Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Front Pharmacol ; 13: 1066988, 2022.
Article in English | MEDLINE | ID: mdl-36467081

ABSTRACT

Attention-deficit/hyperactivity disorder (ADHD) is one of the most common neurodevelopmental disorders having a high influence on social interactions. The number of approved treatments and clinical trials for ADHD have increased markedly during the recent decade. This analytical review provides a quantitative overview of the existing pharmacological and non-pharmacological methods of ADHD treatments investigated in clinical trials during 1999-2021. A total of 695 interventional trials were manually assessed from clinicaltrial.gov with the search term « ADHD¼, and trial data has been used for analysis. A clear majority of the studies investigated non-pharmacological therapies (∼80%), including many behavioral options, such as social skills training, sleep and physical activity interventions, meditation and hypnotherapy. Devices, complementary and other alternative methods of ADHD treatment are also gaining attention. The pharmacological group accounts for ∼20% of all the studies. The most common drug classes include central nervous system stimulants (e.g., methylphenidate hydrochloride, lisdexamfetamine dimesylate, amphetamine sulfate, mixed amphetamine salts, a combination of dexmethylphenidate hydrochloride and serdexmethylphenidate chloride), selective noradrenaline reuptake inhibitors (atomoxetine, viloxazine), and alpha2 adrenergic receptor agonists (guanfacine hydrochloride, clonidine hydrochloride). Several studies investigated antidepressants (e.g., bupropion hydrochloride, vortioxetine), and atypical antipsychotics (e.g., quetiapine, aripiprazole) but these are yet not approved by the FDA for ADHD treatment. We discuss the quantitative trends in clinical trials and provide an overview of the new drug agents and non-pharmacological therapies, drug targets, and novel treatment options.

2.
Pharmacol Rev ; 74(3): 506-551, 2022 07.
Article in English | MEDLINE | ID: mdl-35710135

ABSTRACT

Acylcarnitines are fatty acid metabolites that play important roles in many cellular energy metabolism pathways. They have historically been used as important diagnostic markers for inborn errors of fatty acid oxidation and are being intensively studied as markers of energy metabolism, deficits in mitochondrial and peroxisomal ß -oxidation activity, insulin resistance, and physical activity. Acylcarnitines are increasingly being identified as important indicators in metabolic studies of many diseases, including metabolic disorders, cardiovascular diseases, diabetes, depression, neurologic disorders, and certain cancers. The US Food and Drug Administration-approved drug L-carnitine, along with short-chain acylcarnitines (acetylcarnitine and propionylcarnitine), is now widely used as a dietary supplement. In light of their growing importance, we have undertaken an extensive review of acylcarnitines and provided a detailed description of their identity, nomenclature, classification, biochemistry, pathophysiology, supplementary use, potential drug targets, and clinical trials. We also summarize these updates in the Human Metabolome Database, which now includes information on the structures, chemical formulae, chemical/spectral properties, descriptions, and pathways for 1240 acylcarnitines. This work lays a solid foundation for identifying, characterizing, and understanding acylcarnitines in human biosamples. We also discuss the emerging opportunities for using acylcarnitines as biomarkers and as dietary interventions or supplements for many wide-ranging indications. The opportunity to identify new drug targets involved in controlling acylcarnitine levels is also discussed. SIGNIFICANCE STATEMENT: This review provides a comprehensive overview of acylcarnitines, including their nomenclature, structure and biochemistry, and use as disease biomarkers and pharmaceutical agents. We present updated information contained in the Human Metabolome Database website as well as substantial mapping of the known biochemical pathways associated with acylcarnitines, thereby providing a strong foundation for further clarification of their physiological roles.


Subject(s)
Carnitine , Insulin Resistance , Biomarkers , Carnitine/analogs & derivatives , Carnitine/chemistry , Carnitine/metabolism , Carnitine/therapeutic use , Fatty Acids/metabolism , Humans , Insulin Resistance/physiology
3.
Cell Mol Biol Lett ; 26(1): 5, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33588752

ABSTRACT

BACKGROUND: The synaptic vesicle glycoprotein 2 (SV2) family is essential to the synaptic machinery involved in neurotransmission and vesicle recycling. The isoforms SV2A, SV2B and SV2C are implicated in neurological diseases such as epilepsy, Alzheimer's and Parkinson's disease. Suitable cell systems for studying regulation of these proteins are essential. Here we present gene expression data of SV2A, SV2B and SV2C in two human neuroblastoma cell lines after differentiation. METHODS: Human neuroblastoma cell lines SiMa and IMR-32 were treated for seven days with growth supplements (B-27 and N-2), all-trans-retinoic acid (ATRA) or vasoactive intestinal peptide (VIP) and gene expression levels of SV2 and neuronal targets were analyzed. RESULTS: The two cell lines reacted differently to the treatments, and only one of the three SV2 isoforms was affected at a time. SV2B and choline O-acetyltransferase (CHAT) expression was changed in concert after growth supplement treatment, decreasing in SiMa cells while increasing in IMR-32. ATRA treatment resulted in no detected changes in SV2 expression in either cell line while VIP increased both SV2C and dopamine transporter (DAT) in IMR-32 cells. CONCLUSION: The synergistic expression patterns between SV2B and CHAT as well as between SV2C and DAT mirror the connectivity between these targets found in disease models and knock-out animals, although here no genetic alteration was made. These cell lines and differentiation treatments could possibly be used to study SV2 regulation and function.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation, Neoplastic , Membrane Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Neuroblastoma/genetics , Neuroblastoma/pathology , Binding Sites , Cell Differentiation/drug effects , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism , Transcription Initiation Site , Tretinoin/pharmacology , Vasoactive Intestinal Peptide/pharmacology
5.
BMC Genomics ; 21(1): 331, 2020 Apr 29.
Article in English | MEDLINE | ID: mdl-32349672

ABSTRACT

BACKGROUND: Salivary cell secretion (SCS) plays a critical role in blood feeding by medicinal leeches, making them of use for certain medical purposes even today. RESULTS: We annotated the Hirudo medicinalis genome and performed RNA-seq on salivary cells isolated from three closely related leech species, H. medicinalis, Hirudo orientalis, and Hirudo verbana. Differential expression analysis verified by proteomics identified salivary cell-specific gene expression, many of which encode previously unknown salivary components. However, the genes encoding known anticoagulants have been found to be expressed not only in salivary cells. The function-related analysis of the unique salivary cell genes enabled an update of the concept of interactions between salivary proteins and components of haemostasis. CONCLUSIONS: Here we report a genome draft of Hirudo medicinalis and describe identification of novel salivary proteins and new homologs of genes encoding known anticoagulants in transcriptomes of three medicinal leech species. Our data provide new insights in genetics of blood-feeding lifestyle in leeches.


Subject(s)
Genome , Hirudo medicinalis/genetics , Salivary Proteins and Peptides/genetics , Animals , Anticoagulants/metabolism , Gene Expression Profiling , Gene Expression Regulation , Hirudo medicinalis/metabolism , Leeches/classification , Leeches/genetics , Leeches/metabolism , Proteomics , Saliva/metabolism , Salivary Proteins and Peptides/metabolism
6.
J Psychiatry Neurosci ; 44(5): 324-339, 2019 09 01.
Article in English | MEDLINE | ID: mdl-30994310

ABSTRACT

Background: Anorexia nervosa and bulimia nervosa are complex mental disorders, and their etiology is still not fully understood. This paper reviews the literature on diffusion tensor imaging studies in patients with anorexia nervosa and bulimia nervosa to explore the usefulness of white matter microstructural analysis in understanding the pathophysiology of eating disorders. Methods: We followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines to identify diffusion tensor imaging studies that compared patients with an eating disorder to control groups. We searched relevant databases for studies published from database inception to August 2018, using combinations of select keywords. We categorized white matter tracts according to their 3 main classes: projection (i.e., thalamo­cortical), association (i.e., occipital­parietal­temporal­frontal) and commissural (e.g., corpus callosum). Results: We included 19 papers that investigated a total of 427 participants with current or previous eating disorders and 444 controls. Overall, the studies used different diffusion tensor imaging approaches and showed widespread white matter abnormalities in patients with eating disorders. Despite differences among the studies, patients with anorexia nervosa showed mainly white matter microstructural abnormalities of thalamo­cortical tracts (i.e., corona radiata, thalamic radiations) and occipital­parietal­temporal­frontal tracts (i.e., left superior longitudinal and inferior fronto-occipital fasciculi). It was less clear whether white matter alterations persist after recovery from anorexia nervosa. Available data on bulimia nervosa were partially similar to those for anorexia nervosa. Limitations: Study sample composition and diffusion tensor imaging analysis techniques were heterogeneous. The number of studies on bulimia nervosa was too limited to be conclusive. Conclusion: White matter microstructure appears to be affected in anorexia nervosa, and these alterations may play a role in the pathophysiology of this eating disorder. Although we found white matter alterations in bulimia nervosa that were similar to those in anorexia nervosa, white matter changes in bulimia nervosa remain poorly investigated, and these findings were less conclusive. Further studies with longitudinal designs and multi-approach analyses are needed to better understand the role of white matter changes in eating disorders.


Subject(s)
Anorexia Nervosa/diagnostic imaging , Bulimia Nervosa/diagnostic imaging , Cerebral Cortex/diagnostic imaging , Thalamus/diagnostic imaging , White Matter/diagnostic imaging , Anorexia Nervosa/physiopathology , Bulimia Nervosa/physiopathology , Cerebral Cortex/physiopathology , Diffusion Tensor Imaging , Humans , Neural Pathways/diagnostic imaging , Neural Pathways/physiopathology , Occipital Lobe/diagnostic imaging , Occipital Lobe/physiopathology , Parietal Lobe/diagnostic imaging , Parietal Lobe/physiopathology , Temporal Lobe/diagnostic imaging , Temporal Lobe/physiopathology , Thalamus/physiopathology , White Matter/physiopathology
7.
J Nutr Biochem ; 61: 82-90, 2018 11.
Article in English | MEDLINE | ID: mdl-30189366

ABSTRACT

Omega-3 polyunsaturated fatty acids (ω-3 PUFAs) are relevant to fetal and infant growth and development. Objective: to assess whether long-term exposure to dietary ω-3 PUFA imbalance alters pre- and/or postnatal pups' development and reproductive function later in life. Mice dams were fed with ω-3 PUFA Control (soybean oil, 7%), Deficient (sunflower oil, 7%) or Excess (blend oil; 4.2% cod-liver+2.8% soybean) diet before conception and throughout gestation-lactation and later on, their pups received the same diet from weaning to adulthood. Offspring somatic, neurobiological and reproductive parameters were evaluated. Excess pups were lighter during the preweaning period and shorter in length from postnatal day (PND) 7 to 49, compared to Control pups (P<.05). On PND14, the percentage of pups with eye opening in Excess group was lower than those from Control and Deficient groups (P<.05). In Excess female offspring, puberty onset (vaginal opening and first estrus) occurred significantly later and the percentage of parthenogenetic oocytes on PND63 was higher than Control and Deficient ones (P<.05). Deficient pups were shorter in length (males: on PND14, 21, 35 and 49; females: on PND14, 21 and 42) compared with Control pups (P<.05). Deficient offspring exhibited higher percentage of bending spermatozoa compared to Control and Excess offspring (P<.05). These results show that either an excessively high or insufficient ω-3 PUFA consumption prior to conception until adulthood seems inadvisable because of the potential risks of short-term adverse effects on growth and development of the progeny or long-lasting effects on their reproductive maturation and function.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Reproduction/physiology , Animals , Body Weight , Fatty Acids, Omega-3/adverse effects , Female , Lactation , Male , Mice , Oocytes/physiology , Ovulation/physiology , Pregnancy , Pregnancy Outcome , Progesterone/blood , Puberty , Reproduction/drug effects , Semen/drug effects , Semen/physiology , Testosterone/blood
8.
Reproduction ; 156(2): 121-132, 2018 08.
Article in English | MEDLINE | ID: mdl-29794024

ABSTRACT

It has been demonstrated that food intake and reproductive physiology are both simultaneously modulated to optimize reproductive success under fluctuating metabolic conditions. Ghrelin (GHRL) is an orexigenic peptide identified as the endogenous ligand of the growth hormone secretagogue receptor that is being investigated for its potential role on reproduction. Considering that data available so far are still limited and characterization of GHRL action mechanism on the reproductive system has not been fully elucidated, we studied the participation of hypothalamus in GHRL effects on sperm functional activity, plasma levels of gonadotropins and histological morphology in mice testes after hypothalamic infusion of 0.3 or 3.0 nmol/day GHRL or artificial cerebrospinal fluid (ACSF) at different treatment periods. We found that GHRL 3.0 nmol/day administration for 42 days significantly reduced sperm concentration (GHRL 3.0 nmol/day = 14.05 ± 2.44 × 106/mL vs ACSF = 20.33 ± 1.35 × 106/mL, P < 0.05) and motility (GHRL 3.0 nmol/day = 59.40 ± 4.20% vs ACSF = 75.80 ± 1.40%, P < 0.05). In addition, histological studies showed a significant decrease percentage of spermatogonia (GHRL 3.0 nmol/day = 6.76 ± 0.68% vs ACSF = 9.56 ± 0.41%, P < 0.05) and sperm (GHRL 3.0 nmol/day = 24.24 ± 1.92% vs ACSF = 31.20 ± 3.06%, P < 0.05). These results were associated with a significant reduction in luteinizing hormone and testosterone plasma levels (P < 0.05). As GHRL is an orexigenic peptide, body weight and food intake were measured. Results showed that GHRL increases both parameters; however, the effect did not last beyond the first week of treatment. Results presented in this work confirm that central GHRL administration impairs spermatogenesis and suggest that this effect is mediated by inhibition of hypothalamic-pituitary-gonadal axis.


Subject(s)
Ghrelin/physiology , Gonadotropins, Pituitary/blood , Hypothalamus/physiology , Spermatogenesis , Testosterone/blood , Animals , Body Weight , Eating , Male , Mice , Spermatozoa/physiology , Testis/ultrastructure
9.
PLoS One ; 12(3): e0172129, 2017.
Article in English | MEDLINE | ID: mdl-28248991

ABSTRACT

Few studies have used diffusion tensor imaging (DTI) to investigate the micro-structural alterations of WM in patients with restrictive eating disorders (rED), and longitudinal data are lacking. Twelve patients with rED were scanned at diagnosis and after one year of family-based treatment, and compared to twenty-four healthy controls (HCs) through DTI analysis. A tract-based spatial statistics procedure was used to investigate diffusivity parameters: fractional anisotropy (FA) and mean, radial and axial diffusivities (MD, RD and AD, respectively). Reduced FA and increased RD were found in patients at baseline in the corpus callosum, corona radiata and posterior thalamic radiation compared with controls. However, no differences were found between follow-up patients and controls, suggesting a partial normalization of the diffusivity parameters. In patients, trends for a negative correlation were found between the baseline FA of the right anterior corona radiata and the Eating Disorder Examination Questionnaire total score, while a positive trend was found between the baseline FA in the splenium of corpus callosum and the weight loss occurred between maximal documented weight and time of admission. A positive trend for correlation was also found between baseline FA in the right anterior corona radiata and the decrease in the Obsessive-Compulsive Inventory Revised total score over time. Our results suggest that the integrity of the limbic-thalamo-cortical projections and the reward-related circuitry are important for cognitive control processes and reward responsiveness in regulating eating behavior.


Subject(s)
Cerebral Cortex , Diffusion Tensor Imaging , Feeding Behavior , Feeding and Eating Disorders , Limbic System , Thalamus , Adolescent , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/physiopathology , Feeding and Eating Disorders/diagnostic imaging , Feeding and Eating Disorders/physiopathology , Female , Humans , Limbic System/diagnostic imaging , Limbic System/physiopathology , Longitudinal Studies , Thalamus/diagnostic imaging , Thalamus/physiopathology
10.
Sleep Med ; 23: 111-118, 2016 07.
Article in English | MEDLINE | ID: mdl-27539026

ABSTRACT

BACKGROUND: The use of electronic devices emitting blue light during evening hours has been associated with sleep disturbances in humans, possibly due to the blue light-mediated suppression of the sleep-promoting hormone melatonin. However, experimental results have been mixed. The present study therefore sought to investigate if reading on a self-luminous tablet during evening hours would alter sleepiness, melatonin secretion, nocturnal sleep, as well as electroencephalographic power spectral density during early slow-wave sleep. METHODS: Following a constant bright light exposure over 6.5 hours (~569 lux), 14 participants (six females) read a novel either on a tablet or as physical book for two hours (21:00-23:00). Evening concentrations of saliva melatonin were repeatedly measured. Sleep (23:15-07:15) was recorded by polysomnography. Sleepiness was assessed before and after nocturnal sleep. About one week later, experiments were repeated; participants who had read the novel on a tablet in the first experimental session continued reading the same novel in the physical book, and vice versa. RESULTS: There were no differences in sleep parameters and pre-sleep saliva melatonin levels between the tablet reading and physical book reading conditions. CONCLUSIONS: Bright light exposure during daytime has previously been shown to abolish the inhibitory effects of evening light stimulus on melatonin secretion. Our results could therefore suggest that exposure to bright light during the day - as in the present study - may help combat sleep disturbances associated with the evening use of electronic devices emitting blue light. However, this needs to be validated by future studies with larger sample populations.


Subject(s)
Computers, Handheld , Light/adverse effects , Reading , Sleep/radiation effects , Adult , Cross-Over Studies , Humans , Male , Melatonin/analysis , Melatonin/physiology , Phototherapy , Polysomnography , Saliva/chemistry , Sleep/physiology , Sleep Latency/physiology , Sleep Latency/radiation effects
11.
Psychopharmacology (Berl) ; 233(6): 1077-86, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26700241

ABSTRACT

RATIONALE: In response to stress, corticotropin releasing hormone (CRH) and vasopressin (AVP) are released from the hypothalamus, activate their receptors (CRHR1, CRHR2 or AVPr1b), and synergistically act to induce adrenocorticotropic hormone (ACTH) release from the anterior pituitary. Overstimulation of this system has been frequently associated with major depression states. OBJECTIVE: The objective of the study is to assess the role of AVP and CRH receptors in fluoxetine and venlafaxine effects on the expression of depression-related behavior. METHODS: In an animal model of depression (olfactory bulbectomy in mice, OB), we evaluated the effects of fluoxetine or venlafaxine (both 10 mg/kg/day) chronic administration on depression-related behavior in the tail suspension test. Plasma levels of AVP, CRH, and ACTH were determined as well as participation of their receptors in the expression of depression related-behavior and gene expression of AVP and CRH receptors (AVPr1b, CRHR1, and CRHR2) in the pituitary gland. RESULTS: The expression of depressive-like behavior in OB animals was reversed by treatment with both antidepressants. Surprisingly, OB-saline mice exhibited increased AVP and ACTH plasma levels, with no alterations in CRH levels when compared to sham mice. Chronic fluoxetine or venlafaxine reversed these effects. In addition, a significant increase only in AVPr1b gene expression was found in OB-saline. CONCLUSION: The antidepressant therapy used seems to be more likely related to a reduced activation of AVP rather than CRH receptors, since a positive correlation between AVP levels and depressive-like behavior was observed in OB animals. Furthermore, a full restoration of depressive behavior was observed in OB-fluoxetine- or venlafaxine-treated mice only when AVP was centrally administered but not CRH.


Subject(s)
Antidepressive Agents/therapeutic use , Depressive Disorder/drug therapy , Fluoxetine/therapeutic use , Receptors, Vasopressin/metabolism , Venlafaxine Hydrochloride/therapeutic use , Adrenocorticotropic Hormone/blood , Animals , Antidepressive Agents/pharmacology , Arginine Vasopressin/blood , Behavior, Animal/drug effects , Corticotropin-Releasing Hormone/blood , Depressive Disorder/etiology , Depressive Disorder/metabolism , Disease Models, Animal , Fluoxetine/pharmacology , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Mice , Olfactory Bulb/surgery , Receptors, Corticotropin-Releasing Hormone/metabolism , Signal Transduction/drug effects , Venlafaxine Hydrochloride/pharmacology
12.
Mol Endocrinol ; 29(9): 1303-19, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26168034

ABSTRACT

Several genome-wide association studies have linked the Nudix hydrolase family member nucleoside diphosphate-linked moiety X motif 3 (NUDT3) to obesity. However, the manner of NUDT3 involvement in obesity is unknown, and NUDT3 expression, regulation, and signaling in the central nervous system has not been studied. We performed an extensive expression analysis in mice, as well as knocked down the Drosophila NUDT3 homolog Aps in the nervous system, to determine its effect on metabolism. Detailed in situ hybridization studies in the mouse brain revealed abundant Nudt3 mRNA and protein expression throughout the brain, including reward- and feeding-related regions of the hypothalamus and amygdala, whereas Nudt3 mRNA expression was significantly up-regulated in the hypothalamus and brainstem of food-deprived mice. Knocking down Aps in the Drosophila central nervous system, or a subset of median neurosecretory cells, known as the insulin-producing cells (IPCs), induces hyperinsulinemia-like phenotypes, including a decrease in circulating trehalose levels as well as significantly decreasing all carbohydrate levels under starvation conditions. Moreover, lowering Aps IPC expression leads to a decreased ability to recruit these lipids during starvation. Also, loss of neuronal Aps expression caused a starvation susceptibility phenotype while inducing hyperphagia. Finally, the loss of IPC Aps lowered the expression of Akh, Ilp6, and Ilp3, genes known to be inhibited by insulin signaling. These results point toward a role for this gene in the regulation of insulin signaling, which could explain the robust association with obesity in humans.


Subject(s)
Acid Anhydride Hydrolases/genetics , Insulin/metabolism , Obesity/genetics , Pyrophosphatases/genetics , Starvation/genetics , Acid Anhydride Hydrolases/biosynthesis , Acid Anhydride Hydrolases/metabolism , Amygdala/metabolism , Animals , Cell Line, Tumor , Drosophila , Drosophila Proteins/biosynthesis , Gene Knockdown Techniques , HCT116 Cells , HeLa Cells , Humans , Hyperinsulinism/genetics , Hyperphagia/genetics , Hypothalamus/metabolism , Insect Hormones/biosynthesis , Insulin/genetics , Insulin-Secreting Cells/metabolism , Intercellular Signaling Peptides and Proteins/biosynthesis , MCF-7 Cells , Male , Mice , Mice, Inbred C57BL , Oligopeptides/biosynthesis , Pyrrolidonecarboxylic Acid/analogs & derivatives , RNA, Messenger/biosynthesis , Signal Transduction/genetics , Somatomedins/biosynthesis , Trehalose/blood
13.
PLoS One ; 10(6): e0122061, 2015.
Article in English | MEDLINE | ID: mdl-26047506

ABSTRACT

G protein-coupled receptors (GPCRs) are a class of integral membrane proteins mediating physiological functions fundamental for survival, including energy homeostasis. A few years ago, an amino acid sequence of a novel GPCR gene was identified and named GPR178. In this study, we provide new insights regarding the biological significance of Gpr178 protein, investigating its evolutionary history and tissue distribution as well as examining the relationship between its expression level and feeding status. Our phylogenetic analysis indicated that GPR178 is highly conserved among all animal species investigated, and that GPR178 is not a member of a protein family. Real-time PCR and in situ hybridization revealed wide expression of Gpr178 mRNA in both the brain and periphery, with high expression density in the hypothalamus and brainstem, areas involved in the regulation of food intake. Hence, changes in receptor expression were assessed following several feeding paradigms including starvation and overfeeding. Short-term starvation (12-48h) or food restriction resulted in upregulation of Gpr178 mRNA expression in the brainstem, hypothalamus and prefrontal cortex. Conversely, short-term (48h) exposure to sucrose or Intralipid solutions downregulated Gpr178 mRNA in the brainstem; long-term exposure (10 days) to a palatable high-fat and high-sugar diet resulted in a downregulation of Gpr178 in the amygdala but not in the hypothalamus. Our results indicate that hypothalamic Gpr178 gene expression is altered during acute exposure to starvation or acute exposure to palatable food. Changes in gene expression following palatable diet consumption suggest a possible involvement of Gpr178 in the complex mechanisms of feeding reward.


Subject(s)
Eating , Receptors, G-Protein-Coupled/metabolism , Amygdala/metabolism , Animals , Biological Evolution , Brain/metabolism , Hypothalamus/metabolism , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Phylogeny , Prefrontal Cortex/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/classification , Receptors, G-Protein-Coupled/genetics
14.
Obes Surg ; 25(11): 2071-7, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25724814

ABSTRACT

BACKGROUND: A recent study in children demonstrated that the rs9939609 single-nucleotide polymorphism in the fat mass and obesity (FTO) gene influences prospective weight gain, however, only in those who were vitamin D-deficient. If this might also be the case for Roux-en-Y gastric bypass (RYGB), surgery-induced weight loss is however unknown. The objective of this study is to examine if the magnitude of RYGB surgery-induced weight loss after 2 years depends on patients' FTO rs9939609 genotype (i.e., TT, AT, and AA) and presurgery vitamin D status (<50 nmol/L equals deficiency). METHODS: Before and at 24 months after RYGB surgery, BMI was measured in 210 obese patients (mean BMI 45 kg/m(2), 72 % females). Serum 25-hydroxyvitamin D3 levels were also repeatedly measured. Following surgery, vitamin D was supplemented. Possible weight loss differences between genotypes were tested with multiple linear regressions. RESULTS: The per-allele effect of each FTO A-allele on excessive BMI loss (EBMIL) was 3 % (P = 0.02). When split by baseline status, the EBMIL of vitamin D-deficient patients carrying AA exceeded that of vitamin D-deficient patients carrying TT by ~14 % (P = 0.03). No such genotypic differences were found in patients without presurgery vitamin D deficiency. Post-surgery serum levels of vitamin D did not differ between groups. CONCLUSIONS: Our data suggest that presurgery vitamin D levels influence the size of genotype effects of FTO rs9939609 on RYGB surgery-induced weight loss in obese patients.


Subject(s)
Anastomosis, Roux-en-Y , Gastric Bypass , Obesity, Morbid/surgery , Proteins/physiology , Vitamin D/blood , Weight Loss , Adult , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Dietary Supplements , Female , Genotype , Humans , Male , Obesity, Morbid/blood , Obesity, Morbid/complications , Obesity, Morbid/genetics , Polymorphism, Single Nucleotide/physiology , Postoperative Period , Prospective Studies , Proteins/genetics , Vitamin D Deficiency/blood , Vitamin D Deficiency/complications , Vitamin D Deficiency/genetics , Weight Loss/genetics
15.
Gene ; 553(1): 1-6, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25275856

ABSTRACT

The Rhodopsin family is a class of integral membrane proteins belonging to G protein-coupled receptors (GPCRs). To date, several orphan GPCRs are still uncharacterized and in this study we present an anatomical characterization of the GPR162 protein and an attempt to describe its functional role. Our results show that GPR162 is widely expressed in GABAergic as well as other neurons within the mouse hippocampus, whereas extensive expression is observed in areas related to energy homeostasis and hedonic feeding such as hypothalamus, amygdala and ventral tegmental area, regions known to be involved in the regulation of palatable food consumption.


Subject(s)
Amygdala/metabolism , Central Nervous System/metabolism , Hypothalamus/metabolism , Receptors, G-Protein-Coupled/metabolism , Ventral Tegmental Area/metabolism , Amygdala/physiology , Animals , Blotting, Western , Cell Line , Feeding Behavior , Hypothalamus/physiology , Mice , Mice, Inbred C57BL , RNA, Small Interfering/genetics , Receptors, G-Protein-Coupled/genetics , Ventral Tegmental Area/physiology
16.
PLoS Genet ; 10(9): e1004499, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25187989

ABSTRACT

In all animals managing the size of individual meals and frequency of feeding is crucial for metabolic homeostasis. In the current study we demonstrate that the noradrenalin analogue octopamine and the cholecystokinin (CCK) homologue Drosulfakinin (Dsk) function downstream of TfAP-2 and Tiwaz (Twz) to control the number of meals in adult flies. Loss of TfAP-2 or Twz in octopaminergic neurons increased the size of individual meals, while overexpression of TfAP-2 significantly decreased meal size and increased feeding frequency. Of note, our study reveals that TfAP-2 and Twz regulate octopamine signaling to initiate feeding; then octopamine, in a negative feedback loop, induces expression of Dsk to inhibit consummatory behavior. Intriguingly, we found that the mouse TfAP-2 and Twz homologues, AP-2ß and Kctd15, co-localize in areas of the brain known to regulate feeding behavior and reward, and a proximity ligation assay (PLA) demonstrated that AP-2ß and Kctd15 interact directly in a mouse hypothalamus-derived cell line. Finally, we show that in this mouse hypothalamic cell line AP-2ß and Kctd15 directly interact with Ube2i, a mouse sumoylation enzyme, and that AP-2ß may itself be sumoylated. Our study reveals how two obesity-linked homologues regulate metabolic homeostasis by modulating consummatory behavior.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Drosophila melanogaster/physiology , Feeding Behavior/physiology , Meals/physiology , Obesity/metabolism , Obesity/physiopathology , Animals , Cell Line , Feedback , Homeostasis/physiology , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Octopamine/metabolism , Potassium Channels/metabolism , Transcription Factor AP-2/metabolism
17.
Age (Dordr) ; 35(4): 1495-505, 2013 Aug.
Article in English | MEDLINE | ID: mdl-22791395

ABSTRACT

In the present study, we tested whether elderly with a high dietary intake of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) would have higher cognitive test scores and greater brain volume than those with low dietary intake of these fatty acids. Data were obtained from the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) cohort. The dietary intake of EPA and DHA was determined by a 7-day food protocol in 252 cognitively healthy elderly (122 females) at the age of 70 years. At age 75, participants' global cognitive function was examined, and their brain volumes were measured by magnetic resonance imaging (MRI). Three different multivariate linear regression models were applied to test our hypothesis: model A (adjusted for gender and age), model B (additionally controlled for lifestyle factors, e.g., education), and model C (further controlled for cardiometabolic factors, e.g., systolic blood pressure). We found that the self-reported 7-day dietary intake of EPA and DHA at the age of 70 years was positively associated with global gray matter volume (P < 0.05, except for model C) and increased global cognitive performance score (P < 0.05). However, no significant associations were observed between the dietary intake of EPA and DHA and global white matter, total brain volume, and regional gray matter, respectively. Further, no effects were observed when examining cognitively impaired (n = 27) elderly as separate analyses. These cross-sectional findings suggest that dietary intake of EPA and DHA may be linked to improved cognitive health in late life but must be confirmed in patient studies.


Subject(s)
Aging/drug effects , Cerebral Cortex/anatomy & histology , Cognition/physiology , Dietary Supplements , Docosahexaenoic Acids/administration & dosage , Eicosapentaenoic Acid/administration & dosage , Magnetic Resonance Imaging/methods , Aged , Aged, 80 and over , Aging/physiology , Cerebral Cortex/drug effects , Cognition/drug effects , Female , Humans , Male , Organ Size/drug effects , Prospective Studies
18.
PLoS Genet ; 8(3): e1002568, 2012.
Article in English | MEDLINE | ID: mdl-22438821

ABSTRACT

Neurobeachin (Nbea) regulates neuronal membrane protein trafficking and is required for the development and functioning of central and neuromuscular synapses. In homozygous knockout (KO) mice, Nbea deficiency causes perinatal death. Here, we report that heterozygous KO mice haploinsufficient for Nbea have higher body weight due to increased adipose tissue mass. In several feeding paradigms, heterozygous KO mice consumed more food than wild-type (WT) controls, and this consumption was primarily driven by calories rather than palatability. Expression analysis of feeding-related genes in the hypothalamus and brainstem with real-time PCR showed differential expression of a subset of neuropeptide or neuropeptide receptor mRNAs between WT and Nbea+/- mice in the sated state and in response to food deprivation, but not to feeding reward. In humans, we identified two intronic NBEA single-nucleotide polymorphisms (SNPs) that are significantly associated with body-mass index (BMI) in adult and juvenile cohorts. Overall, data obtained in mice and humans suggest that variation of Nbea abundance or activity critically affects body weight, presumably by influencing the activity of feeding-related neural circuits. Our study emphasizes the importance of neural mechanisms in body weight control and points out NBEA as a potential risk gene in human obesity.


Subject(s)
Body Mass Index , Carrier Proteins/genetics , Carrier Proteins/metabolism , Feeding Behavior , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Obesity/genetics , Adipose Tissue/metabolism , Adolescent , Animals , Brain Stem/metabolism , Child , Food Deprivation , Gene Expression Regulation/genetics , Genetic Association Studies , Humans , Hypothalamus/metabolism , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Polymorphism, Single Nucleotide
19.
Pharmacol Biochem Behav ; 100(3): 581-6, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21925200

ABSTRACT

Melanin concentrating hormone (MCH) stimulates feeding driven by energy needs and reward and modifies anxiety behavior. Orexigenic peptides of similar characteristics, including nociceptin/orphanin FQ, Agouti-related protein and opioids, increase consumption also by reducing avoidance of potentially tainted food in animals displaying a conditioned taste aversion (CTA). Herein, using real-time PCR, we assessed whether expression levels of genes encoding MCH and its receptor, MCHR1, were affected in CTA in the rat. We also investigated whether injecting MCH intracerebroventricularly (ICV) during the acquisition and retrieval of LiCl-induced CTA, would alleviate aversive responses. MCHR1 gene was upregulated in the hypothalamus and brain stem of aversive animals, MCH mRNA was significantly higher in the hypothalamus, whereas a strong trend suggesting upregulation of MCH and MCHR1 genes was detected in the amygdala. Despite these expression changes associated with aversion, MCH injected prior to the induction of CTA with LiCl as well as later, during the CTA retrieval upon subsequent presentations of the aversive tastant, did not reduce the magnitude of CTA. We conclude that MCH and its receptor form an orexigenic system whose expression is affected in CTA. This altered MCH expression may contribute to tastant-targeted hypophagia in CTA. However, changing the MCH tone in the brain by exogenous peptide was insufficient to prevent the onset or facilitate extinction of LiCl-induced CTA. This designates MCH as one of many accessory molecules associated with shaping an aversive response, but not a critical one for LiCl-dependent CTA to occur.


Subject(s)
Brain/metabolism , Dysgeusia/metabolism , Gene Expression Regulation , Hypothalamic Hormones/metabolism , Melanins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Pituitary Hormones/metabolism , Receptors, Somatostatin/metabolism , Animals , Brain Stem/metabolism , Conditioning, Psychological , Dysgeusia/drug therapy , Hypothalamic Hormones/administration & dosage , Hypothalamic Hormones/genetics , Hypothalamic Hormones/therapeutic use , Hypothalamus/metabolism , Injections, Intraventricular , Male , Melanins/administration & dosage , Melanins/genetics , Melanins/therapeutic use , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/therapeutic use , Organ Specificity , Pituitary Hormones/administration & dosage , Pituitary Hormones/genetics , Pituitary Hormones/therapeutic use , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, Somatostatin/genetics , Up-Regulation
20.
BMC Neurosci ; 12: 117, 2011 Nov 16.
Article in English | MEDLINE | ID: mdl-22087873

ABSTRACT

BACKGROUND: The Fat mass and obesity gene (FTO) has been identified through genome wide association studies as an important genetic factor contributing to a higher body mass index (BMI). However, the molecular context in which this effect is mediated has yet to be determined. We investigated the potential molecular network for FTO by analyzing co-expression and protein-protein interaction databases, Coxpresdb and IntAct, as well as the functional coupling predicting multi-source database, FunCoup. Hypothalamic expression of FTO-linked genes defined with this bioinformatics approach was subsequently studied using quantitative real time-PCR in mouse feeding models known to affect FTO expression. RESULTS: We identified several candidate genes for functional coupling to FTO through database studies and selected nine for further study in animal models. We observed hypothalamic expression of Profilin 2 (Pfn2), cAMP-dependent protein kinase catalytic subunit beta (Prkacb), Brain derived neurotrophic factor (Bdnf), neurotrophic tyrosine kinase, receptor, type 2 (Ntrk2), Signal transducer and activator of transcription 3 (Stat3), and Btbd12 to be co-regulated in concert with Fto. Pfn2 and Prkacb have previously not been linked to feeding regulation. CONCLUSIONS: Gene expression studies validate several candidates generated through database studies of possible FTO-interactors. We speculate about a wider functional role for FTO in the context of current and recent findings, such as in extracellular ligand-induced neuronal plasticity via NTRK2/BDNF, possibly via interaction with the transcription factor CCAAT/enhancer binding protein ß (C/EBPß).


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Hypothalamus/metabolism , Membrane Glycoproteins/genetics , Mixed Function Oxygenases/genetics , Obesity/genetics , Oxo-Acid-Lyases/genetics , Protein-Tyrosine Kinases/genetics , Signal Transduction/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Animals , Computational Biology/methods , Genome-Wide Association Study/methods , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL