Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Brain Behav Immun ; 73: 504-519, 2018 10.
Article in English | MEDLINE | ID: mdl-29928964

ABSTRACT

Interleukin 10 (IL-10) is antinociceptive in various animal models of pain without induction of tolerance, and its mechanism of action was generally believed to be mediated by inhibition of neuroinflammation. Here we reported that intrathecal IL-10 injection dose dependently attenuated mechanical allodynia and thermal hyperalgesiain male and female neuropathic rats, with ED50 values of 40.8 ng and 24 ng, and Emax values of 61.5% MPE and 100% MPE in male rats. Treatment with IL-10 specifically increased expression of the ß-endorphin (but not prodynorphin) gene and protein in primary cultures of spinal microglia but not in astrocytes or neurons. Intrathecal injection of IL-10 stimulated ß-endorphin expression from microglia but not neurons or astrocytes in both contralateral and ipsilateral spinal cords of neuropathic rats. However, intrathecal injection of the ß-endorphin neutralizing antibody, opioid receptor antagonist naloxone, or µ-opioid receptor antagonist CTAP completely blocked spinal IL-10-induced mechanical antiallodynia, while the microglial inhibitor minocycline and specific microglia depletor reversed spinal IL-10-induced ß-endorphin overexpression and mechanical antiallodynia. IL-10 treatment increased spinal microglial STAT3 phosphorylation, and the STAT3 inhibitor NSC74859 completely reversed IL-10-increased spinal expression of ß-endorphin and neuroinflammatory cytokines and mechanical antiallodynia. Silence of the Bcl3 and Socs3 genes nearly fully reversed IL-10-induced suppression of neuroinflammatory cytokines (but not expression of ß-endorphin), although it had no effect on mechanical allodynia. In contrast, disruption of the POMC gene completely blocked IL-10-stimulated ß-endorphin expression and mechanical antiallodynia, but had no effect on IL-10 inhibited expression of neuroinflammatory cytokines. Thus this study revealed that IL-10 produced antinociception through spinal microglial ß-endorphin expression, but not inhibition of neuroinflammation.


Subject(s)
Hyperalgesia/drug therapy , Interleukin-10/pharmacology , beta-Endorphin/metabolism , Analgesics/pharmacology , Animals , Astrocytes , Cytokines/metabolism , Female , Hyperalgesia/metabolism , Injections, Spinal , Interleukin-10/metabolism , Male , Microglia/drug effects , Microglia/metabolism , Microglia/physiology , Minocycline/pharmacology , Naloxone/pharmacology , Neuralgia/metabolism , Neurons , Primary Cell Culture , Rats , Rats, Wistar , Spinal Cord/drug effects , Spinal Cord/metabolism , Spine/drug effects , Spine/metabolism , beta-Endorphin/drug effects
2.
Brain Res ; 1663: 1-8, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28284896

ABSTRACT

With the decline of ovarian steroids levels at menopause, many women experience an increase in anxiety and stress sensitivity. The locus coeruleus (LC), a central source of noradrenaline (NE), is activated by stress and is inhibited by ß-endorphin. Moreover, increased NE has been implicated in pathological anxiety syndromes. Hormone replacement therapy (HRT) in menopause appears to decrease anxiety and vulnerability to stress. Therefore, we questioned the effect of HRT on the inhibitory ß-endorphin innervation of the LC. In addition, we found that progesterone protects serotoninergic neurons in monkeys, leading us to question whether ovarian steroids are also neuroprotective in LC neurons in monkeys. Adult Rhesus monkeys (Macaca mulatta) were ovariectomized, and either treated with Silastic capsules that contained estradiol, estradiol+progesterone, progesterone alone or that were empty (ovariectomized; control). After 1month, the LC was obtained and processed for immunohistochemistry for ß-endorphin and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling (TUNEL). The density of ß-endorphin axons was determined with image analysis using ImageJ. The TUNEL-positive neurons were counted in the entire LC. Progesterone-alone significantly increased the density of the ß-endorphin axons in the LC (p<0.01). No significant differences between groups in the number of TUNEL-positive cells in the LC were found. In conclusion, we found that HRT increases the inhibitory influence of ß-endorphin in the LC, which could, in turn, contribute to reduce anxiety and increase stress resilience. In addition, we did not find compelling evidence of neurodegeneration or neuroprotection by HRT in the LC of Rhesus monkeys.


Subject(s)
Locus Coeruleus/drug effects , beta-Endorphin/drug effects , Adrenergic Neurons/drug effects , Adrenergic Neurons/physiology , Animals , Estradiol/pharmacology , Estrogens/pharmacology , Female , Haplorhini , Hormone Replacement Therapy , In Situ Nick-End Labeling , Locus Coeruleus/physiology , Macaca mulatta/physiology , Menopause/drug effects , Models, Animal , Neurons/drug effects , Norepinephrine/metabolism , Norepinephrine/pharmacology , Ovariectomy , Ovary , Progesterone/metabolism , Progesterone/pharmacology , Steroids , beta-Endorphin/metabolism
3.
Psychopharmacology (Berl) ; 218(1): 229-39, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21597991

ABSTRACT

RATIONALE: The endogenous opioid and corticotropin-releasing hormone (CRH) systems, present in the central amygdala (CeA), are implicated in alcohol consumption. OBJECTIVES: The purpose of this study is to investigate the hypothesis that, in CeA, alcohol stimulates CRH release, which then stimulates ß-endorphin release. MATERIALS AND METHODS: Rats were unilaterally implanted with a guide cannula to aim microdialysis probes in CeA. Experiment 1: rats received an intraperitoneal (IP) injection of various ethanol doses (0.0, 2.0, 2.4, or 2.8 g ethanol/kg body weight) and microdialysates were sampled at 30-min intervals to determine the effects over time of acute alcohol on the extracellular CRH concentrations in CeA. Experiment 2: phosphate-buffered saline, CRH, or CRH receptor (CRHR) antagonists (antalarmin or anti-sauvagine-30) was microinjected into CeA followed by a saline or 2.8 g/kg ethanol IP injection to determine the effects of CRHR activation or blockade in CeA on the basal and alcohol-stimulated release of ß-endorphin. CRH and ß-endorphin dialysate contents were determined using specific radioimmunoassays. RESULTS: Acute alcohol induced a delayed increase in the extracellular CRH levels in CeA. Behavioural data showed no difference in locomotion between alcohol- and saline-treated rats. However, a transient increase in grooming was observed which did not correspond with alcohol-induced changes in CRH. Local CRH microinjections increased the extracellular ß-endorphin concentrations in CeA. CRHR1 and CRHR2 blockade with microinjections of antalarmin and anti-sauvagine-30, respectively, attenuated the alcohol-induced increase of extracellular ß-endorphin in CeA. CONCLUSIONS: Acute alcohol exerts indirect actions on CRH release and induced interactions of the CRH and ß-endorphin systems in CeA.


Subject(s)
Amygdala/drug effects , Corticotropin-Releasing Hormone/drug effects , Ethanol/pharmacology , beta-Endorphin/drug effects , Amygdala/metabolism , Animals , Behavior, Animal/drug effects , Corticotropin-Releasing Hormone/administration & dosage , Corticotropin-Releasing Hormone/metabolism , Dose-Response Relationship, Drug , Ethanol/administration & dosage , Injections, Intraperitoneal , Male , Microdialysis , Microinjections , Peptide Fragments/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/drug effects , Receptors, Corticotropin-Releasing Hormone/metabolism , beta-Endorphin/metabolism
4.
Pain ; 149(2): 254-262, 2010 May.
Article in English | MEDLINE | ID: mdl-20206445

ABSTRACT

Endothelin-1 (ET-1) produced by various cancers is known to be responsible for inducing pain. While ET-1 binding to ETAR on peripheral nerves clearly mediates nociception, effects from binding to ETBR are less clear. The present study assessed the effects of ETBR activation and the role of endogenous opioid analgesia in carcinoma pain using an orthotopic cancer pain mouse model. mRNA expression analysis showed that ET-1 was nearly doubled while ETBR was significantly down-regulated in a human oral SCC cell line compared to normal oral keratinocytes (NOK). Squamous cell carcinoma (SCC) cell culture treated with an ETBR agonist (10(-4)M, 10(-5)M, and 10(-6) M BQ-3020) significantly increased the production of beta-endorphin without any effects on leu-enkephalin or dynorphin. Cancer inoculated in the hind paw of athymic mice with SCC induced significant pain, as indicated by reduction of paw withdrawal thresholds in response to mechanical stimulation, compared to sham-injected and NOK-injected groups. Intratumor administration of 3mg/kg BQ-3020 attenuated cancer pain by approximately 50% up to 3h post-injection compared to PBS-vehicle and contralateral injection, while intratumor ETBR antagonist BQ-788 treatment (100 and 300microg/kg and 3mg/kg) had no effects. Local naloxone methiodide (500microg/kg) or selective mu-opioid receptor antagonist (CTOP, 500microg/kg) injection reversed ETBR agonist-induced antinociception in cancer animals. We propose that these results demonstrate that peripheral ETBR agonism attenuates carcinoma pain by modulating beta-endorphins released from the SCC to act on peripheral opioid receptors found in the cancer microenvironment.


Subject(s)
Endothelin-1/metabolism , Nociceptors/metabolism , Opioid Peptides/metabolism , Pain/metabolism , Receptor, Endothelin B/metabolism , Sensory Receptor Cells/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Down-Regulation/genetics , Endothelin-1/genetics , Endothelins/pharmacology , Humans , Mice , Mice, Nude , Narcotic Antagonists/pharmacology , Neoplasm Transplantation/methods , Neoplasms/complications , Neoplasms/metabolism , Neoplasms/physiopathology , Nociceptors/drug effects , Oligopeptides/pharmacology , Opioid Peptides/drug effects , Pain/drug therapy , Pain/etiology , Peptide Fragments/pharmacology , Piperidines/pharmacology , Receptor, Endothelin B/agonists , Receptors, Opioid/drug effects , Receptors, Opioid/metabolism , Sensory Receptor Cells/drug effects , Up-Regulation/genetics , beta-Endorphin/drug effects , beta-Endorphin/metabolism
5.
Am J Manag Care ; 16 Suppl Management: S48-54, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20297877

ABSTRACT

The major clinical guidelines recommend the use of acetaminophen (acetyl-para-aminophenol [APAP]) for the treatment of mild-to-moderate symptoms of osteoarthritis (OA) and only recommend the use of nonsteroidal anti-inflammatory drugs (NSAIDs) after APAP failure. This recommendation is based on the efficacy of APAP in treating OA and its relatively benign side-effect profile compared with NSAIDs. NSAIDs are associated with a high risk of adverse events, particularly those of the gastrointestinal (GI) tract. A large number of studies in OA have compared APAP with a variety of selective and nonselective NSAIDs and typically found greater efficacy with NSAIDs. This advantage, however, is mainly the result of increased efficacy in patients with more severe disease, and is viewed as a relatively small analgesic advantage in some studies and meta-analyses. Many of these same studies have reported little or no difference in safety between APAP and NSAIDs, but these results are typically based on short-term studies. Results from meta-analyses on the safety of NSAIDs almost unanimously confirm elevated risk of GI complications. The analgesic mechanism of APAP is still not well understood. However, the notion that APAP has no anti-inflammatory effect has been challenged in recent years with increasing data that suggest it may have an effect on inflammation distinct from that seen with NSAIDs. A variety of mechanistic hypotheses have been proposed.


Subject(s)
Acetaminophen/therapeutic use , Analgesics, Non-Narcotic/therapeutic use , Osteoarthritis/drug therapy , Acetaminophen/pharmacology , Analgesics, Non-Narcotic/pharmacology , Animals , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Humans , Nitric Oxide/metabolism , Substance P/drug effects , beta-Endorphin/drug effects
6.
Psychopharmacology (Berl) ; 208(1): 121-30, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19940981

ABSTRACT

RATIONALE: The selectively bred lines of alcohol-preferring alko alcohol (AA) and alcohol-avoiding alko nonalcohol (ANA) rats have been used to demonstrate differences in relevant neurotransmitters which could account for their difference in alcohol consumption. Studies have demonstrated differences in distinct components of the endogenous opioid system in various brain regions associated with the process of reinforcement between the AA and ANA lines of rats. OBJECTIVES: The goal of this current study was to investigate the hypotheses that the AA and ANA rats will show differences in the release of beta-endorphin at the level of nucleus accumbens (NAC) and in locomotor activity in response to acute systemic administration of ethanol. MATERIALS AND METHODS: AA and ANA rats were unilaterally implanted with a guide cannula to aim microdialysis probes at the level of NAC. Intraperitoneal injections of 0.0, 1.5, 2.0, and 2.5 g ethanol/kg body weight were administered. Dialysate samples were collected at 30-min intervals prior to and following the injection. Radioimmunoassay specific for beta-endorphin was used to determine the dialysate beta-endorphin content. RESULTS: The 2.5-g/kg ethanol dose induced a transient increase in extracellular beta-endorphin at the level of NAC of AA but not of ANA rats. The 2.5-g/kg ethanol dose also attenuated locomotor activity in the AA but not in the ANA rats. CONCLUSIONS: The lack of an increase in the beta-endorphin concentration in the NAC of ANA rats in response to ethanol may partially account for their lower alcohol consumption and lower alcohol-induced attenuation of locomotor activity compared to AA rats.


Subject(s)
Alcohol Drinking , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , beta-Endorphin/drug effects , Animals , Central Nervous System Depressants/administration & dosage , Dose-Response Relationship, Drug , Ethanol/administration & dosage , Injections, Intraperitoneal , Male , Microdialysis , Motor Activity/drug effects , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Radioimmunoassay , Rats , beta-Endorphin/metabolism
7.
Menopause ; 16(5): 1037-43, 2009.
Article in English | MEDLINE | ID: mdl-19295450

ABSTRACT

OBJECTIVE: The natural selective estrogen receptor modulator DT56a (Femarelle), derived from soybean, has been shown to relieve menopausal vasomotor symptoms with no effect on sex steroid hormone levels or endometrial thickness.The purpose of the present study was to evaluate the neuroendocrine effect of DT56a administration through the evaluation of brain content of allopregnanolone (AP), an endogenous neurosteroid gamma-aminobutyric acid agonist with anxiolytic properties, and through the assessment of beta-endorphin (beta-END), the endogenous opioid implicated in pain mechanism, emotional state, and autonomic control. METHODS: Five groups of Wistar ovariectomized (OVX) rats received one of the following treatments: oral DT56a administration at doses of 6, 12, 60, and 120 mg kg(-1) day(-1) or estradiol valerate (E2V) at a dose of 0.05 mg kg(-1) day(-1) for 14 days. One group of fertile and one group of OVX rats receiving placebo were used as controls. The concentration of AP was assessed in the frontal and parietal cortex, hippocampus, hypothalamus, anterior pituitary, and serum, whereas the content of beta-END was evaluated in the frontal and parietal cortex, hippocampus, hypothalamus, neurointermediate lobe, anterior pituitary, and plasma. RESULTS: DT56a increased AP levels in all brain areas analyzed and in serum, with a classical dose-related curve in comparison with OVX rats. In some brain areas, such as the frontal cortex, the parietal cortex, and the anterior pituitary, positive results were found even with the administration of a lower DT56a dose of 60 mg kg(-1) day(-1), attaining AP levels in the range of those in animals treated with E2V. Similarly, beta-END levels were enhanced in selected brain areas such as the hippocampus, the hypothalamus, the neurointermediate lobe, and the anterior pituitary in comparison with those in OVX rats, in which the increase of the opioid was dose related and in the range of those in rats treated with E2V. CONCLUSIONS: This study demonstrated that DT56a positively affects brain neurosteroidogenesis and the opiatergic system: DT56a exerts an estrogen-like effect on selective areas related to mood, cognition, and homeostasis control, presenting a specific pattern of interaction with the brain function. These findings may, in part, explain the clinical effect of DT56a on menopausal symptoms.


Subject(s)
Brain Chemistry/drug effects , Menopause , Ovariectomy , Plant Extracts/administration & dosage , Pregnanolone/analysis , beta-Endorphin/analysis , Administration, Oral , Analysis of Variance , Animals , Brain Chemistry/physiology , Cerebral Cortex/chemistry , Cerebral Cortex/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Replacement Therapy/methods , Female , Hot Flashes/drug therapy , Hot Flashes/etiology , Menopause/drug effects , Menopause/physiology , Neurosecretory Systems/chemistry , Neurosecretory Systems/drug effects , Phytotherapy/methods , Plant Extracts/pharmacology , Pregnanolone/physiology , Rats , Rats, Wistar , beta-Endorphin/drug effects , beta-Endorphin/physiology
8.
Am J Drug Alcohol Abuse ; 34(4): 355-81, 2008.
Article in English | MEDLINE | ID: mdl-18584566

ABSTRACT

AIMS: Psychiatric pharmacogenetics involves the use of genetic tests that can predict the effectiveness of treatments for individual patients with mental illness such as drug dependence. This review aims to cover these developments in the pharmacotherapy of alcohol and opiates, two addictive drugs for which we have the majority of our FDA approved pharmacotherapies. METHODS: We conducted a literature review using Medline searching terms related to these two drugs and their pharmacotherapies crossed with related genetic studies. RESULTS: Alcohol's physiological and subjective effects are associated with enhanced beta-endorphin release. Naltrexone increases baseline beta-endorphin release blocking further release by alcohol. Naltrexone's action as an alcohol pharmacotherapy is facilitated by a putative functional single nucleotide polymorphism (SNP) in the opioid mu receptor gene (Al18G) which alters receptor function. Patients with this SNP have significantly lower relapse rates to alcoholism when treated with naltrexone. Caucasians with various forms of the CYP2D6 enzyme results in a 'poor metabolizer' phenotype and appear to be protected from developing opioid dependence. Others with a "ultra-rapid metabolizer" phenotype do poorly on methadone maintenance and have frequent withdrawal symptoms. These patients can do well using buprenorphine because it is not significantly metabolized by CYP2D6. CONCLUSIONS: Pharmacogenetics has great potential for improving treatment outcome as we identify gene variants that affect pharmacodynamic and pharmacokinetic factors. These mutations guide pharmacotherapeutic agent choice for optimum treatment of alcohol and opiate abuse and subsequent relapse.


Subject(s)
Alcoholism/drug therapy , Alcoholism/genetics , Buprenorphine/pharmacology , Buprenorphine/therapeutic use , Naltrexone/pharmacology , Naltrexone/therapeutic use , Narcotic Antagonists/pharmacology , Narcotic Antagonists/therapeutic use , Opioid-Related Disorders/drug therapy , Opioid-Related Disorders/genetics , Alcoholism/diagnosis , Cytochrome P-450 CYP2D6/drug effects , Cytochrome P-450 CYP2D6/genetics , Humans , Opioid-Related Disorders/diagnosis , Polymorphism, Single Nucleotide/drug effects , Polymorphism, Single Nucleotide/genetics , Predictive Value of Tests , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/genetics , beta-Endorphin/blood , beta-Endorphin/drug effects
9.
Naunyn Schmiedebergs Arch Pharmacol ; 377(4-6): 529-40, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18080810

ABSTRACT

In the present study, we investigated the mechanism(s) for glucose-lowering action of andrographolide in streptozotocin-induced diabetic rats (STZ-diabetic rats). Andrographolide lowered plasma glucose concentrations in a dose-dependent manner and increased plasma beta-endorphin-like immunoreactivity (BER) dose-dependently in diabetic rats. Both of these responses to andrographolide were abolished by the pretreatment of animals with prazosin or N-(2-(2-cyclopropylmethoxy) ethyl) 5-choro-alpha-dimethyl-1H-indole-3-thylamine (RS17053) at doses sufficient to block alpha1-adrenoceptors (ARs). Also, andrographolide enhanced BER release from isolated rat adrenal medulla in a concentration-related manner that could be abolished by alpha1-ARs antagonists. Bilateral adrenalectomy in STZ-diabetic rats eliminated the activities of andrographolide, including the plasma glucose-lowering effect and the plasma BER-elevating effect. Andrographolide failed to lower plasma glucose in the presence of opioid micro-receptor antagonists and in the opioid micro-receptor knockout diabetic mice. Treatment of STZ-diabetic rats with andrographolide resulted in the reduced expression of phosphoenolpyruvate carboxykinase (PEPCK) in liver and an increased expression of the glucose transporter subtype 4 (GLUT 4) in soleus muscle. These effects were also blocked by opioid micro-receptor antagonists. In conclusion, our results suggest that andrographolide may activate alpha1-ARs to enhance the secretion of beta-endorphin which can stimulate the opioid micro-receptors to reduce hepatic gluconeogenesis and to enhance the glucose uptake in soleus muscle, resulting in a decrease of plasma glucose in STZ-diabetic rats. However, the roles of other endogenous opioid peptides or the mixture of several opioid peptides in the activation of opioid micro-receptors associated with the plasma glucose-lowering action of andrographolide, should be considered and need more investigation in the future.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Diterpenes/pharmacology , Hypoglycemic Agents/pharmacology , Animals , Blood Glucose/drug effects , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/physiopathology , Diterpenes/administration & dosage , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Glucose Transporter Type 4/genetics , Hypoglycemic Agents/administration & dosage , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Knockout , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Rats , Rats, Wistar , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Streptozocin , beta-Endorphin/drug effects , beta-Endorphin/metabolism
10.
J Clin Anesth ; 19(2): 120-4, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17379124

ABSTRACT

STUDY OBJECTIVE: To investigate the effect of sevoflurane as single anesthetic on melatonin and beta-endorphin plasma levels during the first 24 hours postoperatively. DESIGN: Prospective, open-cohort study. SETTING: University hospital. PATIENTS: 13 ASA physical status I and II, adults, scheduled for dilatation and curettage of the uterus, and 13 healthy volunteers. INTERVENTIONS: Patients received general anesthesia with sevoflurane. MEASUREMENTS: Melatonin and beta-endorphin plasma levels were determined before anesthesia, immediately after, and two, 4, 8, and 24 hours after the end of anesthesia. Melatonin and beta-endorphin were also measured in 13 healthy subjects (controls) not undergoing anesthesia at similar times during the day. Systolic and diastolic blood pressure, heart rate, bispectral index, and oxygen saturation via pulse oximeter (SpO(2)) were recorded before and after anesthesia. Quality of sleep postoperatively was also assessed. MAIN RESULTS: Melatonin levels (pg/mL) in the patients and controls were 8.2 +/- 7.9 versus 15.2 +/- 15.0 before anesthesia and 7.7 +/- 7.9 versus 11.1 +/- 7.0, 6.5 +/- 6.1 versus 15.6 +/- 16.3, and 19.5 +/- 17.9 versus 23.7 +/- 23.3 at the end of anesthesia and 4 and 24 hours after the end of anesthesia, respectively (P = 0.057). At the same time points, beta-endorphin plasma levels (pmol/L) in patients and controls were 5.2 +/- 2.0 versus 4.0 +/- 2.3, 5.4 +/- 3.3 versus 3.9 +/- 2.5, 4.9 +/- 1.2 versus 4.4 +/- 1.7, and 3.7 +/- 2.6 versus 4.2 +/- 1.8, respectively (P= 0.285). The quality of sleep assessed clinically was not altered. CONCLUSION: Sevoflurane as a single anesthetic for minor gynecological procedures did not influence significantly melatonin or beta-endorphin plasma levels. Sleep quality assessed clinically was not influenced.


Subject(s)
Anesthesia, General/methods , Anesthetics, Inhalation/pharmacology , Melatonin/blood , Methyl Ethers/pharmacology , beta-Endorphin/blood , beta-Endorphin/drug effects , Adult , Anesthetics, Inhalation/administration & dosage , Blood Pressure/drug effects , Cohort Studies , Dilatation and Curettage/methods , Electroencephalography/methods , Female , Heart Rate/drug effects , Humans , Methyl Ethers/administration & dosage , Middle Aged , Oximetry/methods , Oxygen/blood , Postoperative Period , Prospective Studies , Reference Values , Sevoflurane , Sleep/drug effects , Time Factors
11.
Clin Rheumatol ; 26(1): 44-9, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16575494

ABSTRACT

The purpose of this study was to evaluate the efficacy of calcitonin on beta-endorphin levels in female patients experiencing back pain associated with postmenopausal osteoporosis. The secondary purpose was to assess the pain and quality of life in these patients. There were 30 patients with a mean age of 58.2+/-5.4 years in the treatment group and 26 patients with a mean age of 58.8+/-5.2 years in the placebo group in this randomized, placebo-controlled study. The patients subcutaneously received 100 IU salmon calcitonin or placebo injections and 1,000 mg elementary calcium for 2 weeks. Baseline plasma beta-endorphin levels were measured and repeated after 2 weeks. Patients' pain and quality of life (QOL) were evaluated by using the Visual Analogue Scale, Modified Face Scale, Beck Depression Index, and Nottingham Health Profile. Patients' global assessment of disease activity was also performed at baseline and at the end of the first and second week. We found that plasma beta-endorphin levels in the treatment group were significantly higher than the placebo group at the end of the second week (p<0.001). Although pain and QOL scores were improved at the end of the second week in both groups (p<0.05), the improvement in the treatment group was more significant when compared with the placebo group (p<0.05). Therefore, calcitonin is an analgesic agent, as it increases the plasma beta-endorphin levels in patients with postmenopausal osteoporosis, which consequently improves QOL.


Subject(s)
Back Pain/drug therapy , Bone Density Conservation Agents/administration & dosage , Calcitonin/administration & dosage , Osteoporosis, Postmenopausal/complications , beta-Endorphin/blood , Aged , Back Pain/etiology , Bone Density/drug effects , Female , Humans , Injections, Subcutaneous , Middle Aged , Placebos , Quality of Life , Single-Blind Method , beta-Endorphin/drug effects
12.
Brain Behav Immun ; 21(5): 544-52, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17174527

ABSTRACT

Endogenous opioids released from leukocytes extravasating into injured tissue can interact with peripheral opioid receptors to inhibit nociception. Animal studies have shown that the homing of opioid-producing leukocytes to the injured site is modulated by spinal blockade of noxious input. This study investigated whether epidural analgesia (EDA) influences the migration of beta-endorphin (END) and/or met-enkephalin (ENK)-containing leukocytes into the subcutaneous wound tissue of patients undergoing abdominal surgery. In part I patients received general anesthesia combined either with intra- and postoperative EDA (with bupivacaine and fentanyl) or with postoperative patient controlled intravenous analgesia (PCIA; with the opioid piritramide). In part II patients received general anesthesia combined with either epidural fentanyl or bupivacaine which was continued postoperatively. Samples of cutanous and subcutanous tissue were taken from the wound site at the beginning, at the end and at various times after surgery, and were examined by immunohistochemistry for the presence of END and ENK. We found that (i) epidural bupivacaine, fentanyl and PCIA provided similar and clinically acceptable postoperative pain relief; (ii) compared to PCIA, epidural bupivacaine or fentanyl did not change the gross inflammatory reaction within the surgical wound; (iii) opioid-containing leukocytes were almost absent in normal subcutaneous tissue but migrated to the inflamed wound tissue in ascending numbers within a few hours, reaching a peak at about 24 h after surgery; (iv) compared to PCIA, EDA resulted in significantly decreased homing of END-containing leukocytes to the injured site at 24 h after surgery; and (v) the magnitude of this decrease was similar regardless of the epidural medication. These findings suggest that nociceptive but not sympathetic neurons are primarily involved in the attraction of opioid-containing leukocytes during early stages of inflammation.


Subject(s)
Analgesics, Opioid/immunology , Cell Movement/drug effects , Enkephalin, Methionine/metabolism , Leukocytes/drug effects , Wound Healing/immunology , beta-Endorphin/metabolism , Adjuvants, Anesthesia/immunology , Adjuvants, Anesthesia/pharmacology , Aged , Analgesia, Patient-Controlled , Analgesics, Opioid/therapeutic use , Anesthesia, Epidural , Anesthetics, Local/immunology , Anesthetics, Local/therapeutic use , Bupivacaine/immunology , Bupivacaine/therapeutic use , Cell Movement/immunology , Enkephalin, Methionine/drug effects , Enkephalin, Methionine/immunology , Female , Fentanyl/immunology , Fentanyl/therapeutic use , Humans , Leukocytes/immunology , Leukocytes/metabolism , Longitudinal Studies , Male , Middle Aged , Nociceptors/drug effects , Nociceptors/immunology , Pain, Postoperative/immunology , Pain, Postoperative/prevention & control , Pirinitramide/therapeutic use , Subcutaneous Tissue/immunology , Sympathetic Fibers, Postganglionic/drug effects , Sympathetic Fibers, Postganglionic/immunology , Wound Healing/drug effects , beta-Endorphin/drug effects , beta-Endorphin/immunology
13.
J Invest Dermatol ; 127(2): 411-20, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16946714

ABSTRACT

The human skin holds the capacity for autocrine processing of the proopiomelanocortin (POMC)-derived peptides. Recent data demonstrated the presence and functionality of ACTH, alpha- and beta-melanocyte-stimulating hormone (MSH), and beta-endorphin in the regulation of skin pigmentation, and a role has been put forward for alpha-MSH as an effective antioxidant. In patients with vitiligo, decreased epidermal POMC processing and low alpha-MSH levels were documented previously. These patients accumulate hydrogen peroxide (H2O2) in the 10(-3) M range in their epidermis. Therefore, we examined the involvement of H2O2 on POMC-derived peptides as possible targets for oxidation by this reactive oxygen species. To address this, we employed immunofluorescence labelling, dot blot analysis, Fourier transform Raman spectroscopy, functionality studies, and computer simulation of the peptide structures. We demonstrate H2O2-mediated oxidation of epidermal ACTH, alpha-MSH, and beta-endorphin in vitiligo owing to oxidation of methionine residues in the sequences of these peptides. Moreover, we show that oxidized beta-endorphin loses its function in the promotion of pigmentation in melanocytes. These changes are reversible upon the reduction of H2O2 levels by a pseudocatalase PC-KUS. Moreover, oxidation of alpha-MSH can be prevented by the formation of a 1:1 complex with the abundant cofactor (6R)-L-erythro-5,6,7,8-tetrahydrobiopterin. Thus, using vitiligo, we demonstrate that H2O2 can affect pigmentation via epidermal POMC peptide redox homeostasis.


Subject(s)
Epidermis/metabolism , Hydrogen Peroxide/metabolism , Oxidative Stress , Pro-Opiomelanocortin/metabolism , Vitiligo/metabolism , Adrenocorticotropic Hormone/chemistry , Adrenocorticotropic Hormone/drug effects , Biopterins/analogs & derivatives , Biopterins/metabolism , Catalase/pharmacology , Cells, Cultured , Computer Simulation , Epidermis/drug effects , Fourier Analysis , Humans , Hydrogen Peroxide/antagonists & inhibitors , Hydrogen Peroxide/pharmacology , Melanins/biosynthesis , Models, Biological , Oxidation-Reduction , Peptide Fragments/metabolism , Skin Pigmentation , Spectrum Analysis, Raman , Vitiligo/physiopathology , alpha-MSH/chemistry , alpha-MSH/drug effects , alpha-MSH/metabolism , beta-Endorphin/chemistry , beta-Endorphin/drug effects , beta-Endorphin/metabolism
14.
Planta Med ; 72(1): 9-13, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16450289

ABSTRACT

We investigated the plasma glucose-lowering mechanism(s) of Rh2, a ginsenoside derived from Panax ginseng, in rats with streptozotocin-induced diabetes (STZ-diabetic rats). After intravenous injection over 120 min into fasting STZ-diabetic rats, Rh2 decreased plasma glucose in a dose-dependent manner. In parallel to the lowering of plasma glucose, an increase of plasma beta-endorphin-like immunoreactivity was observed. In addition, naloxone and naloxonazine at doses sufficient to block opioid mu-receptors inhibited the plasma glucose-lowering action of Rh2 in genetically wild-type, diabetic mice. In contrast, Rh2 failed to lower plasma glucose in opioid mu-receptor knockout diabetic mice. An increase in gene expression at both the mRNA and protein levels of glucose transporter subtype 4 (GLUT 4) was observed in soleus muscle obtained from STZ-diabetic rats treated with Rh2 three times daily for one day; this increase in expression was absent when opioid mu-receptors were blocked. In conclusion, our results suggest that ginsenoside Rh2 may lower plasma glucose in STZ-diabetic rats based on an increase in beta-endorphin secretion that activates opioid mu-receptors thereby resulting in an increased expression of GLUT 4.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Experimental/drug therapy , Ginsenosides/pharmacology , beta-Endorphin/drug effects , Animals , Diabetes Mellitus, Experimental/metabolism , Ginsenosides/therapeutic use , Glucose Transporter Type 4/biosynthesis , Male , Mice , Panax , Phytotherapy , Rats , Rats, Wistar , Receptors, Opioid, mu/metabolism , Streptozocin
16.
Neurosci Lett ; 388(1): 45-8, 2005 Nov 04.
Article in English | MEDLINE | ID: mdl-16009491

ABSTRACT

The aim of this study was to evaluate the beta-endorphin (beta-endorphin) plasma level in Warsaw Low Preferring (WLP) and Warsaw high-preferring (WHP) rats after repeated administration of acamprosate, one of most effective drug in the treatment of alcoholism. Treatment with acamprosate in dose 200mg/kg, p.o. for 10 days induced an increase in plasma beta-endorphin levels. A single injection of ethanol also results in the increase of beta-endorphin level. Moreover, it was found that single injection of ethanol to WHP rats resulted in lower increase of plasma beta-endorphin content in rats earlier treated with acamprosate. In WLP rats, repeated acamprosate treatment prevents the ethanol-induced increase in plasma beta-endorphin level. It may be concluded that acamprosate modulates the endogenous opioid system.


Subject(s)
Alcohol-Induced Disorders, Nervous System/drug therapy , Alcoholism/drug therapy , Brain/drug effects , Taurine/analogs & derivatives , beta-Endorphin/blood , beta-Endorphin/drug effects , Acamprosate , Alcohol Deterrents/administration & dosage , Alcohol Deterrents/therapeutic use , Alcohol-Induced Disorders, Nervous System/blood , Alcohol-Induced Disorders, Nervous System/physiopathology , Alcoholism/blood , Alcoholism/physiopathology , Animals , Brain/metabolism , Brain/physiopathology , Disease Models, Animal , Drug Administration Schedule , Drug Interactions/physiology , Ethanol/antagonists & inhibitors , Female , Genetic Predisposition to Disease/genetics , Opioid Peptides/drug effects , Opioid Peptides/metabolism , Rats , Taurine/administration & dosage , Taurine/therapeutic use , Treatment Outcome , Up-Regulation/drug effects , Up-Regulation/physiology
17.
Intensive Care Med ; 31(3): 380-7, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15714323

ABSTRACT

PURPOSE: Patients in the ICU after long-term administration of an opioid/hypnotic often develop delirium. To assess the nature of this phenomenon, patients in a surgical ICU following ventilatory support and sedation with an opioid/hypnotic/sedative were studied. METHODOLOGY: Following sufentanil/midazolam (group 1; n =14) or sufentanil/propofol (group 2; n =15) sedation, patients were evaluated for changes in mean arterial blood pressure and heart rate, the activity of the central nervous system (sensory evoked potentials, spectral edge frequency of EEG), and the endogenous opioids plasma concentrations (beta-endorphin, met-enkephalin). Data obtained were correlated with the individual intensities of withdrawal symptoms 6-, 12-, and 24 h following sedation. RESULTS: Following a mean duration of ventilation of 7.7 days (+/-3.6 SD) in groups 1 and 3.5 (+/-1.7 SD) in group 2, withdrawal intensities peaked within the 6th hour after cessation. Plasma beta-endorphin and met-enkephalin levels were low during sedation, and only the sufentanil/midazolam group demonstrated a postinhibitory overshoot. Withdrawal symptom intensities demonstrated an inverse correlation with beta-endorphin and met-enkephalin levels, a direct linear correlation with amplitude height of the evoked potential, and blood pressure and heart rate changes. Withdrawal intensities did not correlate with EEG power spectral edge frequency. CONCLUSION: The endorphinergic system is suppressed when a potent exogenous opioid like sufentanil is given over a long period of time. Following sedation, abstinence symptoms seem to be related to postinhibitory increased endorphin synthesis. This is mostly seen in the combination of sufentanil/midazolam. In addition, an increase in the amplitude of the sensory-evoked potential suggests a postinhibitory excitatory state within the nociceptive system.


Subject(s)
Conscious Sedation/adverse effects , Delirium/chemically induced , Hypnotics and Sedatives/adverse effects , Midazolam/adverse effects , Propofol/adverse effects , Substance Withdrawal Syndrome , Sufentanil/adverse effects , Adolescent , Adult , Aged , Analgesics, Opioid/adverse effects , Blood Pressure/drug effects , Critical Care/methods , Delirium/blood , Delirium/diagnosis , Dose-Response Relationship, Drug , Drug Therapy, Combination , Electroencephalography , Enkephalin, Methionine/blood , Enkephalin, Methionine/drug effects , Evoked Potentials, Somatosensory , Female , Heart Rate/drug effects , Humans , Male , Middle Aged , Postoperative Care/adverse effects , beta-Endorphin/blood , beta-Endorphin/drug effects
18.
Neuroendocrinology ; 82(3-4): 185-97, 2005.
Article in English | MEDLINE | ID: mdl-16534240

ABSTRACT

In the present study the effects of intravenously administered corticotropin-releasing hormone (CRH) on the release of proopiomelanocortin (POMC) derivatives such as adrenocorticotropic hormone (ACTH), beta-lipotropin (beta-LPH) and beta-endorphin (beta-END) as well as direct effects of CRH on pain sensitivity were examined. In 16 healthy volunteers we studied the effects of 100 microg intravenously administered CRH in absence or presence of 12 mg naloxone on heat or pressure pain sensitivity, using a double-blind, cross-over and placebo-controlled design. To evaluate analgesic effects of CRH via release of POMC derivatives, we determined plasma concentrations of beta-END-immunoreactive material (IRM), authentic beta-END (beta-END(1-31)) and beta-LPH IRM, in parallel with heat and pressure pain tolerance thresholds before and 15 and 30 min after treatment with CRH (or placebo), and 5 min after naloxone (or placebo) administration which was administered 40 min after CRH (or placebo) injection. CRH increased levels of beta-END IRM, beta-END(1-31) and beta-LPH IRM. As compared to beta-END IRM levels measured by a commercial RIA kit, the beta-END(1-31) levels determined by a highly specific two-site RIA, proved to be remarkably small. Furthermore, CRH did not induce increases of heat pain tolerance thresholds, but of pressure pain tolerance thresholds, which, however, were not reversible by naloxone. Neither beta-END nor beta-LPH IRM nor beta-END(1-31) levels correlated with heat or pressure pain tolerance thresholds. We conclude that CRH does not modulate heat, but pressure pain; POMC derivatives like beta-END IRM, beta-END(1-31) or beta-LPH do not mediate this effect.


Subject(s)
Corticotropin-Releasing Hormone/administration & dosage , Pain Threshold/drug effects , Pain/drug therapy , beta-Endorphin/blood , Adrenocorticotropic Hormone/blood , Adrenocorticotropic Hormone/drug effects , Adult , Cross-Sectional Studies , Female , Hot Temperature , Humans , Injections, Intravenous , Male , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Pain Measurement , Pressure , beta-Endorphin/drug effects , beta-Lipotropin/blood
19.
Neurosci Lett ; 373(2): 85-8, 2005 Jan 10.
Article in English | MEDLINE | ID: mdl-15567558

ABSTRACT

It is known that opioid antagonists reduce the orexigenic effect of neropeptide Y (NPY) in mammals. We studied the effect of three opioid antagonists on NPY-induced feeding in male broiler chicks. Beta-funaltrexamine (beta-FNA), naloxonazine (NAL), ICI-174,864 (ICI) or nor-binaltorphimine (nor-BNI), antagonists of mu-, mu1-, delta- or kappa-receptors, and NPY were co-injected in chicks. Food intake was measured 30 min after treatment. Co-injection of beta-FNA or NAL was effective in reducing NPY-induced feeding, whereas ICI and nor-BNI had little effect on NPY-induced feeding. These data suggest that the mu-opioid receptor, especially the mu1-opioid has some relation to NPY-induced feeding, and implies that an endogenous ligand, such as beta-endorphin, participates in the orexigenic effect of NPY in neonatal chicks.


Subject(s)
Chickens/physiology , Feeding Behavior/drug effects , Narcotic Antagonists/pharmacology , Neuropeptide Y/pharmacology , Animals , Animals, Newborn , Male , Neuropeptide Y/metabolism , Receptors, Opioid/metabolism , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/metabolism , beta-Endorphin/drug effects , beta-Endorphin/metabolism
20.
Eur J Neurosci ; 19(12): 3183-92, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15217374

ABSTRACT

beta-Endorphin is an endogenous opioid that produces behavioral effects similar to heroin and morphine and is released in the nucleus accumbens by cocaine, amphetamine and ethanol, suggesting a general involvement in the reinforcing effects of abused drugs. Here we show that, in rats, Delta-9-tetrahydrocannabinol (THC), the main psychoactive ingredient in cannabis, produces large increases in extracellular levels of beta-endorphin in the ventral tegmental area and lesser increases in the shell of the nucleus accumbens. We then used a two-lever choice THC-discrimination procedure to investigate whether THC-induced changes in endogenous levels of beta-endorphin regulate the discriminative effects of THC. In rats that had learned to discriminate injections of THC from injections of vehicle, the opioid agonist morphine did not produce THC-like discriminative effects but markedly potentiated discrimination of THC. Conversely, the opioid antagonist naloxone reduced the discriminative effects of THC. Bilateral microinjections of beta-endorphin directly into the ventral tegmental area, but not into the shell of the nucleus accumbens, markedly potentiated the discriminative effects of ineffective threshold doses of THC but had no effect when given alone. This potentiation was blocked by naloxone. Together these results indicate that certain psychotropic effects of THC related to drug abuse liability are regulated by THC-induced elevations in extracellular beta-endorphin levels in brain areas involved in opiate reward and reinforcement processes.


Subject(s)
Discrimination Learning/drug effects , Dronabinol/pharmacology , Psychotropic Drugs/pharmacology , Ventral Tegmental Area/drug effects , beta-Endorphin/drug effects , Animals , Discrimination Learning/physiology , Injections, Intraventricular , Male , Microdialysis , Morphine/pharmacology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Narcotics/pharmacology , Rats , Rats, Sprague-Dawley , Ventral Tegmental Area/metabolism , beta-Endorphin/administration & dosage , beta-Endorphin/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL