Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 323
Filter
1.
Neuroscience ; 497: 30-38, 2022 08 10.
Article in English | MEDLINE | ID: mdl-35183689

ABSTRACT

A possible role for the brain ß-endorphin system in memory modulation was proposed by Ivan Izquierdo more than 30 years ago. Along with pharmacologic evidence of the effects of morphine and naloxone administered immediately after training in avoidance tasks and with the demonstration of medial-basal hypothalamus ß-endorphin release after novelty detection, it was hypothesized that an endogenous opioid state present in the labile period of consolidation will be part of the memory of the newly acquired information. The fact that pre-test novelty exposure, through release of ß-endorphin, or the injection of opioids facilitate retrieval supports that. The mechanisms through which novelty exerts its retrieval-enhancing effect were studied; evidence that several forms of amnesia induced by post-training treatments are due to unavailability of retrieval and not to a storage deficit, challenging the memory consolidation framework is discussed. In this review some of the original papers in the subject are revisited. Recent studies on the memory beneficial effects of novelty, both in animal models and in humans, indicate this is line of investigation is worth of pursuing and demonstrate the importance of the seminal work of Ivan Izquierdo in the field of memory modulation.


Subject(s)
Memory , beta-Endorphin , Animals , Avoidance Learning , Brain , Humans , Morphine/pharmacology , Naloxone/pharmacology , beta-Endorphin/physiology
2.
CNS Neurosci Ther ; 27(10): 1157-1172, 2021 10.
Article in English | MEDLINE | ID: mdl-34111331

ABSTRACT

AIM: This study aimed to investigate the regulation of pain hypersensitivity induced by the spinal synaptic transmission mechanisms underlying interleukin (IL)-10 and glucagon-like peptide 1 receptor (GLP-1R) agonist exenatide-induced pain anti-hypersensitivity in neuropathic rats through spinal nerve ligations. METHODS: Neuropathic pain model was established by spinal nerve ligation of L5/L6 and verified by electrophysiological recording and immunofluorescence staining. Microglial expression of ß-endorphin through autocrine IL-10- and exenatide-induced inhibition of glutamatergic transmission were performed by behavioral tests coupled with whole-cell recording of miniature excitatory postsynaptic currents (mEPSCs) and miniature inhibitory postsynaptic currents (mIPSCs) through application of endogenous and exogenous IL-10 and ß-endorphin. RESULTS: Intrathecal injections of IL-10, exenatide, and the µ-opioid receptor (MOR) agonists ß-endorphin and DAMGO inhibited thermal hyperalgesia and mechanical allodynia in neuropathic rats. Whole-cell recordings of bath application of exenatide, IL-10, and ß-endorphin showed similarly suppressed enhanced frequency and amplitude of the mEPSCs in the spinal dorsal horn neurons of laminae II, but did not reduce the frequency and amplitude of mIPSCs in neuropathic rats. The inhibitory effects of IL-10 and exenatide on pain hypersensitive behaviors and spinal synaptic plasticity were totally blocked by pretreatment of IL-10 antibody, ß-endorphin antiserum, and MOR antagonist CTAP. In addition, the microglial metabolic inhibitor minocycline blocked the inhibitory effects of IL-10 and exenatide but not ß-endorphin on spinal synaptic plasticity. CONCLUSION: This suggests that spinal microglial expression of ß-endorphin mediates IL-10- and exenatide-induced inhibition of glutamatergic transmission and pain hypersensitivity via presynaptic and postsynaptic MORs in spinal dorsal horn.


Subject(s)
Exenatide/pharmacology , Interleukin-10 , Microglia , Neuralgia/physiopathology , Neuronal Plasticity/drug effects , Spinal Nerves/physiopathology , beta-Endorphin/physiology , Analgesics, Opioid/pharmacology , Animals , Behavior, Animal/drug effects , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Excitatory Postsynaptic Potentials , Glutamic Acid , Injections, Spinal , Interleukin-10/metabolism , Interleukin-10/pharmacology , Neuralgia/psychology , Patch-Clamp Techniques , Rats , Receptors, Opioid, mu/agonists , Signal Transduction , Synaptic Transmission , beta-Endorphin/pharmacology
3.
Neuropsychobiology ; 78(3): 118-127, 2019.
Article in English | MEDLINE | ID: mdl-31117084

ABSTRACT

BACKGROUND: Alcohol is one of the leading threats to health worldwide. Craving for alcohol makes abstinence a difficult challenge by maintaining alcohol dependence. Many studies suppose the hypothalamic-pituitary-adrenal axis, especially the proopiomelanocortin (POMC)-derived neuropeptides, to mediate craving during withdrawal in alcohol dependence. Evidence is available that the two POMC proteins, α-melanocyte-stimulating hormone (α-MSH) and ß-endorphin (ß-END) are altered by alcohol consumption and influence alcohol consumption, respectively. OBJECTIVES: We investigated the dynamics of α-MSH and ß-END during alcohol withdrawal and the influence of intraperitoneal administration of either α-MSH or ß-END in an established rodent model (Wistar rats) for alcohol dependence. RESULTS: After long-term alcohol self-administration over 12 months and repeated deprivation periods for 3 days, we found a significant decrease in α-MSH levels during withdrawal in rodents (p = 0.006) compared to controls, while ß-END levels remained unchanged. Treatment with intraperitoneally administered α-MSH and ß-END did not affect alcohol drinking behavior after deprivation. CONCLUSION: We demonstrate the effects of alcohol deprivation on α-MSH in alcohol-dependent rodents, which appear to mimic α-MSH alteration found after fasting periods during appetite regulation. Therefore, low α-MSH levels are a possible indicator for craving in alcohol-dependent individuals and hence would be a potential target for anti-craving treatment.


Subject(s)
Alcoholism/physiopathology , Ethanol/administration & dosage , alpha-MSH/physiology , beta-Endorphin/physiology , Alcohol Drinking , Animals , Disease Models, Animal , Male , Rats, Wistar , alpha-MSH/administration & dosage , alpha-MSH/blood , beta-Endorphin/administration & dosage , beta-Endorphin/blood
4.
Andrologia ; 50(7): e13049, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29781087

ABSTRACT

It has been assumed that ß-endorphin, belonging to the family of opiodergic neuropeptides, might facilitate the inhibition of the male sexual response; however, its role in the control of the penile erectile tissue remains to be elucidated. This study aimed to evaluate in healthy men the course of ß-endorphin in the systemic and cavernous blood through different stages of sexual arousal. Thirty-four (34) men were exposed to erotic stimuli to induce penile tumescence and rigidity. Blood was aspirated from the corpus cavernosum and a cubital vein during the penile conditions flaccidity, tumescence, rigidity and detumescence. Plasma levels of ß-endorphin were determined by means of radioimmunometric methods. The effects of ß-endorphin on isolated human penile erectile tissue were investigated in vitro. ß-endorphin did not induce a contractile response of the cavernous tissue or reverse the contraction induced by noradrenaline. ß-endorphin decreased in the systemic blood when the penis became tumescent and rigid and increased during detumescence. In the cavernous blood, no alterations in ß-endorphin concentrations were observed. The drop in ß-endorphin observed during tumescence and rigidity seems likely to reflect the inhibition of the opioidergic input with the beginning of sexual arousal.


Subject(s)
Arousal/physiology , Penile Erection/physiology , Sexual Behavior/physiology , beta-Endorphin/physiology , Adult , Female , Humans , Male , Norepinephrine/pharmacology , Penis/drug effects , Transgender Persons , beta-Endorphin/blood
5.
Alcohol ; 53: 51-60, 2016 06.
Article in English | MEDLINE | ID: mdl-27286936

ABSTRACT

Animal models have long been used to study the mechanisms underlying the complex association between alcohol and stress. Female mice prevented from running on a home-cage activity wheel increase voluntary ethanol consumption. ß-endorphin is an endogenous opioid involved in negatively regulating the stress response and has also been implicated in the risk for excessive drinking. The present study investigates the role of ß-endorphin in moderating free-choice consumption of ethanol in response to a blocked activity wheel. Female, transgenic mice with varying levels of the opioid peptide were given daily 2-h access to 20% ethanol with rotations on a running wheel blocked on alternate days. Subjects with low ß-endorphin exhibited enhanced stress sensitivity by self-administering larger quantities of ethanol on days when wheel running was prevented. ß-endorphin levels did not influence voluntary activity on the running wheel. There were genotypic differences in plasma corticosterone levels as well as corticotropin-releasing hormone mRNA content in multiple brain regions associated with the stress response in these free drinking and running subjects. Susceptibility to stress is enhanced in female mice with low levels of ß-endorphin, and better understanding of the role for this opioid in mitigating the response to stressors may aid in the development of interventions and treatments for excessive use of alcohol in women.


Subject(s)
Alcohol Drinking/metabolism , Brain/metabolism , Physical Conditioning, Animal/physiology , beta-Endorphin/deficiency , Alcohol Drinking/psychology , Animals , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Physical Conditioning, Animal/methods , Self Administration , beta-Endorphin/physiology
6.
Fluids Barriers CNS ; 12: 3, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25879522

ABSTRACT

Beta-endorphin (ß-END) is an opioid neuropeptide which has an important role in the development of hypotheses concerning the non-synaptic or paracrine communication of brain messages. This kind of communication between neurons has been designated volume transmission (VT) to differentiate it clearly from synaptic communication. VT occurs over short as well as long distances via the extracellular space in the brain, as well as via the cerebrospinal fluid (CSF) flowing through the ventricular spaces inside the brain and the arachnoid space surrounding the central nervous system (CNS). To understand how ß-END can have specific behavioral effects, we use the notion behavioral state, inspired by the concept of machine state, coming from Turing (Proc London Math Soc, Series 2,42:230-265, 1937). In section 1.4 the sequential organization of male rat behavior is explained showing that an animal is not free to switch into another state at any given moment. Funneling-constraints restrict the number of possible behavioral transitions in specific phases while at other moments in the sequence the transition to other behavioral states is almost completely open. The effects of ß-END on behaviors like food intake and sexual behavior, and the mechanisms involved in reward, meditation and pain control are discussed in detail. The effects on the sequential organization of behavior and on state transitions dominate the description of these effects.


Subject(s)
Behavior/physiology , Central Nervous System/physiology , beta-Endorphin/physiology , Animals , Behavior, Animal/physiology , Cell Communication , Eating , Extracellular Space/physiology , Humans , Motivation/physiology , Pain/physiopathology , Reward , Sexual Behavior/physiology
7.
Pharmacol Biochem Behav ; 121: 170-83, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24368305

ABSTRACT

Sexual behavior of the male rat can be described as a 'sequence': a series of behavioral transitions eventually leading to a consummatory act: ejaculation. A 'funnel-model' is presented to describe the behavioral progression during the sexual sequence. The ejaculation itself is extensively controlled by the 'spinal ejaculation generator', consisting of several elements with afferent sources of genitosensory information, with ascending projection fibers to inform the brainstem and forebrain as well as with descending afferent fibers providing the supraspinal control mechanisms with the opportunity to restrict ejaculations to the optimal moments and circumstances. The messages ascending from the spinal cord reach several interconnected thalamic, hypothalamic and limbic brain areas and are integrated with olfactory information. These brain areas play a role in mechanisms like 'sexual satiety' or a temporary interruption of sexual activities (post-ejaculatory interval), but the exact facilitatory and inhibitory mechanisms involved have not been elucidated yet. In the 'downward' mechanisms controlling the spinal 'release' of an ejaculation, the medial preoptic nucleus plays an important role in cooperation with a number of brainstem areas. This nucleus is also explicitly involved in the rewarding experiences coming with an ejaculation. Finally, the role of several neurotransmitters and-peptides on male sexual behavior are discussed shortly, because sometimes they show remarkable effects on specific aspects of the behavioral sequence. We conclude that, despite our increased knowledge about the brain mechanisms involved in the control of ejaculation, we are still far away from a complete understanding and quite a few questions remain to be resolved.


Subject(s)
Ejaculation/physiology , Sexual Behavior, Animal/physiology , Animals , Brain/physiology , Dopamine/physiology , Male , Models, Animal , Models, Neurological , Nerve Net/physiology , Norepinephrine/physiology , Oxytocin/physiology , Rats , Serotonin/physiology , Spinal Cord/physiology , beta-Endorphin/physiology
8.
Res Vet Sci ; 95(3): 975-85, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24148869

ABSTRACT

This study evaluated porcine natural killer cell cytotoxicity (NKCC), plasma cytokines including interleukin (IL) 1ß, IL-6, IL-10, IL-12 and tumor necrosis factor-α and plasma stress-related hormones including prolactin (PRL), growth hormone (GH), ß-endorphin (BEND), ACTH and cortisol (COR) during a 4h restraint and recovery phase after saline or naloxone (1mg/kg BW) administration. The restraint preceded with saline altered NKCC and IL-12 concentration (an early from 15 to 60 min increase followed by a decrease) and increased other measured cytokines and hormones concentrations. Naloxone pretreatment blocked the suppressive effects of the restraint on NKCC and IL-12 and altered IL-10, IL-6, TNF-α, PRL and ACTH concentrations. Furthermore, in naloxone-injected pigs, a positive correlation was found between NKCC and all measured cytokines (with the exception of IL-6) and BEND, ACTH and COR. Results suggest that naloxone-sensitive opioid pathways could influence the mechanisms underlying the immune system (including NKCC) response during stress.


Subject(s)
Cytokines/physiology , Killer Cells, Natural/physiology , Naloxone/pharmacology , Narcotic Antagonists , Restraint, Physical/veterinary , Adrenocorticotropic Hormone/blood , Adrenocorticotropic Hormone/physiology , Animals , Cytokines/blood , Growth Hormone/blood , Growth Hormone/physiology , Hydrocortisone/blood , Hydrocortisone/physiology , Interleukin-10/blood , Interleukin-10/physiology , Interleukin-12/blood , Interleukin-12/physiology , Interleukin-1beta/blood , Interleukin-1beta/physiology , Interleukin-6/blood , Interleukin-6/physiology , Male , Prolactin/blood , Prolactin/physiology , Restraint, Physical/physiology , Stress, Physiological/physiology , Swine/physiology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/physiology , beta-Endorphin/blood , beta-Endorphin/physiology
9.
Neuropharmacology ; 73: 290-300, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23770261

ABSTRACT

Substantial evidence has implicated the endogenous opioid system in alcohol reinforcement. However, the role of each opioid peptide in alcohol reinforcement and, particularly, reward is not fully characterized. In this study, using the conditioned place preference (CPP) paradigm as an animal model of reward, we determined the role of endogenous ß-endorphin and enkephalins in the rewarding action of ethanol. Female mice lacking beta-endorphin and/or the proenkephalin gene as well as their respective wild-type controls were tested for baseline place preference on day 1, conditioned with ethanol versus saline on days 2-4 and were then tested under a drug-free state for postconditioning place preference on day 5. On each test day, mice were placed in the central neutral chamber and allowed to freely explore all three CPP chambers. The amount of time that mice spent in each chamber was recorded. We also studied the effect of naloxone, a non-selective opioid receptor antagonist, on ethanol CPP, in which wild-type mice were treated with saline or naloxone 10 min prior to ethanol or saline conditioning. Our results showed that the absence of ß-endorphin or enkephalins alone failed to alter the acquisition of ethanol-induced CPP. However, the absence of both ß-endorphin and enkephalins significantly reduced the CPP response. Interestingly, high but not low dose naloxone blunted ethanol CPP. These findings provide the first evidence illustrating that ethanol induces its rewarding action, at least in part, via a joint action of ß-endorphin and enkephalins, possibly involving both mu and delta opioid receptors.


Subject(s)
Enkephalins/physiology , Ethanol/pharmacology , Protein Precursors/physiology , Reward , beta-Endorphin/physiology , Animals , Choice Behavior/physiology , Conditioning, Psychological/drug effects , Dose-Response Relationship, Drug , Enkephalins/genetics , Ethanol/antagonists & inhibitors , Female , Mice , Mice, Knockout , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Protein Precursors/genetics , beta-Endorphin/genetics
10.
Yakugaku Zasshi ; 133(5): 493-9, 2013.
Article in Japanese | MEDLINE | ID: mdl-23649389

ABSTRACT

Docosahexaenoic acid (DHA), a predominant of n-3 polyunsaturated fatty acids (n-3 PUFA), has numerous beneficial physiological effects, including neuroprotection and cardiovascular protection. Recently, a possible involvement of n-3 PUFA in pain control has gathered considerable attention because numerous studies have reported a regulatory role of n-3 PUFAs. However, the mechanisms underlying how DHA exerts antinociceptive effect remain unknown. Here, we performed elucidation of mechanisms underlying DHA-induced antinociception. DHA administration dose-dependently exerted an antinociceptive effect. This effect was abolished by pretreated with the ß-funaltrexamine (ß-FNA), a µ-opioid receptor antagonist, and the nartrindole (NTI), a δ-opioid receptor antagonist, but not by the nor-binaltorphimine (nor-BNI), a κ-opioid receptor antagonist. In the radioligand binding assay, DHA itself did not have the affinity for µ-, δ- and κ- opioid receptor. Furthermore, the pretreatment of anti ß-endorphin antiserum inhibited DHA-induced antinociception. The plasma levels of ß-endorphin increased 30 min after DHA administration. The ß-endorphin immunoreactivity in the brain increased at 30 min after DHA treatment. Expression of GPR40 protein was widely observed in the brain as well as the spinal cord. The intracerebroventricular but not intrathecal injection of DHA and GW9508, a GPR40/GPR120 agonist, significantly reduced formalin-induced pain behavior. The ß-endorphin immunoreactivity in the brain increased at 10 and 20 min after intracerebroventricular injection of DHA and GW9508. These findings suggest that DHA-induced antinociception via ß-endorphin release may be mediated through GPR40 signaling in the supraspinal area.


Subject(s)
Analgesics/pharmacology , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/physiology , Animals , Brain/metabolism , Docosahexaenoic Acids/metabolism , Dose-Response Relationship, Drug , Fatty Acids, Omega-3 , Mice , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/physiology , Signal Transduction/genetics , Signal Transduction/physiology , beta-Endorphin/metabolism , beta-Endorphin/physiology
11.
Phytother Res ; 27(10): 1564-71, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23339028

ABSTRACT

Although Berberine (BER) is popular in treating gastrointestinal (GI) disorders, its mechanisms are not clear yet. In order to investigate the effects and possible mechanism of BER on GI motility in rodents, we first explored GI motility by recording the myoelectrical activity of jejunum and colon in rats, and upper GI transit with a charcoal marker in mice. Then, the plasma levels of gastrin, motilin, somatostatin and glucagon-like-peptide-1 (Glp-1) were measured by ELISA or radioimmunoassay (RIA). Furthermore, endogenous opioid-peptides (ß-endorphin, dynorphin-A, met-enkephalin) were detected by RIA after treatment with BER. Our results showed that BER concentration-dependently inhibited myoelectrical activity and GI transit, which can be antagonized by opioid-receptor antagonists to different extents. The elevated somatostatin and Glp-1, and decreased gastrin and motilin in plasma, which were caused by BER application, also could be antagonized by the opioid-receptor antagonists. Additionally, plasma level of ß-endorphin, but not dynorphin-A and met-enkephalin, was increased by applying BER. Taken together, these studies show that BER plays inhibiting roles on GI motility and up-regulating roles on somatostatin, Glp-1 and down-regulating roles on gastrin, motilin. The pharmacological mechanisms of BER on GI motility and plasma levels of GI hormones were discovered to be closely related to endogenous opioid system.


Subject(s)
Berberine/pharmacology , Gastrointestinal Hormones/physiology , Gastrointestinal Motility/drug effects , Gastrointestinal Tract/drug effects , Opioid Peptides/physiology , Animals , Colon/drug effects , Colon/physiology , Dynorphins/physiology , Enkephalin, Methionine/physiology , Gastrins/physiology , Gastrointestinal Tract/physiology , Gastrointestinal Transit/drug effects , Gastrointestinal Transit/physiology , Glucagon-Like Peptide 1/physiology , Jejunum/drug effects , Jejunum/physiology , Male , Mice , Mice, Inbred BALB C , Motilin/physiology , Rats , Rats, Sprague-Dawley , Somatostatin/physiology , beta-Endorphin/physiology
12.
Addict Biol ; 18(4): 614-22, 2013 Jul.
Article in English | MEDLINE | ID: mdl-21812868

ABSTRACT

In the present study, we investigated the possible development of tolerance to the antihyperalgesic effect of µ-opioid receptor (MOR) agonists under a neuropathic pain-like state. Repeated treatment with fentanyl, but not morphine or oxycodone, produced a rapid development of tolerance to its antihyperalgesic effect in mice with sciatic nerve ligation. Like the behavioral study, G-protein activation induced by fentanyl was significantly reduced in membranes obtained from the spinal cord of nerve-ligated mice with in vivo repeated injection of fentanyl. In ß-endorphin-knockout mice with nerve ligation, developed tolerance to the antihyperalgesic effect of fentanyl was abolished, and reduced G-protein activation by fentanyl after nerve ligation with fentanyl was reversed to the normal level. The present findings indicate that released ß-endorphin within the spinal cord may be implicated in the rapid development of tolerance to fentanyl under a neuropathic pain-like state.


Subject(s)
Analgesics, Opioid/pharmacology , Drug Tolerance/physiology , Fentanyl/pharmacology , Neuralgia/drug therapy , Receptors, Opioid, mu/agonists , Spinal Cord/metabolism , beta-Endorphin/physiology , Analgesics, Opioid/administration & dosage , Animals , Dose-Response Relationship, Drug , Female , Fentanyl/administration & dosage , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Hot Temperature , Hyperalgesia/drug therapy , Injections, Subcutaneous , Ligation , Male , Mice , Mice, Knockout , Morphine/administration & dosage , Morphine/pharmacology , Neuralgia/metabolism , Oxycodone/administration & dosage , Oxycodone/pharmacology , Pain Measurement/methods , Pain Threshold/drug effects , Radioligand Assay , Receptors, Opioid, mu/physiology , Sciatic Nerve/surgery , Sodium Chloride/administration & dosage , beta-Endorphin/genetics
13.
Eur J Pharmacol ; 686(1-3): 50-4, 2012 Jul 05.
Article in English | MEDLINE | ID: mdl-22575525

ABSTRACT

We have previously shown that ß-endorphin plays a functional role in the rewarding effect of acute cocaine. Considering that ß-endorphin has high affinity for the µ opioid receptor, we determined the role of this receptor in the rewarding action of acute cocaine. For comparison, we assessed the role of the µ opioid receptor in the rewarding effect of acute morphine. We also examined the effect of intracerebroventricular (i.c.v.) administration of ß-funaltrexamine (ß-FNA), an irreversible µ opioid receptor antagonist, on the rewarding action of acute cocaine as well as that of morphine. Using the conditioned place preference (CPP) paradigm as an animal model of reward, we first assessed the rewarding action of cocaine in mice lacking ß-endorphin or the µ opioid receptor and their respective wild-type littermates/controls. Mice were tested for preconditioning place preference on day 1, conditioned once daily with saline/cocaine (30mg/kg, i.p.) or cocaine/saline on days 2 and 3, and then tested for postconditioning place preference on day 4. We next studied the rewarding action of acute morphine in µ knockout mice and their wild-type controls. The CPP was induced by single alternate-day saline/morphine (10mg/kg, s.c.) or morphine/saline conditioning. We finally determined the effect of ß-FNA on CPP induced by cocaine or morphine in wild-type mice, in which mice were treated with saline or ß-FNA (9ug/3µl; i.c.v.) a day prior to the preconditioning test day. Our results revealed that morphine induced a robust CPP in wild-type mice but not in mice lacking the µ opioid receptor or in wild-type mice treated with ß-FNA. In contrast, cocaine induced CPP in µ knockout mice as well as in wild-type mice treated with ß-FNA. On the other hand, cocaine failed to induce CPP in mice lacking ß-endorphin. These results illustrate that ß-endorphin is essential for the rewarding action of acute cocaine, but the µ opioid receptor may not mediate the regulatory action of endogenous ß-endorphin.


Subject(s)
Cocaine/pharmacology , Conditioning, Operant/physiology , Receptors, Opioid, mu/physiology , beta-Endorphin/physiology , Animals , Behavior, Animal/drug effects , Male , Mice , Mice, Knockout , Morphine/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Reward
14.
Brain Res ; 1448: 63-70, 2012 Apr 11.
Article in English | MEDLINE | ID: mdl-22381895

ABSTRACT

We have shown previously that intracerebroventricular (icv) injection of naloxone (a non-selective opioid receptor antagonist) or naloxonazine (a selective µ1-opioid receptor antagonist) at the maintenance phase of hibernation arouses Syrian hamsters from hibernation. This study was designed to clarify the role of ß-endorphin (an endogenous µ-opioid receptor ligand) on regulation of body temperature (T(b)) during the maintenance phase of hibernation. The number of c-Fos-positive cells and ß-endorphin-like immunoreactivity increased in the arcuate nucleus (ARC) after hibernation onset. In contrast, endomorphin-1 (an endogenous µ-opioid receptor ligand)-like immunoreactivity observed on the anterior hypothalamus decreased after hibernation onset. In addition, hibernation was interrupted by icv injection of anti-ß-endorphin antiserum at the maintenance phase of hibernation. The mRNA expression level of proopiomelanocortin (a precursor of ß-endorphin) on ARC did not change throughout the hibernation phase. However, the mRNA expression level of prohormone convertase-1 increased after hibernation onset. [D-Ala2,N-MePhe4,Gly-ol5] enkephalin (DAMGO, a selective µ-opioid receptor agonist) microinjection into the dorsomedial hypothalamus (DMH) elicited the most marked T(b) decrease than other sites such as the preoptic area (PO), anterior hypothalamus (AH), lateral hypothalamus (LH), ventromedial hypothalamus and posterior hypothalamus (PH). However, microinjected DAMGO into the medial septum indicated negligible changes in T(b). These results suggest that ß-endorphin which synthesizes in ARC neurons regulates T(b) during the maintenance phase of hibernation by activating µ-opioid receptors in PO, AH, VMH, DMH and PH.


Subject(s)
Body Temperature Regulation/physiology , Central Nervous System/physiology , Hibernation/physiology , beta-Endorphin/physiology , Analgesics, Opioid/pharmacology , Animals , Brain Chemistry/physiology , Cell Count , Cricetinae , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Hypothalamus/physiology , Hypothermia/physiopathology , Immunohistochemistry , Injections, Intraventricular , Mesocricetus , Oligopeptides/metabolism , Polymerase Chain Reaction , Proprotein Convertase 1/biosynthesis , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Opioid, mu/physiology
15.
Brain Res ; 1432: 74-83, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22137657

ABSTRACT

G-protein receptor (GPR) 40 is known to be activated by docosahexaenoic acid (DHA). However, reports studying the role and functions (including pain regulation) of GPR40 in the brain are lacking. We investigated the involvement of GPR40 in the brain on DHA-induced antinociceptive effects. Expression of GPR40 protein was observed in the olfactory bulb, striatum, hippocampus, midbrain, hypothalamus, medulla oblongata, cerebellum and cerebral cortex in the brain as well as the spinal cord, whereas GPR120 protein expression in these areas was not observed. Intracerebroventricular (i.c.v.), but not intrathecal (i.t.) injection of DHA (25 and 50µg/mouse) and GW9508 (a GPR40- and GPR120-selective agonist; 0.1 and 1.0µg/mouse) significantly reduced formalin-induced pain behavior. These effects were inhibited by pretreatment with the µ opioid receptor antagonist ß-funaltrexamine (ß-FNA), naltrindole (δ opioid receptor antagonist) and anti-ß-endorphin antiserum. The κ opioid receptor antagonist norbinaltorphimine (nor-BNI) did not affect the antinociception of DHA or GW9508. Furthermore, the immunoreactivity of ß-endorphin in the hypothalamus increased at 10 and 20min after i.c.v. injection of DHA and GW9508. These findings suggest that DHA-induced antinociception via ß-endorphin release may be mediated (at least in part) through GPR40 signaling in the supraspinal area, and may provide valuable information on a novel therapeutic approach for pain control.


Subject(s)
Analgesics/pharmacology , Brain/physiology , Docosahexaenoic Acids/physiology , Pain Management/methods , Receptors, G-Protein-Coupled/physiology , Animals , Brain/metabolism , Drug Synergism , Hypothalamus/drug effects , Hypothalamus/metabolism , Injections, Intraventricular/methods , Male , Methylamines/pharmacology , Mice , Narcotic Antagonists/pharmacology , Pain Measurement , Propionates/pharmacology , Receptors, Opioid/drug effects , Receptors, Opioid/physiology , beta-Endorphin/physiology
16.
Brain Res ; 1422: 13-9, 2011 Nov 08.
Article in English | MEDLINE | ID: mdl-21983207

ABSTRACT

BACKGROUND: Endogenous ß-endorphin (ß-EP) in the central nervous system (CNS) is decreased upon opioid addiction. The current study examined whether exogenous ß-EP, delivered using an adenoviral vector into the CNS could attenuate morphine withdrawal syndrome in rats. METHODS: The model of opioid-dependent rats was set up by receiving subcutaneous injection of morphine using an escalating regimen for 6days (5, 10, 20, 40, 50, 60mg/kg, three times/day). The adenovirus mediated ß-EP gene was constructed based on our previous work. The ilea of opioid-dependent rats were isolated and treated with the supernatant of Ad-NEP. The basic and naloxone-induced (4µm/l) contractions of dependent ilea were recorded. The Ad-NEP was injected into the left lateral ventricle of the addition rats. The expression of the ß-EP gene was verified by radioimmunoassay of the cerebrospinal fluid (CSF) and immunocytochemistry for ß-EP. Withdrawal syndrome was evaluated after intraperitoneal injection of naloxone. RESULTS: The contractions of dependent ilea were attenuated with supernatant containing ß-EP expressed by Ad-NEP. Injection of the Ad-NEP resulted in significant increases in ß-EP level in the CSF and ß-EP-positive neurons. Rats receiving adenovirus carrying the ß-EP gene had significantly less severe withdrawal symptoms upon naloxone challenge. CONCLUSIONS: Exogenous ß-EP mediated by adenovirus could attenuate withdrawal syndrome in morphine-dependent rats.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/pharmacology , Morphine Dependence/therapy , Substance Withdrawal Syndrome/therapy , beta-Endorphin/genetics , beta-Endorphin/physiology , Acute Disease , Adenoviridae/genetics , Animals , Disease Models, Animal , Genetic Vectors/genetics , HEK293 Cells , Humans , Male , Morphine/pharmacology , Morphine Dependence/genetics , Morphine Dependence/physiopathology , Rats , Rats, Sprague-Dawley , Substance Withdrawal Syndrome/genetics , Substance Withdrawal Syndrome/physiopathology , beta-Endorphin/antagonists & inhibitors
17.
COPD ; 8(3): 160-6, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21513438

ABSTRACT

BACKGROUND: Endogenous opioids are naturally occurring peptides released by the brain in response to noxious stimuli. Although these naturally occurring peptides modulate pain, it is unknown whether endogenous opioids affect the perception of breathlessness associated with a specific respiratory challenge. The hypothesis is that intravenous administration of naloxone, used to block opioid signaling and inhibit neural pathways, will increase ratings of breathlessness during resistive load breathing (RLB) in patients with chronic obstructive pulmonary disease (COPD). METHODS: Fourteen patients with COPD (age, 64 ± 9 years) inspired through resistances during practice sessions to identify an individualized target load that caused ratings of intensity and/or unpleasantness of breathlessness ≥ 50 mm on a 100 mm visual analog scale. At two intervention visits, serum beta-endorphins were measured, naloxone (10 mg/25 ml) or normal saline (25 ml) was administered intravenously, and patients rated the two dimensions of breathlessness each minute during RLB. RESULTS: Patient ratings of intensity (p = 0.0004) and unpleasantness (p = 0.024) of breathlessness were higher with naloxone compared with normal saline. Eleven patients (79%) reported that it was easier to breathe during RLB with normal saline (p = 0.025). RLB led to significant increases in serum beta-endorphin immunoreactivity and decreases in inspiratory capacity. There were no significant differences in physiological responses between interventions. CONCLUSIONS: Endogenous opioids modulate the intensity and the unpleasantness of breathlessness in patients with COPD. Differences in breathlessness ratings between interventions were clinically relevant based on the patients' global assessment.


Subject(s)
Airway Resistance/drug effects , Dyspnea/drug therapy , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Pulmonary Disease, Chronic Obstructive/physiopathology , Respiration/drug effects , beta-Endorphin/physiology , Aged , Cross-Over Studies , Double-Blind Method , Female , Humans , Inspiratory Capacity/drug effects , Male , Middle Aged , Naloxone/therapeutic use , Narcotic Antagonists/therapeutic use , Pulmonary Disease, Chronic Obstructive/blood , Pulmonary Disease, Chronic Obstructive/drug therapy , Random Allocation , beta-Endorphin/blood
18.
Regul Pept ; 167(2-3): 222-6, 2011 Apr 11.
Article in English | MEDLINE | ID: mdl-21324347

ABSTRACT

In the present study, we investigated the degree to which ß-endorphin plays a role in the alpha 2-adrenergic/imidazoline receptor agonist attenuation of salt appetite. In order to evaluate whether the inhibitory action of clonidine (an α2-adrenergic/imidazoline receptor agonist) on induced sodium intake is mediated by the ß-endorphinergic system, we used a ß-endorphin deficient mouse line. ß-endorphin knockout (ßend(-/-)), heterozygous (ßend(+/-)) and wild-type (ßend(+/+)) mice were submitted to acute sodium depletion by a combined treatment of furosemide and low sodium diet and, 20h later, were administered with clonidine (0.5mg/kg). An hour later, the animals were subjected to a two-bottle choice test (water/2% NaCl). The results indicate that clonidine administration during the first stage of the test exerts an equivalent inhibition on sodium intake regardless of the genotype; however, in the final stage of the test, a reversal of the inhibitory response on induced sodium appetite becomes evident in the mice lacking ß-endorphin. Moreover no differences in dipsogenic response were observed between the genotypes. Considering these results and the fact that plasma half-life of clonidine at the dose administered is approximately 3h, it is possible to speculate that the inhibitory effect of clonidine on sodium appetite may be independent of ß-endorphin modulation during the first stage; however, the long-lasting inhibitory effect of clonidine may be mediated by the ß-endorphinergic system. This evidence supports the existence of adrenergic and ß-endorphinergic system interaction in the osmoregulatory response to achieve sodium balance.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Appetite/drug effects , Clonidine/pharmacology , Imidazoline Receptors/agonists , Sodium Chloride, Dietary/pharmacology , beta-Endorphin/physiology , Animals , Clonidine/administration & dosage , Drinking/physiology , Furosemide/pharmacology , Mice , Mice, Inbred Strains , Receptors, Adrenergic, alpha-2/metabolism
19.
Ir J Med Sci ; 180(2): 319-25, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21076975

ABSTRACT

INTRODUCTION: Depression is a very prevalent mental disorder affecting 340 million people globally and is projected to become the leading cause of disability and the second leading contributor to the global burden of disease by the year 2020. AIM: In this paper, we review the evidence published to date in order to determine whether exercise and physical activity can be used as therapeutic means for acute and chronic depression. Topics covered include the definition, classification criteria and treatment of depression, the link between ß-endorphin and exercise, the efficacy of exercise and physical activity as treatments for depression, properties of exercise stimuli used in intervention programs, as well as the efficacy of exercise and physical activity for treating depression in diseased individuals. CONCLUSIONS: The presented evidence suggests that exercise and physical activity have beneficial effects on depression symptoms that are comparable to those of antidepressant treatments.


Subject(s)
Depression/therapy , Exercise/psychology , Motor Activity/physiology , beta-Endorphin/physiology , Depression/psychology , Exercise/physiology , Humans
20.
Eur J Pharmacol ; 650(2-3): 563-7, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21044625

ABSTRACT

Stress is known to elicit pain relief, a phenomenon referred to as stress-induced analgesia. Based on stress parameters, opioid and non-opioid intrinsic pain inhibitory systems can be activated. In the present study, we assessed whether changing the duration of stress would affect the involvement of endogenous opioids in antinociception elicited by swim in warm water (32 °C), known to be opioid-mediated. Using mice lacking beta-endorphin, enkephalins or dynorphins and their respective wild-type littermates, we assessed the role of each opioid peptide in antinociception induced by a short (3 min) vs. long (15 min) swim. Mice were tested for baseline hot plate latency, exposed to swim (3 or 15 min) in warm water (32 °C) and then tested for antinociception at 5, 15 and 30 min. Our results revealed that both swim paradigms induced significant antinociception in wild-type mice. However, the short swim failed to induce antinociception in beta-endorphin-deficient mice, illustrating that beta-endorphin is important in this form of stress-induced antinociception. On the other hand, antinociception elicited by the long swim was only slightly reduced in beta-endorphin-deficient mice despite pretreatment with naloxone, a non-selective opioid receptor antagonist, significantly attenuated the antinociception elicited by the long swim. Nevertheless, a delayed hyperalgesic response developed in mice lacking beta-endorphin following exposure to either swim paradigm. On the other hand, mice lacking enkephalins or dynorphins and their respective wild-type littermates expressed a comparable antinociceptive response and did not exhibit the delayed hyperalgesic response. Together, our results suggest that the endogenous opioid peptide beta-endorphin not only mediates antinociception induced by the short swim but also prevents the delayed hyperalgesic response elicited by either swim paradigm.


Subject(s)
Analgesia , Analgesics, Opioid/pharmacology , Opioid Peptides/physiology , Stress, Psychological/metabolism , Animals , Dynorphins/genetics , Dynorphins/physiology , Enkephalins/genetics , Enkephalins/physiology , Female , Hot Temperature , Mice , Mice, Inbred C57BL , Mice, Knockout , Naloxone/pharmacology , Nociceptors/physiology , Opioid Peptides/genetics , Pain Measurement , Stress, Psychological/physiopathology , Swimming , Time Factors , beta-Endorphin/genetics , beta-Endorphin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...