Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Endocrinology ; 163(6)2022 06 01.
Article in English | MEDLINE | ID: mdl-35352108

ABSTRACT

INTRODUCTION: Liver-expressed antimicrobial peptide-2 (LEAP2) is an endogenous ghrelin receptor antagonist, which is upregulated in the fed state and downregulated during fasting. We hypothesized that the ketone body beta-hydroxybutyrate (BHB) is involved in the downregulation of LEAP2 during conditions with high circulating levels of BHB. METHODS: Hepatic and intestinal Leap2 expression were determined in 3 groups of mice with increasing circulating levels of BHB: prolonged fasting, prolonged ketogenic diet, and oral BHB treatment. LEAP2 levels were measured in lean and obese individuals, in human individuals following endurance exercise, and in mice after BHB treatment. Lastly, we investigated Leap2 expression in isolated murine hepatocytes challenged with BHB. RESULTS: We confirmed increased circulating LEAP2 levels in individuals with obesity compared to lean individuals. The recovery period after endurance exercise was associated with increased plasma levels of BHB levels and decreased LEAP2 levels in humans. Leap2 expression was selectively decreased in the liver after fasting and after exposure to a ketogenic diet for 3 weeks. Importantly, we found that oral administration of BHB increased circulating levels of BHB in mice and decreased Leap2 expression levels and circulating LEAP2 plasma levels, as did Leap2 expression after direct exposure to BHB in isolated murine hepatocytes. CONCLUSION: From our data, we suggest that LEAP2 is downregulated during different states of energy deprivation in both humans and rodents. Furthermore, we here provide evidence that the ketone body, BHB, which is highly upregulated during fasting metabolism, directly downregulates LEAP2 levels. This may be relevant in ghrelin receptor-induced hunger signaling during energy deprivation.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Diet, Ketogenic , Receptors, Ghrelin , 3-Hydroxybutyric Acid/metabolism , Animals , Ghrelin/metabolism , Liver/metabolism , Mice , Obesity/metabolism , Receptors, Ghrelin/metabolism
2.
Front Endocrinol (Lausanne) ; 12: 734547, 2021.
Article in English | MEDLINE | ID: mdl-34646236

ABSTRACT

Anorexia Nervosa (AN) is a complex disease that impairs the metabolic, mental and physiological health of affected individuals in a severe and sometimes lethal way. Many of the common symptoms in AN patients, such as reduced food intake, anxiety, impaired gut motility or overexercising are connected to both the orexigenic gut hormone ghrelin and the dopaminergic system. Targeting the ghrelin receptor (GhrR) to treat AN seems a promising possibility in current research. However, GhrR signaling is highly complex. First, the GhrR can activate four known intracellular pathways Gαq, Gαi/o, Gα12/13 and the recruitment of ß-arrestin. Biased signaling provides the possibility to activate or inhibit only one or a subset of the intracellular pathways of a pleiotropic receptor. This allows specific targeting of physiological functions without adverse effects. Currently little is known on how biased signaling could specifically modulate GhrR effects. Second, GhrR signaling has been shown to be interconnected with the dopaminergic system, particularly in the context of AN symptoms. This review highlights that a biased agonist for the GhrR may be a promising target for the treatment of AN, however extensive and systematic translational studies are still needed and the connection to the dopaminergic system has to be taken into account.


Subject(s)
Anorexia Nervosa/metabolism , Anorexia Nervosa/psychology , Dopamine/metabolism , Receptors, Ghrelin/metabolism , Animals , Anorexia Nervosa/complications , Anorexia Nervosa/genetics , Eating/physiology , Energy Metabolism/genetics , Feeding Behavior/physiology , Ghrelin/metabolism , Humans , Insulin Resistance/physiology , Signal Transduction/physiology
3.
Mol Metab ; 49: 101207, 2021 07.
Article in English | MEDLINE | ID: mdl-33711555

ABSTRACT

OBJECTIVES: Obesity is a complex disease associated with a high risk of comorbidities. Gastric bypass surgery, an invasive procedure with low patient eligibility, is currently the most effective intervention that achieves sustained weight loss. This beneficial effect is attributed to alterations in gut hormone signaling. An attractive alternative is to pharmacologically mimic the effects of bariatric surgery by targeting several gut hormonal axes. The G protein-coupled receptor 39 (GPR39) expressed in the gastrointestinal tract has been shown to mediate ghrelin signaling and control appetite, food intake, and energy homeostasis, but the broader effect on gut hormones is largely unknown. A potent and efficacious GPR39 agonist (Cpd1324) was recently discovered, but the in vivo function was not addressed. Herein we studied the efficacy of the GPR39 agonist, Cpd1324, on metabolism and gut hormone secretion. METHODS: Body weight, food intake, and energy expenditure in GPR39 agonist-treated mice and GPR39 KO mice were studied in calorimetric cages. Plasma ghrelin, glucose-dependent insulinotropic polypeptide (GIP), glucagon-like peptide-1 (GLP-1), and peptide YY (PYY) levels were measured. Organoids generated from murine and human small intestine and mouse colon were used to study GLP-1 and PYY release. Upon GPR39 agonist administration, dynamic changes in intracellular GLP-1 content were studied via immunostaining and changes in ion transport across colonic mucosa were monitored in Ussing chambers. The G protein activation underlying GPR39-mediated selective release of gut hormones was studied using bioluminescence resonance energy transfer biosensors. RESULTS: The GPR39 KO mice displayed a significantly increased food intake without corresponding increases in respiratory exchange ratios or energy expenditure. Oral administration of a GPR39 agonist induced an acute decrease in food intake and subsequent weight loss in high-fat diet (HFD)-fed mice without affecting their energy expenditure. The tool compound, Cpd1324, increased GLP-1 secretion in the mice as well as in mouse and human intestinal organoids, but not in GPR39 KO mouse organoids. In contrast, the GPR39 agonist had no effect on PYY or GIP secretion. Transepithelial ion transport was acutely affected by GPR39 agonism in a GLP-1- and calcitonin gene-related peptide (CGRP)-dependent manner. Analysis of Cpd1324 signaling properties showed activation of Gαq and Gαi/o signaling pathways in L cells, but not Gαs signaling. CONCLUSIONS: The GPR39 agonist described in this study can potentially be used by oral administration as a weight-lowering agent due to its stimulatory effect on GLP-1 secretion, which is most likely mediated through a unique activation of Gα subunits. Thus, GPR39 agonism may represent a novel approach to effectively treat obesity through selective modulation of gastrointestinal hormonal axes.


Subject(s)
Gastrointestinal Hormones/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Animals , Appetite Regulation , Bariatric Surgery , Body Weight , Eating , Enteroendocrine Cells , Gastric Inhibitory Polypeptide/pharmacology , Ghrelin/metabolism , Glucagon-Like Peptide 1/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Peptide YY/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Gastrointestinal Hormone , Weight Loss
4.
Mol Metab ; 47: 101174, 2021 05.
Article in English | MEDLINE | ID: mdl-33549847

ABSTRACT

OBJECTIVE: The goal of this study was to investigate the importance of central hormone-sensitive lipase (HSL) expression in the regulation of food intake and body weight in mice to clarify whether intracellular lipolysis in the mammalian hypothalamus plays a role in regulating appetite. METHODS: Using pharmacological and genetic approaches, we investigated the role of HSL in the rodent brain in the regulation of feeding and energy homeostasis under basal conditions during acute stress and high-fat diet feeding. RESULTS: We found that HSL, a key enzyme in the catabolism of cellular lipid stores, is expressed in the appetite-regulating centers in the hypothalamus and is activated by acute stress through a mechanism similar to that observed in adipose tissue and skeletal muscle. Inhibition of HSL in rodent models by a synthetic ligand, global knockout, or brain-specific deletion of HSL prevents a decrease in food intake normally seen in response to acute stress and is associated with the increased expression of orexigenic peptides neuropeptide Y (NPY) and agouti-related peptide (AgRP). Increased food intake can be reversed by adeno-associated virus-mediated reintroduction of HSL in neurons of the mediobasal hypothalamus. Importantly, metabolic stress induced by a high-fat diet also enhances the hyperphagic phenotype of HSL-deficient mice. Specific deletion of HSL in the ventromedial hypothalamic nucleus (VMH) or AgRP neurons reveals that HSL in the VMH plays a role in both acute stress-induced food intake and high-fat diet-induced obesity. CONCLUSIONS: Our results indicate that HSL activity in the mediobasal hypothalamus is involved in the acute reduction in food intake during the acute stress response and sensing of a high-fat diet.


Subject(s)
Appetite/physiology , Homeostasis , Hypothalamus/metabolism , Sterol Esterase/metabolism , Agouti-Related Protein/metabolism , Animals , Body Weight , Diet, High-Fat/adverse effects , Eating , Energy Metabolism , Female , Hyperphagia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Neuropeptide Y/metabolism , Obesity/metabolism , RNA Splicing Factors , Sterol Esterase/genetics , Stress, Physiological/genetics , Transcriptome
5.
Acta Physiol (Oxf) ; 228(4): e13437, 2020 04.
Article in English | MEDLINE | ID: mdl-31900990

ABSTRACT

AIM: Neurons in the arcuate nucleus of the hypothalamus are involved in regulation of food intake and energy expenditure, and dysregulation of signalling in these neurons promotes development of obesity. The role of the rate-limiting enzyme in the NAD+ salvage pathway, nicotinamide phosphoribosyltransferase (NAMPT), for regulation energy homeostasis by the hypothalamus has not been extensively studied. METHODS: We determined whether Nampt mRNA or protein levels in the hypothalamus of mice were affected by diet-induced obesity, by fasting and re-feeding, and by leptin and ghrelin treatment. Primary hypothalamic neurons were treated with FK866, a selective inhibitor of NAMPT, or rAAV carrying shRNA directed against Nampt, and levels of reactive oxygen species (ROS) and mitochondrial respiration were assessed. Fasting and ghrelin-induced food intake was measured in mice in metabolic cages after intracerebroventricular (ICV)-mediated FK866 administration. RESULTS: NAMPT levels in the hypothalamus were elevated by administration of ghrelin and leptin. In diet-induced obese mice, both protein and mRNA levels of NAMPT decreased in the hypothalamus. NAMPT inhibition in primary hypothalamic neurons significantly reduced levels of NAD+ , increased levels of ROS, and affected the expression of Agrp, Pomc and genes related to mitochondrial function. Finally, ICV-induced NAMPT inhibition by FK866 did not cause malaise or anhedonia, but completely ablated fasting- and ghrelin-induced increases in food intake. CONCLUSION: Our findings indicate that regulation of NAMPT levels in hypothalamic neurons is important for the control of fasting- and ghrelin-induced food intake.


Subject(s)
Fasting/metabolism , Ghrelin/metabolism , Hypothalamus/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Acrylamides/administration & dosage , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Animals , Cell Line , Eating , Female , Male , Mice , Mice, Inbred C57BL , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Piperidines/administration & dosage , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism
6.
J Neuroendocrinol ; 31(7): e12761, 2019 07.
Article in English | MEDLINE | ID: mdl-31237372

ABSTRACT

Dopamine-producing tyrosine hydroxylase (TH) neurones in the hypothalamic arcuate nucleus (ARC) have recently been shown to be involved in ghrelin signalling and body weight homeostasis. In the present study, we investigate the role of the intracellular regulator RhoA in hypothalamic TH neurones in response to peripheral hormones. Diet-induced obesity was found to be associated with increased phosphorylation of TH in ARC, indicating obesity-associated increased activity of ARC TH neurones. Mice in which RhoA was specifically knocked out in TH neurones (TH-RhoA-/- mice) were more sensitive to the orexigenic effect of peripherally administered ghrelin and displayed an abolished response to the anorexigenic hormone leptin. When TH-RhoA-/- mice were challenged with a high-fat high-sucrose (HFHS) diet, they became hyperphagic and gained more body weight and fat mass compared to wild-type control mice. Importantly, lack of RhoA prevented development of ghrelin resistance, which is normally observed in wild-type mice after long-term HFHS diet feeding. Patch-clamp electrophysiological analysis demonstrated increased ghrelin-induced excitability of TH neurones in lean TH-RhoA-/- mice compared to lean littermate control animals. Additionally, increased expression of the orexigenic hypothalamic neuropeptides agouti-related peptide and neuropeptide Y was observed in TH-RhoA-/- mice. Overall, our data indicate that TH neurones in ARC are important for the regulation of body weight homeostasis and that RhoA is both a central effector in these neurones and important for the development of obesity-induced ghrelin resistance. The obese phenotype of TH-RhoA-/- mice may be a result of increased sensitivity to ghrelin and decreased sensitivity to leptin, resulting in increased food intake.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Body Weight , Eating , Ghrelin/metabolism , Neurons/metabolism , Tyrosine 3-Monooxygenase/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Female , Gene Expression , Male , Mice, Knockout , Obesity/metabolism , RNA, Messenger/metabolism , rhoA GTP-Binding Protein/genetics
7.
Diabetes ; 68(6): 1329-1340, 2019 06.
Article in English | MEDLINE | ID: mdl-30936142

ABSTRACT

Neurotensin (NT), a gut hormone and neuropeptide, increases in circulation after bariatric surgery in rodents and humans and inhibits food intake in mice. However, its potential to treat obesity and the subsequent metabolic dysfunctions have been difficult to assess owing to its short half-life in vivo. Here, we demonstrate that a long-acting, pegylated analog of the NT peptide (P-NT) reduces food intake, body weight, and adiposity in diet-induced obese mice when administered once daily for 6 days. Strikingly, when P-NT was combined with the glucagon-like peptide 1 mimetic liraglutide, the two peptides synergized to reduce food intake and body weight relative to each monotherapy, without inducing a taste aversion. Further, P-NT and liraglutide coadministration improved glycemia and reduced steatohepatitis. Finally, we show that the melanocortin pathway is central for P-NT-induced anorexia and necessary for the full synergistic effect of P-NT and liraglutide combination therapy. Overall, our data suggest that P-NT and liraglutide combination therapy could be an enhanced treatment for obesity with improved tolerability compared with liraglutide monotherapy.


Subject(s)
Adiposity/drug effects , Blood Glucose/drug effects , Body Weight/drug effects , Eating/drug effects , Hypoglycemic Agents/pharmacology , Liraglutide/pharmacology , Neurotensin/pharmacology , Obesity/metabolism , Animals , Blood Glucose/metabolism , Delayed-Action Preparations , Drug Synergism , Fatty Liver/metabolism , Fatty Liver/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Melanocortins/metabolism , Mice , Mice, Knockout , Polyethylene Glycols
8.
Phys Chem Chem Phys ; 20(45): 28678-28684, 2018 Nov 21.
Article in English | MEDLINE | ID: mdl-30406792

ABSTRACT

A model system of the flavin chromophore was synthesized and investigated for its intrinsic optical properties by gas phase action spectroscopy using an ion storage ring. An ammonium group was anchored to this flavin chromophore to allow its transfer to the gas phase by electrospray ionization and for studying the influence of hydrogen bonding and a nearby positive charge. According to calculations one of the hydrogen atoms of the ammonium group favorably forms an intramolecular ionic hydrogen bond to one of the oxygen atoms of the flavin chromophore, and this interaction was found to cause a blueshift of the S0 → S1 transition and a redshift of the S0 → S2 transition. For comparison, the S0 → S1 transition shows little solvent dependence (only in regard to the degree of fine structure). In addition, the influence of protonation of the flavin chromophore was elucidated by experimental and theoretical studies of a simple flavin system. While the position of the S0 → S1 absorption was at identical positions in the gas phase for the intramolecularly hydrogen-bonded and protonated flavin systems, the S0 → S2 absorption was further redshifted for the protonated species. This redshift resulting from protonation was also observed in solution.

9.
Proc Natl Acad Sci U S A ; 115(43): E10255-E10264, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30301804

ABSTRACT

Biased signaling has been suggested as a means of selectively modulating a limited fraction of the signaling pathways for G-protein-coupled receptor family members. Hence, biased ligands may allow modulation of only the desired physiological functions and not elicit undesired effects associated with pharmacological treatments. The ghrelin receptor is a highly sought antiobesity target, since the gut hormone ghrelin in humans has been shown to increase both food intake and fat accumulation. However, it also modulates mood, behavior, growth hormone secretion, and gastric motility. Thus, blocking all pathways of this receptor may give rise to potential side effects. In the present study, we describe a highly promiscuous signaling capacity for the ghrelin receptor. We tested selected ligands for their ability to regulate the various pathways engaged by the receptor. Among those, a biased ligand, YIL781, was found to activate the Gαq/11 and Gα12 pathways selectively without affecting the engagement of ß-arrestin or other G proteins. YIL781 was further characterized for its in vivo physiological functions. In combination with the use of mice in which Gαq/11 was selectively deleted in the appetite-regulating AgRP neurons, this biased ligand allowed us to demonstrate that selective blockade of Gαq/11, without antagonism at ß-arrestin or other G-protein coupling is sufficient to decrease food intake.


Subject(s)
Ghrelin/metabolism , Receptors, Ghrelin/metabolism , Animals , Eating/drug effects , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Ligands , Male , Mice , Piperidines/pharmacology , Quinazolinones/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , beta-Arrestins/metabolism
10.
Eur J Med Chem ; 144: 887-897, 2018 Jan 20.
Article in English | MEDLINE | ID: mdl-29329072

ABSTRACT

Neuromedin U (NMU) is a highly conserved endogenous peptide that is involved in a wide range of physiological processes such as regulation of feeding behavior, the stress response and nociception. The major limitation to use NMU as a therapeutic is its short half-life. Here, we describe the development of a set of novel NMU-analogs based on NMU-8, by introducing unnatural amino acids into the native sequence. This approach shows that it is possible to generate molecules with increased potency and improved plasma stability without major changes of the peptidic nature or the introduction of large conjugates. When compared to the native NMU-8 peptide, compounds 16, 18 and 20 have potent agonist activity and affinity for both NMU receptors. Selectivity towards NMUR1 was observed when the Phe residue in position 4 was modified, whereas higher potencies at NMUR2 were found when substitutions of the Pro residue in position 6 were executed. To study the effect of the modifications on the proteolytic stability of the molecules, an in vitro stability assay in human plasma at 37 °C was performed. All analyzed analogs possessed an increased resistance against enzymatic degradation in human plasma resulting in half-lifes from 4 min for NMU-8, up to more than 23 h for compound 42.


Subject(s)
Neuropeptides/pharmacology , Proteolysis/drug effects , Receptors, Neurotransmitter/agonists , Dose-Response Relationship, Drug , Humans , Molecular Structure , Neuropeptides/chemical synthesis , Neuropeptides/chemistry , Structure-Activity Relationship
11.
Int J Mol Sci ; 18(5)2017 Apr 26.
Article in English | MEDLINE | ID: mdl-28445429

ABSTRACT

Ghrelin receptor (Ghr-R) signaling in neurons of the ventral tegmental area (VTA) can modulate dopaminergic function and the reward-related effects of both palatable foods and drugs of abuse. In this study, we re-introduced the Ghr-R in VTA neurons in Ghr-R knockout mice (Ghr-RVTA mice) to specifically study the importance of the constitutively active Ghr-R for VTA neuronal signaling. Our results showed that re-introduction of the Ghr-R in the VTA had no impact on body weight or food intake under basal conditions. However, during novel environment stress Ghr-RVTA mice showed increased food intake and energy expenditure compared to Ghr-R knockout mice, demonstrating the significance of Ghr-R signaling in the response to stress. Ghr-RVTA mice also showed increased cocaine-induced locomotor activity compared to Ghr-R knockout mice, highlighting the importance of ghrelin signaling for the reward-related effects of activation of VTA neurons. Overall, our data suggest that re-introduction of the Ghr-R in the mesolimbic reward system of Ghr-R knockout mice increases the level of activation induced by both cocaine and novelty stress.


Subject(s)
Behavior, Animal/physiology , Receptors, Ghrelin/metabolism , Ventral Tegmental Area/metabolism , Animals , Body Weight , Dependovirus/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Eating , Energy Metabolism , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Locomotion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygen Consumption , Receptors, Dopamine D2/metabolism , Receptors, Ghrelin/deficiency , Receptors, Ghrelin/genetics , Tyrosine 3-Monooxygenase/metabolism
12.
J Immunol ; 198(8): 3118-3126, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28258200

ABSTRACT

Congenital heart defects are a major cause of perinatal mortality and morbidity, affecting >1% of all live births in the Western world, yet a large fraction of such defects have an unknown etiology. Recent studies demonstrated surprising dual roles for immune-related molecules and their effector mechanisms during fetal development and adult homeostasis. In this article, we describe the function of an endogenous complement inhibitor, mannan-binding lectin (MBL)-associated protein (MAp)44, in regulating the composition of a serine protease-pattern recognition receptor complex, MBL-associated serine protease (MASP)-3/collectin-L1/K1 hetero-oligomer, which impacts cardiac neural crest cell migration. We used knockdown and rescue strategies in zebrafish, a model allowing visualization and assessment of heart function, even in the presence of severe functional defects. Knockdown of embryonic expression of MAp44 caused impaired cardiogenesis, lowered heart rate, and decreased cardiac output. These defects were associated with aberrant neural crest cell behavior. We found that MAp44 competed with MASP-3 for pattern recognition molecule interaction, and knockdown of endogenous MAp44 expression could be rescued by overexpression of wild-type MAp44. Our observations provide evidence that immune molecules are centrally involved in the orchestration of cardiac tissue development.


Subject(s)
Heart/embryology , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Animals , Gene Knockdown Techniques , Humans , In Situ Hybridization , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Zebrafish , Zebrafish Proteins/metabolism
13.
Forensic Sci Int ; 268: 39-45, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27685474

ABSTRACT

Reference concentrations are needed to evaluate postmortem toxicology results and usually femoral blood is the specimen of choice. However, brain tissue has been suggested as a viable alternative specimen, since postmortem blood concentrations can be difficult to interpret due to postmortem redistribution, among other factors. Here we present reference concentrations of postmortem brain and femoral blood of the nitrobenzodiazepines clonazepam, flunitrazepam, and nitrazepam that are of particular interest since they commonly are converted to their corresponding 7-aminometabolites in the postmortem situation. The drugs and metabolites were quantified in both matrices using LC-MS-MS in 69 cases. In 63 cases the compounds were judged not to have been of significance for the death (C cases), whereas they were considered to have been a contributing factor in 6 cases (B cases). No cases were observed with a nitrobenzodiazepine being the sole cause of death (A cases). The brain-blood ratios for clonazepam and nitrazepam were 5.5 and 4.7, respectively, while the brain-blood ratios for the 7-aminometabolites ranged from 0.4 to 0.5. Flunitrazepam only occurred as the 7-aminometabolite. A positive correlation between brain and blood concentrations was found with Spearman's rank correlation coefficients (rs) ranging from 0.77 to 0.96. The measured femoral blood concentrations agree with literature values, but only few brain concentrations were available for comparison. The drug-metabolite ratios for clonazepam and nitrazepam were 10-12 times higher in brain than in blood. The pre-analytical variation in brain of 5.9% was fairly low, suggesting that brain tissue is a useful alternative to blood. The reported brain and femoral blood concentrations serve as reference values in postmortem investigations.


Subject(s)
Brain Chemistry , Clonazepam/analysis , Flunitrazepam/analysis , Nitrazepam/analysis , Postmortem Changes , Adolescent , Adult , Aged , Chromatography, Liquid , Clonazepam/analogs & derivatives , Female , Forensic Toxicology , Humans , Male , Middle Aged , Reference Values , Tandem Mass Spectrometry , Young Adult
14.
Endocrinology ; 157(9): 3482-92, 2016 09.
Article in English | MEDLINE | ID: mdl-27580810

ABSTRACT

Neurotensin (NT) is a peptide expressed in the brain and in the gastrointestinal tract. Brain NT inhibits food intake, but the effects of peripheral NT are less investigated. In this study, peripheral NT decreased food intake in both mice and rats, which was abolished by a NT antagonist. Using c-Fos immunohistochemistry, we found that peripheral NT activated brainstem and hypothalamic regions. The anorexigenic effect of NT was preserved in vagotomized mice but lasted shorter than in sham-operated mice. This in combination with a strong increase in c-Fos activation in area postrema after ip administration indicates that NT acts both through the blood circulation and the vagus. To improve the pharmacokinetics of NT, we developed a pegylated NT peptide, which presumably prolonged the half-life, and thus, the effect on feeding was extended compared with native NT. On a molecular level, the pegylated NT peptide increased proopiomelanocortin mRNA in the arcuate nucleus. We also investigated the importance of NT for the decreased food intake after gastric bypass surgery in a rat model of Roux-en-Y gastric bypass (RYGB). NT was increased in plasma and in the gastrointestinal tract in RYGB rats, and pharmacological antagonism of NT increased food intake transiently in RYGB rats. Taken together, our data suggest that NT is a metabolically active hormone, which contributes to the regulation of food intake.


Subject(s)
Appetite Regulation/drug effects , Gastric Bypass , Neurotensin/administration & dosage , Animals , Eating/drug effects , Female , Male , Mice, Inbred C57BL , Neurotensin/antagonists & inhibitors , Neurotensin/blood , Rats, Sprague-Dawley , Sucrose , Vagotomy
15.
J Anal Toxicol ; 40(7): 529-36, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27416838

ABSTRACT

To interpret postmortem toxicology results, reference concentrations for non-toxic and toxic levels are needed. Usually, measurements are performed in blood, but because of postmortem redistribution phenomena this may not be optimal. Rather, measurement in the target organ of psychoactive drugs, the brain, might be considered. Here we present reference concentrations of femoral blood and brain tissue of selected benzodiazepines (BZDs). Using LC-MS/MS, we quantified alprazolam, bromazepam, chlordiazepoxide, diazepam, and the metabolites desmethyldiazepam, oxazepam and temazepam in postmortem femoral blood and brain tissue in 104 cases. BZDs were judged to be unrelated to the cause of death in 88 cases and contributing to death in 16 cases. No cases were found with cause of death solely attributed to BZD poisoning. All BZDs investigated tended to have higher concentrations in brain than in blood with median brain-blood ratios ranging from 1.1 to 2.3. A positive correlation between brain and blood concentrations was found with R(2) values from 0.51 to 0.95. Our reported femoral blood concentrations concur with literature values, but sparse information on brain concentration was available. Drug-metabolite ratios were similar in brain and blood for most compounds. Duplicate measurements of brain samples showed that the pre-analytical variation in brain (5.9%) was relatively low, supporting the notion that brain tissue is a suitable postmortem specimen. The reported concentrations in both brain and blood can be used as reference values when evaluating postmortem cases.


Subject(s)
Alprazolam/metabolism , Blood-Brain Barrier/metabolism , Bromazepam/metabolism , Chlordiazepoxide/metabolism , Diazepam/metabolism , Forensic Toxicology , Humans
16.
Article in English | MEDLINE | ID: mdl-26578081

ABSTRACT

BACKGROUND: Besides the well-known effects of ghrelin on adiposity and food intake regulation, the ghrelin system has been shown to regulate aspects of behavior including anxiety and stress. However, the effect of virus-mediated overexpression of the ghrelin receptor in the amygdala has not previously been addressed directly. METHODS: First, we examined the acute effect of peripheral ghrelin administration on anxiety- and depression-like behavior using the open field, elevated plus maze, forced swim, and tail suspension tests. Next, we examined the effect of peripheral ghrelin administration and ghrelin receptor deficiency on stress in a familiar and social environment using the Intellicage system. Importantly, we also used a novel approach to study ghrelin receptor signaling in the brain by overexpressing the ghrelin receptor in the amygdala. We examined the effect of ghrelin receptor overexpression on anxiety-related behavior before and after acute stress and measured the modulation of serotonin receptor expression. RESULTS: We found that ghrelin caused an anxiolytic-like effect in both the open field and elevated plus maze tests. Additionally, it attenuated air-puff-induced stress in the social environment, while the opposite was shown in ghrelin receptor deficient mice. Finally, we found that overexpression of the ghrelin receptor in the basolateral division of the amygdala caused an anxiolytic-like effect and decreased the 5HT1a receptor expression. CONCLUSIONS: Ghrelin administration and overexpression of the ghrelin receptor in the amygdala induces anxiolytic-like behavior. Since the ghrelin receptor has high constitutive activity, ligand-independent signaling in vivo may be important for the observed anxiolytic-like effects. The anxiolytic effects seem to be mediated independently from the HPA axis, potentially engaging the central serotonin system.


Subject(s)
Amygdala/drug effects , Anti-Anxiety Agents/pharmacology , Anxiety/prevention & control , Behavior, Animal/drug effects , Ghrelin/pharmacology , Receptors, Ghrelin/agonists , Signal Transduction/drug effects , Amygdala/metabolism , Amygdala/physiopathology , Animals , Anxiety/genetics , Anxiety/metabolism , Anxiety/psychology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Hindlimb Suspension , Humans , Locomotion/drug effects , Male , Maze Learning/drug effects , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Ghrelin/genetics , Receptors, Ghrelin/metabolism , Social Behavior , Stress, Psychological/complications , Stress, Psychological/metabolism , Stress, Psychological/psychology , Swimming , Time Factors
17.
J Anal Toxicol ; 39(7): 557-61, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26159868

ABSTRACT

Brain tissue is a useful alternative to blood in postmortem forensic investigations, but scarcity of information on reference concentrations in brain tissue makes interpretation challenging. Here we present a study of 43 cases where the antipsychotic drug quetiapine was quantified in brain tissue and related to concentrations in postmortem blood. For cases, where quetiapine was unrelated to the cause of death (N = 36), the 10-90 percentiles for quetiapine concentrations in brain tissue were 0.030-1.54 mg/kg (median 0.48 mg/kg, mean 0.79 mg/kg). Corresponding blood 10-90 percentile values were 0.007-0.39 mg/kg (median 0.15 mg/kg, mean 0.19 mg/kg), giving brain-blood ratio 10-90 percentiles of 2.31-6.54 (median 3.87, mean 4.32). Both correspond well to the limited amount of data found in the literature. For cases where quetiapine was a contributing factor to death (N = 5), the median value in brain tissue of 8.02 mg/kg (range 2.69-22.98 mg/kg) was more than 15 times higher than the median of the nontoxic values, and about the same relationship occurred for blood with a median of 3.19 mg/kg (range 1.00-6.90 mg/kg). The brain-blood ratios for toxic concentrations were in the range of 2.08-6.05, which correspond to those of the nontoxic concentrations. A single case, where quetiapine was ruled as the sole cause of death, a suicide by quetiapine overdose, had an even higher value of 25.74 mg/kg in brain tissue. The blood concentration was 8.99 mg/kg, giving a brain-blood ratio of 2.86. Thus, on average the brain concentrations were about four times the blood concentrations. The brain concentrations of quetiapine observed in cases, where quetiapine was unrelated to death, may serve as a reference, when evaluating postmortem cases with no blood available. The recorded concentrations, where quetiapine was contributing to death, give an indication of likely toxic concentrations.


Subject(s)
Antipsychotic Agents/blood , Brain/metabolism , Drug Overdose/blood , Forensic Toxicology/standards , Quetiapine Fumarate/blood , Substance Abuse Detection/standards , Antipsychotic Agents/poisoning , Autopsy , Calibration , Cause of Death , Chromatography, High Pressure Liquid/standards , Drug Overdose/diagnosis , Drug Overdose/mortality , Humans , Predictive Value of Tests , Quetiapine Fumarate/poisoning , Reference Standards , Suicide , Tandem Mass Spectrometry/standards
18.
Am J Physiol Regul Integr Comp Physiol ; 308(11): R973-82, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25855307

ABSTRACT

To characterize mechanisms responsible for fat accumulation we used a selectively bred obesity-prone (OP) and obesity-resistant (OR) rat model where the rats were fed a Western diet for 76 days. Body composition was assessed by magnetic resonance imaging scans, and as expected, the OP rats developed a higher degree of fat accumulation compared with OR rats. Indirect calorimetry showed that the OP rats had higher respiratory exchange ratio (RER) compared with OR rats, indicating an impaired ability to oxidize fat. The OP rats had lower expression of carnitine palmitoyltransferase 1b in intra-abdominal fat, and higher expression of stearoyl-CoA desaturase 1 in subcutaneous fat compared with OR rats, which could explain the higher fat accumulation and RER values. Basal metabolic parameters were also examined in juvenile OP and OR rats before and during the introduction of the Western diet. Juvenile OP rats likewise had higher RER values, indicating that this trait may be a primary and contributing factor to their obese phenotype. When the adult obese rats were exposed to the orexigenic and adipogenic hormone ghrelin, we observed increased RER values in both OP and OR rats, while OR rats were more sensitive to the orexigenic effects of ghrelin as well as ghrelin-induced attenuation of activity and energy expenditure. Thus increased fat accumulation characterizing obesity may be caused by impaired oxidative capacity due to decreased carnitine palmitoyltransferase 1b levels in the white adipose tissue, whereas ghrelin sensitivity did not seem to be a contributing factor.


Subject(s)
Adiposity , Carnitine O-Palmitoyltransferase/metabolism , Energy Metabolism , Intra-Abdominal Fat/enzymology , Obesity/enzymology , Subcutaneous Fat/enzymology , Adiposity/drug effects , Animals , Calorimetry, Indirect , Carnitine O-Palmitoyltransferase/genetics , Diet, High-Fat , Disease Models, Animal , Down-Regulation , Eating , Energy Metabolism/drug effects , Gene Expression Regulation , Ghrelin/administration & dosage , Hypothalamus/enzymology , Insulin/blood , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/physiopathology , Leptin/blood , Magnetic Resonance Imaging , Male , Muscle, Skeletal/enzymology , Obesity/blood , Obesity/etiology , Obesity/genetics , Obesity/physiopathology , Oxidation-Reduction , Rats , Subcutaneous Fat/drug effects , Subcutaneous Fat/physiopathology , Time Factors , Weight Gain
19.
FEBS J ; 282(4): 803-16, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25557436

ABSTRACT

Calmodulin (CaM) is the central mediator of intracellular Ca(2+) signalling in cardiomyocytes, where it conveys the intricate Ca(2+) transients to the proteins controlling cardiac contraction. We recently linked two separate mutations in CaM (N53I and N97S) to dominantly inherited catecholaminergic polymorphic ventricular tachycardia (CPVT), an arrhythmic disorder in which exercise or acute emotion can lead to syncope and sudden cardiac death. Given the ubiquitous presence of CaM in all eukaryote cells, it is particular intriguing that carriers of either mutation show no additional symptoms. Here, we investigated the effects of the CaM CPVT mutations in a zebrafish animal model. Three-day-old embryos injected with either CaM mRNA showed no detectable pathologies or developmental abnormalities. However, embryos injected with CPVT CaM mRNA displayed increased heart rate compared to wild-type CaM mRNA under ß-adrenergic stimulation, demonstrating a conserved dominant cardiac specific effect between zebrafish and human carriers of these mutations. Motivated by the highly similar physiological phenotypes, we compared the effects of the N53I and N97S mutations on the biophysical and functional properties of CaM. Surprisingly, the mutations have opposing effects on CaM C-lobe Ca(2+) binding affinity and kinetics, and changes to the CaM N-lobe Ca(2+) binding are minor and specific to the N53I mutation. Furthermore, both mutations induce differential perturbations to structure and stability towards unfolding. Our results suggest different molecular disease mechanisms for the CPVT (N53I and N97S mutations) and strongly support that cardiac contraction is the physiological process most sensitive to CaM integrity.


Subject(s)
Calmodulin/chemistry , Calmodulin/metabolism , Tachycardia, Ventricular/metabolism , Zebrafish Proteins/chemistry , Zebrafish Proteins/metabolism , Animals , Calcium Signaling/genetics , Calcium Signaling/physiology , Calmodulin/genetics , Mutation , Protein Folding , Tachycardia, Ventricular/genetics , Zebrafish , Zebrafish Proteins/genetics
20.
J Anal Toxicol ; 39(1): 41-4, 2015.
Article in English | MEDLINE | ID: mdl-25342720

ABSTRACT

Postmortem femoral blood concentrations of the antipsychotic drugs aripiprazole, chlorprothixene and its metabolite, and quetiapine were determined by LC-MS-MS in 25 cases for aripiprazole and 60 cases each for chlorprothixene and quetiapine. For cases where the cause of death was not related to the considered drugs, the following blood concentration intervals (10-90 percentiles) were observed: 0.049-0.69 mg/kg for aripiprazole, 0.006-0.24 mg/kg for chlorprothixene, and 0.006-0.37 mg/kg for quetiapine. These concentration ranges largely correspond to therapeutic plasma levels observed in vivo suggesting no or only limited postmortem redistribution for aripiprazole, chlorprothixene with metabolite, and quetiapine in these cases. One fatality caused by chlorprothixene with a blood level of 0.90 mg/kg was recorded, and in six cases chlorprothixene was judged to be contributing to death with concentrations 0.43-0.91 mg/kg. No fatalities exclusively ascribed to the two other drugs were observed, but aripiprazole was considered to be contributing to death in one case (1.9 mg/kg) and quetiapine in seven cases with concentrations 0.35-10.0 mg/kg. The presented values may serve as a reference for judgment of postmortem cases with presence of these antipsychotics.


Subject(s)
Autopsy , Chlorprothixene/blood , Dibenzothiazepines/blood , Piperazines/blood , Quinolones/blood , Antipsychotic Agents/blood , Aripiprazole , Chromatography, Liquid , Humans , Quetiapine Fumarate , Reference Values , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...