Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 762
Filter
1.
Mol Neurobiol ; 58(11): 5459-5472, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34331656

ABSTRACT

Corticotropin-releasing factor (CRF) orchestrates our body's response to stressful stimuli. Pain is often stressful and counterbalanced by activation of CRF receptors along the nociceptive pathway, although the involvement of the CRF receptor subtypes 1 and/or 2 (CRF-R1 and CRF-R2, respectively) in CRF-induced analgesia remains controversial. Thus, the aim of the present study was to examine CRF-R1 and CRF-R2 expression within the spinal cord of rats with Freund's complete adjuvant-induced unilateral inflammation of the hind paw using reverse transcriptase polymerase chain reaction, Western blot, radioligand binding, and immunofluorescence confocal analysis. Moreover, the antinociceptive effects of intrathecal (i.t.) CRF were measured by paw pressure algesiometer and their possible antagonism by selective antagonists for CRF-R1 and/or CRF-R2 as well as for opioid receptors. Our results demonstrated a preference for the expression of CRF-R2 over CRF-R1 mRNA, protein, binding sites and immunoreactivity in the dorsal horn of the rat spinal cord. Consistently, CRF as well as CRF-R2 agonists elicited potent dose-dependent antinociceptive effects which were antagonized by the i.t. CRF-R2 selective antagonist K41498, but not by the CRF-R1 selective antagonist NBI35965. In addition, i.t. applied opioid antagonist naloxone dose-dependently abolished the i.t. CRF- as well as CRF-R2 agonist-elicited inhibition of somatic pain. Importantly, double immunofluorescence confocal microscopy of the spinal dorsal horn showed CRF-R2 on enkephalin (ENK)-containing inhibitory interneurons in close opposition of incoming mu-opioid receptor-immunoreactive nociceptive neurons. CRF-R2 was, however, not seen on pre- or on postsynaptic sensory neurons of the spinal cord. Taken together, these findings suggest that i.t. CRF or CRF-R2 agonists inhibit somatic inflammatory pain predominantly through CRF-R2 receptors located on spinal enkephalinergic inhibitory interneurons which finally results in endogenous opioid-mediated pain inhibition.


Subject(s)
Pain/physiopathology , Receptors, Corticotropin-Releasing Hormone/physiology , Spinal Cord/chemistry , Acenaphthenes/pharmacology , Amphibian Proteins/pharmacology , Animals , Arthritis, Experimental/physiopathology , Corticotropin-Releasing Hormone/pharmacology , Enkephalins/physiology , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Interneurons/physiology , Male , Naloxone/pharmacology , Nociception/physiology , Peptide Hormones/pharmacology , Posterior Horn Cells/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptors, Corticotropin-Releasing Hormone/biosynthesis , Receptors, Corticotropin-Releasing Hormone/genetics , Spinal Cord/physiopathology , Urocortins/pharmacology
2.
J Orthop Surg Res ; 15(1): 162, 2020 Apr 25.
Article in English | MEDLINE | ID: mdl-32334633

ABSTRACT

BACKGROUND: This article reports the effects of proenkephalin (PENK) on osteosarcoma (OS) cell migration. METHODS: A Gene Expression Omnibus (GEO) dataset was used to identify differentially expressed genes (DEGs) in OS tumor samples and normal human osteoblasts. Tumor tissue and adjacent normal tissue were collected from 40 OS patients. MG63 cells were transfected with si-PENK. Transwell migration assays and wound healing assays were performed to compare the effect of PENK on migration. Moreover, LY294002 was used to identify the potential mechanism. Gene expression was examined via qRT-PCR and Western blotting. RESULTS: Bioinformatic analysis revealed that PENK was downregulated in OS tumor samples compared with normal human osteoblasts. Moreover, PENK was identified as the hub gene of the DEGs. The PI3K/Akt signaling pathway was significantly enriched in the DEGs. Moreover, PENK was downregulated in OS and MG63 cells compared with the corresponding control cells. Silencing PENK promoted MG63 cell migration; however, treatment with LY294002 partially attenuated PENK silencing-induced OS cell migration. CONCLUSION: PENK inhibits OS cell migration by activating the PI3K/Akt signaling pathway.


Subject(s)
Bone Neoplasms/metabolism , Cell Movement , Enkephalins/physiology , Osteosarcoma/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Precursors/physiology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Adult , Blotting, Western , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement/physiology , Enkephalins/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Osteoblasts/metabolism , Osteosarcoma/pathology , Protein Precursors/metabolism , Real-Time Polymerase Chain Reaction , Young Adult
3.
Vitam Horm ; 111: 105-129, 2019.
Article in English | MEDLINE | ID: mdl-31421697

ABSTRACT

After millennia of knowledge of opium, it was only recently that endogenous substances called opioids with similar properties to opium and derivatives were discovered. The first to be discovered were enkephalins. In addition to the regulation of their synthesis and expression of receptors, an important mechanism for the regulation of their functions carried out by multiple proteolytic enzymes acting at all levels of their structure is described. The action of such enzymes, known as enkephalinases, is also regulated by endogenous and exogenous factors which ultimately affect the control of the enkephalins's action. For therapeutic purposes, it is not only necessary to develop specific inhibitors but also to acquire a deep knowledge of the influence that such factors exert on their activities. This knowledge could help us to establish adapted therapeutic strategies in the treatment of pain or other processes in which enkephalinases are involved. In this chapter, some of these regulatory factors are discussed, such as regional and subcellular distribution, developmental changes, diurnal variations, hormonal influences, stress, dietary factors or interactions with other neurotransmitters.


Subject(s)
Neprilysin/metabolism , Animals , Brain/growth & development , Brain/ultrastructure , Brain Chemistry/physiology , Circadian Rhythm/physiology , Diet , Endocrine System/physiology , Enkephalins/physiology , Female , Homeostasis , Humans , Male , Neprilysin/analysis , Pain Management/methods , Stress, Psychological/enzymology , Subcellular Fractions/chemistry , Tissue Distribution
4.
Vitam Horm ; 111: 147-193, 2019.
Article in English | MEDLINE | ID: mdl-31421699

ABSTRACT

The pentapeptides methionine-enkephalin and leucine-enkephalin belong to the opioid family of peptides, and the non-opiate peptide adrenocorticotropin hormone (ACTH) to the melanocortin peptide family. Enkephalins/ACTH are derived from pro-enkephalin, pro-dynorphin or pro-opiomelanocortin precursors and, via opioid and melanocortin receptors, are responsible for many biological activities. Enkephalins exhibit the highest affinity for the δ receptor, followed by the µ and κ receptors, whereas ACTH binds to the five subtypes of melanocortin receptor, and is the only member of the melanocortin family of peptides that binds to the melanocortin-receptor 2 (ACTH receptor). Enkephalins/ACTH and their receptors exhibit a widespread anatomical distribution. Enkephalins are involved in analgesia, angiogenesis, blood pressure, embryonic development, emotional behavior, feeding, hypoxia, limbic system modulation, neuroprotection, peristalsis, and wound repair; as well as in hepatoprotective, motor, neuroendocrine and respiratory mechanisms. ACTH plays a role in acetylcholine release, aggressive behavior, blood pressure, bone maintenance, hyperalgesia, feeding, fever, grooming, learning, lipolysis, memory, nerve injury repair, neuroprotection, sexual behavior, sleep, social behavior, tissue growth and stimulates the synthesis and secretion of glucocorticoids. Enkephalins/ACTH are also involved in many pathologies. Enkephalins are implicated in alcoholism, cancer, colitis, depression, heart failure, Huntington's disease, influenza A virus infection, ischemia, multiple sclerosis, and stress. ACTH plays a role in Addison's disease, alcoholism, cancer, Cushing's disease, dermatitis, encephalitis, epilepsy, Graves' disease, Guillain-Barré syndrome, multiple sclerosis, podocytopathies, and stress. In this review, we provide an updated description of the enkephalinergic and ACTH systems.


Subject(s)
Adrenocorticotropic Hormone/physiology , Enkephalins/physiology , Nervous System Physiological Phenomena , Adrenocorticotropic Hormone/analysis , Adrenocorticotropic Hormone/genetics , Amino Acid Sequence , Animals , Enkephalins/analysis , Enkephalins/genetics , Humans , Nervous System , Protein Precursors/metabolism , Receptors, Corticotropin/physiology , Receptors, Opioid/physiology , Signal Transduction , Tissue Distribution
5.
Vitam Horm ; 111: 313-337, 2019.
Article in English | MEDLINE | ID: mdl-31421706

ABSTRACT

Endogenous opioids (enkephalins, endorphins and dynorphins) are small peptides that play a main role in pain perception and analgesia, as well as in alcohol (ethanol) reinforcement and reward. Alcohol reinforcement involves the ethanol-induced activation of the endogenous opioid system, a process that may augment the hedonic value and the reinforcing properties of the drug, which in turn increases substance consumption. Changes in opioidergic transmission may contribute to alcohol intoxication and to the neuroadaptive responses produced by the long-lasting exposure to ethanol. Opioidergic transmission may be altered by ethanol at distinct levels, including the expression of precursor mRNAs, biosynthesis and release of opioid peptides, as well as ligand binding to opioid receptors. In adult rats, ß-endorphinergic and enkephalinergic transmission, through activation of mu and delta opioid receptors, mediate ethanol reinforcement and high alcohol drinking behavior. Prenatal ethanol exposure (PEE) selectively modifies Methionine-enkephalin (Met-enk) content in several brain regions of infant and adolescent rats, particularly those of the reward circuits. In preweanling rats, Met-enk content is decreased in the ventral tegmental area but is increased in the prefrontal cortex and the nucleus accumbens and other brain areas, as a consequence of a short and moderate ethanol exposure during late gestation. PEE also increases Met-enk levels in the prefrontal cortex and other brain regions of 30-day-old adolescent rats. These findings suggest that mesocorticolimbic enkephalins are essential in ethanol reinforcement in offspring, as previously reported in adult rats.


Subject(s)
Enkephalins/analysis , Enkephalins/physiology , Ethanol/pharmacology , Prenatal Exposure Delayed Effects , Synaptic Transmission/drug effects , Alcohol Drinking , Animals , Brain/drug effects , Brain/metabolism , Brain Chemistry/drug effects , Dopamine/physiology , Ethanol/administration & dosage , Ethanol/adverse effects , Female , Nucleus Accumbens/metabolism , Opioid Peptides/metabolism , Prefrontal Cortex/metabolism , Pregnancy , Rats , Receptors, Opioid/metabolism , Reinforcement, Psychology , Ventral Tegmental Area/metabolism
6.
Peptides ; 107: 10-16, 2018 09.
Article in English | MEDLINE | ID: mdl-30040980

ABSTRACT

Kyotorphin is a unique biologically active neuropeptide (l-tyrosine-l-arginine), which is reported to have opioid-like analgesic actions through a release of Met-enkephalin from the brain slices. N-methyl-l-tyrosine-l-arginine (NMYR), an enzymatically stable mimetic of kyotorphin, successfully caused potent analgesic effects in thermal and mechanical nociception tests in mice when it was given through systemic routes. NMYR analgesia was abolished in µ-opioid receptor-deficient (MOP-KO) mice, and by intracerebroventricular (i.c.v.) injection of naloxone and of N-methyl l-leucine-l-arginine (NMLR), a kyotorphin receptor antagonist. In the Ca2+-mobilization assay using CHO cells expressing Gαqi5 and hMOPr or hDOPr, however, the addition of kyotorphin neither activated MOPr-mechanisms, nor affected the concentration-dependent activation of DAMGO- or Met-Enkephalin-induced MOPr activation, and Met-enkephalin-induced DOPr activation. NMYR-analgesia was significantly attenuated in preproenkephalin (PENK)- or proopioimelanocortin (POMC)-KO mice. The systemic administration of arginine, which is reported to elevate the level of endogenous kyotorphin selectively in midbrain and medulla oblongata, pain-related brain regions, caused significant analgesia, and the analgesia was reversed by i.c.v. injection of NMLR or naloxone. In addition, PENK- and POMC-KO mice also attenuated the arginine-induced analgesia. All these findings suggest that NMYR and arginine activate brain kyotorphin receptor in direct and indirect manner, respectively and both compounds indirectly cause the opioid-like analgesia through the action of endogenous opioid peptides.


Subject(s)
Arginine/pharmacology , Enkephalins/genetics , Neuropeptides/pharmacology , Pain/genetics , Pro-Opiomelanocortin/genetics , Protein Precursors/genetics , Analgesics/pharmacology , Animals , CHO Cells , Cricetulus , Enkephalins/physiology , Gene Knockout Techniques , Mice , Mice, Inbred C57BL , Pain/metabolism , Pain Management , Pro-Opiomelanocortin/physiology , Protein Precursors/physiology
7.
Anesthesiology ; 128(5): 967-983, 2018 05.
Article in English | MEDLINE | ID: mdl-29334500

ABSTRACT

BACKGROUND: The current study used recombinant herpes simplex virus type I to increase expression of µ-opiate receptors and the opioid ligand preproenkephalin in peripheral nerve fibers in a mouse model of neuropathic pain. It was predicted that viral vector delivery of a combination of genes encoding the µ-opioid receptor and preproenkephalin would attenuate neuropathic pain and enhance opioid analgesia. The behavioral effects would be paralleled by changes in response properties of primary afferent neurons. METHODS: Recombinant herpes simplex virus type 1 containing cDNA sequences of the µ-opioid receptor, human preproenkephalin, a combination, or Escherichia coli lacZ gene marker (as a control) was used to investigate the role of peripheral opioids in neuropathic pain behaviors. RESULTS: Inoculation with the µ-opioid receptor viral vector (n = 13) reversed mechanical allodynia and thermal hyperalgesia and produced leftward shifts in loperamide (ED50 = 0.6 ± 0.2 mg/kg vs. ED50 = 0.9 ± 0.2 mg/kg for control group, n = 8, means ± SD) and morphine dose-response curves (ED50 = 0.3 ± 0.5 mg/kg vs. ED50 = 1.1 ± 0.1 mg/kg for control group). In µ-opioid receptor viral vector inoculated C-fibers, heat-evoked responses (n = 12) and ongoing spontaneous activity (n = 18) were decreased after morphine application. Inoculation with both µ-opioid receptor and preproenkephalin viral vectors did not alter mechanical and thermal responses. CONCLUSIONS: Increasing primary afferent expression of opioid receptors can decrease neuropathic pain-associated behaviors and increase systemic opioid analgesia through inhibition of peripheral afferent fiber activity.


Subject(s)
Analgesics, Opioid/pharmacology , Enkephalins/physiology , Neuralgia/prevention & control , Neurons, Afferent/physiology , Receptors, Opioid, mu/physiology , Analgesia , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Herpesvirus 1, Human/genetics , Male , Mice , Morphine/pharmacology , Proto-Oncogene Proteins c-fos/analysis , Receptors, Opioid, mu/analysis
8.
Handb Exp Pharmacol ; 247: 53-70, 2018.
Article in English | MEDLINE | ID: mdl-27417433

ABSTRACT

Very few discoveries in the neurosciences have triggered clinical speculation and experimentation regarding the etiology of psychiatric illness to the same extent as that following identification of the opiate receptor(s) and subsequent isolation of endogenous morphine-like peptides. There is overwhelming evidence in animals and in human that opioids are involved in behaviorally relevant issues such as the modulation of pain, the response to stress, motivation, addiction, sexuality, food intake, etc., but our knowledge on the possible relation between opioids and mental illness is still very limited.These responses could be explored eitheir by using higlhy selective delta agonist or by emphasizing the effects of phasically secreted endogenous opioid peptides, enkephalin. Both approaches were investigated in particular through protection of enkephalin degradation by dual enkephalinase ihibitors DENKIs such as RB101, PL37 or PL265.


Subject(s)
Enkephalins/physiology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Neprilysin/antagonists & inhibitors , Receptors, Opioid, delta/drug effects , Animals , Humans , Mental Disorders/drug therapy , Mental Disorders/physiopathology , Pain/drug therapy , Pain/physiopathology
9.
Yakugaku Zasshi ; 137(4): 459-475, 2017.
Article in Japanese | MEDLINE | ID: mdl-28381725

ABSTRACT

In the beginning of the 1970s, only two chemical substances, acetylcholine and γ-aminobutyric acid (GABA), had been definitely established as neurotransmitters. Under such circumstances, I started my scientific career in Professor Masanori Otsuka's lab searching for the transmitter of primary sensory neurons. Until 1976, lines of evidence had accumulated indicating that the undecapeptide substance P could be released as a transmitter from primary afferent fibers into spinal synapses, although the substance P-mediated synaptic response had yet to be identified. Peripheral synapses could serve as a good model and thus, it was demonstrated in the prevertebral sympathetic ganglia by1985 that substance P released from axon collaterals of primary sensory neurons acts as the transmitter mediating non-cholinergic slow excitatory postsynaptic potential (EPSP). At that time, we also found that autonomic synapses were useful to uncover the transmitter role of the opioid peptide enkephalins, whose functions had been unknown since their discovery in 1975. Accordingly, enkephalins were found to serve a transmitter role in mediating presynaptic inhibition of cholinergic fast and non-cholinergic slow transmission in the prevertebral sympathetic ganglia. In 1990s, we attempted to devise a combined technique of brain slices and patch-clamp recordings. We applied it to study the regulatory mechanisms that operate around cerebellar GABAergic inhibitory synapses, because most of the studies then had centered on excitatory synapses and because inhibitory synapses are crucially involved in brain functions and disorders. Consequently, we discovered novel forms of heterosynaptic interactions, dual actions of a single transmitter, and receptor crosstalk, the details of which are described in this review.


Subject(s)
Neurotransmitter Agents/physiology , Sensory Receptor Cells/physiology , Synapses/physiology , Animals , Axons/metabolism , Enkephalins/physiology , Excitatory Postsynaptic Potentials/physiology , Ganglia, Sympathetic/physiology , Humans , Neurons, Afferent/metabolism , Neurons, Afferent/physiology , Patch-Clamp Techniques , Receptor Cross-Talk/physiology , Spinal Cord/cytology , Substance P/metabolism , Substance P/physiology , Synapses/metabolism , gamma-Aminobutyric Acid/physiology
10.
Neuroscience ; 326: 10-21, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27045594

ABSTRACT

The dorsal horn (DH) of the spinal cord contains a heterogenous population of neurons that process incoming sensory signals before information ascends to the brain. We have recently characterized calretinin-expressing (CR+) neurons in the DH and shown that they can be divided into excitatory and inhibitory subpopulations. The excitatory population receives high-frequency excitatory synaptic input and expresses delayed firing action potential discharge, whereas the inhibitory population receives weak excitatory drive and exhibits tonic or initial bursting discharge. Here, we characterize inhibitory synaptic input and neuromodulation in the two CR+ populations, in order to determine how each is regulated. We show that excitatory CR+ neurons receive mixed inhibition from GABAergic and glycinergic sources, whereas inhibitory CR+ neurons receive inhibition, which is dominated by glycine. Noradrenaline and serotonin produced robust outward currents in excitatory CR+ neurons, predicting an inhibitory action on these neurons, but neither neuromodulator produced a response in CR+ inhibitory neurons. In contrast, enkephalin (along with selective mu and delta opioid receptor agonists) produced outward currents in inhibitory CR+ neurons, consistent with an inhibitory action but did not affect the excitatory CR+ population. Our findings show that the pharmacology of inhibitory inputs and neuromodulator actions on CR+ cells, along with their excitatory inputs can define these two subpopulations further, and this could be exploited to modulate discrete aspects of sensory processing selectively in the DH.


Subject(s)
Calbindin 2/metabolism , Inhibitory Postsynaptic Potentials , Posterior Horn Cells/physiology , Synaptic Transmission , Animals , Enkephalins/administration & dosage , Enkephalins/physiology , Female , GABA-A Receptor Antagonists/administration & dosage , Glycine/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Miniature Postsynaptic Potentials , Norepinephrine/administration & dosage , Norepinephrine/physiology , Posterior Horn Cells/cytology , Posterior Horn Cells/drug effects , Posterior Horn Cells/metabolism , Receptors, GABA-A/physiology , Serotonin/administration & dosage , Serotonin/physiology , gamma-Aminobutyric Acid/physiology
11.
Clin Exp Dermatol ; 41(2): 124-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26011402

ABSTRACT

BACKGROUND: Psoriasis is a common chronic skin disease that can also affect the mucous membranes and joints. It is multifactorial in origin, occurring in genetically predisposed individuals, and triggered by various endogenous and exogenous factors. Proenkephalin (PENK) is an endogenous opioid polypeptide hormone that acts on specific opiate receptors found on nerve and mucosal cells, and on various cells in the immune system. PENK receptors are expressed on skin cells, and their activation can regulate keratinocyte and melanocyte activities. PENK expression has been found to be increased in keratinocytes in psoriatic skin, and together with its inflammatory basis, this suggests that PENK may be regulated by inflammatory stimuli. AIM: To assess the possible role of PENK in the pathogenesis of psoriasis and to assess if it is related to the severity of psoriatic lesions. METHODS: Serum and tissue PENK levels were estimated in 20 patients with psoriasis vulgaris, and compared with those of 20 healthy controls (HCs). RESULTS: PENK levels were found to be significantly increased both in serum and in psoriatic lesions in patients compared with HCs. No significant correlation was found between PENK levels and patient age, disease duration or disease severity (Psoriasis Area and Severity Index). CONCLUSION: Our results support the role of PENK in the aetiopathogenesis of psoriasis, and indicate that giving anti-PENK drugs in addition to current antipsoriatic therapies might be of value in treating this common chronic skin disease.


Subject(s)
Enkephalins/physiology , Protein Precursors/physiology , Psoriasis/metabolism , Adolescent , Adult , Aged , Case-Control Studies , Enkephalins/blood , Female , Humans , Male , Middle Aged , Protein Precursors/blood , Risk Factors , Severity of Illness Index , Skin/metabolism , Young Adult
12.
J Clin Invest ; 125(10): 3782-94, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26426077

ABSTRACT

Primary pain and touch sensory neurons not only detect internal and external sensory stimuli, but also receive inputs from other neurons. However, the neuronal derived inputs for primary neurons have not been systematically identified. Using a monosynaptic rabies viruses-based transneuronal tracing method combined with sensory-specific Cre-drivers, we found that sensory neurons receive intraganglion, intraspinal, and supraspinal inputs, the latter of which are mainly derived from the rostroventral medulla (RVM). The viral-traced central neurons were largely inhibitory but also consisted of some glutamatergic neurons in the spinal cord and serotonergic neurons in the RVM. The majority of RVM-derived descending inputs were dual GABAergic and enkephalinergic (opioidergic). These inputs projected through the dorsolateral funiculus and primarily innervated layers I, II, and V of the dorsal horn, where pain-sensory afferents terminate. Silencing or activation of the dual GABA/enkephalinergic RVM neurons in adult animals substantially increased or decreased behavioral sensitivity, respectively, to heat and mechanical stimuli. These results are consistent with the fact that both GABA and enkephalin can exert presynaptic inhibition of the sensory afferents. Taken together, this work provides a systematic view of and a set of tools for examining peri- and extrasynaptic regulations of pain-afferent transmission.


Subject(s)
Afferent Pathways/physiology , Efferent Pathways/physiology , Nerve Net/physiology , Nociception/physiology , Sensory Receptor Cells/physiology , Spinal Cord Dorsal Horn/cytology , Animals , Defective Viruses/physiology , Enkephalins/physiology , Forelimb/innervation , GABAergic Neurons/physiology , GABAergic Neurons/virology , Ganglia, Spinal/cytology , Hyperalgesia/physiopathology , Interneurons/physiology , Interneurons/virology , Nerve Tissue Proteins/analysis , Neural Conduction , Neurons, Afferent/physiology , Neurons, Afferent/virology , Neurons, Efferent/physiology , Neurons, Efferent/virology , Nociceptors/physiology , Posterior Horn Cells/physiology , Posterior Horn Cells/virology , Presynaptic Terminals/physiology , Rabies virus/physiology , Sensory Receptor Cells/classification , Sensory Receptor Cells/virology , Skin/innervation , Spinal Cord Dorsal Horn/physiology , Spinal Cord Dorsal Horn/ultrastructure , Virus Replication , gamma-Aminobutyric Acid/physiology
13.
Alcohol ; 48(7): 657-64, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25241055

ABSTRACT

Different alcohol drinking patterns, involving either small and frequent drinking bouts or large and long-lasting bouts, are found to differentially affect the risk for developing alcohol-related diseases, suggesting that they have different underlying mechanisms. Such mechanisms may involve orexigenic peptides known to stimulate alcohol intake through their actions in the hypothalamic paraventricular nucleus (PVN). These include orexin (OX), which is expressed in the perifornical lateral hypothalamus, and galanin (GAL) and enkephalin (ENK), which are expressed within as well as outside the PVN. To investigate the possibility that these peptides affect different aspects of consumption, a microstructural analysis of ethanol drinking behavior was performed in male, Sprague-Dawley rats trained to drink 7% ethanol and implanted with guide shafts aimed at the PVN. While housed in specialized cages containing computerized intake monitors (BioDAQ Laboratory Intake Monitoring System, Research Diets Inc., New Brunswick, NJ) that measure bouts of ethanol drinking, these rats were given PVN injections of OX (0.9 nmol), GAL (1.0 nmol), or the ENK analog D-Ala2-met-enkephalinamide (DALA) (14.2 nmol), as compared to saline vehicle. Results revealed clear differences between the effects of these peptides. While all 3 stimulated ethanol intake, they had distinct effects on patterns of drinking, with OX increasing the number of drinking bouts, GAL increasing the size of the drinking bouts, and DALA increasing both the size and duration of the bouts. In contrast, these peptides had little impact on water or food intake. These results support the idea that different peptides can increase ethanol consumption by promoting distinct aspects of the ethanol drinking response. The stimulatory effect of OX on drinking frequency may be related to its neuronally stimulatory properties, while the stimulatory effect of GAL and ENK on bout size and duration may reflect a suppressive effect of these neuronally inhibitory peptides on the satiety-controlling PVN.


Subject(s)
Alcohol Drinking/physiopathology , Enkephalin, Methionine/analogs & derivatives , Galanin/pharmacology , Intracellular Signaling Peptides and Proteins/pharmacology , Neuropeptides/pharmacology , Animals , Enkephalin, Methionine/pharmacology , Enkephalins/physiology , Galanin/physiology , Injections, Intraventricular , Intracellular Signaling Peptides and Proteins/physiology , Male , Neuropeptides/physiology , Orexins , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/physiopathology , Rats , Rats, Sprague-Dawley
14.
Anal Chem ; 86(15): 7806-12, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-24967837

ABSTRACT

Methionine-enkephalin (M-ENK) and leucine-enkephalin (L-ENK) are small endogenous opioid peptides that have been implicated in a wide variety of complex physiological functions, including nociception, reward processing, and motivation. However, our understanding of the role that these molecules play in modulating specific brain circuits remains limited, largely due to challenges in determining where, when, and how specific neuropeptides are released in tissue. Background-subtracted fast-scan cyclic voltammetry coupled with carbon-fiber microelectrodes has proven to be sensitive and selective for detecting rapidly fluctuating neurochemicals in vivo; however, many challenges exist for applying this approach to the detection of neuropeptides. We have developed and characterized a novel voltammetric waveform for the selective quantification of small tyrosine-containing peptides, such as the ENKs, with rapid temporal (subsecond) and precise spatial (10s of micrometers) resolution. We have established that the main contributor to the electrochemical signal inherent to M-ENK is tyrosine and that conventional waveforms provide poor peak resolution and lead to fouling of the electrode surface. By employing two distinct scan rates in each anodic sweep of this analyte-specific waveform, we have selectively distinguished M-ENK from common endogenous interfering agents, such as ascorbic acid, pH shifts, and even L-ENK. Finally, we have used this approach to simultaneously quantify catecholamine and M-ENK fluctuations in live tissue. This work provides a foundation for real-time measurements of endogenous ENK fluctuations in biological locations, and the underlying concept of using multiple scan rates is adaptable to the voltammetric detection of other tyrosine-containing neuropeptides.


Subject(s)
Enkephalins/physiology , Animals , Electrodes , In Vitro Techniques , Male , Rats , Rats, Sprague-Dawley
15.
Nihon Arukoru Yakubutsu Igakkai Zasshi ; 49(5): 227-37, 2014 Oct.
Article in Japanese | MEDLINE | ID: mdl-25651617

ABSTRACT

Nicotine (NIC) regulates various cellular functions acting on the nicotinic acetylcholine receptor (nAChR). And nAChR consists of ligand-gated cation channels with pentameric structure and composed of α and ß subunits. In the central nervous system, α 4 ß 2 and α 7 nAChRs are the most abundantly expressed as nAChR subtypes. There are several lines of evidence indicating that systemic administration of NIC elicits the release of endogenous opioids, such as, endorphins, enkephalins and dynorphins, in the brain. NIC exerts numerous acute effects, for example, antinociceptive effects and the activating effects of the hypothalamic-pituitary-adrenal (HPA) axis. In these effects, NIC-induced antinociception, but not HPA axis activation, was inhibited by opioid receptor antagonist, naloxone (NLX), and was also suppressed in morphine tolerated mice, indicating the participation of the endogenous opioid system in NIC-induced antinociception, but not HPA axis activation. Moreover, NIC-induced antinociception was antagonized by both α 4 ß 2 and α 7 nAChR antagonists, while NIC-induced HPA axis activation was antagonized by α 4 ß 2 nAChR antagonist, but not by α 7 nAChR antagonist. These results suggest that the endogenous opioid system may not be located on the downstream of α 4 ß 2 nAChR. On the other hand, NIC has substantial physical dependence liability. NLX elicits NIC withdrawal after repeated NIC administration evaluated by corticosterone increase as a withdrawal sign, and NLX-precipitated NIC withdrawal is inhibited by concomitant administration of other opioid receptor antagonist, naltrexone, indicating the participation of endogenous opioid system in the development of physical dependence on NIC. NLX-precipitated NIC withdrawal was also inhibited by concomitant administration of an α 7 nAChR antagonist, but not an α 4 ß 2 nAChR antagonist. Taken together, these findings suggest that the endogenous opioid system may be located on the downstream of α 7 nAChR and participates in the development of physical dependence on NIC.


Subject(s)
Nicotine/pharmacology , Opioid Peptides/physiology , Tobacco Use Disorder/genetics , alpha7 Nicotinic Acetylcholine Receptor/drug effects , alpha7 Nicotinic Acetylcholine Receptor/physiology , Analgesics , Animals , Brain/metabolism , Dynorphins/metabolism , Dynorphins/physiology , Endorphins/metabolism , Endorphins/physiology , Enkephalins/metabolism , Enkephalins/physiology , Humans , Hypothalamo-Hypophyseal System/drug effects , Mice , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nicotine/antagonists & inhibitors , Nociception/drug effects , Opioid Peptides/metabolism , Pituitary-Adrenal System/drug effects , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors
16.
J Clin Invest ; 123(12): 5334-41, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24231353

ABSTRACT

Negative affect is critical for conferring vulnerability to opiate addiction as reflected by the high comorbidity of opiate abuse with major depressive disorder (MDD). Rodent models implicate amygdala prodynorphin (Pdyn) as a mediator of negative affect; however, evidence of PDYN involvement in human negative affect is limited. Here, we found reduced PDYN mRNA expression in the postmortem human amygdala nucleus of the periamygdaloid cortex (PAC) in both heroin abusers and MDD subjects. Similar to humans, rats that chronically self-administered heroin had reduced Pdyn mRNA expression in the PAC at a time point associated with a negative affective state. Using the in vivo functional imaging technology DREAMM (DREADD-assisted metabolic mapping, where DREADD indicates designer receptors exclusively activated by designer drugs), we found that selective inhibition of Pdyn-expressing neurons in the rat PAC increased metabolic activity in the extended amygdala, which is a key substrate of the extrahypothalamic brain stress system. In parallel, PAC-specific Pdyn inhibition provoked negative affect-related physiological and behavioral changes. Altogether, our translational study supports a functional role for impaired Pdyn in the PAC in opiate abuse through activation of the stress and negative affect neurocircuitry implicated in addiction vulnerability.


Subject(s)
Amygdala/metabolism , Depressive Disorder, Major/metabolism , Enkephalins/physiology , Heroin Dependence/metabolism , Protein Precursors/physiology , Adult , Amygdala/chemistry , Amygdala/diagnostic imaging , Animals , Clozapine/analogs & derivatives , Clozapine/pharmacology , Corticosterone/blood , Depressive Disorder, Major/genetics , Designer Drugs/pharmacokinetics , Enkephalins/analysis , Enkephalins/biosynthesis , Enkephalins/deficiency , Enkephalins/genetics , Female , Fluorine Radioisotopes , Fluorodeoxyglucose F18 , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Heroin Dependence/genetics , Humans , Hungary , Limbic System/chemistry , Limbic System/diagnostic imaging , Limbic System/metabolism , Male , Middle Aged , Neuroimaging/methods , Neurons/metabolism , Positron-Emission Tomography/methods , Protein Precursors/analysis , Protein Precursors/biosynthesis , Protein Precursors/deficiency , Protein Precursors/genetics , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Radiopharmaceuticals , Rats , Rats, Long-Evans , Recombinant Fusion Proteins/metabolism , United States
17.
J Parkinsons Dis ; 3(3): 341-9, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23948996

ABSTRACT

BACKGROUND: In Parkinson's disease (PD), bradykinesia, or slowness of movement, only appears after a large striatal dopamine depletion. Compensatory mechanisms probably play a role in this delayed appearance of symptoms. OBJECTIVE: Our hypothesis is that the striatal direct and indirect pathways participate in these compensatory mechanisms. METHODS: We used the unilateral 6-hydroxydopamine (6-OHDA) rat model of PD and control animals. Four weeks after the lesion, the spontaneous locomotor activity of the rats was measured and then the animals were killed and their brain extracted. We quantified the mRNA expression of markers of the striatal direct and indirect pathways as well as the nigral expression of dopamine transporter (DAT) and tyrosine hydroxylase (TH) mRNA. We also carried out an immunohistochemistry for the striatal TH protein expression. RESULTS: As expected, the unilateral 6-OHDA rats presented a tendency to an ipsilateral head turning and a low locomotor velocity. In 6-OHDA rats only, we observed a significant and positive correlation between locomotor velocity and both D1-class dopamine receptor (D1R) (direct pathway) and enkephalin (ENK) (indirect pathway) mRNA in the lesioned striatum, as well as between D1R and ENK mRNA. CONCLUSIONS: Our results demonstrate a strong relationship between both direct and indirect pathways and spontaneous locomotor activity in the parkinsonian rat model. We suggest a synergy between both pathways which could play a role in compensatory mechanisms and may contribute to the delayed appearance of bradykinesia in PD.


Subject(s)
Gene Expression/genetics , Gene Expression/physiology , Motor Activity/physiology , Neostriatum/metabolism , Parkinson Disease, Secondary/genetics , Parkinson Disease, Secondary/physiopathology , Signal Transduction/genetics , Signal Transduction/physiology , Animals , Dopamine Plasma Membrane Transport Proteins/metabolism , Dynorphins/biosynthesis , Dynorphins/genetics , Enkephalins/biosynthesis , Enkephalins/genetics , Enkephalins/physiology , Female , Glutamate Decarboxylase/metabolism , Hydroxydopamines , Immunohistochemistry , In Situ Hybridization , Neural Pathways , Parkinson Disease, Secondary/chemically induced , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/physiology , Tyrosine 3-Monooxygenase/metabolism
18.
Neuropharmacology ; 73: 290-300, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23770261

ABSTRACT

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


Subject(s)
Enkephalins/physiology , Ethanol/pharmacology , Protein Precursors/physiology , Reward , beta-Endorphin/physiology , Animals , Choice Behavior/physiology , Conditioning, Psychological/drug effects , Dose-Response Relationship, Drug , Enkephalins/genetics , Ethanol/antagonists & inhibitors , Female , Mice , Mice, Knockout , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Protein Precursors/genetics , beta-Endorphin/genetics
19.
Fertil Steril ; 98(3): 617-625.e3, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22749218

ABSTRACT

OBJECTIVE: To verify the presence of protein precursor pro-enkephalin (PENK) and met-enkephalin in human spermatozoa and to characterize the effects of exogenous and endogenous enkephalins on sperm motility. DESIGN: We carried out expression assays for met-enkephalin and its protein precursor PENK by reverse transcription-polymerase chain reaction (RT-PCR), Western blot, and immunofluorescence techniques in sperm cells and motility analysis after incubation of semen samples with met-enkephlin enzyme inhibitors and the opioid receptor antagonist naloxone. Met-enkephalin secretion was analyzed by flow cytometry. SETTING: Assisted reproduction unit and academic research laboratory. PATIENT(S): Semen from 50 normozoospermic healthy human donors. INTERVENTION(S): Spermatozoa isolated from semen on discontinuous Percoll gradient (40%-80%) followed by a swim-up was used for all techniques. MAIN OUTCOME MEASURE(S): Immunoblotting blots, indirect immunofluorescence antibody assays, RT-PCR blots, flow cytometry plots, and percentage of motile sperm. RESULT(S): We found by RT-PCR and immunofluorescence that met-enkephalin and its protein precursor PENK are present in the head of human sperm cells. Endogenous met-enkephalin increased sperm motility, whereas the addition of exogenous met-enkephalin had a biphasic effect on motility, likely due to the activation of distinct receptor subtypes. CONCLUSION(S): We provide evidence for a new role of met-enkephalin as an endogenous mediator of sperm motility. This autocrine regulation of sperm function by the opioid system represents a new mechanism of regulation of male factor fertility and could be useful as an emerging target for male contraception.


Subject(s)
Enkephalin, Methionine/physiology , Sperm Motility , Adolescent , Adult , Enkephalin, Methionine/analysis , Enkephalin, Methionine/pharmacology , Enkephalins/analysis , Enkephalins/physiology , Fluorescent Antibody Technique , Humans , Male , Protein Precursors/analysis , Protein Precursors/physiology , Reverse Transcriptase Polymerase Chain Reaction , Sperm Motility/drug effects
20.
Synapse ; 65(11): 1196-203, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21638337

ABSTRACT

Dystonia is regarded as a basal ganglia disorder. In the dt(sz) hamster, a genetic animal model of paroxysmal dystonia, previous studies demonstrated a reduced density of striatal GABAergic interneurons which inhibit striatal GABAergic projection neurons. Although the disinhibition of striatal GABAergic projection neurons was evidenced in the dt(sz) hamster, alterations in their density have not been elucidated so far. Therefore, in the present study, the density of striatal methionin-(met-) enkephalin (ENK) immunoreactive GABAergic neurons, which project to the globus pallidus (indirect pathway), was determined in dt(sz) and control hamsters to clarify a possible role of an altered ratio between striatal interneurons and projection neurons. Furthermore, the immunoreactivity of dynorphin A (DYN), which is expressed in entopeduncular fibers of striatal neurons of the direct pathway, was verified by gray level measurements to illuminate the functional relevance of an enhanced striato-entopeduncular neuronal activity previously found in dt(sz) hamsters. While the density of striatal ENK immunoreactive (ENK(+) ) neurons did not significantly differ between mutant and control hamsters, there was a significantly enhanced ratio between the DYN immunoreactive area and the whole area of the EPN in dt(sz) hamsters compared to controls. These results support the hypothesis that a disbalance between a reduced density of striatal interneurons and an unchanged density of striatal projection neurons causes imbalances in the basal ganglia network. The consequentially enhanced striato-entopeduncular inhibition leads to an already evidenced reduced activity and an altered firing pattern of entopeduncular neurons in the dt(sz) hamster.


Subject(s)
Basal Ganglia/chemistry , Dynorphins/genetics , Dynorphins/metabolism , Dystonia/genetics , Enkephalins/metabolism , Neurons/chemistry , Neurons/metabolism , Animals , Animals, Genetically Modified , Basal Ganglia/metabolism , Cricetinae , Disease Models, Animal , Dynorphins/biosynthesis , Dystonia/metabolism , Dystonia/pathology , Enkephalins/physiology , Female , Interneurons/chemistry , Interneurons/metabolism , Interneurons/pathology , Male , Mesocricetus , Neurons/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...