Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Cell ; 186(2): 413-427.e17, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36638794

ABSTRACT

Opioids are effective analgesics, but their use is beset by serious side effects, including addiction and respiratory depression, which contribute to the ongoing opioid crisis. The human opioid system contains four opioid receptors (µOR, δOR, κOR, and NOPR) and a set of related endogenous opioid peptides (EOPs), which show distinct selectivity toward their respective opioid receptors (ORs). Despite being key to the development of safer analgesics, the mechanisms of molecular recognition and selectivity of EOPs to ORs remain unclear. Here, we systematically characterize the binding of EOPs to ORs and present five structures of EOP-OR-Gi complexes, including ß-endorphin- and endomorphin-bound µOR, deltorphin-bound δOR, dynorphin-bound κOR, and nociceptin-bound NOPR. These structures, supported by biochemical results, uncover the specific recognition and selectivity of opioid peptides and the conserved mechanism of opioid receptor activation. These results provide a structural framework to facilitate rational design of safer opioid drugs for pain relief.


Subject(s)
Receptors, Opioid , Humans , Analgesics, Opioid/pharmacology , Opioid Peptides , Receptors, Opioid, mu/metabolism , Receptors, Opioid/chemistry
2.
J Allergy Clin Immunol ; 149(3): 1085-1096, 2022 03.
Article in English | MEDLINE | ID: mdl-34411589

ABSTRACT

BACKGROUND: Mechanical alloknesis (or innocuous mechanical stimuli-evoked itch) often occurs in dry skin-based disorders such as atopic dermatitis and psoriasis. However, the molecular and cellular mechanisms underlying mechanical alloknesis remain unclear. We recently reported the involvement of CD26 in the regulation of psoriatic itch. This molecule exhibits dipeptidyl peptidase IV (DPPIV) enzyme activity and exerts its biologic effects by processing various substances, including neuropeptides. OBJECTIVE: The aim of the present study was to investigate the peripheral mechanisms of mechanical alloknesis by using CD26/DPPIV knockout (CD26KO) mice. METHODS: We applied innocuous mechanical stimuli to CD26KO or wild-type mice. The total number of scratching responses was counted as the alloknesis score. Immunohistochemical and behavioral pharmacologic analyses were then performed to examine the physiologic activities of CD26/DPPIV or endomorphins (EMs), endogenous agonists of µ-opioid receptors. RESULTS: Mechanical alloknesis was more frequent in CD26KO mice than in wild-type mice. The alloknesis score in CD26KO mice was significantly reduced by the intradermal administration of recombinant DPPIV or naloxone methiodide, a peripheral µ-opioid receptor antagonist, but not by that of mutant DPPIV without enzyme activity. EMs (EM-1 and EM-2), selective ligands for µ-opioid receptors, are substrates for DPPIV. Immunohistochemically, EMs were located in keratinocytes, fibroblasts, and peripheral sensory nerves. Behavioral analyses revealed that EMs preferentially provoked mechanical alloknesis over chemical itch. DPPIV-digested forms of EMs did not induce mechanical alloknesis. CONCLUSION: The present results suggest that EMs induce mechanical alloknesis at the periphery under the enzymatic control of CD26/DPPIV.


Subject(s)
Dermatitis, Atopic , Dipeptidyl Peptidase 4 , Psoriasis , Animals , Dipeptidyl Peptidase 4/genetics , Keratinocytes , Mice , Pruritus
3.
Arch Biochem Biophys ; 727: 109305, 2022 09 30.
Article in English | MEDLINE | ID: mdl-35660411

ABSTRACT

Reduction of Cu(II) to Cu(I) in an oxidizing extracellular environment is a potential risk factor for neurodegenerative diseases, because the re-oxidation of Cu(I) to Cu(II) can be coupled to generation of reactive oxygen species. However, little is known about how the brain is protected from the copper-induced oxidative stress. In the present study, interactions of the endogenous opioid peptide endomorphin-1 (EM1, Tyr-Pro-Trp-Phe-NH2) with ionic copper were investigated. EM1 cannot bind copper with ordinary metal coordination chemistry, since the chelate complex formation of EM1 with the metal ion is inhibited by the proline residue in the second position. In the presence of SDS micelles, however, a significant quenching of fluorescence of the tryptophan side chain of EM1 was observed on addition of copper ion, either Cu(II) or Cu(I). The spectral changes of the UV absorption of the tryptophan, which are diagnostic of cation-π interaction, were also brought about by addition of copper to EM1 only in the presence of micelles. The copper-induced spectral changes of both fluorescence and UV absorption disappeared upon the substitution of Tyr1 with alanine. The obtained results indicated that EM1 binds the copper ion through the π-electrons of aromatic side chains of Tyr1 and Trp3, which are in close contact each other in the micelle-associated form. The copper-catalyzed oxidation/reduction reaction process converting dopamine to neuromelanin, which involves potentially neurotoxic intermediates, is inhibited by EM1. Owing to the ability to bind both Cu(II) and Cu(I), EM1 may have the potential to suppress the copper-mediated oxidative stress in the brain. The present results suggest an antioxidative effect of EM1, distinct from its known analgesic effect.


Subject(s)
Copper , Micelles , Copper/chemistry , Oligopeptides , Oxidation-Reduction , Tryptophan/chemistry
4.
Pharmacol Res ; 182: 106322, 2022 08.
Article in English | MEDLINE | ID: mdl-35750299

ABSTRACT

Recent studies have proposed that heteromers of µ-opioid receptors (MORs) and galanin Gal1 receptors (Gal1Rs) localized in the mesencephalon mediate the dopaminergic effects of opioids. The present study reports converging evidence, using a peptide-interfering approach combined with biophysical and biochemical techniques, including total internal reflection fluorescence microscopy, for a predominant homodimeric structure of MOR and Gal1R when expressed individually, and for their preference to form functional heterotetramers when co-expressed. Results show that a heteromerization-dependent change in the Gal1R homodimeric interface leads to a switch in G-protein coupling from inhibitory Gi to stimulatory Gs proteins. The MOR-Gal1R heterotetramer, which is thus bound to Gs via the Gal1R homodimer and Gi via the MOR homodimer, provides the framework for a canonical Gs-Gi antagonist interaction at the adenylyl cyclase level. These novel results shed light on the intense debate about the oligomeric quaternary structure of G protein-coupled receptors, their predilection for heteromer formation, and the resulting functional significance.


Subject(s)
Analgesics, Opioid , Galanin , Analgesics, Opioid/pharmacology , Mesencephalon , Peptides , Receptors, Opioid
5.
Clin Exp Pharmacol Physiol ; 48(8): 1162-1170, 2021 08.
Article in English | MEDLINE | ID: mdl-33851456

ABSTRACT

PK20 is an anti-inflammatory hybrid compound, composed of an endomorphin-2-like and neurotensin-like fragments. The aim of the present study is to assess the contribution of particular pharmacophores to the activity of the hybrid tested. For this purpose, airway hyperresponsiveness, accumulation of inflammatory cells in bronchoalveolar lavage fluid (BALF), concentration of mouse mast cell protease, malondialdehyde and secretory phospholipase 2 activity in lung tissue, as well as production of pro-inflammatory cytokines in BALF and lung were determined by using murine model of non-atopic asthma. Blocking either neurotensin receptors or mu opioid receptors did not alter the potential of PK20 in reducing airway hyperresponsiveness. In studies of inflammatory cells, the beneficial effect of the entire peptide occurs to be mediated by the stimulation of neurotensin receptors. However, regarding cytokine and biochemical assays, pretreatment with both receptor antagonists resulted in a different effect on its activity depending on the parameter studied. To conclude, the activation of both the opioid and neurotensin receptors seems to be necessary to induce the full anti-inflammatory activity of the hybrid compound.


Subject(s)
Analgesics, Opioid , Neurotensin , Receptors, Neurotensin , Animals , Anti-Inflammatory Agents , Mice
6.
Glia ; 68(7): 1513-1530, 2020 07.
Article in English | MEDLINE | ID: mdl-32065429

ABSTRACT

The generation of fully functional oligodendrocytes, the myelinating cells of the central nervous system, is preceded by a complex maturational process. We previously showed that the timing of oligodendrocyte differentiation and rat brain myelination were altered by perinatal exposure to buprenorphine and methadone, opioid analogs used for the management of pregnant addicts. Those observations suggested the involvement of the µ-opioid receptor (MOR) and the nociceptin/orphanin FQ receptor (NOR). However, it remained to be determined if these receptors and their endogenous ligands could indeed control the timing of myelination under normal physiological conditions of brain development. We now found that the endogenous MOR ligand endomorphin-1 (EM-1) exerts a striking stimulatory action on cellular and morphological maturation of rat pre-oligodendrocytes, but unexpectedly, these effects appear to be restricted to the cells from the female pups. Critically, this stimulation is abolished by coincubation with the endogenous NOR ligand nociceptin. Furthermore, NOR antagonist treatment of 9-day-old female pups results in accelerated brain myelination. Interestingly, the lack of sex-dependent differences in developmental brain levels of EM-1 and nociceptin, or oligodendroglial expression of MOR and NOR, suggests that the observed sex-specific responses may be highly dependent on important intrinsic differences between the male and female oligodendrocytes. The discovery of a significant effect of EM-1 and nociceptin in the developing female oligodendrocytes and brain myelination, underscores the need for further studies investigating brain sex-related differences and their implications in opioid use and abuse, pain control, and susceptibility and remyelinating capacity in demyelinating disease as multiple sclerosis.


Subject(s)
Brain/metabolism , Oligodendroglia/metabolism , Opioid Peptides/metabolism , Sex Factors , Analgesics, Opioid/metabolism , Analgesics, Opioid/pharmacology , Animals , Brain/growth & development , Rats, Sprague-Dawley , Receptors, Opioid/metabolism , Receptors, Opioid, mu/metabolism
7.
Bioorg Med Chem ; 28(9): 115438, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32199689

ABSTRACT

Endomorphin (EM)-1 and EM-2 are the most effective endogenous analgesics with efficient separation of analgesia from the risk of adverse effects. Poor metabolic stability and ineffective analgesia after peripheral administration were detrimental for the use of EMs as novel clinical analgesics. Therefore, here, we aimed to establish new EM analogs via introducing different bifunctional d-amino acids at position 2 of [(2-furyl)Map4]EMs. The combination of [(2-furyl)Map4]EMs with D-Arg2 or D-Cit2 yielded analogs with enhanced binding affinity to the µ-opioid receptor (MOR) and increased stability against enzymatic degradation (t1/2 > 300 min). However, the agonistic activities of these analogs toward MOR were slightly reduced. Similar to morphine, peripheral administration of the analog [D-Cit2, (2-furyl)Map4]EM-1 (10) significantly inhibited the pain behavior of mice in multiple pain models. In addition, this EM-1 analog was associated with reduced tolerance, less effect on gastrointestinal mobility, and no significant motor impairment. Compared to natural EMs, the EM analogs synthesized herein had enhanced metabolic stability, bioavailability, and analgesic properties.


Subject(s)
Analgesics, Opioid/pharmacology , Drug Design , Oligopeptides/pharmacology , Pain/drug therapy , Receptors, Opioid, mu/agonists , Animals , Dose-Response Relationship, Drug , Formaldehyde , Male , Mice , Mice, Inbred Strains , Molecular Structure , Pain/chemically induced , Pain Measurement , Structure-Activity Relationship
8.
Pain Med ; 21(5): 992-1004, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31165885

ABSTRACT

OBJECTIVE: Although mu-opioid receptor agonists have been the mainstay of analgesic regimens for moderate to severe pain, they are associated with serious side effects, risks, and limitations. We evaluate the most serious risks associated with conventional opioids and compare these with the pharmacology of CYT-1010, a prototypical endomorphin and mu-opioid receptor agonist. RESULTS: Addiction and respiratory depression are serious risks of traditional mu-opioid analgesics. Mitigation strategies have been inadequate at addressing the opioid crisis and may interfere with the effective treatment of pain. Improved understanding of mu-opioid receptor biology and the discovery in 1997 of an additional and unique family of endogenous opioid peptides (endomorphins) have provided a pathway for dissociating analgesia from opioid-related adverse events and developing new classes of mu-opioid receptor agonists that use biased signaling and/or target novel sites to produce analgesia with reduced side effect liability. Endomorphin-1 and -2 are endogenous opioid peptides highly selective for mu-opioid receptors that exhibit potent analgesia with reduced side effects. CYT-1010 is a cyclized, D-lysine-containing analog of endomorphin-1 with a novel mechanism of action targeting traditional mu- and exon 11/truncated mu-opioid receptor 6TM variants. CYT-1010 preclinical data have demonstrated reduced abuse potential and analgesic potency exceeding that of morphine. In an initial phase 1 clinical study, CYT-1010 demonstrated significant analgesia vs baseline and no respiratory depression at the dose levels tested. CONCLUSIONS: CYT-1010 and other novel mu-opioid receptor agonists in clinical development are promising alternatives to conventional opioids that may offer the possibility of safer treatment of moderate to severe pain.


Subject(s)
Receptors, Opioid, mu , Respiratory Insufficiency , Analgesics/therapeutic use , Analgesics, Opioid/therapeutic use , Humans , Morphine/therapeutic use , Pain/drug therapy , Receptors, Opioid, mu/therapeutic use , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/drug therapy
9.
Int J Mol Sci ; 21(23)2020 Nov 25.
Article in English | MEDLINE | ID: mdl-33255594

ABSTRACT

The opioid-induced analgesia is associated with a number of side effects such as addiction, tolerance and respiratory depression. The involvement of neuropeptide FF (NPFF) in modulation of pain perception, opioid-induced tolerance and dependence was well documented in contrast to respiratory depression. Therefore, the aim of the present study was to examine the potency of NPFF to block post-opioid respiratory depression, one of the main adverse effects of opioid therapy. Urethane-chloralose anaesthetized Wistar rats were injected either intravenously (iv) or intracerebroventricularly (icv) with various doses of NPFF prior to iv endomorphin-1 (EM-1) administration. Iv NPFF diminished the number of EM-1-induced apneas without affecting their length and without influence on the EM-1 induced blood pressure decline. Icv pretreatment with NPFF abolished the occurrence of post-EM-1 apneas and reduced also the maximal drop in blood pressure and heart rate. These effects were completely blocked by the NPFF receptor antagonist RF9, which was given as a mixture with NPFF before systemic EM-1 administration. In conclusion, our results showed that centrally administered neuropeptide FF is effective in preventing apnea evoked by stimulation of µ-opioid receptors and the effect was due to activation of central NPFF receptors. Our finding indicates a potential target for reversal of opioid-induced respiratory depression.


Subject(s)
Apnea/drug therapy , Cardiovascular Diseases/drug therapy , Oligopeptides/pharmacology , Receptors, Opioid, mu/genetics , Analgesia/adverse effects , Analgesics, Opioid/adverse effects , Animals , Apnea/chemically induced , Apnea/genetics , Apnea/pathology , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/genetics , Disease Models, Animal , Humans , Infusions, Intraventricular , Oligopeptides/adverse effects , Oligopeptides/genetics , Pain Perception/drug effects , Rats , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Neuropeptide/genetics , Receptors, Opioid, mu/antagonists & inhibitors , Transcriptional Activation/drug effects
10.
J Neuroinflammation ; 16(1): 100, 2019 May 21.
Article in English | MEDLINE | ID: mdl-31109346

ABSTRACT

BACKGROUND: Numerous studies have identified the proinflammatory, pronociceptive effects of morphine which ultimately exacerbate pain. Our novel endomorphin analog ZH853 does not produce proinflammatory effects on its own and gives potent, long-lasting analgesia. This study investigates whether ZH853's lack of interaction with the neuroimmune system reduces the risk of prolonged pain. METHODS: Adult male Sprague-Dawley rats were subjected to one of two treatment paradigms. Either (1) chronic pain followed by chronic treatment with morphine, ZH853 or vehicle, or (2) chronic drug administered prior to pain induction. Complete Freund's adjuvant (CFA) was injected or paw incision surgery was performed on the left hind plantar foot pad. Drugs were administered through Alzet osmotic minipumps at a rate of 1 µl/h for 5 days at appropriate doses based on prior experiments. Animals were tested for mechanical allodynia and thermal hyperalgesia using von Frey filaments and the Hargreaves apparatus, respectively. Additionally, several gait parameters were measured using the CatWalk XT. When all animals had recovered from pain, 1 mg/kg of naltrexone was administered to test for development of latent sensitization (LS). A second set of animals was used to investigate dorsal horn inflammation following CFA and drug treatment. ANOVAs were used to assess differences between drug treatment groups. RESULTS: As expected, morphine increased and prolonged pain in all experiments compared to vehicle treatment. However, ZH853 treatment reduced the overall time spent in pain and the severity of pain scores compared to morphine. ZH853 not only reduced inflammation versus morphine treatment but also, in some instances, acted as an anti-inflammatory drug compared to vehicle treatment. Finally, ZH853 prevented the development of LS while vehicle- and morphine-treated animals showed robust relapse to pain. CONCLUSIONS: ZH853 has a favorable side effect profile versus morphine and provides superior analgesia in a number of pain states. We now know that chronic use of this compound reduces time spent in a chronic pain state, the opposite of common opioids like morphine, and reduces the risk of LS, making ZH853 an excellent candidate for clinical development in humans for inflammatory and postoperative pain.


Subject(s)
Analgesics, Opioid/therapeutic use , Analgesics/therapeutic use , Immunomodulation/drug effects , Morphine/therapeutic use , Pain, Postoperative/drug therapy , Peptides, Cyclic/therapeutic use , Analgesics/pharmacology , Analgesics, Opioid/pharmacology , Animals , Immunomodulation/physiology , Inflammation/drug therapy , Inflammation/immunology , Male , Morphine/pharmacology , Pain Measurement/drug effects , Pain Measurement/methods , Pain, Postoperative/immunology , Peptides, Cyclic/pharmacology , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Recovery of Function/physiology
11.
Bioorg Med Chem ; 27(4): 630-643, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30626554

ABSTRACT

It is well known that opioid analgesics produce side effects including tolerance and constipation. Since neuropeptide FF (NPFF) receptor antagonists reversed opioid-induced hyperalgesia and analgesic tolerance, the present work was performed to synthetize two branched peptidomimetics, EKR and RKE, containing the opioid peptide endomorphin-2 (EM-2) and the NPFF receptor antagonist RF9. Our data obtained from the in vitro cyclic adenosine monophosphate experiment demonstrated that EKR functioned as a mixed mu-, delta-opioid receptors agonist and NPFF1 receptor antagonist/NPFF2 receptor partial agonist, whereas RKE acted as a multi-functional peptidomimetic with the mu-opioid agonism and the NPFF1 antagonism/NPFF2 partial agonism. Furthermore, EKR and RKE completely blocked the NPFF2 receptor-mediated neurite outgrowth of Neuro 2A cells. In vivo antinociception studies found that supraspinal administration of EKR and RKE dose-dependently produced potent antinociception via the mu-opioid receptor in the tail-flick test. In carrageenan inflammatory pain model, spinal administration of EKR and RKE induced dose-related analgesia, which was significantly reduced by the opioid antagonist naloxone and the NPFF antagonist RF9. Notably, compared with morphine, intracerebroventricular repeated administration of EKR and RKE maintained prolonged antinociceptive effectiveness. In addition, at the antinociceptive doses, these two branched peptidomimetics did not significantly inhibit gastrointestinal transit. Taken together, the present work suggest that EKR and RKE behave as multi-functional ligands with the opioid agonism and the NPFF1 antagonism/NPFF2 partial agonism, and produce prolonged antinociception with limited side effects. Moreover, our results imply that EKR and RKE might be interesting pharmacological tools for further investigating the biological function of the NPFF and opioid systems.


Subject(s)
Analgesics, Opioid/pharmacology , Drug Discovery , Peptidomimetics/pharmacology , Analgesics, Opioid/chemical synthesis , Analgesics, Opioid/therapeutic use , Animals , Cell Line, Tumor , Drug Partial Agonism , Gastrointestinal Transit/drug effects , HEK293 Cells , Humans , Hyperalgesia/drug therapy , Male , Mice , Morphine/pharmacology , Naloxone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Neuronal Outgrowth/drug effects , Peptidomimetics/chemical synthesis , Peptidomimetics/therapeutic use , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism
12.
Chem Pharm Bull (Tokyo) ; 67(9): 977-984, 2019.
Article in English | MEDLINE | ID: mdl-31474737

ABSTRACT

Endomorphin-1 (Tyr-Pro-Trp-Phe-NH2, EM-1), an endogenous µ-opioid receptor ligand with strong antinociceptive activity, is not in clinical use because of its limited metabolic stability and membrane permeability. In this study, we develop a short-peptide self-delivery system for brain targets with the capability to deliver EM-1 without vehicle. Two amphiphilic EM-1 derivatives, C18-SS-EM1 and C18-CONH-EM1, were synthesized by attaching a stearyl moiety to EM-1 via a disulfide and amide bond, respectively. The amphiphilicity of EM-1 derivatives enabled self-assembling into nanoparticles for brain delivery. The study assessed morphology, circular dichroism, and metabolic stability of the formulations, as well as their pharmacodynamics and in vivo distribution, directly monitored by near-IR fluorescence imaging in mouse brains. In aqueous solution, the C18-SS-EM1 derivative self-assembled into spherical nanostructures with a diameter of 10-20 nm. Near-IR fluorescence analysis visualized the accumulation of the peptides in the brain. Importantly, the analgesic effect of C18-SS-EM1 nanoparticles was significantly stronger as compared to that of unmodified EM-1 or C18-CONH-EM1 nanoparticles. An in vitro release study demonstrated that self-assembled C18-SS-EM1 nanoparticles possessed reduction-responsive behavior. In summary, self-assembling C18-SS-EM1 nanoparticles, which integrate the advantages of lipidization, nanoscale characteristics and, labile disulfide bonds, represent a promising strategy for brain delivery of short peptides.


Subject(s)
Brain/metabolism , Nanomedicine , Oligopeptides/pharmacokinetics , Animals , Blood-Brain Barrier/metabolism , Brain/diagnostic imaging , Circular Dichroism , Drug Carriers/chemistry , Half-Life , Male , Mice , Nanoparticles/chemistry , Oligopeptides/blood , Oligopeptides/chemistry , Spectroscopy, Near-Infrared
13.
Bull Exp Biol Med ; 168(1): 55-57, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31741246

ABSTRACT

We studied the effect of endomorphin-1 on isolated mesenteric lymphatic vessels in rats. It was found that endomorphin-1 caused a dose-dependent increase in the contractile activity of lymphangions, which was associated with stimulation of intracellular calcium depots. The observed effect of endomorphin-1 in isolated lymphatic vessels has a complex mechanism; it depends on the concentration of the applied peptide and is probably determined by its interaction with non-opioid receptors.


Subject(s)
Calcium/metabolism , Lymphatic Vessels/drug effects , Lymphatic Vessels/metabolism , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Oligopeptides/pharmacology , Receptors, Opioid, mu/metabolism , Animals , Male , Rats
14.
J Neurosci ; 37(46): 11181-11191, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29025923

ABSTRACT

We previously showed that intrathecal application of endomorphin 2 [EM2; the highly specific endogenous µ-opioid receptor (MOR) ligand] induces antinociception that varies with stage of the rat estrous cycle: minimal during diestrus and prominent during proestrus. Earlier studies, however, did not identify proestrus-activated signaling strategies that enable spinal EM2 antinociception. We now report that in female rats, increased spinal dynorphin release and κ-opioid receptor (KOR) signaling, as well as the emergence of glutamate-activated metabotropic glutamate receptor 1 (mGluR1) signaling, are critical to the transition from an EM2 nonresponsive state (during diestrus) to an analgesically responsive state (during proestrus). Differential signaling by mGluR1, depending on its activation by membrane estrogen receptor α (mERα; during diestrus) versus glutamate (during proestrus), concomitant with the ebb and flow of spinal dynorphin/KOR signaling, functions as a switch, preventing or promoting, respectively, spinal EM2 antinociception. Importantly, EM2 and glutamate-containing varicosities appose spinal neurons that express MOR along with mGluRs and mERα, suggesting that signaling mechanisms regulating analgesic effectiveness of intrathecally applied EM2 also pertain to endogenous EM2. Regulation of spinal EM2 antinociception by both the nature of the endogenous mGluR1 activator (i.e., endogenous biased agonism at mGluR1) and changes in spinal dynorphin/KOR signaling represent a novel mechanism for modulating analgesic responsiveness to endogenous EM2 (and perhaps other opioids). This points the way for developing noncanonical pharmacological approaches to pain management by harnessing endogenous opioids for pain relief.SIGNIFICANCE STATEMENT The current prescription opioid abuse epidemic underscores the urgency to develop alternative pharmacotherapies for managing pain. We find that the magnitude of spinal endomorphin 2 (EM2) antinociception not only varies with stage of reproductive cycle, but is also differentially regulated during diestrus and proestrus. This finding highlights the need for sex-specific and cycle-specific approaches to pain management. Additionally, our finding that spinal EM2 antinociception in female rats is regulated by both the ebb and flow of spinal dynorphin/κ-opioid receptor signaling over the estrous cycle, as well as the nature of the endogenous mGluR1 activator, could encourage noncanonical pharmacological approaches to pain management, such as harnessing endogenous opioids for pain relief.


Subject(s)
Estrogen Receptor alpha/metabolism , Estrous Cycle/metabolism , Oligopeptides/metabolism , Receptors, Metabotropic Glutamate/metabolism , Receptors, Opioid, kappa/metabolism , Spinal Cord/metabolism , Animals , Estrogen Receptor alpha/agonists , Female , Neuronal Plasticity/physiology , Nociception/physiology , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/agonists , Receptors, Opioid, kappa/agonists , Reproduction/physiology , Signal Transduction/physiology
15.
J Neuroinflammation ; 15(1): 320, 2018 Nov 15.
Article in English | MEDLINE | ID: mdl-30442166

ABSTRACT

BACKGROUND: Preemptive administration of analgesic drugs reduces perceived pain and prolongs duration of antinociceptive action. Whereas several lines of evidence suggest that endomorphins, the endogenous mu-opioid agonists, attenuate acute and chronic pain at the spinal level, their preemptive analgesic effects remain to be determined. In this study, we evaluated the anti-allodynic activities of endomorphins and explored their mechanisms of action after preemptive administration in a mouse model of inflammatory pain. METHODS: The anti-allodynic activities of preemptive intrathecal administration of endomorphin-1 and endomorphin-2 were investigated in complete Freund's adjuvant (CFA)-induced inflammatory pain model and paw incision-induced postoperative pain model. The modulating effects of endomorphins on the expression of p38 mitogen-activated protein kinase (p38 MAPK) and inflammatory mediators in dorsal root ganglion (DRG) of CFA-treated mice were assayed by real-time reverse transcription-polymerase chain reaction (RT-PCR), Western blotting, or immunofluorescence staining. RESULTS: Preemptive intrathecal injection of endomorphins dose-dependently attenuated CFA-induced mechanical allodynia via the mu-opioid receptor and significantly reversed paw incision-induced allodynia. In addition, CFA-caused increase of phosphorylated p38 MAPK in DRG was dramatically reduced by preemptive administration of endomorphins. Repeated intrathecal application of the specific p38 MAPK inhibitor SB203580 reduced CFA-induced mechanical allodynia as well. Further RT-PCR assay showed that endomorphins regulated the mRNA expression of inflammatory cytokines in DRGs induced by peripheral inflammation. CONCLUSIONS: Our findings reveal a novel mechanism by which preemptive treatment of endomorphins attenuates inflammatory pain through regulating the production of inflammatory cytokines in DRG neurons via inhibition of p38 MAPK phosphorylation.


Subject(s)
Analgesics, Opioid/therapeutic use , Oligopeptides/therapeutic use , Pain/drug therapy , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Disease Models, Animal , Freund's Adjuvant , Ganglia, Spinal/cytology , Gene Expression Regulation/drug effects , Inflammation/chemically induced , Inflammation/complications , Injections, Spinal , Male , Mice , Neurons/drug effects , Opioid Peptides , Pain/etiology , Pain/pathology , Pain Threshold , Signal Transduction/drug effects , Time Factors
16.
Neurosignals ; 26(1): 43-57, 2018.
Article in English | MEDLINE | ID: mdl-29554653

ABSTRACT

BACKGROUND/AIMS: Spinal dorsal horn (SDH) is one of the most important regions for analgesia produced by endomorphin-2 (EM2), which has a higher affinity and specificity for the µ-opioid receptor (MOR) than morphine. Many studies have focused on substantia gelatinosa (SG, lamina II) neurons to elucidate the cellular basis for its antinociceptive effects. However, the complicated types and local circuits of interneurons in the SG make it difficult to understand the real effects of EM2. Therefore, in the present study, we examined the effects of EM2 on projection neurons (PNs) in lamina I. METHODS: Tracing, immunofluoresence, and immunoelectron methods were used to examine the morphological connections between EM2-immunoreactive (-ir) terminals and PNs. By using in vitro whole cell patch clamp recording technique, we investigated the functional effects of EM2 on PNs. RESULTS: EM2-ir afferent terminals directly contacted PNs projecting to the parabrachial nucleus in lamina I. Their synaptic connections were further confirmed by immunoelectron microscopy, most of which were asymmetric synapses. It was found that EM2 had a strong inhibitory effect on the frequency, but not amplitude, of the spontaneous excitatory postsynaptic current (sEPSC) of the spinoparabrachial PNs in lamina I, which could be reversed by MOR antagonist CTOP. However, their spontaneous inhibitory postsynaptic current (sIPSC) and intrinsic properties were not changed after EM2 application. CONCLUSION: Applying EM2 to the SDH could produce analgesia through inhibiting the activities of the spinoparabrachial PNs in lamina I by reducing presynaptic neurotransmitters release from the primary afferent terminals.

17.
Amino Acids ; 49(10): 1719-1731, 2017 10.
Article in English | MEDLINE | ID: mdl-28752396

ABSTRACT

In this study semisolid formulations containing AcYPFF (N-acetyl-Tyr-Pro-Phe-Phe-NH2) tetrapeptide were obtained and characterized in terms of rheology, stability by multiple light scattering and particle size distribution by laser diffraction. Additionally, the release studies of tetrapeptide from formulations obtained were performed. The influence of different factors such as semisolid and membrane type on tetrapeptide release rate was examined. The release experiments of tetrapeptide modified with palmitoyl group (PalmYPFF) were also carried out. The results proved that formulation type and its rheological properties strongly determined the permeation process of the tetrapeptide. The fastest release of tetrapeptide was observed from hydrogel that had the lowest viscosity. The kinetic data of tetrapeptide released from oil-in-water (o/w) and water-in-oil (w/o) emulsions prepared at elevated temperature showed good fit to the Higuchi equation, whereas when AcYPFF was released from oil-in-water (o/w) emulsion prepared with the addition of auto-emulsifier high linearity with Korsmeyer-Peppas model was observed. While when tetrapeptide was released from Hydrogel the most suitable model was the first-order kinetics. It was suggested that mechanism that led to the release of tetrapeptide from all formulations was non-Fickian diffusion transport. The presence of palmitoyl group changed the solubility of tetrapeptide both in formulation and receptor fluid and thus the release rate of active compound was modified.


Subject(s)
Oligopeptides/chemistry , Oligopeptides/pharmacokinetics , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics
18.
J Exp Biol ; 220(Pt 18): 3381-3390, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28931721

ABSTRACT

Trichoplax adhaerens is a flat, millimeter-sized marine animal that adheres to surfaces and grazes on algae. Trichoplax displays a repertoire of different feeding behaviors despite the apparent absence of a true nervous system with electrical or chemical synapses. It glides along surfaces to find food, propelled by beating cilia on cells at its ventral surface, and pauses during feeding by arresting ciliary beating. We found that when endomorphin-like peptides are applied to an animal, ciliary beating is arrested, mimicking natural feeding pauses. Antibodies against these neuropeptides label cells that express the neurosecretory proteins and voltage-gated calcium channels implicated in regulated secretion. These cells are embedded in the ventral epithelium, where they comprise only 4% of the total, and are concentrated around the edge of the animal. Each bears a cilium likely to be chemosensory and used to detect algae. Trichoplax pausing during feeding or spontaneously in the absence of food often induce their neighbors to pause as well, even neighbors not in direct contact. Pausing behavior propagates from animal to animal across distances much greater than the signal that diffuses from just one animal, so we presume that the peptides secreted from one animal elicit secretion from nearby animals. Signal amplification by peptide-induced peptide secretion explains how a small number of sensory secretory cells lacking processes and synapses can evoke a wave of peptide secretion across the entire animal to globally arrest ciliary beating and allow pausing during feeding.


Subject(s)
Neuropeptides/genetics , Placozoa/physiology , Animals , Epithelium , Feeding Behavior , Neuropeptides/metabolism , Placozoa/genetics , Sequence Analysis, DNA
19.
Bioorg Med Chem Lett ; 27(7): 1557-1560, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28256374

ABSTRACT

A new class of endomorphin-1 analogues was synthesized by combining successful chemical modifications including N-terminal guanidino modification, Phe4 was chlorinated, D-Ala-Gly Substituted L-Pro2. Their bioactivities were measured by radioligand binding assay, metabolic stability and the tail-flick test. In radioligand binding assays, analogue GAGPC (Nα-Amidino-Tyr-D-Ala-Gly-Trp-p-Cl-Phe-NH2), shown a µ-opioid receptor affinity about 1.42-fold higher and a 2.51-fold higher δ-opioid receptor affinity than EM-1. In the metabolic stability assays, GAGPC had the longest half-lives which was 284min and 53-fold higher than that of EM-1. In the tail-flick test in mice, GAGPC chloride modification increases the lipid content of the drug, thus increases the permeability of the blood brain barrier, and has a higher analgesic activity. It might be of importance in potential application as drug candidates as analgesic.


Subject(s)
Analgesics, Opioid/pharmacology , Oligopeptides/pharmacology , Analgesics, Opioid/chemical synthesis , Analgesics, Opioid/metabolism , Analgesics, Opioid/pharmacokinetics , Animals , Blood-Brain Barrier/metabolism , Cell Membrane/metabolism , Half-Life , Male , Mice , Naloxone/administration & dosage , Naloxone/pharmacology , Oligopeptides/chemical synthesis , Oligopeptides/metabolism , Oligopeptides/pharmacokinetics , Radioligand Assay , Rats, Wistar , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism
20.
Bioorg Med Chem Lett ; 27(10): 2119-2123, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28377055

ABSTRACT

To search a novel analgesic characterizes the effects on human sperm motility as minimal as possible. A new class of endomorphin-1 (EM-1) analogues was synthesized by combining successful chemical modifications including N-terminal guanidino modification, Phe4 was chlorinated, replaced of l-Pro2-Trp3 by d-Ala2-Gly3 or d-Pro2-Gly3 at position 2 and 3. Their bioactivities were measured by radioligand binding assay, metabolic stability, antinociception activity and sperm motility effects. In radioligand binding assays, analogue GAGP shown a µ-opioid receptor affinity about 17.7-fold higher and a 57.3-fold higher δ-opioid receptor affinity than EM-1. In the metabolic stability assays, GAGP had the longest half-lives and 16.6-fold higher than EM-1. In the tail-flick test in mice, GAGP showed the best analgesia. In sperm motility assays, the group of GAGP (10-5, 10-7mol/L) decreased of the percentage of a+b grade, and no significant when compared with initial value. In GAGP (10-6mol/L) group, sperm motility was progressively increased, although it was not statistically significant. But at the groups of morphine (10-7mol/L) and GAGD (10-7mol/L), these caused significant reduction between 0 and 90 min. We found that analogues GAGP, activating µ-opioid receptor and partial δ-opioid receptor, exhibit good analgesic effects with minimal implications for human sperm motility. It might be important in potential application as drug candidates of analgesic without implications for human sperm motility.


Subject(s)
Analgesics, Opioid/chemistry , Oligopeptides/chemistry , Amino Acid Sequence , Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/toxicity , Animals , Half-Life , Humans , Mice , Oligopeptides/pharmacokinetics , Oligopeptides/toxicity , Receptors, Opioid, delta/chemistry , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/metabolism , Sperm Motility/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL