Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
Addict Biol ; 26(2): e12945, 2021 03.
Article in English | MEDLINE | ID: mdl-32761675

ABSTRACT

A pronounced decrease of oxytocin and increase of oxytocin receptor binding sites were recently reported in male alcohol dependent rats and male alcohol dependent patients. Here we comment on this and emphasize that in female alcohol dependent rats and humans no changes occur in the oxytocin system. We therefore suggest specific intervention with oxytocin only in male subjects.


Subject(s)
Alcoholism/pathology , Ethanol/pharmacology , Oxytocin/drug effects , Receptors, Oxytocin/drug effects , Animals , Female , Humans , Male , Psychopharmacology , Rats , Sex Factors
2.
Horm Behav ; 122: 104753, 2020 06.
Article in English | MEDLINE | ID: mdl-32302594

ABSTRACT

Sildenafil is a phosphodiesterase type 5 inhibitor used to treat male erectile dysfunction and pulmonary hypertension. A potential side effect of sildenafil is a noticeable decrease in seizure threshold. Oxytocin (OXT) secretion and the subsequent cAMP-responsive element-binding (CREB) phosphorylation are involved in proconvulsant effects of sildenafil in experimental models. The aim of the present study was to investigate the potential role of OXT receptors and their downstream calcineurin (CN)/inducible nitric oxide synthase (iNOS) pathways in proconvulsant effects of sildenafil. The pentylenetetrazole (PTZ)-induced seizure was used as a standard convulsion model in this study. Cortical CN activity, hippocampal nitrite levels, and proinflammatory cytokine content were measured. Our results indicated that following PTZ administration, sildenafil significantly increased CN activity at 40 mg/kg, respectively, in the control group. The combination of sildenafil and OXT receptor antagonist, atosiban (10 µg/kg, i.c.v) 30 min before sildenafil administration significantly reduced the CN activity. Also, the subeffective dose of CN inhibitor cyclosporine (5 mg/kg) 30 min before the administration of effective dose of sildenafil (40 mg/kg) reversed proconvulsant actions of sildenafil. This effect was iNOS-dependent because pretreatment of a low dose of aminoguanidine (20 mg/kg) 15 min before the administration of a low dose of cyclosporine (1 mg/kg) reversed the proconvulsant action of sildenafil (40 mg/kg). Finally, sildenafil induced the elevation of tumor necrosis factor alpha (TNF-α) and the nitrite level was blocked by the administration of cyclosporine in PTZ-treated mice. Collectively, our data provide insights into the role of OXT receptor/CN/iNOS pathway in the proconvulsant aspect of sildenafil.


Subject(s)
Convulsants , Oxytocin/metabolism , Receptors, Oxytocin/metabolism , Seizures/chemically induced , Sildenafil Citrate/adverse effects , Animals , Calcineurin/metabolism , Convulsants/pharmacology , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Oxytocin/drug effects , Phosphodiesterase 5 Inhibitors/adverse effects , Phosphodiesterase 5 Inhibitors/pharmacology , Receptors, Oxytocin/antagonists & inhibitors , Receptors, Oxytocin/drug effects , Seizures/metabolism , Seizures/physiopathology , Signal Transduction/drug effects , Sildenafil Citrate/pharmacology
3.
Mar Drugs ; 18(3)2020 Mar 06.
Article in English | MEDLINE | ID: mdl-32155768

ABSTRACT

Cone snails produce a fast-acting and often paralyzing venom, largely dominated by disulfide-rich conotoxins targeting ion channels. Although disulfide-poor conopeptides are usually minor components of cone snail venoms, their ability to target key membrane receptors such as GPCRs make them highly valuable as drug lead compounds. From the venom gland transcriptome of Conus miliaris, we report here on the discovery and characterization of two conopressins, which are nonapeptide ligands of the vasopressin/oxytocin receptor family. These novel sequence variants show unusual features, including a charge inversion at the critical position 8, with an aspartate instead of a highly conserved lysine or arginine residue. Both the amidated and acid C-terminal analogues were synthesized, followed by pharmacological characterization on human and zebrafish receptors and structural investigation by NMR. Whereas conopressin-M1 showed weak and only partial agonist activity at hV1bR (amidated form only) and ZFV1a1R (both amidated and acid form), both conopressin-M2 analogues acted as full agonists at the ZFV2 receptor with low micromolar affinity. Together with the NMR structures of amidated conopressins-M1, -M2 and -G, this study provides novel structure-activity relationship information that may help in the design of more selective ligands.


Subject(s)
Conotoxins/chemistry , Conotoxins/pharmacology , Conus Snail/chemistry , Amino Acid Sequence , Animals , Conotoxins/chemical synthesis , Disulfides/chemistry , Disulfides/pharmacology , Humans , Molecular Conformation , Mollusk Venoms/chemistry , Neurophysins/antagonists & inhibitors , Protein Precursors/antagonists & inhibitors , Receptors, Oxytocin/drug effects , Receptors, Vasopressin/drug effects , Structure-Activity Relationship , Transcriptome , Vasopressins/antagonists & inhibitors , Zebrafish
4.
J Neurophysiol ; 123(2): 587-599, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31891523

ABSTRACT

Oxytocin (OT) is a neuroactive peptide that influences the processing of fearful stimuli in the amygdala. In the central nucleus of the amygdala, the activation of OT receptors alters neural activity and ultimately suppresses the behavioral response to a fear conditioned stimulus. Receptors for OT are also found in the lateral amygdala (LA), and infusion of OT into the basolateral amygdala complex affects the formation and consolidation of fear memories. Yet, how OT receptor activation alters neurons and neural networks in the LA is unknown. In this study we used whole cell electrophysiological recordings to determine how OT-receptor activation changes synaptic transmission and synaptic plasticity in the LA of Sprague-Dawley rats. Our results demonstrate that OT-receptor activation results in a 200% increase in spontaneous inhibitory transmission in the LA that leads to the activation of presynaptic GABAB receptors. The activation of these receptors inhibits excitatory transmission in the LA, blocking long-term potentiation of cortical inputs onto LA neurons. Hence, this study provides the first demonstration that OT influences synaptic transmission and plasticity in the LA, revealing a mechanism that could explain how OT regulates the formation and consolidation of conditioned fear memories in the amygdala.NEW & NOTEWORTHY This study investigates modulation of synaptic transmission by oxytocin (OT) in the lateral amygdala (LA). We demonstrate that OT induces transient increases in spontaneous GABAergic transmission by activating interneurons in the basolateral amygdala. The resultant increase in GABA release in the LA activates presynaptic GABAB receptors on both inhibitory and excitatory inputs onto LA neurons, reducing release probability at these synapses. We subsequently demonstrate that OT modulates synaptic plasticity at cortical inputs to the LA.


Subject(s)
Basolateral Nuclear Complex/metabolism , GABAergic Neurons/metabolism , Interneurons/metabolism , Neuronal Plasticity/physiology , Oxytocin/physiology , Receptors, GABA-B/metabolism , Receptors, Oxytocin/metabolism , Synaptic Transmission/physiology , Animals , Basolateral Nuclear Complex/drug effects , GABAergic Neurons/drug effects , Interneurons/drug effects , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Neuronal Plasticity/drug effects , Oxytocin/administration & dosage , Oxytocin/antagonists & inhibitors , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, GABA-B/drug effects , Receptors, Oxytocin/drug effects , Synaptic Transmission/drug effects
5.
Comput Math Methods Med ; 2019: 8276715, 2019.
Article in English | MEDLINE | ID: mdl-31379974

ABSTRACT

This paper develops a mathematical model describing the potential buildup of high oxytocin concentrations in the maternal circulation during labor in terms of continuous Pitocin infusion rate, half-life, and maternal weight. Oxytocin override of the degradation of oxytocin by placental oxytocinase is introduced to model the potential transfer of oxytocin from the maternal circulation across the placenta into the fetal circulation and from there into the brain of the fetus. The desensitization unit D equal to 1.8E6 (pg·min)/ml is employed to establish a desensitization threshold and by extension, a downregulation threshold as a function of oxytocin override concentration and continuous Pitocin infusion time, that could be a factor in the subsequent development of autism among offspring. Epidemiological studies by Duke University [1], Yale University [2], and Harvard University [3] are discussed regarding Pitocin use and offspring autism development for an explanation of the weak correlations they identified. The findings of the Harvard epidemiological study are reinterpreted regarding Pitocin use and its conclusion questioned. Further evaluations of the findings of these three epidemiological studies are called for to incorporate medical information on quantity of Pitocin used, continuous Pitocin infusion rate, length of labor, and maternal weight to determine if a correlation can be established with offspring autism development above an empirically determined desensitization threshold for Pitocin use. Suggestions for research are discussed, including an alternative to continuous Pitocin infusion, pulsatile infusion of Pitocin during labor induction, which may mitigate possible offspring autism development.


Subject(s)
Autistic Disorder/etiology , Brain/embryology , Labor, Induced/adverse effects , Oxytocin/adverse effects , Receptors, Oxytocin/drug effects , Algorithms , Autistic Disorder/epidemiology , Brain/drug effects , Cystinyl Aminopeptidase/metabolism , Data Interpretation, Statistical , Female , Humans , Labor, Obstetric , Models, Theoretical , Pregnancy , Prenatal Exposure Delayed Effects , United States
6.
Physiol Behav ; 209: 112599, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31276680

ABSTRACT

Medial prefrontal cortex (mPFC), a forebrain structure, is involved in many brain functions such as learning and memory. In the present study, the effect of intra-mPFC microinjection of oxytocin, atosiban, morphine and naloxone was investigated on memory processing. Two guide cannulas were implanted into the right and left sides of the mPFC in ketamine and xylazine-anesthetized rats. To assess spatial memory function MWM test was performed by four training sessions of four trials. On day 5, a probe test was conducted after drugs microinjection. Significant differences were observed in learning activities during training days before microinjection of drugs. Intra-mPFC microinjections of oxytocin (5 and 10 ng/site) significantly increased memory related activities. This effect of oxytocin was inhibited by prior microinjection of atosiban (20 ng/site). On the other hand, morphine microinjection at doses of 5 and 10 µg/site into the mPFC significantly decreased memory related activities that were prevented by prior administration of naloxone (5 µg/site) and oxytocin (5 and 10 ng/site). In addition, intra-mPFC combined microinjections of low doses of oxytocin (2.5 ng/site) and naloxone (1 µg/site) improved memory function. By increasing the doses of oxytocin (5 ng/site) and naloxone (5 µg/site), a more documented improving effect was observed. These results showed that memory performance was impaired by activation of mPFC opioid receptors in rats. In addition, oxytocin in the mPFC improved memory function and prevented memory impairment-induced by morphine. Moreover, an interaction between oxytocin and opioid systems was also appeared in the present study.


Subject(s)
Oxytocin/pharmacology , Prefrontal Cortex/metabolism , Receptors, Opioid/physiology , Receptors, Oxytocin/metabolism , Spatial Memory/physiology , Animals , Hormone Antagonists/pharmacology , Male , Maze Learning/drug effects , Microinjections , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Oxytocin/administration & dosage , Prefrontal Cortex/drug effects , Psychomotor Performance/drug effects , Rats , Rats, Wistar , Receptors, Opioid/drug effects , Receptors, Oxytocin/drug effects , Spatial Memory/drug effects , Swimming/psychology , Vasotocin/analogs & derivatives , Vasotocin/pharmacology
7.
J Med Chem ; 62(10): 4991-5005, 2019 05 23.
Article in English | MEDLINE | ID: mdl-31022340

ABSTRACT

The vasopressin analogue desmopressin (desamino-d-arginine8 vasopressin, dDAVP, 1) is a potent vasopressin 2 (V2) receptor (V2R) agonist approved in many countries for the treatment of diabetes insipidus, primary nocturnal enuresis, nocturia, and coagulation disorders. Since 1 is primarily excreted via the kidneys, an age-related decline in kidney function leads to slower elimination, prolonged antidiuresis, and hyponatremia. In search of novel, potent, selective, and short-acting peptidic V2R agonists, we synthesized a series of C-terminally truncated analogues of [Val4]dDAVP, 2, modified in positions 2, 3, and 7 and/or at the disulfide bridge. The peptides were evaluated for in vitro potency at the human V2 receptor, selectivity versus the related receptors (human vasopressin 1a receptor, human vasopressin 1b receptor, and human oxytocin receptor), and pharmacokinetic profiles in rodents and other higher species. The truncated analogues show excellent potency at the V2R, increased systemic clearance, and shorter half-life in rats. Two compounds 19 (c(Bua-Cpa-Thi-Val-Asn-Cys)-Pro-Agm) and 38 (c(Bua-Cpa-Thi-Val-Asn-Cys)-Pro-d-Arg-NEt2) have been selected for clinical development for nocturia.


Subject(s)
Antidiuretic Agents/chemical synthesis , Antidiuretic Agents/pharmacology , Receptors, Vasopressin/agonists , Animals , Antidiuretic Agents/pharmacokinetics , Deamino Arginine Vasopressin/analogs & derivatives , Deamino Arginine Vasopressin/chemical synthesis , Deamino Arginine Vasopressin/pharmacology , Dose-Response Relationship, Drug , Drug Design , Drug Discovery , Half-Life , Humans , Nocturia/drug therapy , Rats , Receptors, Oxytocin/drug effects , Renal Agents/chemical synthesis , Renal Agents/pharmacology , Structure-Activity Relationship
8.
Front Neuroendocrinol ; 50: 107-117, 2018 07.
Article in English | MEDLINE | ID: mdl-29859882

ABSTRACT

A roadblock to successful treatment for anxiety and depression is the high proportion of individuals that do not respond to existing treatments. Different underlying neurobiological mechanisms may drive similar symptoms, so a more personalized approach to treatment could be more successful. There is increasing evidence that sex is an important biological variable modulating efficacy of antidepressants and anxiolytics. We review evidence for sex-specific effects of traditional monoamine based antidepressants and newer pharmaceuticals targeting kappa opioid receptors (KOR), oxytocin receptors (OTR), and N-methyl-D-aspartate receptors (ketamine). In some cases, similar behavioral effects are observed in both sexes while in other cases strong sex-specific effects are observed. Most intriguing are cases such as ketamine which has similar behavioral effects in males and females, perhaps through sex-specific neurobiological mechanisms. These results show how essential it is to include both males and females in both clinical and preclinical evaluations of novel antidepressants and anxiolytics.


Subject(s)
Antidepressive Agents/pharmacology , Depressive Disorder/drug therapy , Ketamine/pharmacology , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, Opioid, kappa/drug effects , Receptors, Oxytocin/drug effects , Sex Characteristics , Animals , Depressive Disorder/metabolism , Female , Humans , Male
9.
Front Neuroendocrinol ; 51: 14-24, 2018 10.
Article in English | MEDLINE | ID: mdl-29054552

ABSTRACT

Oxytocin (OT) and arginine-vasopressin (AVP) act in the brain to regulate social cognition/social behavior and in the periphery to influence a variety of physiological processes. Although the chemical structures of OT and AVP as well as their receptors are quite similar, OT and AVP can have distinct or even opposing actions. Here, we review the increasing body of evidence that exogenously administered and endogenously released OT and AVP can activate each other's canonical receptors (i.e., cross-talk) and examine the possibility that receptor cross-talk following the synaptic and non-synaptic release of OT and AVP contributes to their distinct roles in the brain and periphery. Understanding the consequences of cross-talk between OT and AVP receptors will be important in identifying how these peptides control social cognition and behavior and for the development of drugs to treat a variety of psychiatric disorders.


Subject(s)
Arginine Vasopressin/metabolism , Mental Disorders/metabolism , Oxytocin/metabolism , Receptors, Oxytocin/metabolism , Receptors, Vasopressin/metabolism , Social Behavior , Animals , Arginine Vasopressin/pharmacology , Female , Humans , Male , Mental Disorders/drug therapy , Oxytocin/pharmacology , Receptors, Oxytocin/drug effects , Receptors, Vasopressin/drug effects
10.
Eur J Pain ; 22(3): 511-526, 2018 03.
Article in English | MEDLINE | ID: mdl-29082571

ABSTRACT

BACKGROUND: Vasopressin (AVP) seems to play a role as an antinociceptive neurohormone, but little is known about the peripheral site of action of its antinociceptive effects. Moreover, AVP can produce motor impairment that could be confused with behavioural antinociception. Finally, it is not clear which receptor is involved in the peripheral antinociceptive AVP effects. METHODS: In anaesthetized rats with end-tidal CO2 monitoring, extracellular unitary recordings were performed, measuring the evoked activity mediated by Aß-, Aδ-, C-fibres and post-discharge. Behavioural nociception and motor impairment were evaluated under subcutaneous AVP (0.1-10 µg) using formalin and rotarod tests. Selective antagonists to vasopressin (V1A R) or oxytocin receptors (OTR) were used. Additionally, vasopressin and oxytocin receptors were explored immunohistochemically in skin tissues. RESULTS: Subcutaneous AVP (1 and 10 µg/paw) induced antinociception and a transitory reduction of the end-tidal CO2 . The neuronal activity associated with Aδ- and C-fibre activation was diminished, but no effect was observed on Aß-fibres. AVP also reduced paw flinches in the formalin test and a transitory locomotor impairment was also found. The AVP-induced antinociception was blocked by the selective antagonist to V1A R (SR49059) or OTR (L368,899). Immunohistochemical evidence of skin VP and OT receptors is given. CONCLUSIONS: Subcutaneous AVP produces antinociception and behavioural analgesia. Both V1a and OTR participate in those effects. Our findings suggest that antinociception could be produced in a local manner using a novel vasopressin receptor located in cutaneous sensorial fibres. Additionally, subcutaneous AVP also produces important systemic effects such as respiratory and locomotor impairment. SIGNIFICANCE: Our findings support that AVP produces peripheral antinociception and behavioural analgesia in a local manner; nevertheless, systemic effects are also presented. Additionally, this is the first detailed electrophysiological analysis of AVP antinociceptive action after subcutaneous administration. The results are reasonably explained by the demonstration of V1A R and OTR in cutaneous fibres.


Subject(s)
Evoked Potentials/drug effects , Nerve Fibers, Myelinated/drug effects , Nerve Fibers, Unmyelinated/drug effects , Nociception/drug effects , Receptors, Oxytocin/drug effects , Receptors, Vasopressin/drug effects , Vasopressins/pharmacology , Analgesics/pharmacology , Animals , Antidiuretic Hormone Receptor Antagonists/pharmacology , Behavior, Animal/drug effects , Camphanes/pharmacology , Indoles/pharmacology , Injections, Subcutaneous , Locomotion/drug effects , Male , Pain Measurement , Piperazines/pharmacology , Pyrrolidines/pharmacology , Rats , Receptors, Oxytocin/antagonists & inhibitors
11.
Exp Physiol ; 103(2): 222-235, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29143998

ABSTRACT

NEW FINDINGS: What is the central question of this study? We hypothesized that potential anti-tumour effects of exercise training might be mediated by oxytocin and explored the underlying mechanisms in a mouse model of breast cancer. What is the main finding and its importance? Interval exercise training, by inducing oxytocin secretion, may reduce the activity of the PI3K/Akt and ERK pathways, and consequently, results in a smaller tumour volume in a mouse model of breast cancer. Exercise training can affect the growth of breast tumours. We hypothesized that exercise training might reduce breast tumour growth by inducing oxytocin (OT) secretion and its related signalling pathways, such as PI3K/Akt and ERK. Therefore, 56 BALB/c mice were equally divided into seven groups to study the effects of OT and atosiban (an oxytocin receptor antagonist) together with interval exercise training on mammary tumour growth, as well as tumour-related signalling pathways, including PI3K/Akt and ERK. Animal weight, OT plasma concentration, tumour weight and volume were measured at the end of the study. PI3K/Akt and ERK were evaluated by Western blot and qPCR assays. The results showed that OT plasma concentration was significantly increased in trained animals. The volume and weight of tumours were decreased significantly after both exercise training and OT administration. The expression of genes involved in tumour cell proliferation, such as PI3KR2, Akt and mTOR, was notably lower in the exercise-trained and OT-treated groups. Furthermore, the expression of genes involved in cell apoptosis, such as caspase-3 and Bax, was significantly increased in the tumour tissues. In addition, Western blot results showed that phosphorylated Akt and ERK were significantly decreased in the exercise training and OT groups compared with the tumour group. Interestingly, atosiban reversed these effects. These results indicated that interval exercise training, acting via OT secretion, may reduce PI3K/Akt and ERK axis activities, and consequently, decrease tumour volume and weight in a mouse model of breast cancer.


Subject(s)
Hormone Antagonists/pharmacology , Oxytocin/pharmacology , Receptors, Oxytocin/drug effects , Vasotocin/analogs & derivatives , Animals , Mice, Inbred BALB C , Oxytocin/blood , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Physical Conditioning, Animal/methods , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Oxytocin/metabolism , TOR Serine-Threonine Kinases/drug effects , Vasotocin/pharmacology
13.
Behav Pharmacol ; 28(6): 450-457, 2017 09.
Article in English | MEDLINE | ID: mdl-28590943

ABSTRACT

To better understand the role of the neuropeptide oxytocin in autism spectrum disorder (ASD), we investigated potential deficits in social play behaviour and oxytocin receptor (OXTR) density alterations in the amygdala in a rodent model of ASD. Pregnant rats were injected daily with 20 or 100 mg/kg valproic acid (VPA) or saline from day 12 until the end of pregnancy. The number of pinning and pouncing events was assessed at postnatal days 29-34. Brains from male offspring (n=7/group) were removed at postnatal day 50. We performed quantitative autoradiography with an OXTR radioligand, the [I]-ornithine vasotocin analogue, in brain slices from the amygdala and other limbic brain regions involved in rat social behaviour. The results demonstrated a significant reduction in pinning behaviour and decreased OXTR density in the central nucleus of the amygdala in the 20 mg/kg VPA group. However, the 100 mg/kg VPA group had no significant changes in the number of play behaviour-related events or OXTR binding in the central nucleus of the amygdala. The reduction in OXTR density in the amygdala may be a critical disrupting mechanism affecting social behaviour in pervasive disorders such as ASD.


Subject(s)
Oxytocin/drug effects , Valproic Acid/adverse effects , Valproic Acid/pharmacology , Amygdala/drug effects , Amygdala/metabolism , Animals , Behavior, Animal/drug effects , Brain/metabolism , Female , Male , Oxytocin/metabolism , Play and Playthings/psychology , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Rats , Rats, Wistar , Receptors, Oxytocin/drug effects , Social Behavior
14.
Horm Behav ; 93: 94-98, 2017 07.
Article in English | MEDLINE | ID: mdl-28512038

ABSTRACT

Exploration of novel environments, stimuli, and conspecifics is highly adaptive during the juvenile period, as individuals transition from immaturity to adulthood. We recently showed that juvenile rats prefer to interact with a novel individual over a familiar cage mate. However, the neural mechanisms underlying this juvenile social novelty-seeking behavior remain largely unknown. One potential candidate is the oxytocin (OXT) system, given its involvement in various motivated social behaviors. Here, we show that administration of the specific oxytocin receptor antagonist desGly-NH2,d(CH2)5-[Tyr(Me)2,Thr4]OVT reduces social novelty seeking-behavior in juvenile male rats when injected into the nucleus accumbens (10ng/0.5µl/side). The same drug dose was ineffective at altering social novelty-seeking behavior when administered into the lateral septum or basolateral amygdala. These results are the first to suggest the involvement of the OXT system in the nucleus accumbens in the regulation of juvenile social novelty-seeking behavior.


Subject(s)
Exploratory Behavior/drug effects , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Oxytocin/pharmacology , Receptors, Oxytocin/metabolism , Social Behavior , Age Factors , Animals , Behavior, Animal/drug effects , Hormone Antagonists/pharmacology , Male , Motivation , Oxytocin/metabolism , Rats , Rats, Wistar , Receptors, Oxytocin/drug effects
15.
Am J Physiol Endocrinol Metab ; 312(3): E183-E189, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28049625

ABSTRACT

It has been demonstrated that the neuropeptide oxytocin (OT) attenuates oxidative stress and inflammation in macrophages. In the current study, we examined the role of inflammation on the expression of the oxytocin receptor (OXTR). We hypothesized that OXTR expression is increased during the inflammation through a nuclear factor-κB (NF-κB)-mediated pathway, thus responding as an acute-phase protein. Inflammation was induced by treating macrophages (human primary, THP-1, and murine) with lipopolysaccharide (LPS) and monitored by expression of IL-6. Expression of OXTR and vasopressin receptors was assessed by qPCR, and OXTR expression was confirmed by immunoblotting. Inflammation upregulated OXTR transcription 10- to 250-fold relative to control in THP-1 and human primary macrophages and increased OXTR protein expression. In contrast, vasopressin receptor-2 mRNA expression was reduced following LPS treatment. Blocking NF-κB activation prevented the increase in OXTR transcription. OT treatment of control cells and LPS-treated cells increased ERK1/2 phosphorylation, demonstrating activation of the OXTR/Gαq/11 signaling pathway. OT activation of OXTR reduced secretion of IL-6 in LPS-activated macrophages. Collectively, these findings suggest that OXTR is an acute-phase protein and that its increased expression is regulated by NF-κB and functions to attenuate cellular inflammatory responses in macrophages.


Subject(s)
Macrophages/metabolism , Receptors, Oxytocin/genetics , Animals , Blotting, Western , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gene Expression Regulation/drug effects , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages, Peritoneal , Male , Mice , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/drug effects , NF-kappa B/metabolism , Oxytocics/pharmacology , Oxytocin/pharmacology , Phosphorylation/drug effects , Polymerase Chain Reaction , Real-Time Polymerase Chain Reaction , Receptors, Oxytocin/drug effects , Receptors, Vasopressin/drug effects , Receptors, Vasopressin/genetics
16.
Am J Obstet Gynecol ; 216(3): 283.e1-283.e14, 2017 03.
Article in English | MEDLINE | ID: mdl-27567564

ABSTRACT

BACKGROUND: The ability to provide safe and effective pharmacotherapy during obstetric complications, such as preterm labor or postpartum hemorrhage, is hampered by the systemic toxicity of therapeutic agents leading to adverse side effects in the mother and fetus. Development of novel strategies to target tocolytic and uterotonic agents specifically to uterine myocytes would improve therapeutic efficacy while minimizing the risk of side effects. Ligand-targeted liposomes have emerged as a reliable and versatile platform for targeted drug delivery to specific cell types, tissues or organs. OBJECTIVE: Our objective was to develop a targeted drug delivery system for the uterus utilizing an immunoliposome platform targeting the oxytocin receptor. STUDY DESIGN: We conjugated liposomes to an antibody that recognizes an extracellular domain of the oxytocin receptor. We then examined the ability of oxytocin receptor-targeted liposomes to deliver contraction-blocking (nifedipine, salbutamol and rolipram) or contraction-enhancing (dofetilide) agents to strips of spontaneously contracting myometrial tissue in vitro (human and mouse). We evaluated the ability of oxytocin receptor-targeted liposomes to localize to uterine tissue in vivo, and assessed if targeted liposomes loaded with indomethacin were capable of preventing lipopolysaccharide-induced preterm birth in mice. RESULTS: Oxytocin receptor-targeted liposomes loaded with nifedipine, salbutamol or rolipram consistently abolished human myometrial contractions in vitro, while oxytocin receptor-targeted liposomes loaded with dofetilide increased contraction duration. Nontargeted control liposomes loaded with these agents had no effect. Similar results were observed in mouse uterine strips. Following in vivo administration to pregnant mice, oxytocin receptor-targeted liposomes localized specifically to the uterine horns and mammary tissue. Targeting increased localization to the uterus 7-fold. Localization was not detected in the maternal brain or fetus. Targeted and nontargeted liposomes also localized to the liver. Oxytocin receptor-targeted liposomes loaded with indomethacin were effective in reducing rates of preterm birth in mice, whereas nontargeted liposomes loaded with indomethacin had no effect. CONCLUSION: Our results demonstrate that oxytocin receptor-targeted liposomes can be used to either inhibit or enhance human uterine contractions in vitro. In vivo, the liposomes localized to the uterine tissue of pregnant mice and were effective in delivering agents for the prevention of inflammation-induced preterm labor. The potential clinical advantage of targeted liposomal drug delivery to the myometrium is reduced dose and reduced toxicity to both mother and fetus.


Subject(s)
Premature Birth/prevention & control , Receptors, Oxytocin/drug effects , Uterine Contraction/drug effects , Uterus/drug effects , Albuterol/administration & dosage , Albuterol/pharmacokinetics , Animals , Drug Delivery Systems , Female , Indomethacin/administration & dosage , Liposomes/immunology , Mice , Myometrium/drug effects , Myometrium/metabolism , Nifedipine/administration & dosage , Nifedipine/pharmacokinetics , Phenethylamines/administration & dosage , Phenethylamines/pharmacokinetics , Pregnancy , Rolipram/administration & dosage , Rolipram/pharmacokinetics , Sulfonamides/administration & dosage , Sulfonamides/pharmacokinetics , Tissue Distribution , Uterine Contraction/immunology , Uterus/immunology
17.
Schmerz ; 30(5): 457-469, 2016 Oct.
Article in German | MEDLINE | ID: mdl-27681779

ABSTRACT

BACKGROUND: Over the years the effect of the neuropeptide oxytocin and its possible utilization for pain management has been increasingly more investigated and discussed. Initial results emphasized the effects of oxytocin with respect to labor and breastfeeding. Diverse animals studies were also able to demonstrate the effectiveness of the peptide in attachment behavior and pain perception; however, it is still unclear how oxytocin affects pain perception in humans. The potential therapeutic effectiveness of oxytocin could be particularly important for primary and secondary treatment of pain patients because chronification of pain can occur more frequently in this area. METHODS: For this review the databases PubMed, Medline und PsycINFO were searched using the terms oxytocin, pain, human and analgesic. The search resulted in a total of 89 original articles after excluding articles regarding labor pain, breastfeeding and animal studies. Only those studies were included which were carried out between 1994 and 2015. A total of 17 articles remained for inclusion in this review and included 13 studies on the exogenous application of oxytocin and 4 on measurement of oxytocin levels in plasma. CONCLUSION: This review article gives a summary of the current state of research on oxytocin and its direct and indirect association with human pain perception and emphasizes its relevance for the multimodal management of pain.


Subject(s)
Oxytocin/physiology , Oxytocin/therapeutic use , Pain Perception/drug effects , Pain Perception/physiology , Affect/drug effects , Affect/physiology , Animals , Brain/drug effects , Brain/physiopathology , Breast Feeding/psychology , Chronic Pain/drug therapy , Chronic Pain/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Labor Pain/physiopathology , Labor Pain/psychology , Mice , Nociceptors/drug effects , Nociceptors/physiology , Pain Management/methods , Pain Threshold/drug effects , Pain Threshold/physiology , Pregnancy , Randomized Controlled Trials as Topic , Rats , Receptors, Oxytocin/drug effects , Receptors, Oxytocin/physiology
18.
Toxicol Lett ; 256: 44-52, 2016 Aug 10.
Article in English | MEDLINE | ID: mdl-27220266

ABSTRACT

Sildenafil is a phosphodiesterase type 5 inhibitor mainly used for male erectile dysfunction. One of rare yet serious adverse effects of Sildenafil is its potential to decrease seizure threshold. Ample evidence suggests that Sildenafil exerts central effects through induction of Oxytocin (OT) secretion and CREB phosphorylation. The aim of the present study is to evaluate potential roles of OT and CREB in the proconvulsant effects of Sildenafil. The Pentylenetetrazole-induced seizure was used as a standard convulsion model in this study. OT release and pCREB expression were evaluated in the hippocampus of mice using ELISA and western blot assays, respectively. Our results showed that Sildenafil at the dose of 10mgkg(-1) or higher, significantly decreased seizure threshold. Pretreatment with a non-effective dose of OT, potentiated while OT receptor antagonist, Atosiban, reversed fully the proconvulsant effects of Sildenafil (5mgkg(-1)). At biochemical inspection, Sildenafil markedly increased CREB which was attenuated by coadministration of Atosiban. The present study shows for the first time that OT release and the subsequent CREB phosphorylation are involved in the proconvulsant effects of acute Sildenafil treatment in an experimental model of seizure.


Subject(s)
CREB-Binding Protein/metabolism , Hippocampus/drug effects , Oxytocin/metabolism , Phosphodiesterase 5 Inhibitors/toxicity , Seizures/chemically induced , Sildenafil Citrate/toxicity , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Hippocampus/metabolism , Hippocampus/physiopathology , Hormone Antagonists/pharmacology , Male , Mice , Pentylenetetrazole , Phosphorylation , Receptors, Oxytocin/drug effects , Receptors, Oxytocin/metabolism , Seizures/metabolism , Seizures/physiopathology , Signal Transduction/drug effects , Time Factors , Vasotocin/analogs & derivatives , Vasotocin/pharmacology
19.
Endocrinology ; 157(2): 692-704, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26671185

ABSTRACT

To determine the comprehensive G protein-coupled receptor (GPCR) expression profile in ghrelin-producing cells and to elucidate the role of GPCR-mediated signaling in the regulation of ghrelin secretion, we determined GPCR expression profiles by RNA sequencing in the ghrelin-producing cell line MGN3-1 and analyzed the effects of ligands for highly expressed receptors on intracellular signaling and ghrelin secretion. Expression of selected GPCRs was confirmed in fluorescence-activated cell-sorted fluorescently tagged ghrelin-producing cells from ghrelin-promoter CreERT2/Rosa-CAG-LSL-ZsGreen1 mice. Expression levels of GPCRs previously suggested to regulate ghrelin secretion including adrenergic-ß1 receptor, GPR81, oxytocin receptor, GPR120, and somatostatin receptor 2 were high in MGN3-1 cells. Consistent with previous reports, isoproterenol and oxytocin stimulated the Gs and Gq pathways, respectively, whereas lactate, palmitate, and somatostatin stimulated the Gi pathway, confirming the reliability of current assays. Among other highly expressed GPCRs, prostaglandin E receptor 4 agonist prostaglandin E2 significantly stimulated the Gs pathway and ghrelin secretion. Muscarine, the canonical agonist of cholinergic receptor muscarinic 4, stimulated both the Gq and Gi pathways. Although muscarine treatment alone did not affect ghrelin secretion, it did suppress forskolin-induced ghrelin secretion, suggesting that the cholinergic pathway may play a role in counterbalancing the stimulation of ghrelin by Gs (eg, by adrenaline). In addition, GPR142 ligand tryptophan stimulated ghrelin secretion. In conclusion, we determined the comprehensive expression profile of GPCRs in ghrelin-producing cells and identified two novel ghrelin regulators, prostaglandin E2 and tryptophan. These results will lead to a greater understanding of the physiology of ghrelin and facilitate the development of ghrelin-modulating drugs.


Subject(s)
Gastric Mucosa/metabolism , Ghrelin/metabolism , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Adrenergic beta-Agonists/pharmacology , Animals , Cell Line, Tumor , Colforsin/pharmacology , Dinoprostone/pharmacology , Gastric Mucosa/cytology , Gastric Mucosa/drug effects , Gene Expression Profiling , Ghrelin/drug effects , Hormones/pharmacology , Immunohistochemistry , Isoproterenol/pharmacology , Lactic Acid/pharmacology , Mice , Mice, Transgenic , Muscarine/pharmacology , Muscarinic Agonists/pharmacology , Oxytocics/pharmacology , Oxytocin/pharmacology , Palmitates/pharmacology , Receptor, Muscarinic M4/agonists , Receptors, Adrenergic, beta-1/drug effects , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-1/metabolism , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, Oxytocin/drug effects , Receptors, Oxytocin/genetics , Receptors, Oxytocin/metabolism , Receptors, Prostaglandin E, EP4 Subtype/agonists , Receptors, Somatostatin/drug effects , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Sequence Analysis, RNA , Somatostatin/pharmacology , Tryptophan/pharmacology
20.
J Neuroendocrinol ; 26(6): 356-69, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24888645

ABSTRACT

Oxytocin is a nonapeptide hormone that has a central role in the regulation of parturition and lactation. In this review, we address oxytocin receptor (OTR) signalling and its role in the myometrium during pregnancy and in labour. The OTR belongs to the rhodopsin-type (Class 1) of the G-protein coupled receptor superfamily and is regulated by changes in receptor expression, receptor desensitisation and local changes in oxytocin concentration. Receptor activation triggers a number of signalling events to stimulate contraction, primarily by elevating intracellular calcium (Ca(2+) ). This includes inositol-tris-phosphate-mediated store calcium release, store-operated Ca(2+) entry and voltage-operated Ca(2+) entry. We discuss each mechanism in turn and also discuss Ca(2+) -independent mechanisms such as Ca(2+) sensitisation. Because oxytocin induces contraction in the myometrium, both the activation and the inhibition of its receptor have long been targets in the management of dysfunctional and preterm labours, respectively. We discuss current and novel OTR agonists and antagonists and their use and potential benefit in obstetric practice. In this regard, we highlight three clinical scenarios: dysfunctional labour, postpartum haemorrhage and preterm birth.


Subject(s)
Myometrium/physiology , Oxytocin/physiology , Receptors, Oxytocin/physiology , Signal Transduction/physiology , Adult , Female , Humans , Male , Oxytocics/pharmacology , Oxytocin/pharmacology , Parturition/drug effects , Pregnancy , Receptors, Oxytocin/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...