Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Behav Brain Res ; 469: 115027, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38697302

ABSTRACT

Tamsulosin is an α1-adrenoceptor antagonist used to treat benign prostatic hyperplasia. This drug exhibits high affinity for α1A- and α1D-adrenoceptor subtypes, which are also expressed in the brain. While dementia symptoms have been reported after administration of tamsulosin in humans, studies on its effects on the rodent brain are still rare. The present study investigated the effects of tamsulosin (and biperiden, an amnesic drug) on cognitive performance in the object recognition task (ORT). Tamsulosin (0.001-0.01 mg/kg) was orally administrated in mice at three distinct time points: pre-training, post-training and pre-test session. Tamsulosin 0.01 mg/kg impaired object recognition regardless of when it was injected, whereas at lower doses did not affect mouse performance in the ORT. Biperiden also impaired acquisition and consolidation of object recognition in mice. Furthermore, the effects of tamsulosin on locomotion, motivation and anxiety were excluded as potential confounding factors. At all doses tested, tamsulosin did not alter distance moved, time spent exploring objects in the ORT, and anxiety-related behaviors in the elevated plus-maze test. By contrast, diazepam evoked a significant reduction of anxiety-like behaviours. In conclusion, tamsulosin impaired memory acquisition, consolidation and retrieval in an object recognition task in mice, thus affecting memory performance in a non-specific phase manner. These findings contribute to our understanding of the potential adverse effects of tamsulosin, and shed light on the role played by α1-adrenoceptors, particularly α1A- subtype, in cognitive processes.

2.
Psychopharmacology (Berl) ; 241(5): 1001-1010, 2024 May.
Article in English | MEDLINE | ID: mdl-38270614

ABSTRACT

RATIONALE: Recently, we demonstrated that the activation of the nociceptin/orphanin FQ (N/OFQ) receptor (NOP) signaling facilitates depressive-like behaviors. Additionally, literature findings support the ability of the N/OFQ-NOP system to modulate the hypothalamic-pituitary-adrenal (HPA) axis. OBJECTIVES: Considering that dysfunctional HPA axis is strictly related to stress-induced psychopathologies, we aimed to study the role of the HPA axis in the pro-depressant effects of NOP agonists. METHODS: Mice were treated prior to stress with the NOP agonist Ro 65-6570, and immobility time in the forced swimming task and corticosterone levels were measured. Additionally, the role of endogenous glucocorticoids and CRF was investigated using the glucocorticoid receptor antagonist mifepristone and the CRF1 antagonist antalarmin in the mediation of the effects of Ro 65-6570. RESULTS: The NOP agonist in a dose-dependent manner further increased the immobility of mice in the second swimming session compared to vehicle. By contrast, under the same conditions, the administration of the NOP antagonist SB-612111 before stress reduced immobility, while the antidepressant nortriptyline was inactive. Concerning in-serum corticosterone in mice treated with vehicle, nortriptyline, or SB-612111, a significant decrease was observed after re-exposition to stress, but no differences were detected in Ro 65-6570-treated mice. Administration of mifepristone or antalarmin blocked the Ro 65-6570-induced increase in the immobility time in the second swimming session. CONCLUSIONS: Present findings suggest that NOP agonists increase vulnerability to depression by hyperactivating the HPA axis and then increasing stress circulating hormones and CRF1 receptor signaling.


Subject(s)
Cycloheptanes , Imidazoles , Opioid Peptides , Piperidines , Receptors, Opioid , Spiro Compounds , Mice , Animals , Receptors, Opioid/physiology , Opioid Peptides/metabolism , Glucocorticoids/pharmacology , Nortriptyline/pharmacology , Nociceptin Receptor , Corticosterone/pharmacology , Hypothalamo-Hypophyseal System/metabolism , Mifepristone/pharmacology , Pituitary-Adrenal System/metabolism
3.
Neuropharmacology ; 212: 109077, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35513173

ABSTRACT

Nociceptin/orphanin FQ (N/OFQ) is the endogenous ligand of an inhibitory G protein coupled receptor named N/OFQ peptide receptor (NOP). Clinical and preclinical findings suggest that the blockade of the NOP signaling induces antidepressant-like effects. Additionally, the blockade of the NOP receptor during inescapable stress exposure prevented the acquisition of the helplessness phenotype, suggesting that NOP antagonists are able to increase stress resilience. BTRX-246040 (aka LY2940094) is a NOP receptor antagonist with high affinity, potency and selectivity for the NOP over classical opioid receptors. BTRX-246040 is under development for the treatment of depression, eating disorders and alcohol abuse and it already entered clinical trials. In the present study, the antidepressant effects of BTRX-246040 were evaluated in mice subjected to the forced swimming test and to the learned helplessness model of depression. Additionally, the ability of BTRX-246040 to prevent the development of the helpless behavior and to modulate adult hippocampal neurogenesis has been investigated. BTRX-246040 (30 mg/kg, i.p.) produced antidepressant-like effects in the forced swimming test and in the learned helplessness model. More interestingly, when given before the stress induction sessions it was able to prevent the development of the helplessness behavior. Under these experimental conditions, BTRX-246040 did not modulate adult hippocampal neurogenesis, neither in naive nor in stressed mice. This study, performed with a clinically viable ligand, further corroborates growing evidence indicating that the blockade of the NOP signaling may provide an innovative strategy for the treatment of stress related psychopathologies.


Subject(s)
Opioid Peptides , Receptors, Opioid , Animals , Antidepressive Agents/pharmacology , Hippocampus/metabolism , Ligands , Mice , Neurogenesis , Opioid Peptides/metabolism , Receptors, Opioid/metabolism
4.
Brain Res Bull ; 178: 29-36, 2022 01.
Article in English | MEDLINE | ID: mdl-34798218

ABSTRACT

The benign prostatic hyperplasia (BPH) is the main source of lower urinary tract symptoms. The BPH is a common age-dependent disease and tamsulosin is an α1-adrenoceptor blocker widely prescribed for BPH. Beyond the common adverse effects of tamsulosin, increased diagnosis of dementia after prescription was observed. Importantly, a clinical study suggested that tamsulosin may exert antidepressant effects in BPH patients. Considering the expression of α1-adrenoceptors in the brain, this study aimed to investigate the effects of tamsulosin in the forced swimming and open field tests in mice. For this, tamsulosin (0.001-1 mg/kg) was orally administered subacutely (1, 5 and 23 hr) and acutely (60 min) before tests. Mifepristone (10 mg/kg), a glucocorticoid receptor antagonist, and aminoglutethimide (10 mg/kg), a streoidogenesis inhibitor, were intraperitoneally injected before tamsulosin to investigate the role of the hypothalamic-pituitary-adrenal axis in the mediation of tamsulosin-induced effects. Subacute and acute administrations of tamsulosin increased the immobility time in the first exposition to an inescapable stressful situation. In the re-exposition to the swim task, controls displayed a natural increase in the immobility time, and the treatment with tamsulosin further increased this behavioral parameter. Tamsuslosin did not affect spontaneous locomotion neither in naïve nor in stressed mice. Our findings also showed that mifepristone and aminoglutethimide prevented the tamsulosin-induced increase in the immobility time in the first and second swimming sessions, respectively. In conclusion, tamsulosin may contribute to increased susceptibility to depressive-like behaviors, by facilitating the acquisition of a passive stress-copying strategy. These effects seem to be dependent on endogenous glucocorticoids.


Subject(s)
Adaptation, Psychological/drug effects , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Aromatase Inhibitors/pharmacology , Depression/chemically induced , Hormone Antagonists/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Receptors, Glucocorticoid/antagonists & inhibitors , Tamsulosin/pharmacology , Adrenergic alpha-1 Receptor Antagonists/administration & dosage , Aminoglutethimide/pharmacology , Animals , Aromatase Inhibitors/administration & dosage , Behavior, Animal/drug effects , Disease Models, Animal , Hormone Antagonists/administration & dosage , Mice , Mifepristone/pharmacology , Tamsulosin/administration & dosage
5.
Pharmaceuticals (Basel) ; 14(8)2021 Aug 06.
Article in English | MEDLINE | ID: mdl-34451872

ABSTRACT

Parkinson disease (PD) is a neurodegenerative disease mainly characterized by the loss of nigral dopaminergic neurons in the substantia nigra pars compacta. Patients suffering from PD develop severe motor dysfunctions and a myriad of non-motor symptoms. The treatment mainly consists of increasing central dopaminergic neurotransmission and alleviating motor symptoms, thus promoting severe side effects without modifying the disease's progress. A growing body of evidence suggests a close relationship between neuropeptide S (NPS) and its receptor (NPSR) system in PD: (i) double immunofluorescence labeling studies showed that NPSR is expressed in the nigral tyrosine hydroxylase (TH)-positive neurons; (ii) central administration of NPS increases spontaneous locomotion in naïve rodents; (iii) central administration of NPS ameliorates motor and nonmotor dysfunctions in animal models of PD; (iv) microdialysis studies showed that NPS stimulates dopamine release in naïve and parkinsonian rodents; (v) central injection of NPS decreases oxidative damage to proteins and lipids in the rodent brain; and, (vi) 7 days of central administration of NPS protects from the progressive loss of nigral TH-positive cells in parkinsonian rats. Taken together, the NPS/NPSR system seems to be an emerging therapeutic strategy for alleviating motor and non-motor dysfunctions of PD and, possibly, for slowing disease progress.

6.
Peptides ; 141: 170548, 2021 07.
Article in English | MEDLINE | ID: mdl-33862163

ABSTRACT

The ability to successfully cope with stress is known as 'resilience', and resilient individuals are less prone to develop psychopathologies. Understanding the neurobiological mechanisms of resilience may be instrumental to improve current therapies and benefit high-risk subjects. This review summarizes the complex interplay that exists between physiological and pathological responses to stressful events and the nociceptin/orphanin FQ (N/OFQ) - N/OFQ receptor (NOP) system, including: the effects of stress in regulating N/OFQ release and NOP expression; the ability of the N/OFQ-NOP system to modulate the hypothalamic-pituitary-adrenal axis; behavioral studies; and evidence in humans correlating this peptidergic system with psychopathologies. Available findings support the view that N/OFQ signaling stimulates the hypothalamic-pituitary-adrenal axis, thus increasing stress circulating hormones and corticotropin-releasing factor signaling. Additionally, activation of the NOP receptor inhibits monoamine transmission, including 5-HT, and this may contribute to maladaptive outcomes of stress. Ultimately, the N/OFQ system seems to have an important role in stress vulnerability, and blockade of NOP signaling may provide an innovative strategy for the treatment of stress related psychopathologies.


Subject(s)
Opioid Peptides/physiology , Receptors, Opioid/physiology , Stress, Psychological/etiology , Animals , Humans , Hypothalamo-Hypophyseal System/physiology , Narcotic Antagonists/pharmacology , Pituitary-Adrenal System/physiology , Stress, Psychological/drug therapy , Nociceptin Receptor , Nociceptin
7.
Neurobiol Stress ; 13: 100255, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33344710

ABSTRACT

The role of stress in the etiology of depression has been largely reported. In this line, exogenous glucocorticoids are employed to mimic the influence of stress on the development of depression. The N/OFQ-NOP receptor system has been implicated in the modulation of stress and emotional behaviors. In fact, the blockade of NOP receptors induces antidepressant effects and increases resilience to acute stress. This study investigated the effects of the NOP receptor blockade on dexamethasone-treated mice exposed to acute and prolonged swimming stress. Swiss and NOP(+/+) and NOP(-/-) mice were treated with dexamethasone, and the protective effects of the NOP antagonist SB-612111 (10 mg/kg, ip) or imipramine (20 mg/kg, ip) were investigated in three swimming sessions. The re-exposure to swim stress increased immobility time in Swiss and NOP(+/+), but not in NOP(-/-) mice. Acute and repeated dexamethasone administration induced a further increase in the immobility time, and facilitated body weight loss in Swiss mice. Single administration of SB-612111, but not imipramine, prevented swimming stress- and dexamethasone-induced increase in the immobility time. Repeated administrations of SB-612111 prevented the deleterious effects of 5 days of dexamethasone treatment. Imipramine also partially prevented the effects of repeated glucocorticoid administration on the immobility time, but did not affect the body weight loss. NOP(-/-) mice were more resistant than NOP(+/+) mice to inescapable swimming stress, but not dexamethasone-induced increase in the immobility time and body weight loss. In conclusion, the blockade of the NOP receptor facilitates an active stress copying response and attenuates body weight loss due to repeated stress.

8.
Neuropeptides ; 82: 102059, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32600667

ABSTRACT

Bipolar disorder is a psychiatric pathology characterized by biphasic mood episodes of mania or hypomania and depression. The pharmacotherapy of bipolar disorder has significant adverse effects impairing treatment adherence and patient quality of life. The N/OFQ-NOP receptor system has been widely implicated with mood disorders. Clinical and preclinical findings suggest antidepressants actions for NOP antagonists. More recently, the administration of NOP agonists has shown to promote depressant states. The present study aimed to investigate the effects of non-peptide NOP ligands in methylphenidate-induced manic-like behavior in mice. The NOP agonist Ro 65-6570 (0.01-1 mg/kg, ip), at the higher dose, did not affect spontaneous locomotion per se, but prevented the methylphenidate (10 mg/kg, sc)-induced hyperlocomotion. The NOP partial agonist AT-090 (0.001-0.03 mg/kg, ip) and the NOP antagonist SB-612111 (1-10 mg/kg, ip) did not significantly affect the psychostimulant-induced hyperactivity. Experiments performed with mice lacking the NOP receptor (NOP(-/-)) demonstrated that the treatment with methylphenidate induced similar hyperlocomotion in NOP(-/-) and NOP(+/+) mice. In conclusion, these findings suggest a potential role for NOP agonists in the prevention of manic states, especially by counteracting the hyperactivity symptom of bipolar patients. However, more studies are necessary in order to evaluate these compounds in other features of bipolar disorder.


Subject(s)
Antimanic Agents/administration & dosage , Bipolar Disorder/physiopathology , Hyperkinesis/physiopathology , Imidazoles/administration & dosage , Methylphenidate/administration & dosage , Receptors, Opioid/physiology , Spiro Compounds/administration & dosage , Animals , Female , Hyperkinesis/chemically induced , Mice , Receptors, Opioid/agonists , Valproic Acid/administration & dosage , Nociceptin Receptor
9.
Psychopharmacology (Berl) ; 237(6): 1633-1642, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32095915

ABSTRACT

RATIONALE: Depression and anxiety frequently co-occur, and this has important clinical implications. Previous studies showed that activation of the nociceptin/orphanin FQ receptor (NOP) elicits anxiolytic effects, while its blockade promotes consistent antidepressant actions. NOP antagonists are effective in reversing footshock-induced depressive-like behaviors, but their effects on stress-induced anxiety are still unclear. OBJECTIVE: This study aimed to investigate the effects of the NOP antagonist SB-612111 on footshock stress-induced anxiety behaviors. METHODS: Male Swiss mice were exposed to inescapable electric footshock stress, and behavioral phenotype was screened based on the ability to escape from footshock (i.e., helpless or non-helpless). Animals were then treated with diazepam (1 mg/kg) and SB-612111 (0.1-10 mg/kg), and their behavior was assessed in the elevated plus-maze (EPM) and open field test. RESULTS: When compared with non-stressed mice, helpless, but not non-helpless, animals displayed significant reductions in the time spent in and entries into open arms in the EPM. Diazepam significantly increased open arms exploration in helpless, non-helpless, and non-stressed mice. However, treatment with the NOP antagonist SB-612111 was inactive in naive mice, while it reversed anxiogenic-related behaviors in helpless mice and increased anxiety states in non-helpless mice. No effects on locomotion were observed. CONCLUSION: Helpless mice displayed increased anxiety compared to non-stressed and non-helpless animals, thus supporting use of this approach as an animal model to investigate anxiety/depression comorbidity. Additionally, SB-612111 modulated anxiety-like behaviors in male mice depending on individual stress susceptibility. Ultimately, NOP antagonists could be useful for treating anxiety in depressed patients.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Cycloheptanes/therapeutic use , Piperidines/therapeutic use , Receptors, Opioid/physiology , Stress, Psychological/drug therapy , Animals , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Anxiety/psychology , Depression/drug therapy , Depression/psychology , Emotions/drug effects , Emotions/physiology , Male , Mice , Stress, Psychological/psychology , Nociceptin Receptor
10.
Acta Neuropsychiatr ; : 1-6, 2020 Jan 29.
Article in English | MEDLINE | ID: mdl-31992385

ABSTRACT

OBJECTIVE: Accumulating evidence from preclinical and clinical studies indicates that prenatal exposure to stress impairs the development of the offspring brain and facilitates the emergence of mental illness. This study aims to describe the impact of prenatal restraint stress on cognition and exploration to an unfamiliar environment at adulthood in an outbred strain of mice. METHODS: Late pregnant mice were exposed to restraint stress and adult offspring (60 days of age) behaviours were assessed in the object recognition task and open field test. FINDINGS: Prenatal stress (PNS) impaired new object recognition in male and female mice. Importantly, the learning deficits in female PNS mice were linked to their estrous cycle. Actually, PNS females in metestrus/diestrus but not in proestrus/estrus phases displayed recognition deficits compared to controls. Concerning locomotion in an unfamiliar environment, male but not female PNS mice displayed significant increase, but showed no differences in the distance travelled within the centre zone of the arena. CONCLUSION: Present findings support the view that maternal restraint-stress during late pregnancy impairs recognition memory in both male and female offspring, and in females, this cognitive deficit is dependent on the estrous cycle phase. Ultimately, these data reinforce that PNS is an aetiological component of psychiatric disorders associated with memory deficits.

11.
Eur J Pharmacol ; 859: 172557, 2019 Sep 15.
Article in English | MEDLINE | ID: mdl-31326375

ABSTRACT

Neuropeptide S (NPS) is the endogenous ligand of a G-protein coupled receptor named NPS receptor. The NPS system controls several biological functions, including anxiety, wakefulness, locomotor activity, food intake, and pain transmission. A growing body of evidence supports facilitatory effects for NPS over dopaminergic neurotransmission. The present study was aimed to investigate the role of dopamine receptors signaling in the antinociceptive effects of NPS in the mouse formalin test. The following dopamine receptor antagonists were employed: SCH 23390 (selective dopamine D1 antagonist, 0.05 mg/kg, ip), haloperidol (non-selective dopamine D2-like receptor antagonist; 0.03 mg/kg, ip), and sulpiride (selective dopamine D2-like receptor antagonist; 25 mg/kg, ip). Mice were pretreated with dopamine antagonists before the supraspinal administration of NPS (0.1 nmol, icv). Morphine (5 mg/kg, sc) and indomethacin (10 mg/kg, ip) were used as positive controls to set up the experimental conditions. Morphine-induced antinociceptive effects were observed during phases 1 and 2 of the test, while indomethacin was only active at phase 2. Central NPS significantly reduced formalin-induced nociception during both phases. The systemic administration of SCH 23390 slightly blocked the effects of NPS only during phase 2. Haloperidol prevented NPS-induced antinociceptive effects. Similar to haloperidol, sulpiride also counteracted the antinociceptive effects of NPS in both phases of the formalin test. In conclusion, the present findings suggest that the analgesic effects of NPS are linked with dopaminergic neurotransmission mainly through dopamine D2-like receptor signaling.


Subject(s)
Analgesics/pharmacology , Formaldehyde/adverse effects , Neuropeptides/pharmacology , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Animals , Dopamine/metabolism , Dose-Response Relationship, Drug , Humans , Male , Mice , Signal Transduction/drug effects
12.
J Psychopharmacol ; 33(12): 1540-1549, 2019 12.
Article in English | MEDLINE | ID: mdl-31337258

ABSTRACT

BACKGROUND: The peptide nociceptin/orphanin FQ (N/OFQ) and its receptor (NOP) are implicated in the modulation of emotional states. Previous human and rodent findings support NOP antagonists as antidepressants. However, the role played by the N/OFQ-NOP receptor system in resilience to stress is unclear. AIMS: The present study investigated the effects of activation or blockade of NOP receptor signaling before exposure to acute stress. METHODS: The behavioral effects of the administration before stress of the NOP agonists Ro 65-6570 (0.01-1 mg/kg) and MCOPPB (0.1-10 mg/kg), and the NOP antagonist SB-612111 (1-10 mg/kg) were assessed in mice exposed to inescapable electric footshock and forced swim as stressors. The behavioral phenotype of mice lacking the NOP receptor (NOP(-/-)) exposed to inescapable electric footshock was also investigated. RESULTS: The activation of NOP receptor signaling with the agonists increased the percentage of mice developing helpless behavior and facilitated immobile posture. In contrast, the blockade of NOP receptor reduced the acquisition of depressive-like phenotypes, and similar resistance to develop helpless behaviors was observed in NOP(-/-) mice. Under the same stressful conditions, the antidepressant nortriptyline (20 mg/kg) did not change the acquisition of helpless behavior and immobile posture. CONCLUSIONS: These findings support the view that NOP activation during acute stress facilitates the development of depressive-related behaviors, whereas NOP blockade has a protective outcome. This study showed for first time that NOP antagonists are worthy of investigation as preemptive treatments in patients with severe risk factors for depression.


Subject(s)
Opioid Peptides/metabolism , Receptors, Opioid/metabolism , Resilience, Psychological/drug effects , Stress, Psychological/drug therapy , Animals , Behavior, Animal/drug effects , Benzimidazoles/administration & dosage , Benzimidazoles/pharmacology , Cycloheptanes/administration & dosage , Cycloheptanes/pharmacology , Depression/drug therapy , Depression/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Imidazoles/administration & dosage , Imidazoles/pharmacology , Male , Mice , Mice, Knockout , Nortriptyline/pharmacology , Piperidines/administration & dosage , Piperidines/pharmacology , Receptors, Opioid/drug effects , Receptors, Opioid/genetics , Spiro Compounds/administration & dosage , Spiro Compounds/pharmacology , Stress, Psychological/physiopathology , Nociceptin Receptor , Nociceptin
13.
Exp Clin Psychopharmacol ; 27(5): 433-442, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30714753

ABSTRACT

Major depression can be triggered by stressful events that promote deregulation of the hypothalamic-pituitary-adrenal axis response and, in some circumstances, persistent elevation of circulating glucocorticoid levels. Animal models are widely used to investigate the mechanisms responsible for the etiology and treatment of major depression. However, to mimic the dysfunction of the hypothalamic-pituitary-adrenal axis in rodents, animals should be exposed to sustained physical and psychological stressful situations. These animal models of depression are labor intensive and impact individual animals differently. Aiming to add evidence for a new acute neuroendocrine model of depression, male and female mice were treated with a single administration of dexamethasone, and behavioral effects were evaluated in the presence and absence of the antidepressants nortriptyline and venlafaxine. Male and female Swiss mice were treated with dexamethasone (0.07 mg/kg, subcutaneously) and the mouse behavior was assessed in the tail suspension and open field tests at 4 h, 24 h, and 7 days after administration. Dexamethasone induced depressogenic-like states in both sexes at 4 and 24 h after injection. Additionally, acute dexamethasone increased latency to body fur licking, thus corroborating the depressive-like behavior. The treatment with nortriptyline and venlafaxine (both at 30 mg/kg, intraperitoneally) blocked dexamethasone-induced increase in the immobility time and the latency to self-care. In conclusion, the present findings suggest that a single administration of dexamethasone induces depressive-like states in male and female mice, and these behavioral alterations are counteracted by conventional antidepressants. Ultimately, these data provide new evidence for an acute neuroendocrine model of depression. (PsycINFO Database Record (c) 2019 APA, all rights reserved).


Subject(s)
Depression/drug therapy , Dexamethasone/pharmacology , Nortriptyline/therapeutic use , Venlafaxine Hydrochloride/therapeutic use , Animals , Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , Depression/chemically induced , Female , Hypothalamo-Hypophyseal System/drug effects , Male , Mice , Pituitary-Adrenal System/drug effects
14.
Behav Brain Res ; 356: 120-126, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30142397

ABSTRACT

Aggressive behaviors can be considered symptoms of bipolar disorder, schizophrenia, post-traumatic stress, intermittent explosive, and personality disorders. Nociceptin/orphanin FQ (N/OFQ) is a peptide acting as endogenous ligand of the NOP receptor. Preclinical and clinical findings suggest the NOP receptor as an innovative target for the treatment of psychopathologies, such as anxiety, depression, and drug abuse. This study investigated the effects of NOP ligands and the behavioral phenotype of mice lacking the NOP receptor in an animal model of aggressiveness, the resident-intruder test. Mood stabilizers, such as valproate, lithium, and carbamazepine reduced aggressive behaviors of resident mice, while diazepam was inactive. In contrast, para-chlorophenylalanine (PCPA), an inhibitor of 5-HT synthesis, increased aggressiveness in mice. Similar to PCPA, the treatment with the NOP agonists Ro 65-6570 and AT-090 also increased aggressive behaviors. The systemic administration of the NOP antagonist SB-612111 did not modify the behavior of resident mice, but it prevented the aggressive behavior of Ro 65-6570. NOP receptor knockout mice did not display any behavioral difference compared to wild-type animals in the resident-intruder test. None of the treatments affected non-agonistic behaviors and spontaneous locomotion. In conclusion, NOP receptor agonists increased aggressiveness, while the pharmacological and genetic blockade of NOP receptor signaling did not modify agonistic behaviors. Ultimately, the aggressive profile of action of NOP agonists should be taken into account in the development of innovative psychiatric drugs targeting the NOP receptor.


Subject(s)
Aggression/physiology , Agonistic Behavior/physiology , Receptors, Opioid/physiology , Animals , Anxiety , Bipolar Disorder , Carbamazepine/pharmacology , Cycloheptanes/pharmacology , Depression , Depressive Disorder , Disease Models, Animal , Fenclonine/pharmacology , Lithium/pharmacology , Male , Mice , Mice, Knockout , Opioid Peptides/metabolism , Piperidines/pharmacology , Receptors, Opioid/agonists , Receptors, Opioid/genetics , Valproic Acid/pharmacology , Nociceptin Receptor , Nociceptin
15.
Peptides ; 112: 101-105, 2019 02.
Article in English | MEDLINE | ID: mdl-30550769

ABSTRACT

Cebranopadol is a mixed NOP/opioid receptor agonist currently under development as innovative analgesic. In this study the liability of cebranopadol to produce opioid-type physical dependence has been evaluated in comparison with morphine in wild type mice and in mice knockout for the NOP receptor gene (NOP(-/-)). Mice were treated twice a day for 5 days with increasing doses of cebranopadol or morphine (cumulative doses 10.2 and 255 mg/kg, respectively) and the number of jumping in response to naloxone 10 mg/kg were measured after 2 h from the last injection. In wild type mice naloxone evoked a similar withdrawal jumping behavior in animal pretreated with morphine or cebranopadol. In NOP(-/-) mice morphine treatment produced the same signs of withdrawal as in NOP(+/+) animals, while cebranopadol treatment elicited a stronger withdrawal syndrome in NOP(-/-) than of NOP(+/+) mice. These results demonstrated that the activation of the NOP receptor reduces the liability of cebranopadol to produce opioid-like physical dependence. Thus, the simultaneous activation of NOP and opioid receptors can be an effective pharmacological strategy to counteract physical dependence to opioid drugs.


Subject(s)
Indoles/pharmacology , Opioid-Related Disorders/prevention & control , Receptors, Opioid/agonists , Spiro Compounds/pharmacology , Analgesics/pharmacology , Animals , Female , Indoles/therapeutic use , Male , Mice , Mice, Knockout , Morphine/pharmacology , Spiro Compounds/therapeutic use , Nociceptin Receptor
16.
Handb Exp Pharmacol ; 254: 233-257, 2019.
Article in English | MEDLINE | ID: mdl-30535941

ABSTRACT

Many studies point toward the nociceptin/orphanin FQ (N/OFQ) and the N/OFQ peptide receptor (NOP) as targets for the development of innovative drugs for treating anxiety- and mood-related disorders. Evidence supports the view that the activation of NOP receptors with agonists elicits anxiolytic-like effects, while its blockade with NOP antagonists promotes antidepressant-like actions in rodents. Genetic studies showed that NOP receptor knockout mice display an antidepressant-like phenotype, and NOP antagonists are inactive in these animals. In contrast, the genetic blockade of NOP receptor signaling generally displays an increase of anxiety states in the elevated plus-maze test. In this chapter we summarized the most relevant findings of NOP receptor ligands in the modulation of anxiety and mood disorders, and the putative mechanisms of action are discussed.


Subject(s)
Antidepressive Agents/pharmacology , Mood Disorders , Opioid Peptides/pharmacology , Receptors, Opioid/metabolism , Animals , Antidepressive Agents/therapeutic use , Anxiety , Ligands , Mice , Opioid Peptides/metabolism , Receptors, Opioid/chemistry , Receptors, Opioid/drug effects , Nociceptin Receptor
17.
Naunyn Schmiedebergs Arch Pharmacol ; 391(12): 1301-1318, 2018 12.
Article in English | MEDLINE | ID: mdl-30338342

ABSTRACT

Caffeine is the most consumed psychoactive substance in the world; in general, it is not associated to potentially harmful effects. Nevertheless, few studies were performed attempting to investigate the caffeine addiction. The present review was mainly aimed to answer the following question: is caffeine an abuse drug? To adress this point, the effects of caffeine in preclinical and clinical studies were summarized and critically analyzed taking account the abuse disorders described in the Diagnostic and Statistical Manual of Mental Disorders (DSM-V). We concluded that the diagnostic criteria evidenced on DSM-V to intoxication-continued use and abstinence are not well supported by clinical studies. The fact that diagnostic criteria is not widely supported by preclinical or clinical studies may be due specially to a controversy in its exactly mechanism of action: recent literature point to an indirect, rather than direct modulation of dopamine receptors, and auto-limitant consumption due to adverse sensations in high doses. On the other hand, it reports clear withdrawal-related symptoms. Thus, based on a classical action on reward system, caffeine only partially fits its mechanism of action as an abuse drug, especially because previous research does not report a clear effect of dopaminergic activity enhance on nucleus accumbens; despite this, there are reports concerning dopaminergic modulation by caffeine on the striatum. However, based on human and animal research, caffeine withdrawal evokes signals and symptoms, which are relevant enough to include this substance among the drugs of abuse.


Subject(s)
Caffeine , Central Nervous System Stimulants , Illicit Drugs , Adenosine/metabolism , Animals , Caffeine/chemistry , Caffeine/pharmacokinetics , Caffeine/pharmacology , Central Nervous System Stimulants/chemistry , Central Nervous System Stimulants/pharmacokinetics , Central Nervous System Stimulants/pharmacology , Craving , Dopamine/metabolism , Humans , Illicit Drugs/chemistry , Illicit Drugs/pharmacokinetics , Illicit Drugs/pharmacology
18.
Psychopharmacology (Berl) ; 235(11): 3093-3102, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30145654

ABSTRACT

RATIONALE: Nociceptin/orphanin FQ (N/OFQ) is the endogenous ligand of a Gi protein-coupled receptor named NOP. Both N/OFQ and NOP receptor are widely expressed in brain areas involved in the control of emotional processes. Clinical and preclinical studies support antidepressant effects due to the blockade of NOP receptor signaling. By contrast, NOP receptor activation did not evoke any change in behavioral despair tests. OBJECTIVES: The present study aimed to investigate the effects of the co-administration of NOP agonists and classic antidepressant drugs in the forced swimming test (FST) and learned helplessness model (LH) in mice. METHODS: Male Swiss mice were co-administered with NOP agonists (N/OFQ and Ro 65-6570) and antidepressants (nortriptyline, fluoxetine, and R-ketamine) or SB-612111 (NOP antagonist) and the behavioral effects were assessed in the FST and LH tests. RESULTS: Fluoxetine, nortriptyline, R-ketamine and the NOP antagonist SB-612111 displayed antidepressant-like effects in the FST. The administration of the NOP agonists N/OFQ and Ro 65-6570 did not induce any behavioral change. However, co-administration of NOP agonists blocked the antidepressant effects of SB-612111, fluoxetine, and nortriptyline, but not R-ketamine in the FST. Similarly, in the LH, the systemic injection of SB-612111, nortriptyline, and R-ketamine reversed helplessness. The co-administration of Ro 65-6570 blocked the antidepressant-like effects of SB-612111 and nortriptyline, but not R-ketamine. CONCLUSIONS: NOP receptor activation inhibits the acute antidepressant effects of nortriptyline and fluoxetine, but not R-ketamine. The present findings contribute to further understand the role played by the N/OFQ-NOP receptor system in regulating mood states.


Subject(s)
Antidepressive Agents/pharmacology , Fluoxetine/pharmacology , Helplessness, Learned , Ketamine/pharmacology , Nortriptyline/pharmacology , Opioid Peptides/agonists , Anesthetics, Dissociative/pharmacology , Animals , Brain/drug effects , Brain/physiology , Emotions/drug effects , Emotions/physiology , Fluoxetine/antagonists & inhibitors , Imidazoles/pharmacology , Male , Mice , Nortriptyline/antagonists & inhibitors , Opioid Peptides/metabolism , Receptors, Opioid/agonists , Receptors, Opioid/metabolism , Spiro Compounds/pharmacology , Swimming/physiology , Swimming/psychology , Nociceptin
19.
Peptides ; 99: 195-204, 2018 01.
Article in English | MEDLINE | ID: mdl-29031796

ABSTRACT

Based on their high selectivity of action and low toxicity, naturally occurring peptides have great potential in terms of drug development. However, the pharmacokinetic properties of peptides, in particular their half life, are poor. Among different strategies developed for reducing susceptibility to peptidases, and thus increasing the duration of action of peptides, the generation of branched peptides has been described. However, the synthesis and purification of branched peptides are extremely complicated thus limiting their druggability. Here we present a novel and facile synthesis of tetrabranched peptides acting as GPCR ligands and their in vitro and vivo pharmacological characterization. Tetrabranched derivatives of nociceptin/orphanin FQ (N/OFQ), N/OFQ related peptides, opioid peptides, tachykinins, and neuropeptide S were generated with the strategy named peptide welding technology (PWT) and characterized by high yield and purity of the desired final product. In general, PWT derivatives displayed a pharmacological profile similar to that of the natural sequence in terms of affinity, pharmacological activity, potency, and selectivity of action in vitro. More importantly, in vivo studies demonstrated that PWT peptides are characterized by increased potency associated with long lasting duration of action. In conclusion, PWT derivatives of biologically active peptides can be viewed as innovative pharmacological tools for investigating those conditions and states in which selective and prolonged receptor stimulation promotes beneficial effects.


Subject(s)
Opioid Peptides , Protein Engineering/methods , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Animals , Humans , Opioid Peptides/chemical synthesis , Opioid Peptides/chemistry , Opioid Peptides/pharmacology
20.
Neurosci Lett ; 660: 79-85, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28893593

ABSTRACT

The genus Passiflora is popularly used to treat anxiety. Recent studies showed antidepressant-like effects of two varieties of P. edulis (edulis and flavicarpa) in mice. However, the mechanisms of antidepressant actions are still unknown. Here, the effects of P. edulis fo. edulis aqueous extract (AE, 100-300mg/kg, po), and ethyl acetate (AcOEt, 25-50mg/kg, po), butanol (BuOH, 25-50mg/kg, po) and residual aqueous (25-100mg/kg, po) fractions were investigated in the mouse forced swimming test. In addition, the involvement of monoamines in the P. edulis fractions-induced antidepressant actions was approached. HPLC analyses showed that AcOEt and BuOH, but not residual, fractions shared with AE the main peaks between 25 and 70min (UV 340nm), which are suggestive of flavonoids. Nortriptyline and fluoxetine reduced the immobility time and similar results were observed for AE, AcOEt and BuOH but not residual fractions. PCPA (inhibitor of 5-HT synthesis), AMPT (inhibitor of catecholamine synthesis) and sulpiride (selective D2 receptor antagonist), but not DSP-4 (noradrenergic neurotoxin), blocked the antidepressant actions of AcOEt and BuOH. In conclusion, AcOEt and BuOH fractions shared with AE similar phytochemical composition and antidepressant actions. Preserved 5-HT and dopamine transmissions were required for the antidepressant effects of P. edulis fractions.


Subject(s)
Antidepressive Agents/administration & dosage , Biogenic Monoamines/metabolism , Depression/metabolism , Passiflora/chemistry , Plant Extracts/administration & dosage , Synaptic Transmission , Acetates/administration & dosage , Animals , Antidepressive Agents/isolation & purification , Antidepressive Agents, Second-Generation/administration & dosage , Antidepressive Agents, Tricyclic/administration & dosage , Behavior, Animal , Benzylamines/administration & dosage , Butanols/administration & dosage , Catecholamines/antagonists & inhibitors , Catecholamines/metabolism , Depression/drug therapy , Dopamine Antagonists/administration & dosage , Fluoxetine/administration & dosage , Male , Mice , Nortriptyline/administration & dosage , Plant Extracts/isolation & purification , Sulpiride/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...