | MEDLINE | ID: mdl-29089612

ABSTRACT

OBJECTIVE: To explore the association between emotion-driven impulsiveness, cognitive inflexibility, decision-making and weight status as reflected in body mass index (BMI) z-score (zBMI) in European adolescents. METHODS: In total, 3354 adolescents aged between 12 and 18 years from the I.Family cohort completed the questionnaire-based negative urgency subscale from the UPPS-P Impulsive Behavior Scale to measure emotion-driven impulsiveness in 2013/2014. Furthermore, 1584 adolescents completed the computer-based Bergs Card Sorting Test to measure cognitive inflexibility, and 1780 adolescents completed the Hungry Donkey Test to assess decision-making ability. Anthropometric variables were measured objectively; confounding variables (age, sex, socioeconomic status and country) were assessed using a questionnaire. Mixed-effect regression analyses were conducted for each outcome of the test or questionnaire as a predictor with standardised BMI (zBMI) as the dependent variable in order to investigate association between markers of cognitive functioning and zBMI. RESULTS: After controlling for confounders, results showed that emotion-driven impulsiveness (ß=0.18, 95% confidence interval (CI): 0.13 to 0.24, P<0.001) and cognitive inflexibility (ß=0.01, 95% CI: 0.002 to 0.02, P=0.016) were positively associated with zBMI. However, decision-making ability was not significantly related to zBMI (ß=0.001, 95% CI: -0.001 to 0.003, P=0.47). CONCLUSIONS: More emotion-driven impulsiveness and reduced cognitive flexibility were associated with a higher zBMI in adolescents across Europe. These results may indicate that being impulsive in negative situations and having difficulties changing mental sets increase the susceptibility for unhealthy weight development. Reducing impulsivity and training cognitive flexibility seem promising targets for the prevention and intervention programmes of obesity.


Subject(s)
Adolescent Behavior/psychology , Body Weight/physiology , Decision Making/physiology , Emotions/physiology , Impulsive Behavior/physiology , Adolescent , Body Mass Index , Child , Cognition , Cross-Sectional Studies , Europe/epidemiology , Female , Humans , Male , Surveys and Questionnaires
4.
Int J Obes (Lond) ; 41(7): 1131-1140, 2017 07.
Article in English | MEDLINE | ID: mdl-28321131

ABSTRACT

BACKGROUND: Dopamine (DA) signalling in the brain is necessary for feeding behaviour, and alterations in the DA system have been linked to obesity. However, the precise role of DA in the control of food intake remains debated. On the one hand, food reward and motivation are associated with enhanced DA activity. On the other hand, psychostimulant drugs that increase DA signalling suppress food intake. This poses the questions of how endogenous DA neuronal activity regulates feeding, and whether enhancing DA neuronal activity would either promote or reduce food intake. METHODS: Here, we used designer receptors exclusively activated by designer drugs (DREADD) technology to determine the effects of enhancing DA neuronal activity on feeding behaviour. We chemogenetically activated selective midbrain DA neuronal subpopulations and assessed the effects on feeding microstructure in rats. RESULTS: Treatment with the psychostimulant drug amphetamine or the selective DA reuptake inhibitor GBR 12909 significantly suppressed food intake. Selective chemogenetic activation of DA neurons in the ventral tegmental area (VTA) was found to reduce meal size, but had less impact on total food intake. Targeting distinct VTA neuronal pathways revealed that specific activation of the mesolimbic pathway towards nucleus accumbens (NAc) resulted in smaller and shorter meals. In addition, the meal frequency was increased, rendering total food intake unaffected. The disrupted feeding patterns following activation of VTA DA neurons or VTA to NAc projection neurons were accompanied by locomotor hyperactivity. Activation of VTA neurons projecting towards prefrontal cortex or amygdala, or of DA neurons in the substantia nigra, did not affect feeding behaviour. CONCLUSIONS: Chemogenetic activation of VTA DA neurons or VTA to NAc pathway disrupts feeding patterns. Increased activity of mesolimbic DA neurons appears to both promote and reduce food intake, by facilitating both the initiation and cessation of feeding behaviour.


Subject(s)
Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/physiology , Feeding Behavior/drug effects , Feeding Behavior/physiology , Mesencephalon/cytology , Mesencephalon/physiology , Amphetamine/pharmacology , Animals , Disease Models, Animal , Dopamine Uptake Inhibitors/pharmacology , Eating/drug effects , Eating/physiology , Male , Neural Pathways/drug effects , Neural Pathways/physiology , Piperazines/pharmacology , Rats , Reward
5.
Am J Physiol Regul Integr Comp Physiol ; 312(6): R938-R947, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28356295

ABSTRACT

The adipocyte-derived hormone leptin is a peripheral signal that informs the brain about the metabolic status of an organism. Although traditionally viewed as an appetite-suppressing hormone, studies in the past decade have highlighted the role of leptin in energy expenditure. Leptin has been shown to increase energy expenditure in particular through its effects on the cardiovascular system and brown adipose tissue (BAT) thermogenesis via the hypothalamus. The current review summarizes the role of leptin signaling in various hypothalamic nuclei and its effects on the sympathetic nervous system to influence blood pressure, heart rate, and BAT thermogenesis. Specifically, the role of leptin signaling on three different hypothalamic nuclei, the dorsomedial hypothalamus, the ventromedial hypothalamus, and the arcuate nucleus, is reviewed. It is known that all of these brain regions influence the sympathetic nervous system activity and thereby regulate BAT thermogenesis and the cardiovascular system. Thus the current work focuses on how leptin signaling in specific neuronal populations within these hypothalamic nuclei influences certain aspects of energy expenditure.


Subject(s)
Adipose Tissue, Brown/innervation , Cardiovascular System/innervation , Energy Metabolism , Hypothalamus/metabolism , Leptin/metabolism , Sympathetic Nervous System/metabolism , Animals , Blood Pressure , Heart Rate , Humans , Nutritional Status , Signal Transduction , Thermogenesis
6.
Int J Obes (Lond) ; 39(12): 1742-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26183405

ABSTRACT

BACKGROUND/OBJECTIVES: The rewarding value of palatable foods contributes to overconsumption, even in satiated subjects. Midbrain dopaminergic activity in response to reward-predicting environmental stimuli drives reward-seeking and motivated behavior for food rewards. This mesolimbic dopamine (DA) system is sensitive to changes in energy balance, yet it has thus far not been established whether reward signaling of DA neurons in vivo is under control of hormones that signal appetite and energy balance such as ghrelin and leptin. SUBJECTS/METHODS: We trained rats (n=11) on an operant task in which they could earn two different food rewards. We then implanted recording electrodes in the ventral tegmental area (VTA), and recorded from DA neurons during behavior. Subsequently, we assessed the effects of mild food restriction and pretreatment with the adipose tissue-derived anorexigenic hormone leptin or the orexigenic hormone ghrelin on VTA DA reward signaling. RESULTS: Animals showed an increase in performance following mild food restriction (P=0.002). Importantly, food-cue induced DA firing increased when animals were food restricted (P=0.02), but was significantly attenuated after leptin pretreatment (P=0.00). While ghrelin did affect baseline DA activity (P=0.025), it did not affect cue-induced firing (P⩾0.353). CONCLUSIONS: Metabolic signals, such as leptin, affect food seeking, a process that is dependent on the formation of cue-reward outcomes and involves midbrain DA signaling. These data show that food restriction engages the encoding of food cues by VTA DA neurons at a millisecond level and leptin suppresses this activity. This suggests that leptin is a key in linking metabolic information to reward signaling.


Subject(s)
Dopamine/metabolism , Dopaminergic Neurons/pathology , Ghrelin/metabolism , Leptin/metabolism , Obesity/pathology , Ventral Tegmental Area/pathology , Animals , Appetite , Cues , Disease Models, Animal , Feeding Behavior , Male , Overnutrition , Rats , Rats, Wistar , Reward , Signal Transduction
7.
Mol Psychiatry ; 19(10): 1085-94, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24514567

ABSTRACT

Anorexia nervosa (AN) is a complex and heritable eating disorder characterized by dangerously low body weight. Neither candidate gene studies nor an initial genome-wide association study (GWAS) have yielded significant and replicated results. We performed a GWAS in 2907 cases with AN from 14 countries (15 sites) and 14 860 ancestrally matched controls as part of the Genetic Consortium for AN (GCAN) and the Wellcome Trust Case Control Consortium 3 (WTCCC3). Individual association analyses were conducted in each stratum and meta-analyzed across all 15 discovery data sets. Seventy-six (72 independent) single nucleotide polymorphisms were taken forward for in silico (two data sets) or de novo (13 data sets) replication genotyping in 2677 independent AN cases and 8629 European ancestry controls along with 458 AN cases and 421 controls from Japan. The final global meta-analysis across discovery and replication data sets comprised 5551 AN cases and 21 080 controls. AN subtype analyses (1606 AN restricting; 1445 AN binge-purge) were performed. No findings reached genome-wide significance. Two intronic variants were suggestively associated: rs9839776 (P=3.01 × 10(-7)) in SOX2OT and rs17030795 (P=5.84 × 10(-6)) in PPP3CA. Two additional signals were specific to Europeans: rs1523921 (P=5.76 × 10(-)(6)) between CUL3 and FAM124B and rs1886797 (P=8.05 × 10(-)(6)) near SPATA13. Comparing discovery with replication results, 76% of the effects were in the same direction, an observation highly unlikely to be due to chance (P=4 × 10(-6)), strongly suggesting that true findings exist but our sample, the largest yet reported, was underpowered for their detection. The accrual of large genotyped AN case-control samples should be an immediate priority for the field.


Subject(s)
Anorexia Nervosa/genetics , Asian People/genetics , Calcineurin/genetics , Carrier Proteins/genetics , Case-Control Studies , Cullin Proteins/genetics , Female , Genome-Wide Association Study , Guanine Nucleotide Exchange Factors/genetics , Humans , Japan , Male , Meta-Analysis as Topic , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide , White People/genetics
8.
Gene Ther ; 21(2): 205-11, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24385145

ABSTRACT

RNA interference (RNAi) is a powerful strategy for unraveling gene function and for drug target validation, but exogenous expression of short hairpin RNAs (shRNAs) has been associated with severe side effects. These may be caused by saturation of the microRNA pathway. This study shows degenerative changes in cell morphology and intrusion of blood vessels after transduction of the ventromedial hypothalamus (VMH) of rats with a shRNA expressing adeno-associated viral (AAV) vector. To investigate whether saturation of the microRNA pathway has a role in the observed side effects, expression of neuronal microRNA miR-124 was used as a marker. Neurons transduced with the AAV vector carrying the shRNA displayed a decrease in miR-124 expression. The decreased expression was unrelated to shRNA sequence or target and observed as early as 1 week after injection. In conclusion, this study shows that the tissue response after AAV-directed expression of a shRNA to the VMH is likely to be caused by shRNA-induced saturation of the microRNA pathway. We recommend controlling for miR-124 expression when using RNAi as a tool for studying (loss of) gene function in the brain as phenotypic effects caused by saturation of the RNAi pathway might mask true effects of specific downregulation of the shRNA target.


Subject(s)
Genetic Vectors/adverse effects , MicroRNAs/genetics , Neurons/metabolism , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Animals , Dependovirus/genetics , Gene Expression Regulation , Genetic Vectors/administration & dosage , MicroRNAs/toxicity , Organ Specificity , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Ventromedial Hypothalamic Nucleus/metabolism
9.
Int J Obes (Lond) ; 38(5): 643-9, 2014 May.
Article in English | MEDLINE | ID: mdl-23979221

ABSTRACT

OBJECTIVES: Rats subjected to a free-choice high-fat high-sugar (fcHFHS) diet persistently overeat, exhibit increased food-motivated behavior and become overtly obese. Conversely, several studies using a non-choice (nc) high-energy diet showed only an initial increase in food intake with unaltered or reduced food-motivated behavior. This raises the question of the importance of choice in the persistence of hyperphagia in rats on a fcHFHS diet. SUBJECTS: Meal patterns, food intake and body weight gain were studied in male Wistar rats on free-choice diets with fat and/or sugar and in rats on nc diets with fat and sugar (custom made with ingredients similar to the fcHFHS diet). RESULTS: Rats on a ncHFHS diet initially overconsumed, but reduced intake thereafter, whereas rats on a fcHFHS diet remained hyperphagic. Because half of the sugar intake in the fcHFHS group occurred during the inactive period, we next determined whether sugar intake during the light phase was a necessary requirement for hyperphagia, by restricting access to liquid sugar to either the light or dark period with unlimited access to fat and chow. Results showed that hyperphagia occurred irrespective of the timing of sugar intake. Meal pattern analysis revealed consumption of larger but fewer meals in the ncHFHS group, as well as the fcHF group. Interestingly, meal number was increased in all rats drinking liquid sugar (whether on a fcHFHS or a fcHS diet), whereas a compensatory decrease in meal size was only observed in the fcHS group, but not the fcHFHS group. CONCLUSION: We hereby show the importance of choice in the observation of fcHFHS diet-induced hyperphagia, which results in increases in meal number due to sugar drinking without any compensatory decrease in meal size. We thus provide a novel dietary model in rats that mimics important features of human overconsumption that have been ignored in rodent models of obesity.


Subject(s)
Hyperphagia/pathology , Obesity/pathology , Snacks , Weight Gain , Animals , Body Weight , Choice Behavior , Dietary Fats , Dietary Sucrose , Disease Models, Animal , Eating , Energy Intake , Feeding Behavior , Hyperphagia/blood , Male , Obesity/blood , Rats , Rats, Wistar
10.
Int J Neuropsychopharmacol ; 17(1): 169-86, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24103788

ABSTRACT

The second-generation antipsychotic drug olanzapine has become a widely prescribed drug in the treatment of schizophrenia and bipolar disorder. Unfortunately, its therapeutic benefits are partly outweighed by significant weight gain and other metabolic side effects, which increase the risk for diabetes and cardiovascular disease. Because olanzapine remains superior to other antipsychotic drugs that show less weight gain liability, insight into the mechanisms responsible for olanzapine-induced weight gain is crucial if it is to be effectively addressed. Over the past few decades, several groups have investigated the effects of olanzapine on energy balance using rat models. Unfortunately, results from different studies have not always been consistent and it remains to be determined which paradigms should be used in order to model olanzapine-induced weight gain most accurately. This review summarizes the effects of olanzapine on energy balance observed in different rat models and discusses some of the factors that appear to contribute to the inconsistencies in observed effects. In addition it compares the effects reported in rats with clinical findings to determine the predictive validity of different paradigms.


Subject(s)
Benzodiazepines/adverse effects , Weight Gain/drug effects , Animals , Disease Models, Animal , Energy Intake/drug effects , Energy Metabolism/drug effects , Humans , Models, Biological , Olanzapine , Predictive Value of Tests , Rats
11.
Mol Psychiatry ; 18(12): 1294-301, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23070073

ABSTRACT

Obesity is a global problem with often strong neurobiological underpinnings. The cannabinoid 1 receptor (CB1R) was put forward as a promising drug target for antiobesity medication. However, the first marketed CB1R antagonist/inverse agonist rimonabant was discontinued, as its use was occasionally associated with negative affect and suicidality. In artificial cell systems, CB1Rs can become constitutively active in the absence of ligands. Here, we show that such constitutive CB1R activity also regulates GABAergic and glutamatergic neurotransmission in the ventral tegmental area and basolateral amygdala, regions which regulate motivation and emotions. We show that CB1R inverse agonists like rimonabant suppress the constitutive CB1R activity in such regions, and cause anxiety and reduced motivation for reward. The neutral CB1R antagonist NESS0327 does not suppress constitutive activity and lacks these negative effects. Importantly, however, both rimonabant and NESS0327 equally reduce weight gain and food intake. Together, these findings suggest that neutral CB1R antagonists can treat obesity efficiently and more safely than inverse agonists.


Subject(s)
Obesity/drug therapy , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Amygdala/drug effects , Animals , Anxiety/drug therapy , Anxiety/physiopathology , Dopaminergic Neurons/drug effects , Eating/drug effects , GABAergic Neurons/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Obesity/physiopathology , Piperidines/pharmacology , Piperidines/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/physiology , Rimonabant , Ventral Tegmental Area/drug effects , Weight Gain/drug effects
12.
Int J Obes (Lond) ; 37(7): 1012-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23069665

ABSTRACT

BACKGROUND: Cues that are associated with the availability of food are known to trigger food anticipatory activity (FAA). This activity is expressed as increased locomotor activity and enables an animal to prepare for maximal utilization of nutritional resources. Although the exact neural network that mediates FAA is still unknown, several studies have revealed that the medial hypothalamus is involved. Interestingly, this area is responsive to the anorexigenic hormone leptin and the orexigenic hormone ghrelin that have been shown to modulate FAA. However, how FAA is regulated by neuronal activity and how leptin and ghrelin modulate this activity is still poorly understood. OBJECTIVE: We aimed to examine how the total neuronal population and individual neurons in the medial hypothalamus respond to cue-signaled food availability in awake, behaving rats. In addition, ghrelin and leptin were injected to investigate whether these hormones could have a modulatory role in the regulation of FAA. DESIGN: Using in vivo electrophysiology, neuronal activity was recorded in the medial hypothalamus in freely moving rats kept on a random feeding schedule, in which a light cue signaled upcoming food delivery. Ghrelin and leptin were administered systemically following the behavioral paradigm. RESULTS: The food-predictive cue induced FAA as well as a significant increase in neural activity on a population level. More importantly, a sub-population of medial hypothalamic neurons displayed highly correlated identical responses to both ghrelin and FAA, suggesting that these neurons are part of the network that regulates FAA. CONCLUSION: This study reveals a role for ghrelin, but not leptin, signaling within medial hypothalamus in FAA on both a population level and in single cells, identifying a subset of neurons onto which cue information and ghrelin signaling converge, possibly to drive FAA.


Subject(s)
Feeding Behavior/physiology , Ghrelin/metabolism , Leptin/metabolism , Motor Activity/physiology , Animals , Anticipation, Psychological/drug effects , Behavior, Animal , Cues , Feeding Behavior/drug effects , Ghrelin/pharmacology , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Leptin/pharmacology , Male , Motor Activity/drug effects , Neuropeptide Y/metabolism , Rats , Rats, Wistar
13.
Int J Obes (Lond) ; 36(2): 254-61, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21556042

ABSTRACT

OBJECTIVE: Significant weight gain is a problematic side effect of treatment with the antipsychotic drug olanzapine (OLA). Previous studies in rats suggest that one of the contributing factors is an impairment in satiation that results in increased food intake. However, the mechanisms underlying this impairment in satiation remain largely unclear. METHODS AND RESULTS: In this study, we determined the effect of OLA on levels of leptin, insulin, ghrelin, cholecystokinin (CCK), glucagon-like peptide-1, peptide YY and amylin in male rats that had received a fixed amount of food. OLA did not affect the secretion of any of these hormones, except for ghrelin levels, which were increased compared with controls. Furthermore, when ghrelin levels were determined in rats just before they received their meal, OLA caused a significant increase in ghrelin levels compared with controls, whereas OLA failed to affect baseline ghrelin levels. Next, we investigated the effect of OLA on the efficacy of CCK to reduce meal size. With coadministration, OLA pretreatment counteracted the reduction in meal size by CCK, although there was no significant interaction between the treatments. Finally, telemetry measurements revealed that acute OLA treatment causes a temporary decrease in both locomotor activity and body core temperature. CONCLUSION: Taken together, this study shows that acute injection of OLA selectively increases meal-related ghrelin secretion and this may partially underlie the impairment in satiation by OLA.


Subject(s)
Antipsychotic Agents/pharmacology , Benzodiazepines/pharmacology , Body Temperature/drug effects , Cholecystokinin/drug effects , Ghrelin/drug effects , Motor Activity/drug effects , Peptide YY/drug effects , Analysis of Variance , Animals , Cholecystokinin/metabolism , Eating , Ghrelin/metabolism , Glucagon-Like Peptide 1/drug effects , Glucagon-Like Peptide 1/metabolism , Islet Amyloid Polypeptide/drug effects , Islet Amyloid Polypeptide/metabolism , Male , Olanzapine , Peptide YY/metabolism , Rats , Rats, Wistar , Satiation/drug effects
14.
Sci Rep ; 12(1): 13374, 2022 08 04.
Article in English | MEDLINE | ID: mdl-35927440

ABSTRACT

Leptin exerts its effects on energy balance by inhibiting food intake and increasing energy expenditure via leptin receptors in the hypothalamus. While LepR neurons in the arcuate nucleus of the hypothalamus, the primary target of leptin, have been extensively studied, LepR neurons in other hypothalamic nuclei remain understudied. LepR neurons in the lateral hypothalamus contribute to leptin's effects on food intake and reward, but due to the low abundance of this population it has been difficult to study their molecular profile and responses to energy deficit. We here explore the transcriptome of LepR neurons in the LH and their response to energy deficit. Male LepR-Cre mice were injected in the LH with an AAV carrying Cre-dependent L10:GFP. Few weeks later the hypothalami from fed and food-restricted (24-h) mice were dissected and the TRAP protocol was performed, for the isolation of translating mRNAs from LepR cells in the LH, followed by RNA sequencing. After mapping and normalization, differential expression analysis was performed with DESeq2. We confirm that the isolated mRNA is enriched in LepR transcripts and other known neuropeptide markers of LepRLH neurons, of which we investigate the localization patterns in the LH. We identified novel markers of LepRLH neurons with association to energy balance and metabolic disease, such as Acvr1c, Npy1r, Itgb1, and genes that are differentially regulated by food deprivation, such as Fam46a and Rrad. Our dataset provides a reliable and extensive resource of the molecular makeup of LH LepR neurons and their response to food deprivation.


Subject(s)
Hypothalamic Area, Lateral , Receptors, Leptin , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Energy Metabolism/genetics , Hypothalamic Area, Lateral/metabolism , Hypothalamus/metabolism , Leptin/metabolism , Male , Mice , Neurons/metabolism , Receptors, Leptin/genetics , Receptors, Leptin/metabolism
15.
Int J Obes (Lond) ; 35(4): 595-604, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20714332

ABSTRACT

OBJECTIVES: In diet-induced obesity, it is not clear whether impaired glucose metabolism is caused directly by the diet, or indirectly via obesity. This study examined the effects of different free-choice, high-caloric, obesity-inducing diets on glucose metabolism. In these free-choice diets, saturated fat and/or a 30% sugar solution are provided in an addition to normal chow pellets. METHOD: In the first experiment, male rats received a free-choice high-fat high-sugar (HFHS), free-choice high-fat (HF) or a chow diet. In a second experiment, male rats received a free-choice high-sugar (HS) diet or chow diet. For both experiments, after weeks 1 and 4, an intravenous glucose tolerance test was performed. RESULTS: Both the HFHS and HF diets resulted in obesity with comparable plasma concentrations of free fatty acids. Interestingly, the HF diet did not affect glucose metabolism, whereas the HFHS diet resulted in hyperglycemia, hyperinsulinemia and in glucose intolerance because of a diminished insulin response. Moreover, adiposity in rats on the HF diet correlated positively with the insulin response to the glucose load, whereas adiposity in rats on the HFHS diet showed a negative correlation. In addition, total caloric intake did not explain differences in glucose tolerance. To test whether sugar itself was crucial, we next performed a similar experiment in rats on the HS diet. Rats consumed three times as much sugar when compared with rats on the HFHS diet, which resulted in obesity with basal hyperinsulinemia. Glucose tolerance, however, was not affected. CONCLUSION: Together, these results suggest that not only obesity or total caloric intake, but the diet content also is crucial for the glucose intolerance that we observed in rats on the HFHS diet.


Subject(s)
Blood Glucose/metabolism , Dietary Fats/adverse effects , Dietary Sucrose/adverse effects , Glucose Intolerance/etiology , Obesity/complications , Animals , Dietary Fats/administration & dosage , Dietary Fats/blood , Dietary Sucrose/administration & dosage , Dietary Sucrose/blood , Energy Intake , Glucose Tolerance Test , Insulin/metabolism , Male , Obesity/blood , Rats , Rats, Wistar
16.
Int J Obes (Lond) ; 35(5): 629-41, 2011 May.
Article in English | MEDLINE | ID: mdl-20733584

ABSTRACT

OBJECTIVE: Reduction of melanocortin signaling in the brain results in obesity. However, where in the brain reduced melanocortin signaling mediates this effect is poorly understood. DESIGN: We determined the effects of long-term inhibition of melanocortin receptor activity in specific brain regions of the rat brain. Melanocortin signaling was inhibited by injection of a recombinant adeno-associated viral (rAAV) vector that overexpressed Agouti-related peptide (AgRP) into the paraventricular nucleus (PVN), the ventromedial hypothalamus (VMH), the lateral hypothalamus (LH) or the accumbens shell (Acc). RESULTS: Overexpression of AgRP in the rat PVN, VMH or LH increased bodyweight, the percentage of white adipose tissue, plasma leptin and insulin concentrations and food intake. Food intake was mainly increased because of an increase in meal size in the light and dark phases, after overexpression of AgRP in the PVN, LH or VMH. Overexpression of AgRP in the PVN or VMH reduced average body core temperature in the dark on day 40 post injection, whereas AgRP overexpression in the LH did not affect temperature. In addition, overexpression of AgRP in the PVN, LH or VMH did not significantly alter mRNA expression of AgRP, neuropeptide Y (NPY), pro-opiomelanocortin (POMC) or suppressor of cytokine signaling 3 (SOCS3) in the arcuate. Overexpression of AgRP in the Acc did not have any effect on the measured parameters. CONCLUSIONS: Reduction of melanocortin signaling in several hypothalamic regions increased meal size. However, there were brain area-specific effects on other parameters such as core temperature and plasma leptin concentrations. In a previous study, where NPY was overexpressed with an rAAV vector in the PVN and LH, meal frequency and meal size were increased respectively, whereas locomotor activity was reduced by NPY overexpression at both nuclei. Taken together, AgRP and NPY have complementary roles in energy balance.


Subject(s)
Agouti-Related Protein/metabolism , Body Weight/physiology , Energy Metabolism/physiology , Hypothalamus/metabolism , Obesity/metabolism , Receptors, Melanocortin/physiology , Animals , Cell Line , Eating/physiology , Hypothalamic Area, Lateral/metabolism , Hypothalamus/physiology , Male , Midline Thalamic Nuclei/metabolism , Nucleus Accumbens/metabolism , Obesity/physiopathology , Rats , Rats, Wistar , Receptors, Melanocortin/antagonists & inhibitors , Ventromedial Hypothalamic Nucleus/metabolism
17.
Int J Obes (Lond) ; 34(3): 537-46, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20029382

ABSTRACT

OBJECTIVES: The mechanisms for how saturated fat and sugar-based beverages contribute to human obesity are poorly understood. This paper describes a series of experiments developed to examine the response of hypothalamic neuropeptides to diets rich in sugar and fat, using three different diets: a high-fat high-sugar (HFHS) choice diet with access to chow, saturated fat and a 30% sugar solution; a high-fat (HF) choice diet with access to chow and saturated fat; or to a high-sugar (HS) choice diet with access to chow and a sugar solution. METHOD: We first studied caloric intake, body weight gain, hormonal alterations and hypothalamic neuropeptide expression when male Wistar rats were subjected to an HFHS choice, an HF choice or an HS choice diet for 1 week. Next, we studied caloric intake and body weight gain when rats were subjected to the choice diets for 5 weeks. Finally, we measured neuropeptide expression in hepatic vagotomized rats subjected to an HFHS choice, an HF choice or an HS choice diet for 1 week. RESULTS: In rats on an HF choice diet, plasma leptin concentrations and proopiomelanocortin (POMC) mRNA increased and neuropeptide Y (NPY) mRNA decreased. Rats on an HFHS choice diet showed identical plasma leptin concentrations as rats on an HF choice diet. However, NPY mRNA increased and POMC mRNA decreased. An HS choice diet for 1 week did not alter hypothalamic neuropeptide expression or plasma leptin concentrations. As hormonal changes did not explain the differences in hypothalamic neuropeptide expression between rats on the choice diets, we addressed whether neuronal feedback signals mediated the hypothalamic neuropeptide response. The POMC mRNA response to different diets depended on an intact innervation of liver and upper intestinal tract. CONCLUSION: Our data suggest that the specific combination of saturated fat and a 30% sugar solution results in hyperphagia-induced obesity and alters hypothalamic neuropeptide expression, and that the response of the melanocortin system is mediated by the hepatic vagus.


Subject(s)
Dietary Fats/adverse effects , Dietary Sucrose/adverse effects , Hyperphagia/complications , Neuropeptide Y/blood , Obesity/etiology , Animals , Body Weight/physiology , Dietary Fats/administration & dosage , Dietary Sucrose/administration & dosage , Energy Intake/physiology , Gene Expression , Hyperphagia/blood , Hyperphagia/physiopathology , Leptin/blood , Liver/innervation , Male , Obesity/blood , Obesity/physiopathology , Pro-Opiomelanocortin/blood , Rats , Rats, Wistar
18.
Annu Rev Nutr ; 29: 283-304, 2009.
Article in English | MEDLINE | ID: mdl-19400703

ABSTRACT

Feeding is a physiological process, influenced by genetic factors and the environment. In recent years, many studies have been performed to unravel the involvement of genetics in both eating behavior and its pathological forms: eating disorders and obesity. In this review, we provide a condensed introduction on the neurological aspects of eating and we describe the current status of research into the genetics of eating behavior, primarily focused on specific traits such as taste, satiation, and hunger. This is followed by an overview on the genetic studies done to unravel the heritable background of obesity and eating disorders. We examine the discussion currently taking place in the field of genetics of complex disorders and phenotypes on how to perform good and powerful studies, with the use of large-scale whole-genome association studies as one of the possible solutions. In the final part of this review, we give our view on the latest developments, including endophenotype approaches and animal studies. Studies of endophenotypes of eating behavior may help to identify core traits that are genetically influenced. Such studies would yield important knowledge on the underlying biological scaffold on which diagnostic criteria for eating disorders could be based and would provide information to influence eating behavior toward healthier living.


Subject(s)
Feeding Behavior/physiology , Feeding and Eating Disorders/genetics , Genetic Variation , Obesity/genetics , Animals , Disease Models, Animal , Eating/genetics , Eating/physiology , Genetic Linkage , Genotype , Humans , Hunger/physiology , Phenotype , Satiation/physiology , Taste/genetics , Taste/physiology
19.
Sci Rep ; 9(1): 11146, 2019 07 31.
Article in English | MEDLINE | ID: mdl-31366942

ABSTRACT

Targeting specific neuronal cell types is a major challenge for unraveling their function and utilizing specific cells for gene therapy strategies. Viral vector tools are widely used to target specific cells or circuits for these purposes. Here, we use viral vectors with short promoters of neuropeptide genes to target distinct neuronal populations in the hypothalamus of rats and mice. We show that lowering the amount of genomic copies is effective in increasing specificity of a melanin-concentrating hormone promoter. However, since too low titers reduce transduction efficacy, there is an optimal titer for achieving high specificity and sufficient efficacy. Other previously identified neuropeptide promoters as those for oxytocin and orexin require further sequence optimization to increase target specificity. We conclude that promoter-driven viral vectors should be used with caution in order to target cells specifically.


Subject(s)
Genetic Vectors/administration & dosage , Hypothalamus/drug effects , Neurons/drug effects , Neuropeptides/administration & dosage , Promoter Regions, Genetic/genetics , Animals , Hypothalamic Hormones/genetics , Melanins/genetics , Mice , Mice, Inbred C57BL , Orexins/genetics , Oxytocin/genetics , Pituitary Hormones/genetics , Rats , Rats, Long-Evans , Rats, Wistar
20.
Physiol Behav ; 94(5): 689-95, 2008 Aug 06.
Article in English | MEDLINE | ID: mdl-18495181

ABSTRACT

Up to 80% of patients with Anorexia Nervosa (AN) demonstrate hyperactivity. Hyperactivity counteracts weight gain during treatment and is associated with poor outcome of the disease. We hypothesized that hyperactivity in AN patients has a neurobiological basis and used an animal model-based translational approach to gain insight in mechanisms underlying this hyperactivity. Previously we and others showed that leptin treatment attenuates hyperactivity in the rat activity-based anorexia (ABA) model. The mechanisms involved in this process are, however, unknown. Here we describe potential downstream effector mechanisms involved in the attenuation of hyperactivity by leptin treatment in ABA rats.


Subject(s)
Anorexia Nervosa/metabolism , Hyperkinesis/metabolism , Leptin/physiology , Motor Activity/physiology , Animals , Anorexia Nervosa/complications , Disease Models, Animal , Humans , Hyperkinesis/complications
SELECTION OF CITATIONS
SEARCH DETAIL