Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.280
Filter
Add more filters

Publication year range
1.
Cell ; 157(7): 1500-1, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24949960

ABSTRACT

Excessive sun tanning can result in addictive behavior. In this issue of Cell, Fell et al. utilize a combination of behavioral pharmacology and transgenic mice to demonstrate that chronic UV light exposure recruits p53 signaling in keratinocytes, subsequently increasing ß-endorphin signaling at opioid receptors, and produces an endogenous opioid-dependent state.


Subject(s)
Behavior, Addictive , Skin/radiation effects , beta-Endorphin/metabolism , Animals , Humans
2.
Cell ; 157(7): 1527-34, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24949966

ABSTRACT

UV light is an established carcinogen, yet evidence suggests that UV-seeking behavior has addictive features. Following UV exposure, epidermal keratinocytes synthesize proopiomelanocortin (POMC) that is processed to melanocyte-stimulating hormone, inducing tanning. We show that, in rodents, another POMC-derived peptide, ß-endorphin, is coordinately synthesized in skin, elevating plasma levels after low-dose UV. Increases in pain-related thresholds are observed and reversed by pharmacologic opioid antagonism. Opioid blockade also elicits withdrawal signs after chronic UV exposure. This effect was sufficient to guide operant behavioral choices to avoidance of opioid withdrawal (conditioned place aversion). These UV-induced nociceptive and behavioral effects were absent in ß-endorphin knockout mice and in mice lacking p53-mediated POMC induction in epidermal keratinocytes. Although primordial UV addiction, mediated by the hedonic action of ß-endorphin and anhedonic effects of withdrawal, may theoretically have enhanced evolutionary vitamin D biosynthesis, it now may contribute to the relentless rise in skin cancer incidence in humans.


Subject(s)
Behavior, Addictive , Skin/radiation effects , beta-Endorphin/metabolism , Animals , Humans , Mice , Mice, Inbred C57BL , Models, Animal , Skin/metabolism , Ultraviolet Rays , beta-Endorphin/genetics
3.
Mol Pain ; 20: 17448069241254201, 2024.
Article in English | MEDLINE | ID: mdl-38670551

ABSTRACT

It has been widely recognized that electroacupuncture (EA) inducing the release of ß-endorphin represents a crucial mechanism of EA analgesia. The arcuate nucleus (ARC) in the hypothalamus is a vital component of the endogenous opioid peptide system. Serving as an integration center, the periaqueductal gray (PAG) receives neural fiber projections from the frontal cortex, insular cortex, and ARC. However, the specific mechanisms how EA facilitates the release of ß-endorphin within the ARC, eliciting analgesic effects are yet to be elucidated. In this study, we conducted in vivo and in vitro experiments by transcriptomics, microdialysis, photogenetics, chemical genetics, and calcium imaging, combined with transgenic animals. Firstly, we detected 2 Hz EA at the Zusanli (ST36) increased the level of ß-endorphin and transcriptional level of proopiomelanocortin (POMC). Our transcriptomics profiling demonstrated that 2 Hz EA at the ST36 modulates the expression of c-Fos and Jun B in ARC brain nuclear cluster, and the transcriptional regulation of 2 Hz EA mainly occur in POMC neurons by Immunofluorescence staining verification. Meaning while, 2 Hz EA specifically activated the cAMP-PKA-CREB signaling pathway in ARC which mediating the c-Fos and Jun B transcription, and 2 Hz EA analgesia is dependent on the activation of cAMP-PKA-CREB signaling pathway in ARC. In order to investigate how the ß-endorphin produced in ARC transfer to integration center PAG, transneuronal tracing technology was used to observe the 2 Hz EA promoted the neural projection from ARC to PAG compared to 100 Hz EA and sham mice. Inhibited PAGGABA neurons, the transfer of ß-endorphin from the ARC nucleus to the PAG nucleus through the ARCPOMC-PAGGABA neural circuit. Furthermore, by manipulating the excitability of POMC neurons from ARCPOMC to PAGGABA using inhibitory chemogenetics and optogenetics, we found that this inhibition significantly reduced transfer of ß-endorphin from the ARC nucleus to the PAG nucleus and the effectiveness of 2 Hz EA analgesia in neurological POMC cyclization recombination enzyme (Cre) mice and C57BL/6J mice, which indicates that the transfer of ß-endorphin depends on the activation of POMC neurons prefect from ARCPOMC to PAGGABA. These findings contribute to our understanding of the neural circuitry underlying the EA pain-relieving effects and maybe provide valuable insights for optimizing EA stimulation parameters in clinical pain treatment using the in vivo dynamic visual investigating the central analgesic mechanism.


Subject(s)
Arcuate Nucleus of Hypothalamus , Electroacupuncture , Periaqueductal Gray , Pro-Opiomelanocortin , beta-Endorphin , Animals , Pro-Opiomelanocortin/metabolism , Pro-Opiomelanocortin/genetics , Periaqueductal Gray/metabolism , Arcuate Nucleus of Hypothalamus/metabolism , Electroacupuncture/methods , beta-Endorphin/metabolism , Male , Mice, Transgenic , Mice, Inbred C57BL , Mice , Proto-Oncogene Proteins c-fos/metabolism , Neurons/metabolism
4.
Fish Physiol Biochem ; 50(2): 733-743, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38277042

ABSTRACT

Although the involvement of ß-endorphin (ß-ERP) in vertebrate reproduction has been suggested, its role in testicular activity is not clear in fish. We describe the influence of ß-ERP on spermatogenesis in a cichlid fish in the present paper. In comparison to the control group, the administration of ß-ERP (3 µg) caused a significant increase in the number of spermatogonia-A and spermatids. Following treatment with ß-ERP (6 µg), a significant increase in the number of spermatogonia-A was observed, whereas the numbers of all the other germ cells, excluding spermatogonia-B, significantly decreased in comparison to those in the control group. In addition, treatment of fish with 6 µg ß-ERP resulted in a significant reduction in the dimensions of the lumen and seminiferous lobules, the level of immunopositive androgen receptor (AR) expression in Sertoli cells, and the percentage of luteinizing hormone (LH) immunolabeled in the pituitary compared to those in the control group or the group treated with 3 µg ß-ERP. In contrast, the intensity of AR immunoreactivity and the percentage of LH immunolabeling were substantially increased in fish treated with 3 µg ß-ERP compared to those in the control group. These findings reveal for the first time that a low dose of ß-ERP stimulates the recruitment of spermatogonia as well as spermateleosis, whereas a high concentration affects the recruitment of germ cells prior to meiotic division in tilapia. These results suggest that ß-ERP exerts modulatory effects at the testicular and hypophysial levels through alterations in AR expression and LH secretory activity, respectively, in teleosts.


Subject(s)
Testis , Tilapia , Male , Animals , Testis/metabolism , Tilapia/metabolism , beta-Endorphin/metabolism , beta-Endorphin/pharmacology , Opioid Peptides/metabolism , Opioid Peptides/pharmacology , Spermatogenesis , Luteinizing Hormone/metabolism , Spermatogonia
5.
Int J Mol Sci ; 24(9)2023 Apr 25.
Article in English | MEDLINE | ID: mdl-37175503

ABSTRACT

Epidural motor cortex stimulation (MCS) is an effective treatment for refractory neuropathic pain; however, some individuals are unresponsive. In this study, we correlated the effectiveness of MCS and refractoriness with the expression of cytokines, neurotrophins, and nociceptive mediators in the dorsal root ganglion (DRG), sciatic nerve, and plasma of rats with sciatic neuropathy. MCS inhibited hyperalgesia and allodynia in two-thirds of the animals (responsive group), and one-third did not respond (refractory group). Chronic constriction injury (CCI) increased IL-1ß in the nerve and DRG, inhibited IL-4, IL-10, and IL-17A in the nerve, decreased ß-endorphin, and enhanced substance P in the plasma, compared to the control. Responsive animals showed decreased NGF and increased IL-6 in the nerve, accompanied by restoration of local IL-10 and IL-17A and systemic ß-endorphin. Refractory animals showed increased TNF-α and decreased IFNγ in the nerve, along with decreased TNF-α and IL-17A in the DRG, maintaining low levels of systemic ß-endorphin. Our findings suggest that the effectiveness of MCS depends on local control of inflammatory and neurotrophic changes, accompanied by recovery of the opioidergic system observed in neuropathic conditions. So, understanding the refractoriness to MCS may guide an improvement in the efficacy of the technique, thus benefiting patients with persistent neuropathic pain.


Subject(s)
Analgesia , Neuralgia , Rats , Animals , Interleukin-10/metabolism , Interleukin-17/metabolism , Tumor Necrosis Factor-alpha/metabolism , beta-Endorphin/metabolism , Neuralgia/therapy , Neuralgia/metabolism , Hyperalgesia/therapy , Hyperalgesia/metabolism , Sciatic Nerve/metabolism , Ganglia, Spinal/metabolism
6.
Am J Physiol Regul Integr Comp Physiol ; 322(3): R219-R227, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35043681

ABSTRACT

Anorexia nervosa (AN) is a debilitating eating disorder characterized by severely restricted eating and significant body weight loss. In addition, many individuals also report engaging in excessive exercise. Previous research using the activity-based anorexia (ABA) model has implicated the hypothalamic proopiomelanocortin (POMC) system. Using the ABA model, Pomc mRNA has been shown to be transiently elevated in both male and female rodents undergoing ABA. In addition, the POMC peptide ß-endorphin appears to contribute to food anticipatory activity (FAA), a characteristic of ABA, as both deletion and antagonism of the µ opioid receptor (MOR) that ß-endorphin targets, results in decreased FAA. The role of ß-endorphin in reduced food intake in ABA is unknown and POMC neurons release multiple transmitters in addition to ß-endorphin. In the current study, we set out to determine whether targeted inhibition of POMC neurons themselves rather than their peptide products would lessen the severity of ABA. Inhibition of POMC neurons during ABA via chemogenetic Designer Receptors Exclusively Activated by Designer Drugs (DREADD) technology resulted in reduced FAA in both male and female mice with no significant changes in body weight or food intake. The selective reduction in FAA persisted even in the face of concurrent chemogenetic inhibition of additional cell types in the hypothalamic arcuate nucleus. The results suggest that POMC neurons could be contributing preferentially to excessive exercise habits in patients with AN. Furthermore, the results also suggest that metabolic control during ABA appears to take place via a POMC neuron-independent mechanism.


Subject(s)
Anorexia/metabolism , Body Weight/physiology , Food , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Hypothalamus/metabolism , Mice , beta-Endorphin/metabolism , beta-Endorphin/pharmacology
7.
Curr Diab Rep ; 22(7): 301-310, 2022 07.
Article in English | MEDLINE | ID: mdl-35593927

ABSTRACT

PURPOSE OF REVIEW: We are currently in the midst of a global opioid epidemic. Opioids affect many physiological processes, but one side effect that is not often taken into consideration is the opioid-induced alteration in blood glucose levels. RECENT FINDINGS: This review shows that the vast majority of studies report that opioid stimulation increases blood glucose levels. In addition, plasma levels of the endogenous opioid ß-endorphin rise in response to low blood glucose. In contrast, in hyperglycaemic baseline conditions such as in patients with type 2 diabetes mellitus (T2DM), opioid stimulation lowers blood glucose levels. Furthermore, obesity itself alters sensitivity to opioids, changes opioid receptor expression and increases plasma ß-endorphin levels. Thus, opioid stimulation can have various side effects on glycaemia that should be taken into consideration upon prescribing opioid-based medication, and more research is needed to unravel the interaction between obesity, glycaemia and opioid use.


Subject(s)
Diabetes Mellitus, Type 2 , Epidemics , Analgesics, Opioid/adverse effects , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/epidemiology , Humans , Obesity/epidemiology , beta-Endorphin/metabolism , beta-Endorphin/pharmacology
8.
Int J Mol Sci ; 24(1)2022 Dec 29.
Article in English | MEDLINE | ID: mdl-36614024

ABSTRACT

The opioid peptide ß-endorphin coexists in the pituitary and brain in its αN-acetylated form, which does not bind to opioid receptors. We now report that these neuropeptides exhibited opposite effects in in vivo paradigms, in which ligands of the sigma type 1 receptor (σ1R) displayed positive effects. Thus, αN-acetyl ß-Endorphin reduced vascular infarct caused by permanent unilateral middle cerebral artery occlusion and diminished the incidence of N-methyl-D-aspartate acid-promoted convulsive syndrome and mechanical allodynia caused by unilateral chronic constriction of the sciatic nerve. Moreover, αN-acetyl ß-Endorphin reduced the analgesia of morphine, ß-Endorphin and clonidine but enhanced that of DAMGO. All these effects were counteracted by ß-Endorphin and absent in σ1R-/- mice. We observed that σ1Rs negatively regulate mu-opioid receptor (MOR)-mediated morphine analgesia by binding and sequestering G proteins. In this scenario, ß-Endorphin promoted the exchange of σ2Rs by G proteins at σ1R oligomers and increased the regulation of G proteins by MORs. The opposite was observed for the αN-acetyl derivative, as σ1R oligomerization decreased and σ2R binding was favored, which displaced G proteins; thus, MOR-regulated transduction was reduced. Our findings suggest that the pharmacological ß-Endorphin-specific epsilon receptor is a σ1R-regulated MOR and that ß-Endorphin and αN-acetyl ß-Endorphin are endogenous ligands of σ1R.


Subject(s)
Receptors, Opioid, mu , Receptors, sigma , beta-Endorphin , Animals , Mice , beta-Endorphin/metabolism , GTP-Binding Proteins/metabolism , Ligands , Morphine/pharmacology , Pain , Receptors, Opioid/metabolism , Receptors, Opioid, mu/metabolism , Receptors, sigma/metabolism
9.
Int J Mol Sci ; 23(19)2022 Sep 30.
Article in English | MEDLINE | ID: mdl-36232883

ABSTRACT

Pain is a worldwide public health problem and its treatment is still a challenge since clinically available drugs do not completely reverse chronic painful states or induce undesirable effects. Crotalphine is a 14 amino acids synthetic peptide that induces a potent and long-lasting analgesic effect on acute and chronic pain models, peripherally mediated by the endogenous release of dynorphin A and the desensitization of the transient receptor potential ankyrin 1 (TRPA1) receptor. However, the effects of crotalphine on the central nervous system (CNS) and the signaling pathway have not been investigated. Thus, the central effect of crotalphine was evaluated on the partial sciatic nerve ligation (PSNL)-induced chronic neuropathic pain model. Crotalphine (100 µg/kg, p.o.)-induced analgesia on the 14th day after surgery lasting up to 24 h after administration. This effect was prevented by intrathecal administration of CB1 (AM251) or CB2 (AM630) cannabinoid receptor antagonists. Besides that, crotalphine-induced analgesia was reversed by CTOP, nor-BNI, and naltrindole, antagonists of mu, kappa, and delta-opioid receptors, respectively, and also by the specific antibodies for ß-endorphin, dynorphin-A, and met-enkephalin. Likewise, the analgesic effect of crotalphine was blocked by the intrathecal administration of minocycline, an inhibitor of microglial activation and proliferation. Additionally, crotalphine decreased the PSNL-induced IL-6 release in the spinal cord. Importantly, in vitro, crotalphine inhibited LPS-induced CD86 expression and upregulated CD206 expression in BV-2 cells, demonstrating a polarization of microglial cells towards the M2 phenotype. These results demonstrated that crotalphine, besides activating opioid and cannabinoid analgesic systems, impairs central neuroinflammation, confirming the neuromodulatory mechanism involved in the crotalphine analgesic effect.


Subject(s)
Analgesia , Cannabinoids , Neuralgia , Amino Acids/metabolism , Analgesics/metabolism , Analgesics/pharmacology , Analgesics/therapeutic use , Analgesics, Opioid/metabolism , Ankyrins/metabolism , Cannabinoid Receptor Antagonists/therapeutic use , Cannabinoids/therapeutic use , Dynorphins/metabolism , Enkephalin, Methionine/metabolism , Humans , Interleukin-6/metabolism , Lipopolysaccharides/metabolism , Microglia/metabolism , Minocycline/therapeutic use , Neuralgia/metabolism , Peptides , Phenotype , Receptors, Opioid/metabolism , Spinal Cord , beta-Endorphin/metabolism
10.
J Neurosci ; 40(41): 7965-7979, 2020 10 07.
Article in English | MEDLINE | ID: mdl-32887744

ABSTRACT

Microglia, a type of CNS immune cell, have been shown to contribute to ethanol-activated neuronal death of the stress regulatory proopiomelanocortin (POMC) neuron-producing ß-endorphin peptides in the hypothalamus in a postnatal rat model of fetal alcohol spectrum disorders. We determined whether the microglial extracellular vesicle exosome is involved in the ethanol-induced neuronal death of the ß-endorphin neuron. Extracellular vesicles were prepared from hypothalamic tissues collected from postnatal rats (both males and females) fed daily with 2.5 mg/kg ethanol or control milk formula for 5 d or from hypothalamic microglia cells obtained from postnatal rats, grown in cultures for several days, and then challenged with ethanol or vehicle for 24 h. Nanoparticle tracking analysis and transmission electron microscopy indicated that these vesicles had the size range and shape of exosomes. Ethanol treatments increased the number and the ß-endorphin neuronal killing activity of microglial exosomes both in vivo and in vitro Proteomics analyses of exosomes of cultured microglial cells identified a large number of proteins, including various complements, which were elevated following ethanol treatment. Proteomics data involving complements were reconfirmed using quantitative protein assays. Ethanol treatments also increased deposition of the complement protein C1q in ß-endorphin neuronal cells in both in vitro and in vivo systems. Recombinant C1q protein increased while C1q blockers reduced ethanol-induced C3a/b, C4, and membrane attack complex/C5b9 formations; ROS production; and ultimately cellular death of ß-endorphin neurons. These data suggest that the complement system involving C1q-C3-C4-membrane attack complex and ROS regulates exosome-mediated, ethanol-induced ß-endorphin neuronal death.SIGNIFICANCE STATEMENT Neurotoxic action of alcohol during the developmental period is recognized for its involvement in fetal alcohol spectrum disorders, but the lack of clear understanding of the mechanism of alcohol action has delayed the progress in therapeutic intervention of this disease. Proopiomelanocortin neurons known to regulate stress, energy homeostasis, and immune functions are reported to be killed by developmental alcohol exposure because of activation of microglial immune cells in the brain. While microglia are known to use extracellular vesicles to communicate with neurons for maintaining homeostasis, we show here that ethanol exposure during the developmental period hijacks this system to spread apoptotic factors, including complement protein C1q, to induce the membrane attack complex and reactive super-oxygen species for proopiomelanocortin neuronal killing.


Subject(s)
Central Nervous System Depressants/pharmacology , Complement C1q/pharmacology , Ethanol/pharmacology , Exosomes/drug effects , Fetal Alcohol Spectrum Disorders/pathology , Microglia/drug effects , Pro-Opiomelanocortin/genetics , Animals , Animals, Newborn , Cell Death/drug effects , Cells, Cultured , Female , Fetal Alcohol Spectrum Disorders/metabolism , Hypothalamus/metabolism , Hypothalamus/pathology , Male , Neurons/drug effects , Neurons/metabolism , Pregnancy , Proteomics , Rats , Rats, Sprague-Dawley , beta-Endorphin/metabolism
11.
Brain Behav Immun ; 95: 344-361, 2021 07.
Article in English | MEDLINE | ID: mdl-33862171

ABSTRACT

Gabapentinoids are recommended first-line treatments for neuropathic pain. They are neuronal voltage-dependent calcium channel α2δ-1 subunit ligands and have been suggested to attenuate neuropathic pain via interaction with neuronal α2δ-1 subunit. However, the current study revealed their microglial mechanisms underlying antineuropathic pain. Intrathecal injection of gabapentin, pregabalin and mirogabalin rapidly inhibited mechanical allodynia and thermal hyperalgesia, with projected ED50 values of 30.3, 6.2 and 1.5 µg (or 176.9, 38.9 and 7.2 nmol) and Emax values of 66%, 61% and 65% MPE respectively for mechanical allodynia. Intrathecal gabapentinoids stimulated spinal mRNA and protein expression of IL-10 and ß-endorphin (but not dynorphin A) in neuropathic rats with the time point parallel to their inhibition of allodynia, which was observed in microglia but not astrocytes or neurons in spinal dorsal horns by using double immunofluorescence staining. Intrathecal gabapentin alleviated pain hypersensitivity in male/female neuropathic but not male sham rats, whereas it increased expression of spinal IL-10 and ß-endorphin in male/female neuropathic and male sham rats. Treatment with gabapentin, pregabalin and mirogabalin specifically upregulated IL-10 and ß-endorphin mRNA and protein expression in primary spinal microglial but not astrocytic or neuronal cells, with EC50 values of 41.3, 11.5 and 2.5 µM and 34.7, 13.3 and 2.8 µM respectively. Pretreatment with intrathecal microglial metabolic inhibitor minocycline, IL-10 antibody, ß-endorphin antiserum or µ-opioid receptor antagonist CTAP (but not κ- or δ-opioid receptor antagonists) suppressed spinal gabapentinoids-inhibited mechanical allodynia. Immunofluorescence staining exhibited specific α2δ-1 expression in neurons but not microglia or astrocytes in the spinal dorsal horns or cultured primary spinal cells. Thus the results illustrate that gabapentinoids alleviate neuropathic pain through stimulating expression of spinal microglial IL-10 and consequent ß-endorphin.


Subject(s)
Gabapentin/pharmacology , Interleukin-10 , Microglia/metabolism , Neuralgia , beta-Endorphin , Animals , Female , Hyperalgesia/drug therapy , Interleukin-10/metabolism , Male , Neuralgia/drug therapy , Rats , Rats, Wistar , Spinal Cord , beta-Endorphin/metabolism
12.
Nature ; 519(7541): 45-50, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25707796

ABSTRACT

Hypothalamic pro-opiomelanocortin (POMC) neurons promote satiety. Cannabinoid receptor 1 (CB1R) is critical for the central regulation of food intake. Here we test whether CB1R-controlled feeding in sated mice is paralleled by decreased activity of POMC neurons. We show that chemical promotion of CB1R activity increases feeding, and notably, CB1R activation also promotes neuronal activity of POMC cells. This paradoxical increase in POMC activity was crucial for CB1R-induced feeding, because designer-receptors-exclusively-activated-by-designer-drugs (DREADD)-mediated inhibition of POMC neurons diminishes, whereas DREADD-mediated activation of POMC neurons enhances CB1R-driven feeding. The Pomc gene encodes both the anorexigenic peptide α-melanocyte-stimulating hormone, and the opioid peptide ß-endorphin. CB1R activation selectively increases ß-endorphin but not α-melanocyte-stimulating hormone release in the hypothalamus, and systemic or hypothalamic administration of the opioid receptor antagonist naloxone blocks acute CB1R-induced feeding. These processes involve mitochondrial adaptations that, when blocked, abolish CB1R-induced cellular responses and feeding. Together, these results uncover a previously unsuspected role of POMC neurons in the promotion of feeding by cannabinoids.


Subject(s)
Cannabinoids/pharmacology , Eating/drug effects , Eating/physiology , Hypothalamus/cytology , Neurons/drug effects , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Animals , Energy Metabolism/drug effects , Hypothalamus/drug effects , Hypothalamus/physiology , Ion Channels/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Naloxone/pharmacology , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/metabolism , Satiety Response/drug effects , Satiety Response/physiology , Uncoupling Protein 2 , alpha-MSH/metabolism , beta-Endorphin/metabolism
13.
Arch Gynecol Obstet ; 303(5): 1207-1216, 2021 05.
Article in English | MEDLINE | ID: mdl-33247770

ABSTRACT

PURPOSE: The present study aims to explore whether ß-EP in serum (sß-EP) and follicular fluid (ffß-EP) could predict the in vitro fertilization (IVF) outcomes of patients with polycystic ovary syndrome (PCOS) and diminished ovarian reserve (DOR). METHODS: 90 PCOS women, 50 DOR women, and 100 women with normal ovarian function (control group), who were all undergoing an IVF-embryo transfer trial, were included in the study. Biochemical characteristics, anti-Mullerian hormone (AMH), sß-EP, ffß-EP, embryo formation, and pregnancy indicators were assessed in all women. The correlations of AMH and ß-EP with oocyte quality were analyzed. Population-based and age-category stratified receiver operating characteristic (ROC) curve analysis of AMH and ß-EP for predicting pregnancy and live birth were performed. RESULTS: Compared with the control group, the PCOS group had higher antral follicle count, testosterone, luteinizing hormone, AMH, sß-EP, and ffß-EP, which were lower in the DOR group. Meanwhile, the PCOS and DOR groups had higher cycle cancellation and miscarriage rates, and lower high quality embryo numbers. Correlation analysis showed that the oocyte quality were positively correlated with AMH, sß-EP, and ffß-EP. The population-based and age-stratified ROC curve analysis showed that sß-EP and ffß-EP had high sensitivity and specificity to predict pregnancy and live birth. Meanwhile, age-stratified AMH enhanced the sensitivity for prediction of live birth after IVF. CONCLUSION: sß-EP and ffß-EP are different among women with PCOS, DOR, and normal ovarian function. ß-EP can be used as a good predictor of clinical pregnancy and live birth after IVF.


Subject(s)
Fertilization in Vitro/adverse effects , Ovarian Reserve/genetics , Polycystic Ovary Syndrome/blood , beta-Endorphin/metabolism , Adult , Female , Fertilization in Vitro/methods , Humans , Pregnancy , Pregnancy Outcome
14.
Biochem Biophys Res Commun ; 525(4): 1087-1094, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32184015

ABSTRACT

Lemairamin (also known as wgx-50), is isolated from the pericarps of the Zanthoxylum plants. As an agonist of α7 nicotinic acetylcholine receptors (α7nAChRs), it can reduce neuroinflammation in Alzheimer's disease. This study evaluated its antinociceptive effects in pain hypersensitivity and explored the underlying mechanisms. The data showed that subcutaneous lemairamin injection dose-dependently inhibited formalin-induced tonic pain but not acute nociception in mice and rats, while intrathecal lemairamin injection also dose-dependently produced mechanical antiallodynia in the ipsilateral hindpaws of neuropathic and bone cancer pain rats without affecting mechanical thresholds in the contralateral hindpaws. Multiple bi-daily lemairamin injections for 7 days did not induce mechanical antiallodynic tolerance in neuropathic rats. Moreover, the antinociceptive effects of lemairamin in formalin-induced tonic pain and mechanical antiallodynia in neuropathic pain were suppressed by the α7nAChR antagonist methyllycaconitine. In an α7nAChR antagonist-reversible manner, intrathecal lemairamin also stimulated spinal expression of IL-10 and ß-endorphin, while lemairamin treatment induced IL-10 and ß-endorphin expression in primary spinal microglial cells. In addition, intrathecal injection of a microglial activation inhibitor minocycline, anti-IL-10 antibody, anti-ß-endorphin antiserum or µ-opioid receptor-preferred antagonist naloxone was all able to block lemairamin-induced mechanical antiallodynia in neuropathic pain. These data demonstrated that lemairamin could produce antinociception in pain hypersensitivity through the spinal IL-10/ß-endorphin pathway following α7nAChR activation.


Subject(s)
Acrylamides/pharmacology , Analgesics/pharmacology , Cancer Pain/drug therapy , Hyperalgesia/drug therapy , Microglia/drug effects , Neuralgia/drug therapy , alpha7 Nicotinic Acetylcholine Receptor/agonists , Aconitine/analogs & derivatives , Aconitine/pharmacology , Acrylamides/administration & dosage , Acrylamides/therapeutic use , Analgesics/administration & dosage , Analgesics/therapeutic use , Animals , Female , Formaldehyde , Hyperalgesia/genetics , Hyperalgesia/metabolism , Injections, Spinal , Interleukin-10/genetics , Interleukin-10/metabolism , Male , Mice , Microglia/metabolism , Minocycline/administration & dosage , Naloxone/administration & dosage , Rats , Rats, Wistar , Spinal Cord/metabolism , Zanthoxylum/chemistry , Zanthoxylum/metabolism , alpha7 Nicotinic Acetylcholine Receptor/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism , beta-Endorphin/genetics , beta-Endorphin/metabolism
15.
J Neuroinflammation ; 17(1): 75, 2020 Feb 29.
Article in English | MEDLINE | ID: mdl-32113469

ABSTRACT

BACKGROUND: Cinobufagin is the major bufadienolide of Bufonis venenum (Chansu), which has been traditionally used for the treatment of chronic pain especially cancer pain. The current study aimed to evaluate its antinociceptive effects in bone cancer pain and explore the underlying mechanisms. METHODS: Rat bone cancer model was used in this study. The withdrawal threshold evoked by stimulation of the hindpaw was determined using a 2290 CE electrical von Frey hair. The ß-endorphin and IL-10 levels were measured in the spinal cord and cultured primary microglia, astrocytes, and neurons. RESULTS: Cinobufagin, given intrathecally, dose-dependently attenuated mechanical allodynia in bone cancer pain rats, with the projected Emax of 90% MPE and ED50 of 6.4 µg. Intrathecal cinobufagin also stimulated the gene and protein expression of IL-10 and ß-endorphin (but not dynorphin A) in the spinal cords of bone cancer pain rats. In addition, treatment with cinobufagin in cultured primary spinal microglia but not astrocytes or neurons stimulated the mRNA and protein expression of IL-10 and ß-endorphin, which was prevented by the pretreatment with the IL-10 antibody but not ß-endorphin antiserum. Furthermore, spinal cinobufagin-induced mechanical antiallodynia was inhibited by the pretreatment with intrathecal injection of the microglial inhibitor minocycline, IL-10 antibody, ß-endorphin antiserum and specific µ-opioid receptor antagonist CTAP. Lastly, cinobufagin- and the specific α-7 nicotinic acetylcholine receptor (α7-nAChR) agonist PHA-543613-induced microglial gene expression of IL-10/ß-endorphin and mechanical antiallodynia in bone cancer pain were blocked by the pretreatment with the specific α7-nAChR antagonist methyllycaconitine. CONCLUSIONS: Our results illustrate that cinobufagin produces mechanical antiallodynia in bone cancer pain through spinal microglial expression of IL-10 and subsequent ß-endorphin following activation of α7-nAChRs. Our results also highlight the broad significance of the recently uncovered spinal microglial IL-10/ß-endorphin pathway in antinociception.


Subject(s)
Bufanolides/pharmacology , Cancer Pain/metabolism , Hyperalgesia/metabolism , Microglia/drug effects , Animals , Bone Neoplasms/complications , Female , Interleukin-10/metabolism , Male , Microglia/metabolism , Rats , Rats, Wistar , Spinal Cord/drug effects , Spinal Cord/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , beta-Endorphin/metabolism
16.
Proc Natl Acad Sci U S A ; 114(20): 5300-5305, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28461468

ABSTRACT

There is growing evidence that the number and quality of social relationships have substantial impacts on health, well-being, and longevity, and, at least in animals, on reproductive fitness. Although it is widely recognized that these outcomes are mediated by a number of neuropeptides, the roles these play remain debated. We suggest that an overemphasis on one neuropeptide (oxytocin), combined with a failure to distinguish between different social domains, has obscured the complexity involved. We use variation in 33 SNPs for the receptor genes for six well-known social neuropeptides in relation to three separate domains of sociality (social disposition, dyadic relationships, and social networks) to show that three neuropeptides (ß-endorphin, oxytocin, and dopamine) play particularly important roles, with each being associated predominantly with a different social domain. However, endorphins and dopamine have a much wider compass than oxytocin (whose effects are confined to romantic/reproductive relationships and often do not survive control for other neuropeptides). In contrast, vasopressin, serotonin, and testosterone play only limited roles.


Subject(s)
Receptors, Dopamine/genetics , Receptors, Opioid/genetics , Receptors, Oxytocin/genetics , Adolescent , Adult , Aged , Brain/metabolism , Dopamine/genetics , Dopamine/metabolism , Female , Humans , Male , Middle Aged , Neuropeptides/genetics , Neuropeptides/metabolism , Oxytocin/genetics , Oxytocin/metabolism , Receptors, Dopamine/metabolism , Receptors, Opioid/metabolism , Saliva , Social Behavior , Social Networking , Social Support , beta-Endorphin/genetics , beta-Endorphin/metabolism
17.
Int J Mol Sci ; 22(1)2020 Dec 30.
Article in English | MEDLINE | ID: mdl-33396962

ABSTRACT

ß-Endorphins are peptides that exert a wide variety of effects throughout the body. Produced through the cleavage pro-opiomelanocortin (POMC), ß-endorphins are the primarily agonist of mu opioid receptors, which can be found throughout the body, brain, and cells of the immune system that regulate a diverse set of systems. As an agonist of the body's opioid receptors, ß-endorphins are most noted for their potent analgesic effects, but they also have their involvement in reward-centric and homeostasis-restoring behaviors, among other effects. These effects have implicated the peptide in psychiatric and neurodegenerative disorders, making it a research target of interest. This review briefly summarizes the basics of endorphin function, goes over the behaviors and regulatory pathways it governs, and examines the variability of ß-endorphin levels observed between normal and disease/disorder affected individuals.


Subject(s)
Behavior, Animal , Behavior , Brain/physiopathology , Energy Metabolism , Inflammation/physiopathology , Stress, Physiological , beta-Endorphin/metabolism , Animals , Humans
18.
J Neuroinflammation ; 16(1): 84, 2019 Apr 13.
Article in English | MEDLINE | ID: mdl-30981281

ABSTRACT

BACKGROUND: The G protein-coupled receptor 40 (GPR40), broadly expressed in various tissues such as the spinal cord, exerts multiple physiological functions including pain regulation. This study aimed to elucidate the mechanisms underlying GPR40 activation-induced antinociception in neuropathic pain, particularly related to the spinal glial expression of IL-10 and subsequent ß-endorphin. METHODS: Spinal nerve ligation-induced neuropathic pain model was used in this study. ß-Endorphin and IL-10 levels were measured in the spinal cord and cultured primary microglia, astrocytes, and neurons. Double immunofluorescence staining of ß-endorphin with glial and neuronal cellular biomarkers was also detected in the spinal cord and cultured primary microglia, astrocytes, and neurons. RESULTS: GPR40 was expressed on microglia, astrocytes, and neurons in the spinal cords and upregulated by spinal nerve ligation. Intrathecal injection of the GPR40 agonist GW9508 dose-dependently attenuated mechanical allodynia and thermal hyperalgesia in neuropathic rats, with Emax values of 80% and 100% MPE and ED50 values of 6.7 and 5.4 µg, respectively. Its mechanical antiallodynia was blocked by the selective GPR40 antagonist GW1100 but not GPR120 antagonist AH7614. Intrathecal GW9508 significantly enhanced IL-10 and ß-endorphin immunostaining in spinal microglia and astrocytes but not in neurons. GW9508 also markedly stimulated gene and protein expression of IL-10 and ß-endorphin in cultured primary spinal microglia and astrocytes but not in neurons, originated from 1-day-old neonatal rats. The IL-10 antibody inhibited GW9508-stimulated gene expression of the ß-endorphin precursor proopiomelanocortin (POMC) but not IL-10, whereas the ß-endorphin antibody did not affect GW9508-stimulated IL-10 or POMC gene expression. GW9508 increased phosphorylation of mitogen-activated protein kinases (MAPKs) including p38, extracellular signal-regulated kinase (ERK), and c-Jun N-terminal kinase (JNK), and its stimulatory effects on IL-10 and POMC expression were blocked by each MAPK isoform inhibitor. Spinal GW9508-induced mechanical antiallodynia was completely blocked by intrathecal minocycline, IL-10 neutralizing antibody, ß-endorphin antiserum, and µ-opioid receptor-preferred antagonist naloxone. CONCLUSIONS: Our results illustrate that GPR40 activation produces antinociception via the spinal glial IL-10/ß-endorphin antinociceptive pathway.


Subject(s)
Hyperalgesia/etiology , Hyperalgesia/metabolism , Interleukin-10/metabolism , Neuralgia , Neuroglia/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , beta-Endorphin/metabolism , Animals , Animals, Newborn , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Gene Expression Regulation/drug effects , Hyperalgesia/drug therapy , Interleukin-10/genetics , Male , Methylamines/therapeutic use , Nerve Tissue Proteins/metabolism , Neuralgia/complications , Neuralgia/metabolism , Neuralgia/pathology , Pain Measurement , Propionates/therapeutic use , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction/drug effects
19.
Exp Dermatol ; 28(5): 585-592, 2019 05.
Article in English | MEDLINE | ID: mdl-30903724

ABSTRACT

BACKGROUND: Therapeutic effects of Dead Sea (DS) minerals are well established, and their unique combination is analysed and reported. DS water (DSW) is a key source for DS minerals, and various studies report the capability of DSW to alleviate symptoms of different skin disorders and to contribute to skin maintenance. However, the biological mechanisms beyond reported effects are not fully understood yet. OBJECTIVE: To elucidate the effect of topically applied DSW via the expression of different skin biomarkers related to barrier function, homeostasis, inflammation and irritation. METHODS: In vitro skin equivalents and ex vivo human skin organ culture were used to assess the biological effects of DSW. Epidermal barrier protein expression and DSW ions transdermal penetration were analysed on skin equivalents. ß-endorphin secretion was tested on human skin organ culture. The capability of DSW to protect against skin inflammation and irritation was tested on ex vivo human skin organ culture by lipopolysaccharides and sodium dodecyl sulphate addition, respectively. RESULTS: Topical application of DSW encouraged the expression of the barrier-related proteins: filaggrin, involucrin and transglutaminase, while transdermal penetration of calcium ions was not detected. Additionally, DSW application had increased skin secretion of ß-endorphin and attenuated the expression of inflammatory and irritation-related cytokines. CONCLUSIONS: This study reports new findings of DSW effects on skin. Signalling pathway activation is proposed as a key step that may result in a vast range of proven biological activities following skin exposure to DS minerals, and specifically DSW.


Subject(s)
Minerals/pharmacology , Seawater/chemistry , Skin/drug effects , Skin/metabolism , Biomarkers/metabolism , Calcium/metabolism , Cytokines/metabolism , Epidermis/metabolism , Filaggrin Proteins , Homeostasis , Humans , Inflammation , Ions , Lipopolysaccharides , Microscopy, Fluorescence , Organ Culture Techniques , Skin/pathology , Skin Diseases/drug therapy , Sodium Dodecyl Sulfate , beta-Endorphin/metabolism
20.
Gynecol Endocrinol ; 35(9): 767-771, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30935252

ABSTRACT

ß-endorphin is a neuropeptide involved in several brain functions: its plasma levels are higher in obese women and its release increases after oral glucose tolerance test (OGTT) in normal or obese women. The study included 46 healthy women and evaluated the effect of oral dehydroepiandrosterone [DHEA] (50 mg/day) in early postmenopausal women (50-55 years) both of normal weight (group A, n = 12, BMI = 22.1 ± 0.5) and overweight (group B, n = 12, BMI = 28.2 ± 0.5), and late postmenopausal women (60-65 years) both normal weight (group C, n = 11, BMI = 22.5 ± 0.6) and overweight (group D, n = 11, BMI = 27.9 ± 0.4) undergone OGTT, in order to investigate if DHEA could restore/modify the control of insulin and glucose secretion and ß-endorphin release in response to glucose load. The area under the curve (AUC) of OGTT evaluated plasma levels of different molecules. DHEA, DHEAS, and ß-endorphin plasma levels were lower in baseline conditions in older women than younger women. Considering the AUC of ß-endorphin response to OGTT, all groups showed a progressive significant increase after 3 and also after 6 months of treatment in comparison to baseline and 3 months of treatment.


Subject(s)
Dehydroepiandrosterone/administration & dosage , Glucose/pharmacology , Postmenopause/blood , Postmenopause/drug effects , beta-Endorphin/metabolism , Administration, Oral , Aged , Androgens/blood , Female , Glucose Tolerance Test , Humans , Ideal Body Weight/drug effects , Ideal Body Weight/physiology , Insulin/blood , Middle Aged , Overweight/blood , Overweight/metabolism , Overweight/physiopathology , Sex Hormone-Binding Globulin/metabolism , Testosterone/blood , Time Factors , beta-Endorphin/blood
SELECTION OF CITATIONS
SEARCH DETAIL