Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 570
1.
Neuropharmacology ; 185: 108456, 2021 03 01.
Article En | MEDLINE | ID: mdl-33444637

Recent evidence suggests that kappa opioid receptors (KOR) in limbic brain regions such as the amygdala contribute to pain conditions, but underlying mechanisms remain to be determined. The amygdala is an important player in averse-affective aspects of pain and pain modulation. The central nucleus (CeA) serves output functions through projection neurons that include corticotropin releasing factor (CRF) expressing neurons. The CeA is also rich in KOR. Here we tested the novel hypothesis that KOR activation in the CeA generates pain-like behaviors through a mechanism that involves inhibition of synaptic inhibition (disinhibition) of CRF neurons. Intra-CeA administration of a KOR agonist (U-69,593) increased vocalizations of naïve rats to noxious stimuli, and induced anxiety-like behaviors in the open field test (OFT) and avoidance in the conditioned place preference test, without affecting mechanosensory thresholds. Optogenetic silencing of CeA-CRF neurons blocked the facilitatory effects of systemically applied U-69,593 in naïve rats. Patch-clamp recordings of CRF neurons in rat brain slices found that U-69,593 decreased feedforward inhibitory transmission evoked by optogenetic stimulation of parabrachial afferents, but had no effect on monosynaptic excitatory transmission. U-69,593 decreased frequency, but not amplitude, of inhibitory synaptic currents, suggesting a presynaptic action. Multiphoton imaging of CeA-CRF neurons in rat brain slices showed that U-69,593 increased calcium signals evoked by electrical stimulation of presumed parabrachial input. This study shows for the first time that KOR activation increases activity of amygdala CRF neurons through synaptic disinhibition, resulting in averse-affective pain-like behaviors. Blocking KOR receptors may therefore represent a novel therapeutic strategy.


Amygdala/metabolism , Benzeneacetamides/administration & dosage , Corticotropin-Releasing Hormone/antagonists & inhibitors , Corticotropin-Releasing Hormone/metabolism , Pain/metabolism , Pyrrolidines/administration & dosage , Receptors, Opioid, kappa/metabolism , Amygdala/drug effects , Animals , Male , Pain Measurement/drug effects , Pain Measurement/methods , Rats , Rats, Transgenic , Rats, Wistar , Stereotaxic Techniques , Vocalization, Animal/drug effects , Vocalization, Animal/physiology
2.
Neuropharmacology ; 186: 108463, 2021 03 15.
Article En | MEDLINE | ID: mdl-33460689

CRF is the main activator of the hypothalamic-pituitary-adrenal (HPA) axis in response to stress. CRF neurons are found mainly in the hypothalamus, but CRF positive cells and CRF1 receptors are also found in extrahypothalamic structures, including amygdala (CeA), hippocampus, NAc and VTA. CRF release in the hypothalamus is regulated by inhibitory GABAergic interneurons and extrahypothalamic glutamatergic inputs, and disruption of this balance is found in stress-related disorders and addiction. (3α,5α)3-hydroxypregnan-20-one (3α,5α-THP), the most potent positive modulator of GABAA receptors, attenuates the stress response reducing hypothalamic CRF mRNA expression and ACTH and corticosterone serum levels. In this study, we explored 3α,5α-THP regulation of hypothalamic and extrahypothalamic CRF mRNA and peptide expression, in male and female Sprague Dawley rats, following vehicle or 3α,5α-THP administration (15 mg/kg). In the hypothalamus, we found sex differences in CRF mRNA expression (females +74%, p < 0.01) and CRF peptide levels (females -71%, p < 0.001). 3α,5α-THP administration reduced hypothalamic CRF mRNA expression only in males (-50%, p < 0.05) and did not alter CRF peptide expression in either sex. In hippocampus and CeA, 3α,5α-THP administration reduced CRF peptide concentrations only in the male (hippocampus -29%, p < 0.05; CeA -62%, p < 0.01). In contrast, 3α,5α-THP injection increased CRF peptide concentration in the VTA of both males (+32%, p < 0.01) and females (+26%, p < 0.01). The results show sex and region-specific regulation of CRF signals and the response to 3α,5α-THP administration. This data may be key to successful development of therapeutic approaches for stress-related disorders and addiction.


Corticotropin-Releasing Hormone/antagonists & inhibitors , Corticotropin-Releasing Hormone/biosynthesis , Hypothalamus/drug effects , Hypothalamus/metabolism , Pregnanolone/administration & dosage , Sex Characteristics , Animals , Female , Injections, Intraperitoneal , Male , Pregnanolone/analogs & derivatives , Rats , Rats, Sprague-Dawley
3.
Neuropharmacology ; 170: 108045, 2020 06 15.
Article En | MEDLINE | ID: mdl-32217364

The neuropeptide corticotropin-releasing factor (CRF) is critical in neural circuit function and behavior, particularly in the context of stress, anxiety, and addiction. Despite a wealth of preclinical evidence for the efficacy of CRF receptor 1 antagonists in reducing behavioral pathology associated with alcohol exposure, several clinical trials have had disappointing outcomes, possibly due to an underappreciation of the role of biological variables. Although he National Institutes of Health (NIH) now mandate the inclusion of sex as a biological variable in all clinical and preclinical research, the current state of knowledge in this area is based almost entirely on evidence from male subjects. Additionally, the influence of biological variables other than sex has received even less attention in the context of neuropeptide signaling. Age (particularly adolescent development) and housing conditions have been shown to affect CRF signaling and voluntary alcohol intake, and the interaction between these biological variables is particularly relevant to the role of the CRF system in the vulnerability or resilience to the development of alcohol use disorder (AUD). Going forward, it will be important to include careful consideration of biological variables in experimental design, reporting, and interpretation. As new research uncovers conditions in which sex, age, and environment play major roles in physiological and/or pathological processes, our understanding of the complex interaction between relevant biological variables and critical signaling pathways like the CRF system in the cellular and behavioral consequences of alcohol exposure will continue to expand ultimately improving the ability of preclinical research to translate to the clinic. This article is part of the special issue on Neuropeptides.


Alcohol Drinking/metabolism , Corticotropin-Releasing Hormone/metabolism , Gene-Environment Interaction , Receptors, Corticotropin-Releasing Hormone/metabolism , Sex Characteristics , Social Environment , Age Factors , Alcohol Drinking/drug therapy , Alcohol Drinking/psychology , Animals , Corticotropin-Releasing Hormone/antagonists & inhibitors , Female , Humans , Male , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Stress, Psychological/psychology
4.
J Neurosci ; 40(12): 2519-2537, 2020 03 18.
Article En | MEDLINE | ID: mdl-32054675

The bed nucleus of the stria terminalis (BNST) is a forebrain region highly responsive to stress that expresses corticotropin-releasing hormone (CRH) and is implicated in mood disorders, such as anxiety. However, the exact mechanism by which chronic stress induces CRH-mediated dysfunction in BNST and maladaptive behaviors remains unclear. Here, we first confirmed that selective acute optogenetic activation of the oval nucleus BNST (ovBNST) increases maladaptive avoidance behaviors in male mice. Next, we found that a 6 week chronic variable mild stress (CVMS) paradigm resulted in maladaptive behaviors and increased cellular excitability of ovBNST CRH neurons by potentiating mEPSC amplitude, altering the resting membrane potential, and diminishing M-currents (a voltage-gated K+ current that stabilizes membrane potential) in ex vivo slices. CVMS also increased c-fos+ cells in ovBNST following handling. We next investigated potential molecular mechanism underlying the electrophysiological effects and observed that CVMS increased CRH+ and pituitary adenylate cyclase-activating polypeptide+ (PACAP; a CRH upstream regulator) cells but decreased striatal-enriched protein tyrosine phosphatase+ (a STEP CRH inhibitor) cells in ovBNST. Interestingly, the electrophysiological effects of CVMS were reversed by CRHR1-selective antagonist R121919 application. CVMS also activated protein kinase A (PKA) in BNST, and chronic infusion of the PKA-selective antagonist H89 into ovBNST reversed the effects of CVMS. Coadministration of the PKA agonist forskolin prevented the beneficial effects of R121919. Finally, CVMS induced an increase in surface expression of phosphorylated GluR1 (S845) in BNST. Collectively, these findings highlight a novel and indispensable stress-induced role for PKA-dependent CRHR1 signaling in activating BNST CRH neurons and mediating maladaptive behaviors.SIGNIFICANCE STATEMENT Chronic stress and acute activation of oval bed nucleus of the stria terminalis (ovBNST) induces maladaptive behaviors in rodents. However, the precise molecular and electrophysiological mechanisms underlying these effects remain unclear. Here, we demonstrate that chronic variable mild stress activates corticotropin-releasing hormone (CRH)-associated stress signaling and CRH neurons in ovBNST by potentiating mEPSC amplitude and decreasing M-current in male mice. These electrophysiological alterations and maladaptive behaviors were mediated by BNST protein kinase A-dependent CRHR1 signaling. Our results thus highlight the importance of BNST CRH dysfunction in chronic stress-induced disorders.


Adaptation, Psychological , Corticotropin-Releasing Hormone/physiology , Septal Nuclei/physiology , Signal Transduction/physiology , Stress, Psychological/psychology , Animals , Chronic Disease , Corticotropin-Releasing Hormone/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Electrophysiological Phenomena/physiology , Excitatory Postsynaptic Potentials/physiology , Genes, fos , Male , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Potassium Channels/physiology , Protein Tyrosine Phosphatases/metabolism , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors
5.
J Exp Med ; 216(11): 2479-2491, 2019 11 04.
Article En | MEDLINE | ID: mdl-31467037

Hypothalamic-pituitary-adrenal (HPA) axis dysfunction contributes to numerous human diseases and disorders. We developed a high-affinity monoclonal antibody, CTRND05, targeting corticotropin-releasing factor (CRF). In mice, CTRND05 blocks stress-induced corticosterone increases, counteracts effects of chronic variable stress, and induces other phenotypes consistent with suppression of the HPA axis. CTRND05 induces skeletal muscle hypertrophy and increases lean body mass, effects not previously reported with small-molecule HPA-targeting pharmacologic agents. Multiorgan transcriptomics demonstrates broad HPA axis target engagement through altering levels of known HPA-responsive transcripts such as Fkbp5 and Myostatin and reveals novel HPA-responsive pathways such as the Apelin-Apelin receptor system. These studies demonstrate the therapeutic potential of CTRND05 as a suppressor of the HPA axis and serve as an exemplar of a potentially broader approach to target neuropeptides with immunotherapies, as both pharmacologic tools and novel therapeutics.


Antibodies, Monoclonal/pharmacology , Corticotropin-Releasing Hormone/antagonists & inhibitors , Hypothalamo-Hypophyseal System/drug effects , Pituitary-Adrenal System/drug effects , Stress, Physiological/drug effects , Animals , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Corticosterone/immunology , Corticosterone/metabolism , Corticotropin-Releasing Hormone/immunology , Gene Expression Profiling/methods , Humans , Hypothalamo-Hypophyseal System/immunology , Hypothalamo-Hypophyseal System/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Phenotype , Pituitary-Adrenal System/immunology , Pituitary-Adrenal System/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Stress, Physiological/immunology
6.
BMC Neurosci ; 20(1): 11, 2019 Mar 18.
Article En | MEDLINE | ID: mdl-30885137

BACKGROUND: Pre-treatment with the corticotropin-releasing factor antagonist α-helical CRF9-41 prevents inhibition of gastric emptying by cocaine-and amphetamine-regulated transcript peptide at a dorsal hindbrain level, but its inhibition of sucrose intake is not affected. This is suggestive of separable underlying mechanisms of action in the caudal brainstem for cocaine-and amphetamine-regulated transcript peptide with regard to food intake and gastrointestinal functions. Here we further examine cocaine-and amphetamine-regulated transcript peptide-corticotropin-releasing factor receptor interactions in caudal brainstem controls of solid food intake. Injections of combinations of vehicle, cocaine-and amphetamine-regulated transcript peptide (0.5 µg or 1 µg) or α-helical CRF9-41 were given into the fourth cerebral ventricle of rats. Nocturnal solid food intake was recorded over 22 h. RESULTS: Pre-treatment with α-helical CRF9-41 into the fourth ventricle significantly increased the responsivity to cocaine-and amphetamine-regulated transcript peptide on hypophagia. In a separate control experiment, α-helical CRF9-41 pre-treatment blocked CRF-induced food intake inhibition indicative of its antagonistic effectiveness. CONCLUSIONS: We conclude that an endogenous Corticotropin-releasing factor agonist may modulate suppression of food intake caused by cocaine-and amphetamine-regulated transcript peptide at a dorsal hindbrain level in the absence of stress. A potential caudal brainstem mechanism whereby cocaine-and amphetamine-regulated transcript peptide effects on food intake is attenuated via corticotropin-releasing factor receptor activity causing tonic inhibition, is suggested.


Central Nervous System Agents/pharmacology , Corticotropin-Releasing Hormone/antagonists & inhibitors , Eating/drug effects , Nerve Tissue Proteins/pharmacology , Peptide Fragments/pharmacology , Rhombencephalon/drug effects , Animals , Corticotropin-Releasing Hormone/metabolism , Corticotropin-Releasing Hormone/pharmacology , Eating/physiology , Fourth Ventricle , Injections, Intraventricular , Male , Nerve Tissue Proteins/metabolism , Peptide Fragments/metabolism , Rats, Sprague-Dawley , Rhombencephalon/metabolism
7.
J Physiol Pharmacol ; 69(3)2018 06.
Article En | MEDLINE | ID: mdl-30342430

This study aimed to determine the mechanisms governing Gonadotropin releasing hormone (GnRH) biosynthesis and luteinising hormone (LH) secretion in follicular-phase sheep after infusion of corticotropin releasing hormone (CRH) and/or CRH antagonist corticotropin releasing hormone nist (CRH-A) into the third cerebral ventricle. The study included two experimental approaches: first, we investigated the effect of CRH or CRH-A (α-helical CRH 9-41) on GnRH and GnRH receptor (GnRHR) biosynthesis in the preoptic area (POA), anterior (AH) and ventromedial hypothalamus (VMH), stalk/median eminence (SME), and on GnRHR in the anterior pituitary (AP) using an enzyme-linked immunosorbent assay (ELISA); second, we used real-time PCR to analyse the influence of CRH and CRH-A on the levels of kisspeptin (Kiss1) mRNA in POA and VMH including arcuate nucleus (VMH/ARC), and on Kiss1 receptor (Kiss1r) mRNA abundance in POA-hypothalamic structures. These analyses were supplemented by radioimmunoassay (RIA) and ELISA methods for measurement of LH and cortisol levels in the blood, respectively. Our results show that administration of CRH significantly decreased GnRH biosynthesis in the POA/hypothalamus. CRH also decreased GnRHR abundance in the hypothalamus and in the AP, but increased it in the POA. Furthermore, administration of CRH decreased plasma LH concentration and levels of Kiss1 mRNA in the POA and VMH/ARC as well as Kiss1r mRNA in these structures and in the SME. Significant increase in plasma cortisol concentration in the group treated with CRH was also observed. For CRH-A, all analysed effects were opposite to those induced by CRH. The study demonstrates that intracerebroventricular (i.c.v.) infusion of both CRH and CRH-A affects the GnRH/GnRHR biosynthesis and LH secretion in follicular-phase sheep conceivably via either central and peripheral mechanisms including Kiss1 neurons activity and cortisol signals. It has also been suggested that CRH and CRH-A infusion probably had effects directly at the AP.


Corticotropin-Releasing Hormone/antagonists & inhibitors , Corticotropin-Releasing Hormone/pharmacology , Gonadotropin-Releasing Hormone/biosynthesis , Hypothalamus/metabolism , Receptors, LHRH/metabolism , Animals , Female , Follicular Phase/metabolism , Hydrocortisone/blood , Hypothalamus/drug effects , Kisspeptins/genetics , Luteinizing Hormone/blood , Receptors, Kisspeptin-1/genetics , Sheep
8.
Braz J Med Biol Res ; 51(11): e7541, 2018 Oct 04.
Article En | MEDLINE | ID: mdl-30304131

We previously found that acute exercise inhibited the gastric emptying of liquid in awake rats by causing an acid-base imbalance. In the present study, we investigated the involvement of the nitric oxide-cyclic guanosine monophosphate (NO-cGMP) pathway, vasoactive intestinal peptide (VIP), and corticotropin-releasing factor (CRF) peptide in this phenomenon. Male rats were divided into exercise or sedentary group and were subjected to a 15-min swim session against a load (2.5 or 5% b.w.). The rate of gastric emptying was evaluated after 5, 10, or 20 min postprandially. Separate groups of rats were treated with vehicle (0.9% NaCl, 0.1 mL/100 g, ip) or one of the following agents: atropine (1.0 mg/kg, ip), the NO non-selective inhibitor Nω-nitro-L-arginine methyl ester hydrochloride (L-NAME; 10.0 mg/kg, ip), or the selective cGMP inhibitor 1H-(1,2,4)oxadiazole[4,3-a]quinoxalin-1-one (ODQ; 5.0 mg/kg, ip), the i-NOS non-specific inhibitor (aminoguanidine; 10.0 mg/kg, ip), the corticotropin-releasing factor receptor antagonist (astressin; 100 µg/kg, ip), or the vasoactive intestinal peptide (VIP) receptor antagonist Lys1, Pro2,5, Arg3,4, Tyr6 (100 µg/kg, ip). Compared to sedentary rats, both the 2.5 and 5% exercise groups exhibited higher (P<0.05) values of blood lactate and fractional gastric dye recovery. Corticosterone and NO levels increased (P<0.05) in the 5% exercised rats. Pretreatment with astressin, VIP antagonist, atropine, L-NAME, and ODQ prevented the increase in gastric retention caused by exercise in rats. Acute exercise increased gastric retention, a phenomenon that appears to be mediated by the NO-cGMP pathway, CRF, and VIP receptors.


Corticotropin-Releasing Hormone/metabolism , Gastric Emptying/physiology , Guanosine Monophosphate/metabolism , Nitric Oxide/metabolism , Physical Conditioning, Animal/physiology , Vasoactive Intestinal Peptide/metabolism , Animals , Atropine/pharmacology , Corticosterone/blood , Corticotropin-Releasing Hormone/antagonists & inhibitors , Corticotropin-Releasing Hormone/pharmacology , Enzyme Inhibitors/pharmacology , Gastric Emptying/drug effects , Guanosine Monophosphate/antagonists & inhibitors , Lactic Acid/blood , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/antagonists & inhibitors , Peptide Fragments/pharmacology , Postprandial Period/drug effects , Postprandial Period/physiology , Random Allocation , Rats, Wistar , Reference Values , Reproducibility of Results , Sedentary Behavior , Time Factors , Vasoactive Intestinal Peptide/antagonists & inhibitors
9.
Horm Behav ; 100: 20-28, 2018 04.
Article En | MEDLINE | ID: mdl-29501756

The basolateral nucleus of the amygdala (BLA) plays a significant role in mediating individual differences in the effects of fear memory on sleep. Here, we assessed the effects of antagonizing corticotropin releasing factor receptor 1 (CRFR1) after shock training (ST) on fear-conditioned behaviors and sleep. Outbred Wistar rats were surgically implanted with electrodes for recording EEG and EMG and with bilateral guide cannulae directed at BLA. Data loggers were placed intraperitoneally to record core body temperature. The CRFR1 antagonist, antalarmin (ANT; 4.82 mM) was microinjected into BLA after shock training (ST: 20 footshocks, 0.8 mA, 0.5 s duration, 60 s interstimulus interval), and the effects on sleep, freezing and the stress response (stress-induced hyperthermia, SIH) were examined after ST and fearful context re-exposure alone at 7 days (CTX1) and 21 days (CTX2) post-ST. EEG and EMG recordings were scored for non-rapid eye movement sleep (NREM), rapid eye movement sleep (REM) and wakefulness. The rats were separated into 4 groups: Vehicle-vulnerable (Veh-Vul; n = 10), Veh-resilient (Veh-Res; n = 11), ANT-vulnerable (ANT-Vul; n = 8) and ANT-resilient (ANT-Res; n = 8) based on whether, compared to baseline, the rats showed a decrease or no change/increase in REM during the first 4 h following ST. Post-ST ANT microinjected into BLA attenuated the fear-conditioned reduction in REM in ANT-Vul rats on CTX1, but did not significantly alter REM in ANT-Res rats. However, compared to Veh treated rats, REM was reduced in ANT treated rats on CTX2. There were no group differences in freezing or SIH across conditions. Therefore, CRFR1 in BLA plays a role in mediating individual differences in sleep responses to stress and in the extinction of fear conditioned changes in sleep.


Adaptation, Psychological/drug effects , Basolateral Nuclear Complex/drug effects , Body Temperature/drug effects , Fear/drug effects , Freezing Reaction, Cataleptic/drug effects , Pyrimidines/pharmacology , Pyrroles/pharmacology , Sleep/drug effects , Animals , Basolateral Nuclear Complex/metabolism , Corticotropin-Releasing Hormone/antagonists & inhibitors , Electroencephalography , Fear/psychology , Male , Memory/drug effects , Physical Conditioning, Animal/physiology , Physical Conditioning, Animal/psychology , Rats , Rats, Wistar , Sleep/physiology , Sleep, REM/drug effects
10.
J Biomol Struct Dyn ; 36(7): 1691-1712, 2018 May.
Article En | MEDLINE | ID: mdl-28521603

Despite the various research efforts toward the treatment of stress-related disorders, the drug has not yet launched last 20 years. Corticotropin releasing factor-1 receptor antagonists have been point of great interest in stress-related disorders. In the present study, we have selected benzazole scaffold-based compounds as corticotropin releasing factor-1 antagonists and performed 2D and 3D QSAR studies to identify the structural features to elucidating the binding mechanism prediction. The best 2D QSAR model was obtained through multiple linear regression method with r2 value of .7390, q2 value of .5136 and pred_r2 (predicted square correlation coefficient) value of .88. The contribution of 2D descriptor, T_2_C_1 was 60% (negative contribution) and 4pathClusterCount was 40.24% (positive contribution) in enhancing the activity. Also 3D QSAR model was statistically significant with q2 value of .9419 and q2_se (standard error of internal validation) value of .19. Statistical parameters results prove the robustness and significance of both models. Further, molecular docking and pharmacokinetic analysis was performed to explore the scope of investigation. Docking results revealed that the all benzazole compounds show hydrogen bonding with residue Asn283 and having same hydrophobic pocket (Phe286, Leu213, Ile290, Leu287, Phe207, Arg165, Leu323, Tyr327, Phe284, and Met206). Compound B14 has higher activity compare to reference molecules. Most of the compounds were found within acceptable range for pharmacokinetic parameters. This work provides the extremely useful leads for structural substituents essential for benzimidazole moiety to exhibit antagonistic activity against corticotropin releasing factor-1 receptors.


Benzimidazoles/chemistry , Corticotropin-Releasing Hormone/antagonists & inhibitors , Corticotropin-Releasing Hormone/chemistry , Binding Sites , Computer Simulation , Drug Design , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Molecular Docking Simulation/methods , Protein Binding , Quantitative Structure-Activity Relationship
11.
Neuropsychiatr ; 32(1): 1-8, 2018 Mar.
Article De | MEDLINE | ID: mdl-28639210

One of the crucial purposes of treating alcohol-dependent patients is to enhance their ability to stay abstinent after detoxification therapy. Anxiety and stress vulnerability are the main factors provoking alcohol craving and relapse. In the first months of abstinence, alcohol-dependent patients frequently show sleep disturbances, irritability and depression, indicating chronic activation of stress pathways. In addition, the loss of confidence in interpersonal interactions results in social withdrawal and reduced willingness to participate in therapeutic programs.Current research shows that the peptide hormone oxytocin exerts substantial anxiolytic effects and facilitates prosocial behavior. Oxytocin can be safely applied as intranasal preparation. Oxytocin acts by inhibiting the effects of the corticotropin-releasing factor on GABAergic interneurons in the amygdala and paraventricular nucleus of hypothalamus.Recent research strongly suggests that application of oxytocin may beneficially influence the mechanisms of relapse and craving by reduction of anxiety, stress vulnerability and social withdrawal in abstinent alcohol-dependent patients.This article reviews neurobiological mechanisms of oxytocin effects on stress-related pathways and discusses the potential use of oxytocin in the treatment of alcohol addiction.


Alcoholism/rehabilitation , Craving/drug effects , Oxytocin/administration & dosage , Administration, Intranasal , Adrenocorticotropic Hormone/blood , Alcoholism/physiopathology , Alcoholism/psychology , Amygdala/drug effects , Amygdala/physiopathology , Animals , Anxiety/physiopathology , Anxiety/psychology , Anxiety/rehabilitation , Arousal/drug effects , Arousal/physiology , Corticotropin-Releasing Hormone/antagonists & inhibitors , Humans , Interneurons/physiology , Neural Pathways/drug effects , Neural Pathways/physiopathology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/physiopathology , Stress, Psychological/complications , Stress, Psychological/physiopathology , gamma-Aminobutyric Acid/physiology
12.
Braz. j. med. biol. res ; 51(11): e7541, 2018. tab, graf
Article En | LILACS | ID: biblio-951721

We previously found that acute exercise inhibited the gastric emptying of liquid in awake rats by causing an acid-base imbalance. In the present study, we investigated the involvement of the nitric oxide-cyclic guanosine monophosphate (NO-cGMP) pathway, vasoactive intestinal peptide (VIP), and corticotropin-releasing factor (CRF) peptide in this phenomenon. Male rats were divided into exercise or sedentary group and were subjected to a 15-min swim session against a load (2.5 or 5% b.w.). The rate of gastric emptying was evaluated after 5, 10, or 20 min postprandially. Separate groups of rats were treated with vehicle (0.9% NaCl, 0.1 mL/100 g, ip) or one of the following agents: atropine (1.0 mg/kg, ip), the NO non-selective inhibitor Nω-nitro-L-arginine methyl ester hydrochloride (L-NAME; 10.0 mg/kg, ip), or the selective cGMP inhibitor 1H-(1,2,4)oxadiazole[4,3-a]quinoxalin-1-one (ODQ; 5.0 mg/kg, ip), the i-NOS non-specific inhibitor (aminoguanidine; 10.0 mg/kg, ip), the corticotropin-releasing factor receptor antagonist (astressin; 100 µg/kg, ip), or the vasoactive intestinal peptide (VIP) receptor antagonist Lys1, Pro2,5, Arg3,4, Tyr6 (100 µg/kg, ip). Compared to sedentary rats, both the 2.5 and 5% exercise groups exhibited higher (P<0.05) values of blood lactate and fractional gastric dye recovery. Corticosterone and NO levels increased (P<0.05) in the 5% exercised rats. Pretreatment with astressin, VIP antagonist, atropine, L-NAME, and ODQ prevented the increase in gastric retention caused by exercise in rats. Acute exercise increased gastric retention, a phenomenon that appears to be mediated by the NO-cGMP pathway, CRF, and VIP receptors.


Animals , Male , Corticotropin-Releasing Hormone/metabolism , Guanosine Monophosphate/metabolism , Gastric Emptying/physiology , Nitric Oxide/metabolism , Reference Values , Atropine/pharmacology , Time Factors , Corticosterone/blood , Corticotropin-Releasing Hormone/antagonists & inhibitors , Corticotropin-Releasing Hormone/pharmacology , Random Allocation , Rats, Wistar , Enzyme Inhibitors/pharmacology , Gastric Emptying/drug effects
13.
Int J Mol Sci ; 18(8)2017 Aug 21.
Article En | MEDLINE | ID: mdl-28825666

Neuropeptide FF (NPFF) belongs to the RFamide family and is known as a morphine-modulating peptide. NPFF regulates various hypothalamic functions through two receptors, NPFFR1 and NPFFR2. The hypothalamic-pituitary-adrenal (HPA) axis participates in physiological stress response by increasing circulating glucocorticoid levels and modulating emotional responses. Other RFamide peptides, including neuropeptide AF, neuropeptide SF and RFamide related peptide also target NPFFR1 or NPFFR2, and have been reported to activate the HPA axis and induce anxiety- or depression-like behaviors. However, little is known about the action of NPFF on HPA axis activity and anxiety-like behaviors, and the role of the individual receptors remains unclear. In this study, NPFFR2 agonists were used to examine the role of NPFFR2 in activating the HPA axis in rodents. Administration of NPFFR2 agonists, dNPA (intracerebroventricular, ICV) and AC-263093 (intraperitoneal, IP), time-dependently (in rats) and dose-dependently (in mice) increased serum corticosteroid levels and the effects were counteracted by the NPFF receptor antagonist, RF9 (ICV), as well as corticotropin-releasing factor (CRF) antagonist, α-helical CRF(9-41) (intravenous, IV). Treatment with NPFFR2 agonist (AC-263093, IP) increased c-Fos protein expression in the hypothalamic paraventricular nucleus and induced an anxiogenic effect, which was evaluated in mice using an elevated plus maze. These findings reveal, for the first time, that the direct action of hypothalamic NPFFR2 stimulates the HPA axis and triggers anxiety-like behaviors.


Depressive Disorder/metabolism , Oligopeptides/metabolism , Receptors, Neuropeptide/metabolism , Adrenal Cortex Hormones/blood , Animals , Anxiety , Corticotropin-Releasing Hormone/administration & dosage , Corticotropin-Releasing Hormone/antagonists & inhibitors , Depressive Disorder/blood , Depressive Disorder/physiopathology , Hydrazines/administration & dosage , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Hypothalamus/metabolism , Hypothalamus/pathology , Mice , Oligopeptides/administration & dosage , Peptide Fragments/administration & dosage , Rats , Receptors, Neuropeptide/agonists
14.
Psychopharmacology (Berl) ; 234(18): 2813-2821, 2017 Sep.
Article En | MEDLINE | ID: mdl-28698920

BACKGROUND: A history of brief intermittent social defeat stress can escalate cocaine self-administration and induce long-term adaptations in the mesolimbic dopamine system. Extra-hypothalamic corticotrophin releasing factor (CRF) has been shown to be closely associated with stress-induced escalation of drug use. How repeated stress modulates CRF release in the ventral tegmental area (VTA) and the roles of CRF receptors during different phases of stress-induced cocaine self-administration remain to be defined. OBJECTIVE: The current study examines the roles of CRF and CRF receptor 1 (CRFR1) in escalated intravenous cocaine self-administration after exposure to social defeat stress in mice. METHODS AND RESULTS: First, CRFR1 antagonist (CP 376,395, 15 mg/kg, i.p.) given 30 min prior to each social defeat episode prevented later escalated cocaine self-administration. When CP 376,395 (5 and 15 mg/kg, i.p.) was administered 10 days after the last episode of social stress, the escalation of cocaine intake was dose-dependently reversed. Moreover, socially defeated mice showed increased CRF release in the VTA compared to controls. To further explore the role of CRFR1, CP 376,395 (0.5 and 1 µg/0.2 µl) was infused directly into the VTA before the cocaine self-administration session. Intra-VTA antagonism of CRFR1 was sufficient to reverse social defeat stress-escalated cocaine self-administration. CONCLUSION: These findings suggest that CRF and CRFR1 exert multiple roles in the response to social stress that are relevant to escalated cocaine self-administration.


Cocaine/administration & dosage , Interpersonal Relations , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Stress, Psychological/psychology , Ventral Tegmental Area/drug effects , Animals , Corticotropin-Releasing Hormone/antagonists & inhibitors , Corticotropin-Releasing Hormone/metabolism , Male , Mice , Microdialysis/methods , Random Allocation , Receptors, Corticotropin-Releasing Hormone/metabolism , Self Administration , Ventral Tegmental Area/metabolism
15.
Psychopharmacology (Berl) ; 234(18): 2823-2836, 2017 Sep.
Article En | MEDLINE | ID: mdl-28725939

RATIONALE: Exposure to intermittent social defeat stress elicits corticotropin releasing factor (CRF) release into the VTA and induces long-term modulation of mesocorticolimbic dopamine activity in rats. These adaptations are associated with an intense cocaine-taking phenotype, which is prevented by CRF receptor antagonists. OBJECTIVE: The present studies examine whether infusion of CRF into the VTA is sufficient to escalate cocaine-taking behavior, in the absence of social defeat experience. Additionally, we aimed to characterize changes in cocaine valuation that may promote binge-like cocaine intake. METHODS: Male Long-Evans rats were microinjected into the VTA with CRF (50 or 500 ng/side), vehicle, or subjected to social defeat stress, intermittently over 10 days. Animals were then trained to self-administer IV cocaine (FR5). Economic demand for cocaine was evaluated using a within-session behavioral-economics threshold procedure, which was followed by a 24-h extended access "binge." RESULTS: Rats that experienced social defeat or received intra-VTA CRF microinfusions (50 ng) both took significantly more cocaine than controls over the 24-h binge but showed distinct patterns of intake. Behavioral economic analysis revealed that individual demand for cocaine strongly predicts binge-like consumption, and demand elasticity (i.e. α) is augmented by intra-VTA CRF, but not by social defeat. The effects of CRF on cocaine-taking were also prevented by intra-VTA pretreatment with CP376395, but not Astressin-2B. CONCLUSIONS: Repeated infusion of CRF into the VTA persistently alters cocaine valuation and intensifies binge-like drug intake in a CRF-R1-dependent manner. Conversely, the persistent pattern of cocaine bingeing induced by social defeat stress may suggest impaired inhibitory control, independent of reward valuation.


Cocaine-Related Disorders/prevention & control , Cocaine-Related Disorders/psychology , Cocaine/administration & dosage , Corticotropin-Releasing Hormone/administration & dosage , Stress, Psychological/psychology , Ventral Tegmental Area/drug effects , Aminopyridines/administration & dosage , Animals , Cocaine-Related Disorders/metabolism , Corticotropin-Releasing Hormone/antagonists & inhibitors , Corticotropin-Releasing Hormone/metabolism , Infusions, Intraventricular , Male , Rats , Rats, Long-Evans , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/metabolism , Self Administration , Stress, Psychological/metabolism , Ventral Tegmental Area/metabolism
16.
Eur J Med Chem ; 138: 900-908, 2017 Sep 29.
Article En | MEDLINE | ID: mdl-28750312

Corticotropin-releasing factor (CRF) is an important neuropeptide hormone which controls the body's overall response to stress. It plays a crucial role in regulating the behavioral, cardiovascular, immune and gastrointestinal systems. Over-activation of the CRF system has been implicated in many disorders including anxiety, depression, drug addiction, hypertension, Irritable Bowel Syndrome (IBS), peptic ulcers, inflammation and others. Thus, binding of CRF to its receptors is an attractive target to develop new medications which aim at treating ailments associated with chronic stress. Numerous small-molecule non-peptide CRF receptor antagonists were developed and many are in various stages in clinical trials. Many showed great promise in treatment of anxiety, depression, peptic ulcers, inflammation, IBS and drug addiction. In our recent previous work, the development of two series of pyrimidine and fused pyrimidine CRF antagonists were described. In continuation of our efforts in this direction, in the current manuscript, the synthesis of a third series of CRF receptor antagonists is described. The binding affinities of select compounds for the type 1 receptor of CRF (CRF1R) were determined and compared to a standard CRF antagonist drug antalarmin. A lead compound was identified and further evaluated by measuring its effect on the inhibition of the agonist-stimulated accumulation of second messengers.


Corticotropin-Releasing Hormone/antagonists & inhibitors , Thiazoles/pharmacology , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Molecular Structure , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry
17.
Psychopharmacology (Berl) ; 234(9-10): 1467-1481, 2017 05.
Article En | MEDLINE | ID: mdl-28265716

BACKGROUND: Dr. Athina Markou sought treatments for a common neural substrate shared by depression and drug dependence. Antagonists of corticotropin-releasing factor (CRF) receptors, a target of interest to her, have not reached the clinic despite strong preclinical rationale and sustained translational efforts. METHODS: We explore potential causes for the failure of CRF1 antagonists and review recent findings concerning CRF-CRF1 systems in psychopathology. RESULTS: Potential causes for negative outcomes include (1) poor safety and efficacy of initial drug candidates due to bad pharmacokinetic and physicochemical properties, (2) specificity problems with preclinical screens, (3) the acute nature of screens vs. late-presenting patients, (4) positive preclinical results limited to certain models and conditions with dynamic CRF-CRF1 activation not homologous to tested patients, (5) repeated CRF1 activation-induced plasticity that reduces the importance of ongoing CRF1 agonist stimulation, and (6) therapeutic silencing which may need to address CRF2 receptor or CRF-binding protein molecules, constitutive CRF1 activity, or molecules that influence agonist-independent activity or to target structural regions other than the allosteric site bound by all drug candidates. We describe potential markers of activation towards individualized treatment, human genetic, and functional data that still implicate CRF1 systems in emotional disturbance, sex differences, and suggestive clinical findings for CRF1 antagonists in food craving and CRF-driven HPA-axis overactivation. CONCLUSION: The therapeutic scope of selective CRF1 antagonists now appears narrower than had been hoped. Yet, much remains to be learned about CRF's role in the neurobiology of dysphoria and addiction and the potential for novel anti-CRF therapies therein.


Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Stress, Psychological/drug therapy , Translational Research, Biomedical/methods , Animals , Behavior, Addictive/drug therapy , Behavior, Addictive/metabolism , Corticotropin-Releasing Hormone/metabolism , Depression/drug therapy , Depression/metabolism , Humans , Pyrimidines/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Pyrroles/metabolism , Pyrroles/pharmacology , Pyrroles/therapeutic use , Receptors, Corticotropin-Releasing Hormone/metabolism , Stress, Psychological/metabolism , Substance-Related Disorders/drug therapy , Substance-Related Disorders/metabolism
18.
Curr Med Chem ; 24(23): 2528-2558, 2017.
Article En | MEDLINE | ID: mdl-28302012

BACKGROUND: The term Alcohol Use Disorder (AUD) incorporates different states of disease related to the recurrent use of alcohol and linked to the relevant impairment, disability and failure to perform major responsibilities in different realms. Many neurotransmitter systems are involved in the phases or states of alcoholism from reward mechanisms, associated to binge intoxication, to stress and anxiety linked to relapse and withdrawal. Some neuropeptides play a key function in the control of anxiety and stress, and establish a close relationship with the pathological mechanisms underlying alcohol addiction. Among them, Neuropeptide Y (NPY), Corticotropin-releasing factor (CRF)/Urocortins and Neuropeptide S (NPS) cross-talk, and are responsible for some of the maladaptation processes that the brain exhibits during the progression of the disease. METHOD: In this study, we review the literature mainly focused on the participation of these neuropeptides in the pathophysiology of AUD, as well as on the use of antagonists designed to investigate signaling mechanisms initiated after ligand binding and their connection to biochemical adaptation events coupled to alcohol addiction. The possibility that these systems may serve as therapeutic objectives to mitigate or eliminate the harm that drinking ethanol generates, is also discussed. CONCLUSION: The peptide systems reviewed here, together with other neurotransmitter systems and their mutual relationships, are firm candidates to be targeted to treat AUD.


Alcohol Drinking/drug therapy , Alcohol-Related Disorders/drug therapy , Corticotropin-Releasing Hormone/antagonists & inhibitors , Neuropeptide Y/antagonists & inhibitors , Neuropeptides/antagonists & inhibitors , Urocortins/antagonists & inhibitors , Alcohol Drinking/metabolism , Alcohol-Related Disorders/metabolism , Animals , Corticotropin-Releasing Hormone/metabolism , Humans , Neuropeptide Y/metabolism , Neuropeptides/metabolism , Urocortins/metabolism
19.
Curr Mol Pharmacol ; 10(4): 264-269, 2017.
Article En | MEDLINE | ID: mdl-28103781

It is noteworthy that thirty three years of efforts in corticotropin releasing factor (CRF) research by academia and the pharmaceutical industry resulted in several thousand papers and patents, yet little progress has been made to identify and market diagnostic or therapeutic CRF peptides and small molecule ligands. We document the potential relevance of CRF peptide antagonists to reinvigorate stress/anxiety affected "anatomy systems" in order to boost their efficacy.


Corticotropin-Releasing Hormone/antagonists & inhibitors , Drug Discovery , Peptides/chemistry , Peptides/pharmacology , Amino Acid Sequence , Animals , Anxiety/drug therapy , Anxiety/metabolism , Corticotropin-Releasing Hormone/chemistry , Corticotropin-Releasing Hormone/metabolism , Drug Discovery/methods , Humans , Irritable Bowel Syndrome/drug therapy , Irritable Bowel Syndrome/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Stress, Physiological/drug effects
20.
Pain ; 158(2): 296-305, 2017 02.
Article En | MEDLINE | ID: mdl-27849648

Alterations in amygdala activity are apparent in women who report a history of early life stress (ELS) and those diagnosed with chronic pain disorders. Chronic stress in adulthood induces visceral hypersensitivity by alterations in glucocorticoid receptor (GR) and corticotropin-releasing factor (CRF) expression within the central amygdala (CeA). Here, we hypothesized that unpredictable ELS, previously shown to induce visceral hypersensitivity in adult female rats, alters GR and CRF expression in the CeA. After neonatal ELS, visceral sensitivity and GR and CRF gene expression were quantified in adult female rats. After unpredictable ELS, adult female rats exhibited visceral hypersensitivity and increased expression of GR and CRF in the CeA. After predictable ELS, adult female rats demonstrated normosensitive behavioral pain responses and upregulation of GR but not CRF in the CeA. After the ELS paradigms, visceral sensitivity and gene expression within the CeA were unaffected in adult male rats. The role of GR and CRF in modulating visceral sensitivity in adult female rats after ELS was investigated using oligodeoxynucleotide sequences targeted to the CeA for knockdown of GR or CRF. Knockdown of GR increased visceral sensitivity in all rats but revealed an exaggerated visceral hypersensitivity in females with a history of predictable or unpredictable ELS compared with that of controls. Knockdown of CRF expression or antagonism of CRF1R in the CeA attenuated visceral hypersensitivity after unpredictable ELS. This study highlights a shift in GR and CRF regulation within the CeA after ELS that underlies the development of visceral hypersensitivity in adulthood.


Amygdala/metabolism , Corticotropin-Releasing Hormone/metabolism , Receptors, Glucocorticoid/metabolism , Sex Characteristics , Stress, Psychological/physiopathology , Visceral Pain/pathology , Aminopyridines/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Conditioning, Classical , Corticotropin-Releasing Hormone/antagonists & inhibitors , Disease Models, Animal , Female , Male , Maze Learning , Odorants , Oligodeoxyribonucleotides, Antisense/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Long-Evans , Receptors, Glucocorticoid/antagonists & inhibitors , Up-Regulation/drug effects , Up-Regulation/physiology , Visceral Pain/physiopathology
...