dynorphins and Pain

dynorphins has been researched along with Pain* in 168 studies

Reviews

19 review(s) available for dynorphins and Pain

ArticleYear
Molecular Genetics of Kappa Opioids in Pain and Itch Sensations.
    Handbook of experimental pharmacology, 2022, Volume: 271

    The opioid peptides and their receptors have been linked to multiple key biological processes in the nervous system. Here we review the functions of the kappa opioid receptor (KOR) and its endogenous agonists dynorphins (Goldstein A, Tachibana S, Lowney LI, Hunkapiller M, Hood L, Proc Natl Acad Sci U S A 76:6666-6670, 1979) in modulating itch and pain (nociception). Specifically, we discuss their roles relative to recent findings that tell us more about the cells and circuits which are impacted by this opioid and its receptor and present reanalysis of single-cell sequencing data showing the expression profiles of these molecules. Since the KOR is relatively specifically activated by peptides derived from the prodynorphin gene and other opioid peptides that show lower affinities, this will be the only interactions we consider (Chavkin C, Goldstein A, Nature 291:591-593, 1981; Chavkin C, James IF, Goldstein A, Science 215:413-415, 1982), although it was noted that at higher doses peptides other than dynorphins might stimulate KOR (Lai J, Luo MC, Chen Q, Ma S, Gardell LR, Ossipov MH, Porreca F, Nat Neurosci 9:1534-1540, 2006). This review has been organized based on anatomy with each section describing the effect of the kappa opioid system in a specific location but let us not forget that most of these circuits are interconnected and are therefore interdependent.

    Topics: Analgesics, Opioid; Dynorphins; Humans; Molecular Biology; Pain; Receptors, Opioid, kappa

2022
Inhibitors of neuropeptide peptidases engaged in pain and drug dependence.
    Neuropharmacology, 2020, 09-15, Volume: 175

    Owing to a broad spectrum of functions performed by neuropeptides, this class of signaling molecules attracts an increasing interest. One of the key steps in the regulation of biological activity of neuropeptides is proteolytic conversion or degradation by proteinases that change or terminate biological activity of native peptides. These enzymes, in turn, are regulated by inhibitors, which play integral role in controlling many metabolic pathways. Thus, the search for selective inhibitors and detailed knowledge on the mechanisms of binding of these substances to enzymes, could be of importance for designing new pharmacological approaches. The aim of this review is to summarize the current knowledge on the inhibitors of enzymes that convert selected groups of neuropeptides, such as dynorphins, enkephalins, substance P and NPFF fragments. The importance of these substances in pathophysiological processes involved in pain and drug addiction, have been discussed. This article is part of the special issue on Neuropeptides.

    Topics: Animals; Dynorphins; Enkephalin, Leucine; Enzyme Inhibitors; Humans; Neuropeptides; Pain; Peptide Hydrolases; Protease Inhibitors; Substance-Related Disorders

2020
Nociceptive behavior induced by the endogenous opioid peptides dynorphins in uninjured mice: evidence with intrathecal N-ethylmaleimide inhibiting dynorphin degradation.
    International review of neurobiology, 2009, Volume: 85

    Dynorphins, the endogenous opioid peptides derived from prodynorphin may participate not only in the inhibition, but also in facilitation of spinal nociceptive transmission. However, the mechanism of pronociceptive dynorphin actions, and the comparative potential of prodynorphin processing products to induce these actions were not fully elucidated. In our studies, we examined pronociceptive effects of prodynorphin fragments dynorphins A and B and big dynorphin consisting of dynorphins A and B, and focused on the mechanisms underlying these effects. Our principal finding was that big dynorphin was the most potent pronociceptive dynorphin; when administered intrathecally into mice at extremely low doses (1-10fmol), big dynorphin produced nociceptive behavior through the activation of the NMDA receptor ion-channel complex by acting on the polyamine recognition site. We next examined whether the endogenous dynorphins participate in the spinal nociceptive transmission using N-ethylmaleimide (NEM) that blocks dynorphin degradation by inhibiting cysteine proteases. Similar to big dynorphin and dynorphin A, NEM produced nociceptive behavior mediated through inhibition of the degradation of endogenous dynorphins, presumably big dynorphin that in turn activates the NMDA receptor ion-channel complex by acting on the polyamine recognition site. Our findings support the notion that endogenous dynorphins are critical neurochemical mediators of spinal nociceptive transmission in uninjured animals. This chapter will review above-described phenomena and their mechanism.

    Topics: Animals; Cysteine Endopeptidases; Dynorphins; Ethylmaleimide; Injections, Spinal; Ion Channels; Mice; Models, Biological; Pain; Receptors, N-Methyl-D-Aspartate

2009
Pronociceptive actions of dynorphin via bradykinin receptors.
    Neuroscience letters, 2008, Jun-06, Volume: 437, Issue:3

    The endogenous opioid peptide dynorphin A is distinct from other endogenous opioid peptides in having significant neuronal excitatory and neurotoxic effects that are not mediated by opioid receptors. Some of these non-opioid actions of dynorphin contribute to the development of abnormal pain resulting from a number of pathological conditions. Identifying the mechanisms and the sites of action of dynorphin is essential for understanding the pathophysiology of dynorphin and for exploring novel therapeutic targets for pain. This review will discuss the mechanisms that have been proposed and the recent finding that spinal dynorphin may be an endogenous ligand of bradykinin receptors under pathological conditions to promote pain.

    Topics: Animals; Dynorphins; Humans; Pain; Receptors, Bradykinin; Spinal Cord

2008
Cholecystokinin antagonists a new way to improve the analgesia from old analgesics?
    Current pharmaceutical design, 2004, Volume: 10, Issue:3

    Cholecystokinin, originally thought to be confined only to the gastrointestinal tract, is now known to be co-localised in both the gastrointestinal tract and central nervous system. In animal models levels are increased after neural injury and with opioid administration. This peptide acts as an anti-opioid, and as levels increase, the extent of opioid derived antinociception decreases. Co-administration of a CCK antagonist along with an opioid is associated with an improved level of antinociception. Furthermore CCK antagonists may prevent antinociceptive tolerance with opioids and even reverse established tolerance. Human studies have now confirmed the pro-analgesic effect of some CCK antagonists. Human investigation of the effect of CCK antagonists on analgesic tolerance has yet to be performed. This review examines the available evidence that suggests a role for CCK antagonists in human pain management.

    Topics: Analgesics; Animals; Cholecystokinin; Clinical Trials as Topic; Drug Tolerance; Dynorphins; Enkephalins; Humans; Narcotics; Pain; Time Factors

2004
No DREAM, No pain. Closing the spinal gate.
    Cell, 2002, Feb-08, Volume: 108, Issue:3

    Pain transmission in the spinal cord is regulated by a balance of facilitatory and inhibitory influences operating on the neural circuits of the somatosensory system. The transcriptional repressor DREAM acts constitutively to suppress prodynorphin expression in spinal cord neurons. Knocking out DREAM results in sufficient dynorphin expression to produce a strong reduction in generalized pain behavior, highlighting the role that intracellular molecules play in modulating pain gating in the spinal cord.

    Topics: Animals; Calcium-Binding Proteins; Dynorphins; Humans; Kv Channel-Interacting Proteins; Pain; Receptors, Opioid, kappa; Repressor Proteins; Spinal Cord

2002
Neuropathic pain: the paradox of dynorphin.
    Molecular interventions, 2001, Volume: 1, Issue:3

    One of the curious but common consequences of opioid administration in the clinical setting is the induction, at sites uninvolved in the original presentation of discomfort, of pain itself. The induction of pain is also a reliable, measurable phenomenon in animals receiving continuous delivery of opioid. Such pain induction is associated with the expression of spinal dynorphin, a finding that is especially intriguing in light of dynorphin's ability to recapitulate many of the characteristics of chronic, neuropathic pain when administered intrathecally (i.e., into the spine). The effective treatment of chronic pain syndromes-and of tolerance to antinociceptive therapies-may thus rest on an understanding of the biological roles of dynorphin in neurotransmission.

    Topics: Analgesics, Opioid; Animals; Drug Tolerance; Dynorphins; Humans; Hyperalgesia; Injections, Spinal; Models, Biological; Neurotransmitter Agents; Pain; Receptors, N-Methyl-D-Aspartate; Spinal Cord

2001
Mechanisms of opioid-induced pain and antinociceptive tolerance: descending facilitation and spinal dynorphin.
    Pain, 2001, Volume: 92, Issue:1-2

    Topics: Analgesics, Opioid; Drug Tolerance; Dynorphins; Humans; Nociceptors; Pain; Spinal Cord

2001
Spinal and supraspinal mechanisms of neuropathic pain.
    Annals of the New York Academy of Sciences, 2000, Volume: 909

    Neuropathic pain is associated with abnormal tactile and thermal responses that may be extraterritorial to the injured nerve. Importantly, tactile allodynia and thermal hyperalgesia may involve separate pathways, since complete and partial spinal cord lesions have blocked allodynia, but not hyperalgesia, after spinal nerve ligation (SNL). Furthermore, lesions of the dorsal column, and lidocaine microinjected into dorsal column nuclei block only tactile allodynia. Conversely, thermal hyperalgesia, but not tactile allodynia was blocked by desensitization of C-fibers with resiniferotoxin. Therefore, it seems that tactile allodynia is likely to be mediated by large diameter A beta fibers, and not susceptible to modulation by spinal opioids, whereas hyperalgesia is mediated by unmyelinated C-fibers, and is sensitive to blockade by spinal opioids. Additionally, abnormal, spontaneous afferent drive in neuropathic pain may contribute to NMDA-mediated central sensitization by glutamate and by non-opioid actions of spinal dynorphin. Correspondingly, SNL elicited elevation in spinal dynorphin content in spinal segments at and adjacent to the zone of entry of the injured nerve along with signs of neuropathic pain. Antiserum to dynorphin A(1-17) or MK-801 given spinally blocked thermal hyperalgesia, but not tactile allodynia, after SNL, and also restored diminished morphine antinociception. Finally, afferent drive may induce descending facilitation from the rostroventromedial medulla (RVM). Blocking afferent drive with bupivicaine also restored lost potency of PAG morphine, as did CCK antagonists in the RVM. This observation is consistent with afferent drive activating descending facilitation from the RVM, and thus diminishing opioid activity, and may underlie the clinical observation of limited responsiveness of neuropathic pain to opioids.

    Topics: Animals; Dizocilpine Maleate; Dynorphins; Humans; Hyperalgesia; Lidocaine; Morphine; Pain; Spinal Cord

2000
Dynorphin: friend or foe?
    Pain, 2000, Volume: 87, Issue:3

    Topics: Animals; Dynorphins; Humans; Pain; Receptors, N-Methyl-D-Aspartate

2000
[The brain and the pain: neurotransmitters and neuronal pathways of pain perception and response].
    Orvosi hetilap, 2000, Oct-08, Volume: 141, Issue:41

    The pain, an unpleasant feeling, induces several central nervous system mechanisms, like sensory-discriminative, motivational-affective activities, behavioral changes and it activates various responses, including antinociceptive actions. Accordingly, signals from the nociceptive neurons in the spinal cord and the sensory trigeminal nucleus ascend in various neuronal pathways and target several brain areas. Here, five ascending pain-conducting neuronal pathways and two spinal reflex routes are briefly summarized. The spinal and supraspinal antinociceptive mechanisms are described in more detail. During the past two decades, endogenous opioids, cannabinoids and their receptors have been discovered, localized and cloned. Five groups of endogenous opiates are known: beta-endorphin, enkephalins, dynorphins, endomorphins, and nociceptin. Two endogenous cannabinoids have already been described in the brain: the anandamide and the 2-arachidonyl-glycerol. The site of their antinociceptive (analgesic) actions in the brain are briefly summarized.

    Topics: Afferent Pathways; Animals; Arachidonic Acids; Brain; Cannabinoids; Dynorphins; Efferent Pathways; Endocannabinoids; Endorphins; Glycerides; Humans; Neural Conduction; Neural Pathways; Neurotransmitter Agents; Nociceptin; Opioid Peptides; Pain; Polyunsaturated Alkamides; Receptors, Neurotransmitter; Receptors, Opioid

2000
Reward and its control by dynorphin peptides.
    EXS, 1994, Volume: 71

    Reward is a strong behavioral cue. It is argued that a sense of reward must be strictly controlled in time and magnitude to be functional. Central to the reward system is the mesolimbic dopamine pathway which is titillated by addictive drugs and alcohol. Rat lines or strains that voluntarily drink alcohol have lower dynorphin levels in relevant brain areas than those which avoid alcohol. Reward control may be a protective factor against addiction.

    Topics: Alcohol Drinking; Animals; Brain; Dynorphins; Endorphins; Humans; Pain; Reward

1994
Neurophysiology of opioid poorly responsive pain.
    Cancer surveys, 1994, Volume: 21

    This review has covered the five potential causes for opioid poorly responsive pain, namely (a) a loss of opioid receptors on the spinal terminals of C-fibres as a result of peripheral nerve damage, (b) an accumulation of morphine-3-glucuronide, (c) changes in the non-opioid peptides, F8Fa or CCK, either spinally or supraspinally, (d) actions of the opioid peptide dynorphin and (e) spinally generated hypersensitive states via activation of the NMDA receptor. The loss of opioid receptors is likely to be important where peripheral nerve pathology or compression occurs, but the evidence suggests that increasing the dose will overcome the reduced opioid response. Morphine-3-glucuronide is unlikely to be a factor, nor is dynorphin, but the endogenous peptides CCK and F8Fa may be important. Finally, there is an association between the NMDA receptor and hyperalgesia/allodynia and reduced opioid sensitivity. Dextrorphan and ketamine reduce NMDA mediated events and so are available to test this hypothesis.

    Topics: Analgesics, Opioid; Animals; Central Nervous System; Clonidine; Dextromethorphan; Dextrorphan; Dynorphins; Humans; Morphine; N-Methylaspartate; Neoplasms; Neurophysiology; Neuroprotective Agents; Pain; Rats; Receptors, N-Methyl-D-Aspartate; Receptors, Opioid; Sympatholytics

1994
Cytochemical studies of the neural circuitry underlying pain and pain control.
    Acta neurochirurgica. Supplementum, 1987, Volume: 38

    Topics: Animals; Dynorphins; Enkephalins; Pain; Spinal Cord

1987
Functional response of multiple opioid systems to chronic arthritic pain in the rat.
    Annals of the New York Academy of Sciences, 1986, Volume: 467

    Topics: 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer; Animals; Arthritis; Arthritis, Experimental; Benzomorphans; beta-Endorphin; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalin, Methionine; Morphine; Naloxone; Nociceptors; Pain; Pituitary Gland, Anterior; Protein Precursors; Pyrrolidines; Rats; Receptors, Opioid; Sensory Thresholds; Spinal Cord; Thalamus

1986
The endocrinology of the opioids.
    International review of neurobiology, 1985, Volume: 26

    Topics: Adrenal Glands; Adrenocorticotropic Hormone; Animals; Arginine Vasopressin; beta-Endorphin; Circadian Rhythm; Dynorphins; Endocrine Glands; Endorphins; Enkephalins; Female; Humans; Hypothalamo-Hypophyseal System; Islets of Langerhans; Melanocyte-Stimulating Hormones; Oxytocin; Pain; Pituitary Gland, Anterior; Pituitary Gland, Posterior; Pregnancy; Rats; Receptors, Opioid; Shock; Stress, Physiological

1985
Endogenous pain control systems: brainstem spinal pathways and endorphin circuitry.
    Annual review of neuroscience, 1984, Volume: 7

    Topics: Acetylcholine; Afferent Pathways; Analgesics; Animals; beta-Endorphin; Brain Stem; Dynorphins; Endorphins; Enkephalins; Medulla Oblongata; Models, Biological; Neural Pathways; Neurons; Neurotensin; Norepinephrine; Pain; Peptide Fragments; Periaqueductal Gray; Pituitary Hormones, Anterior; Pro-Opiomelanocortin; Protein Precursors; Spinal Cord; Stress, Physiological

1984
Pain, nociception and spinal opioid receptors.
    Progress in neuro-psychopharmacology & biological psychiatry, 1984, Volume: 8, Issue:4-6

    Opioid peptides derived from proenkephalin and prodynorphin are differentially distributed in the spinal cord. Proenkephalin peptides are preferentially located in the sacral portion of the cord while prodynorphin peptides are concentrated in the cervical spinal cord. Mu opioid receptor are highly concentrated in superficial layers of the dorsal horn in all the spinal cord. Delta opioid receptor are more diffusely distributed in the gray matter of the spinal cord. These sites are principally located in cervical and thoracic portions of the spinal cord. Kappa opioid receptors are highly concentrated in the superficial layers of the lumbo-sacral spinal cord. Its density decreased in the upper levels of the spinal cord. It appears that mu opioid receptors are indifferentially activated by thermal, pressure and visceral nociceptive inputs. Delta receptors are more likely to be involved in thermal nociception while kappa opioid binding sites are associated to visceral pain nociceptive inputs.

    Topics: Animals; Dogs; Dynorphins; Endorphins; Enkephalin, Methionine; Guinea Pigs; Injections, Spinal; Mice; Pain; Rats; Receptors, Opioid; Receptors, Opioid, delta; Receptors, Opioid, kappa; Receptors, Opioid, mu; Spinal Cord; Thermosensing; Touch

1984
Biogenesis, release and inactivation of enkephalins and dynorphins.
    British medical bulletin, 1983, Volume: 39, Issue:1

    Topics: Amino Acid Sequence; Animals; Brain; Dynorphins; Endorphins; Enkephalins; Guinea Pigs; Mice; Pain; Protein Precursors; Rats; Stress, Physiological

1983

Other Studies

149 other study(ies) available for dynorphins and Pain

ArticleYear
Sex-dependent effect of inflammatory pain on negative affective states is prevented by kappa opioid receptors blockade in the nucleus accumbens shell.
    Neuropharmacology, 2024, Jan-01, Volume: 242

    Pain comorbidities include several psychological disorders, such as anxiety and anhedonia. However, the way pain affects male and female individuals and by which mechanism is not well understood. Previous research shows that pain induces alterations in the dynorphinergic pathway within the mesocorticolimbic system (MCLS), together with a relationship between corticotropin-releasing system and dynorphin release in the MCLS. Here, we analyse the sex and time course-dependent effects of pain on negative affect. Additionally, we study the implication of dynorphinergic and corticotropin releasing factor in these pain related behaviours. We used behavioural pharmacology and biochemical tools to characterise negative affective states induced by inflammatory pain in male and female rats, and the alterations in the dynorphinergic and the corticotropin systems within the MCLS. Female rats showed persistent anxiety-like and reversible anhedonia-like behaviours derived from inflammatory pain. Additionally, we found alterations in dynorphin and corticotropin releasing factor in NAc and amygdala, which suggests sex-dependent dynamic adaptations. Finally blockade on the kappa opioid receptor in the NAc confirmed its role in pain-induced anxiety-like behaviour in female rats. Our results show sex and time-dependent anxiety- and anhedonia-like behaviours induced by the presence of pain in female rats. Furthermore, we replicated previous data, pointing to the KOR/DYN recruitment in the NAc as a key neurological substrate mediating pain-induced behavioural alterations. This research studies the mechanisms underlying these behaviours, to better understand the emotional dimension of pain.

    Topics: Adrenocorticotropic Hormone; Anhedonia; Animals; Corticotropin-Releasing Hormone; Dynorphins; Female; Male; Nucleus Accumbens; Pain; Rats; Receptors, Opioid, kappa

2024
Single-nucleus characterization of adult mouse spinal dynorphin-lineage cells and identification of persistent transcriptional effects of neonatal hindpaw incision.
    Pain, 2021, Volume: 162, Issue:1

    Neonatal tissue damage can have long-lasting effects on nociceptive processing in the central nervous system, which may reflect persistent injury-evoked alterations to the normal balance between synaptic inhibition and excitation in the spinal dorsal horn. Spinal dynorphin-lineage (pDyn) neurons are part of an inhibitory circuit which limits the flow of nociceptive input to the brain and is disrupted by neonatal tissue damage. To identify the potential molecular underpinnings of this disruption, an unbiased single-nucleus RNAseq analysis of adult mouse spinal pDyn cells characterized this population in depth and then identified changes in gene expression evoked by neonatal hindpaw incision. The analysis revealed 11 transcriptionally distinct subpopulations (ie, clusters) of dynorphin-lineage cells, including both inhibitory and excitatory neurons. Investigation of injury-evoked differential gene expression identified 15 genes that were significantly upregulated or downregulated in adult pDyn neurons from neonatally incised mice compared with naive littermate controls, with both cluster-specific and pan-neuronal transcriptional changes observed. Several of the identified genes, such as Oxr1 and Fth1 (encoding ferritin), were related to the cellular stress response. However, the relatively low number of injury-evoked differentially expressed genes also suggests that posttranscriptional regulation within pDyn neurons may play a key role in the priming of developing nociceptive circuits by early-life injury. Overall, the findings reveal novel insights into the molecular heterogeneity of a key population of dorsal horn interneurons that has previously been implicated in the suppression of mechanical pain and itch.

    Topics: Animals; Dynorphins; Interneurons; Mice; Neurons; Pain; Posterior Horn Cells; Spinal Cord Dorsal Horn

2021
Structure and dynamics of dynorphin peptide and its receptor.
    Vitamins and hormones, 2019, Volume: 111

    Dynorphin is a neuropeptide involved in pain, addiction and mood regulation. It exerts its activity by binding to the kappa opioid receptor (KOP) which belongs to the large family of G protein-coupled receptors. The dynorphin peptide was discovered in 1975, while its receptor was cloned in 1993. This review will describe: (a) the activities and physiological functions of dynorphin and its receptor, (b) early structure-activity relationship studies performed before cloning of the receptor (mostly pharmacological and biophysical studies of peptide analogues), (c) structure-activity relationship studies performed after cloning of the receptor via receptor mutagenesis and the development of recombinant receptor expression systems, (d) structural biology of the opiate receptors culminating in X-ray structures of the four opioid receptors in their inactive state and structures of MOP and KOP receptors in their active state. X-ray and EM structures are combined with NMR data, which gives complementary insight into receptor and peptide dynamics. Molecular modeling greatly benefited from the availability of atomic resolution 3D structures of receptor-ligand complexes and an example of the strategy used to model a dynorphin-KOP receptor complex using NMR data will be described. These achievements have led to a better understanding of the complex dynamics of KOP receptor activation and to the development of new ligands and drugs.

    Topics: Amino Acid Sequence; Animals; Cloning, Molecular; Dynorphins; Humans; Models, Molecular; Molecular Structure; Mutagenesis, Site-Directed; Pain; Protein Binding; Receptors, Opioid; Structure-Activity Relationship; Substance-Related Disorders

2019
Pain-Induced Negative Affect Is Mediated via Recruitment of The Nucleus Accumbens Kappa Opioid System.
    Neuron, 2019, 05-08, Volume: 102, Issue:3

    Negative affective states affect quality of life for patients suffering from pain. These maladaptive emotional states can lead to involuntary opioid overdose and many neuropsychiatric comorbidities. Uncovering the mechanisms responsible for pain-induced negative affect is critical in addressing these comorbid outcomes. The nucleus accumbens (NAc) shell, which integrates the aversive and rewarding valence of stimuli, exhibits plastic adaptations in the presence of pain. In discrete regions of the NAc, activation of the kappa opioid receptor (KOR) decreases the reinforcing properties of rewards and induces aversive behaviors. Using complementary techniques, we report that in vivo recruitment of NAc shell dynorphin neurons, acting through KOR, is necessary and sufficient to drive pain-induced negative affect. Taken together, our results provide evidence that pain-induced adaptations in the kappa opioid system within the NAc shell represent a functional target for therapeutic intervention that could circumvent pain-induced affective disorders. VIDEO ABSTRACT.

    Topics: Affect; Animals; Dynorphins; Inflammation; Mice; Mood Disorders; Neural Inhibition; Neuronal Plasticity; Neurons; Nucleus Accumbens; Pain; Rats; Receptors, Opioid, kappa

2019
Lateralized kappa opioid receptor signaling from the amygdala central nucleus promotes stress-induced functional pain.
    Pain, 2018, Volume: 159, Issue:5

    The response of diffuse noxious inhibitory controls (DNIC) is often decreased, or lost, in stress-related functional pain syndromes. Because the dynorphin/kappa opioid receptor (KOR) pathway is activated by stress, we determined its role in DNIC using a model of stress-induced functional pain. Male, Sprague-Dawley rats were primed for 7 days with systemic morphine resulting in opioid-induced hyperalgesia. Fourteen days after priming, when hyperalgesia was resolved, rats were exposed to environmental stress and DNIC was evaluated by measuring hind paw response threshold to noxious pressure (test stimulus) after capsaicin injection in the forepaw (conditioning stimulus). Morphine priming without stress did not alter DNIC. However, stress produced a loss of DNIC in morphine-primed rats in both hind paws that was abolished by systemic administration of the KOR antagonist, nor-binaltorphimine (nor-BNI). Microinjection of nor-BNI into the right, but not left, central nucleus of the amygdala (CeA) prevented the loss of DNIC in morphine-primed rats. Diffuse noxious inhibitory controls were not modulated by bilateral nor-BNI in the rostral ventromedial medulla. Stress increased dynorphin content in both the left and right CeA of primed rats, reaching significance only in the right CeA; no change was observed in the rostral ventromedial medulla or hypothalamus. Although morphine priming alone is not sufficient to influence DNIC, it establishes a state of latent sensitization that amplifies the consequences of stress. After priming, stress-induced dynorphin/KOR signaling from the right CeA inhibits DNIC in both hind paws, likely reflecting enhanced descending facilitation that masks descending inhibition. Kappa opioid receptor antagonists may provide a new therapeutic strategy for stress-related functional pain disorders.

    Topics: Analgesics, Opioid; Animals; Capsaicin; Central Amygdaloid Nucleus; Dynorphins; Hypothalamus; Male; Medulla Oblongata; Morphine; Naltrexone; Narcotic Antagonists; Pain; Rats; Rats, Sprague-Dawley; Receptors, Opioid, kappa; Stress, Physiological

2018
Circuit dissection of the role of somatostatin in itch and pain.
    Nature neuroscience, 2018, Volume: 21, Issue:5

    Topics: Animals; Dynorphins; Female; Ganglia, Spinal; Male; Mice; Mice, Inbred C57BL; Neural Pathways; Optogenetics; Pain; Pruritus; Receptors, Atrial Natriuretic Factor; Receptors, Purinergic; Receptors, Somatostatin; Sensory Receptor Cells; Somatostatin; Spinal Cord

2018
Mu and delta opioid receptor knockout mice show increased colonic sensitivity.
    European journal of pain (London, England), 2017, Volume: 21, Issue:4

    Opiates act through opioid receptors to diminish pain. Here, we investigated whether mu (MOR) and delta (DOR) receptor endogenous activity assessed in the whole mouse body or in particular at peripheral receptors on primary nociceptive neurons, control colonic pain.. We compared global MOR and DOR receptor knockout (KO) mice, mice with a conditional deletion of MOR and DOR in Nav1.8-positive nociceptive primary afferent neurons (cKO), and control floxed mice of both genders for visceral sensitivity. Visceromotor responses to colorectal distension (CRD) and macroscopic colon scores were recorded on naïve mice and mice with acute colitis induced by 3% dextran sodium sulphate (DSS) for 5 days. Transcript expression for opioid genes and cytokines was measured by quantitative RT-PCR.. Naïve MOR and DOR global KO mice show increased visceral sensitivity that was not observed in cKO mice. MOR and preproenkephalin (Penk) were the most expressed opioid genes in colon. MOR KO mice had augmented kappa opioid receptor and Tumour-Necrosis-Factor-α and diminished Penk transcript levels while DOR, preprodynorphin and Interleukin-1β were unchanged. Global MOR KO females had a thicker colon than floxed females. No alteration was detected in DOR mutant animals. A 5-day DSS treatment led to comparable hypersensitivity in the different mouse lines.. Our results suggest that mu and delta opioid receptor global endogenous activity but not activity at the peripheral Nav1.8 neurons contribute to visceral sensitivity in naïve mice, and that endogenous MOR and DOR tones were insufficient to elicit analgesia after 5-day DSS-induced colitis.. Knockout mice for mu and delta opioid receptor have augmented colon sensitivity in the CRD assay. It shows endogenous mu and delta opioid analgesia that may be explored as potential targets for alleviating chronic intestinal pain.

    Topics: Analgesics, Opioid; Animals; Colitis; Dextran Sulfate; Dynorphins; Enkephalins; Female; Interleukin-1beta; Male; Mice; Mice, Knockout; Pain; Pain Management; Protein Precursors; Receptors, Opioid, delta; Receptors, Opioid, mu; Tumor Necrosis Factor-alpha

2017
Concurrent bullatine A enhances morphine antinociception and inhibits morphine antinociceptive tolerance by indirect activation of spinal κ-opioid receptors.
    Journal of ethnopharmacology, 2017, Jan-20, Volume: 196

    Bullatine A, a C. To assess the interaction between bullatine A and morphine on antinociception in acute nociception and pain hypersensitivity states, with the exogenous synthetic dynorphin A as a comparison MATERIALS AND METHODS: Spinal nerve ligation-induced neuropathic rats and naïve mice were used for assessing the acute and chronic interactions of bullatine A/dynorphin A with morphine.. Single subcutaneous injection of bullatine A or dynorphin A(1-17) did not either alter formalin- and thermally (hot-plate and water immersion tests)-induced acute nociception or potentiate morphine antinociception in naïve mice. In contrast, bullatine A dose-dependently inhibited formalin-induced tonic pain with the efficacy of 54% inhibition and the half-effective dose of 0.9mg/kg. Concurrent bullatine A additively enhanced morphine antinociception. In neuropathic rats, the antinociceptive effects of multiple bidaily intrathecal injections of bullatine A and dynorphin A remained consistent over 13 days, whereas morphine produced progressive and complete tolerance to antinociception, which was completely inhibited by concurrent bullatine A and dynorphin A. A single intrathecal injection of bullatine A and dynorphin A immediately reversed established morphine tolerance in neuropathic rats, although the blockade was a less degree in the thermally induced mouse acute nociceptive tests. The inhibitory effects of bullatine A and dynorphin A on morphine tolerance were immediately and completely attenuated by intrathecal dynorphin A antibody and/or selective κ-opioid receptor antagonist GNTI.. These results suggest that bullatine A produces antinociception without induction of tolerance and inhibits morphine antinociceptive tolerance, and provide pharmacological basis for concurrent bullatine A and morphine treatment for chronic pain and morphine analgesic tolerance.

    Topics: Alkaloids; Analgesics; Animals; Behavior, Animal; Diterpenes; Dose-Response Relationship, Drug; Drug Combinations; Drug Tolerance; Dynorphins; Formaldehyde; Hot Temperature; Hyperalgesia; Male; Mice; Microglia; Morphine; Pain; Rats, Wistar; Receptors, Opioid, kappa; Spine

2017
The efficacy of Dynorphin fragments at the κ, μ and δ opioid receptor in transfected HEK cells and in an animal model of unilateral peripheral inflammation.
    Peptides, 2017, Volume: 89

    Dynorphin 1-17 is an endogenous peptide that is released at sites of inflammation by leukocytes, binding preferentially to κ-opioid receptors (KOP) to mediate nociception. We have previously shown that dynorphin 1-17 is rapidly biotransformed to smaller peptide fragments in inflamed tissue homogenate. This study aimed to determine the efficacy and potency of selected dynorphin fragments produced in an inflamed environment at the KOP, μ and δ-opioid receptors (MOP and DOP respectively) and in a model of inflammatory pain. Functional activity of Dynorphin 1-17 and fragments (1-6, 1-7 and 1-9) were screened over a range of concentrations against forskolin stimulated human embryonic kidney 293 (HEK) cells stably transfected with one of KOP, MOP or DOP. The analgesic activity of dynorphin 1-7 in a unilateral model of inflammatory pain was subsequently tested. Rats received unilateral intraplantar injections of Freund's Complete Adjuvant to induce inflammation. After six days rats received either dynorphin 1-7, 1-17 or the selective KOP agonist U50488H and mechanical allodynia determined. Dynorphin 1-7 and 1-9 displayed the greatest activity across all receptor subtypes, while dynorphin 1-7, 1-9 and 1-17 displaying a potent activation of both KOP and DOP evidenced by cAMP inihibition. Administration of dynorphin 1-7 and U50488H, but not dynorphin 1-17 resulted in a significant increase in paw pressure threshold at an equimolar dose suggesting the small peptide dynorphin 1-7 mediates analgesia. These results show that dynorphin fragments produced in an inflamed tissue homogenate have changed activity at the opioid receptors and that dynorphin 1-7 mediates analgesia.

    Topics: 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer; Analgesia; Animals; Disease Models, Animal; Dynorphins; HEK293 Cells; Humans; Inflammation; Pain; Rats; Receptors, Opioid, delta; Receptors, Opioid, kappa; Receptors, Opioid, mu; Transfection

2017
Synthesis of mixed MOR/KOR efficacy cyclic opioid peptide analogs with antinociceptive activity after systemic administration.
    European journal of medicinal chemistry, 2016, Feb-15, Volume: 109

    Cyclic pentapeptide Tyr-c[D-Lys-Phe-Phe-Asp]NH2, based on the structure of endomorphin-2 (EM-2), which shows high affinity to the μ-opioid receptor (MOR) and a very strong antinociceptive activity in mice was used as a parent compound for the structure-activity relationship studies. In this report we synthesized analogs of a general sequence Dmt-c[D-Lys-Xaa-Yaa-Asp]NH2, with D-1- or D-2-naphthyl-3-alanine (D-1-Nal or D-2-Nal) in positions 3 or 4. In our earlier papers we have indicated that replacing a phenylalanine residue by the more extended aromatic system of naphthylalanines may result in increased bioactivities of linear analogs. The data obtained here showed that only cyclopeptides modified in position 4 retained the sub-nanomolar MOR and nanomolar κ-opioid receptor (KOR) affinity, similar but not better than that of a parent cyclopeptide. In the in vivo mouse hot-plate test, the most potent analog, Dmt-c[D-Lys-Phe-D-1-Nal-Asp]NH2, exhibited higher than EM-2 but slightly lower than the cyclic parent peptide antinociceptive activity after peripheral (ip) and also central administration (icv). Conformational analyses in a biomimetic environment and molecular docking studies disclosed the structural determinants responsible for the different pharmacological profiles of position 3- versus position 4-modified analogs.

    Topics: Amino Acid Sequence; Analgesics, Opioid; Animals; CHO Cells; Cricetulus; Guinea Pigs; Male; Mice; Molecular Docking Simulation; Oligopeptides; Pain; Peptides, Cyclic; Rats, Wistar; Receptors, Opioid, kappa; Receptors, Opioid, mu; Structure-Activity Relationship

2016
Structure-Activity Relationships of [des-Arg
    Journal of medicinal chemistry, 2016, 11-23, Volume: 59, Issue:22

    Dynorphin A (Dyn A) is an endogenous ligand for the opioid receptors with preference for the κ opioid receptor (KOR), and its structure-activity relationship (SAR) has been extensively studied at the KOR to develop selective potent agonists and antagonists. Numerous SAR studies have revealed that the Arg

    Topics: Animals; Cell Line; CHO Cells; Cricetulus; Dose-Response Relationship, Drug; Dynorphins; Guinea Pigs; Male; Mice; Mice, Inbred ICR; Narcotic Antagonists; Pain; Rats; Receptors, Opioid, kappa; Structure-Activity Relationship

2016
Aconitum-Derived Bulleyaconitine A Exhibits Antihypersensitivity Through Direct Stimulating Dynorphin A Expression in Spinal Microglia.
    The journal of pain, 2016, Volume: 17, Issue:5

    Aconitine and its structurally-related diterpenoid alkaloids have been shown to interact differentially with neuronal voltage-dependent sodium channels, which was suggested to be responsible for their analgesia and toxicity. Bulleyaconitine A (BAA) is an aconitine analogue and has been prescribed for the management of pain. The present study aimed to evaluate the inhibitory effects of BAA on pain hypersensitivity and morphine antinociceptive tolerance, and explore whether the expression of dynorphin A in spinal microglia was responsible for its actions. Single intrathecal or subcutaneous (but not intraventricular or local) injection of BAA blocked spinal nerve ligation-induced painful neuropathy, bone cancer-induced pain, and formalin-induced tonic pain by 60 to 100% with the median effective dose values of 94 to 126 ng per rat (intrathecal) and 42 to 59 μg/kg (subcutaneous), respectively. After chronic treatment, BAA did not induce either self-tolerance to antinociception or cross-tolerance to morphine antinociception, and completely inhibited morphine tolerance. The microglial inhibitor minocycline entirely blocked spinal BAA (but not exogenous dynorphin A) antinociception, but failed to attenuate spinal BAA neurotoxicity. In a minocycline-sensitive and lidocaine- or ropivacaine-insensitive manner, BAA stimulated the expression of dynorphin A in the spinal cord, and primary cultures of microglia but not of neurons or astrocytes. The blockade effects of BAA on nociception and morphine tolerance were totally eliminated by the specific dynorphin A antiserum and/or κ-opioid receptor antagonist. Our results suggest that BAA eliminates pain hypersensitivity and morphine tolerance through directly stimulating dynorphin A expression in spinal microglia, which is not dependent on the interactions with sodium channels.. The newly illustrated mechanisms underlying BAA antinociception help us to better understand and develop novel dynorphin A expression-based painkillers to treat chronic pain.

    Topics: Aconitine; Aconitum; Analgesics; Analysis of Variance; Animals; Animals, Newborn; Bone Neoplasms; CD11b Antigen; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Administration Routes; Dynorphins; Female; Functional Laterality; Gene Expression Regulation; Glial Fibrillary Acidic Protein; Hyperalgesia; Male; Microglia; Microscopy, Confocal; Morphine; Neurons; Pain; Pain Measurement; Phosphopyruvate Hydratase; Rats; Receptors, Opioid, kappa; RNA, Messenger; Spinal Cord; Time Factors

2016
Effect of estrogen on morphine- and oxycodone-induced antinociception in a female femur bone cancer pain model.
    European journal of pharmacology, 2016, Feb-15, Volume: 773

    Although estrous cycle has been reported to influence antiociceptive effect of morphine in several pain conditions, its effect on cancer pain is not well established. We investigated the effect of estrogen on morphine antinociception using a bone cancer pain model and compared its potency with that of oxycodone. Female mice were ovariectomized (OVX) for preparation of a femur bone cancer pain (FBC) model. β-estradiol was subcutaneously (s.c.) administered and antinociceptive effects of opioids was assessed using the von Frey monofilament test. Although morphine (5-20mg/kg, s.c.) did have significant antinociceptive effects in the FBC-OVX group, its effects in the FBC-OVX+β-estradiol (OVX+E) group was limited. Oxycodone (1-5mg/kg, s.c.) exhibited significant effects in both groups. Expression changes in opioid-related genes (μ-, κ-, δ-opioid receptors, prodynorphin, proenkephalin, proopiomelanocortin) in the spinal and supraspinal sites were examined among the sham-OVX, sham-OVX+E, FBC-OVX, and FBC-OVX+E groups by in situ hybridization. These studies detected a significant increase in prodynorphin in the spinal dorsal horn of the FBC-OVX+E group. Spinal injection of a dynorphin-A antibody to FBC-OVX+E mice restored antinociception of morphine. In conclusion, we detected a differential effect of estrogen on morphine- and oxycodone-induced antinociception in a female FBC model. The effect of morphine was limited with estrogen exposure, which may be due to estrogen- and pain-mediated spinal expression of dynorphin-A.

    Topics: Analgesics; Animals; Bone Neoplasms; Cell Line, Tumor; Dynorphins; Estrogens; Estrous Cycle; Female; Femur; Gene Expression Regulation; Male; Mice; Morphine; Ovariectomy; Oxycodone; Pain

2016
The affective dimension of pain as a risk factor for drug and alcohol addiction.
    Alcohol (Fayetteville, N.Y.), 2015, Volume: 49, Issue:8

    Addiction, or substance use disorder (SUD), is a devastating psychiatric disease composed of multiple elemental features. As a biobehavioral disorder, escalation of drug and/or alcohol intake is both a cause and consequence of molecular neuroadaptations in central brain reinforcement circuitry. Multiple mesolimbic areas mediate a host of negative affective and motivational symptoms that appear to be central to the addiction process. Brain stress- and reinforcement-related regions such as the central amygdala (CeA), prefrontal cortex (PFC), and nucleus accumbens (NAc) also serve as central processors of ascending nociceptive input. We hypothesize that a sensitization of brain mechanisms underlying the processing of persistent and maladaptive pain contributes to a composite negative affective state to drive the enduring, relapsing nature of addiction, particularly in the case of alcohol and opioid use disorder. At the neurochemical level, pain activates central stress-related neuropeptide signaling, including the dynorphin and corticotropin-releasing factor (CRF) systems, and by this process may facilitate negative affect and escalated drug and alcohol use over time. Importantly, the widespread prevalence of unresolved pain and associated affective dysregulation in clinical populations highlights the need for more effective analgesic medications with reduced potential for tolerance and dependence. The burgeoning epidemic of prescription opioid abuse also demands a closer investigation into the neurobiological mechanisms of how pain treatment could potentially represent a significant risk factor for addiction in vulnerable populations. Finally, the continuing convergence of sensory and affective neuroscience fields is expected to generate insight into the critical balance between pain relief and addiction liability, as well as provide more effective therapeutic strategies for chronic pain and addiction.

    Topics: Affect; Alcoholism; Amygdala; Animals; Brain; Chronic Pain; Corticotropin-Releasing Hormone; Dynorphins; Humans; Hyperalgesia; Mood Disorders; Neuropeptides; Nucleus Accumbens; Pain; Prefrontal Cortex; Risk Factors; Substance-Related Disorders

2015
Distinct functions of opioid-related peptides and gastrin-releasing peptide in regulating itch and pain in the spinal cord of primates.
    Scientific reports, 2015, Jun-29, Volume: 5

    How neuropeptides in the primate spinal cord regulate itch and pain is largely unknown. Here we elucidate the sensory functions of spinal opioid-related peptides and gastrin-releasing peptide (GRP) in awake, behaving monkeys. Following intrathecal administration, β-endorphin (10-100 nmol) and GRP (1-10 nmol) dose-dependently elicit the same degree of robust itch scratching, which can be inhibited by mu-opioid peptide (MOP) receptor and GRP receptor (BB2) antagonists, respectively. Unlike β-endorphin, which produces itch and attenuates inflammatory pain, GRP only elicits itch without affecting pain. In contrast, enkephalins (100-1000 nmol) and nociceptin-orphanin FQ (3-30 nmol) only inhibit pain without eliciting itch. More intriguingly, dynorphin A(1-17) (10-100 nmol) dose-dependently attenuates both β-endorphin- and GRP-elicited robust scratching without affecting pain processing. The anti-itch effects of dynorphin A can be reversed by a kappa-opioid peptide (KOP) receptor antagonist nor-binaltorphimine. These nonhuman primate behavioral models with spinal delivery of ligands advance our understanding of distinct functions of neuropeptides for modulating itch and pain. In particular, we demonstrate causal links for itch-eliciting effects by β-endorphin-MOP receptor and GRP-BB2 receptor systems and itch-inhibiting effects by the dynorphin A-KOP receptor system. These studies will facilitate transforming discoveries of novel ligand-receptor systems into future therapies as antipruritics and/or analgesics in humans.

    Topics: Analgesics, Opioid; Animals; Behavior, Animal; beta-Endorphin; Dynorphins; Female; Gastrin-Releasing Peptide; Hyperalgesia; Injections, Spinal; Macaca mulatta; Male; Neuropeptides; Pain; Pruritus; Spinal Cord

2015
Development and characterization of functionalized niosomes for brain targeting of dynorphin-B.
    European journal of pharmaceutics and biopharmaceutics : official journal of Arbeitsgemeinschaft fur Pharmazeutische Verfahrenstechnik e.V, 2014, Volume: 87, Issue:1

    A niosomal formulation, functionalized with N-palmitoylglucosamine, was developed as potential brain targeted delivery system of dynorphin-B. In fact, this endogenous neuropeptide, selective agonist of k opioid receptors, is endowed with relevant pharmacological activities on the central nervous system, including a marked antinociceptive effect, but is unable to cross the blood brain barrier (BBB), thus requiring intracerebroventricular administration. Statistical design of experiments was utilized for a systematic evaluation of the influence of variations of the relative amounts of the components of the vesicle membrane (Span 60, cholesterol and SolulanC24) on vesicle mean diameter, polydispersity index and drug entrapment efficiency, chosen as the responses to optimize. A Scheffé simplex-centroid design was used to obtain the coefficients of the postulated mathematical model. The study of the response surface plots revealed that variations of the considered factors had different effects on the selected responses. The desirability function enabled for finding the optimal mixture composition, which represented the best compromise to simultaneously optimize all the three responses. The experimental values obtained with the optimized formulation were very similar to the predicted ones, proving the validity of the proposed regression model. The optimized niosomal formulation of dynorphin-B administered intravenously to mice (100mg/kg) showed a pronounced antinociceptive effect, significantly higher (P<0.05) than that given by i.v. administration of the simple solution of the peptide at the same concentration, proving its effectiveness in enabling the peptide brain delivery. These positive results suggest that the proposed approach could be successfully extended to other neuro-active peptides exerting a strong central action, even at low doses, but unable to cross the BBB.

    Topics: Analgesics; Animals; Blood-Brain Barrier; Brain; Drug Carriers; Drug Compounding; Drug Delivery Systems; Drug Stability; Dynorphins; Endorphins; Glycolipids; Injections, Intravenous; Injections, Intraventricular; Liposomes; Male; Mice; Pain; Receptors, Opioid, kappa

2014
Effect of intravenous general anaesthesia with epidural block on the expression of pre-endogenous opioid peptide genes.
    The Journal of international medical research, 2014, Volume: 42, Issue:3

    To measure the plasma concentrations of three endogenous opioid peptides and the levels of preproenkephalin (PPE) and preprodynorphin (PPD) mRNA in peripheral blood lymphocytes of patients during scheduled surgery performed under intravenous general anaesthesia combined with an epidural block.. Patients were anaesthetized and arterial blood was collected at 0 (baseline), 20, 40, 60, and 80 min during surgery. The plasma concentrations of β-endorphin, leucine-enkephalin and dynorphin A were measured using radioimmunoassay. Reverse transcription-polymerase chain reaction was used to measure the levels of PPD and PPE mRNA in peripheral blood lymphocytes collected during surgery.. Fifteen patients participated in this prospective study. The plasma concentrations of β-endorphin were significantly lower at all time-points compared with the baseline value. The plasma concentrations of leucine-enkephalin and dynorphin A were significantly lower at 40, 60, and 80 min compared with baseline. The PPD/β-actin ratio was significantly lower at 80 min compared with baseline, while the PPE/β-actin ratio showed no significant change.. The level of mRNA from two pre-endogenous opioid peptide genes either decreased or remained unchanged during surgery under intravenous general anaesthesia with epidural block, suggesting that patients remained pain free during surgery.

    Topics: Abdomen; Adult; Anesthesia, Epidural; Anesthesia, General; Anesthetics, Intravenous; beta-Endorphin; Bupivacaine; Dynorphins; Enkephalin, Leucine; Enkephalins; Female; Fentanyl; Gene Expression; Humans; Male; Midazolam; Middle Aged; Pain; Prospective Studies; Protein Precursors; Radioimmunoassay; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Vecuronium Bromide

2014
Identification of spinal circuits transmitting and gating mechanical pain.
    Cell, 2014, Dec-04, Volume: 159, Issue:6

    Pain information processing in the spinal cord has been postulated to rely on nociceptive transmission (T) neurons receiving inputs from nociceptors and Aβ mechanoreceptors, with Aβ inputs gated through feed-forward activation of spinal inhibitory neurons (INs). Here, we used intersectional genetic manipulations to identify these critical components of pain transduction. Marking and ablating six populations of spinal excitatory and inhibitory neurons, coupled with behavioral and electrophysiological analysis, showed that excitatory neurons expressing somatostatin (SOM) include T-type cells, whose ablation causes loss of mechanical pain. Inhibitory neurons marked by the expression of dynorphin (Dyn) represent INs, which are necessary to gate Aβ fibers from activating SOM(+) neurons to evoke pain. Therefore, peripheral mechanical nociceptors and Aβ mechanoreceptors, together with spinal SOM(+) excitatory and Dyn(+) inhibitory neurons, form a microcircuit that transmits and gates mechanical pain. PAPERCLIP:

    Topics: Animals; Dynorphins; Mechanoreceptors; Mice; Neurons; Pain; Pain Perception; Somatostatin; Spinal Cord

2014
Spinal astrocytes produce and secrete dynorphin neuropeptides.
    Neuropeptides, 2013, Volume: 47, Issue:2

    Dynorphin peptide neurotransmitters (neuropeptides) have been implicated in spinal pain processing based on the observations that intrathecal delivery of dynorphin results in proalgesic effects and disruption of extracellular dynorphin activity (by antisera) prevents injury evoked hyperalgesia. However, the cellular source of secreted spinal dynorphin has been unknown. For this reason, this study investigated the expression and secretion of dynorphin-related neuropeptides from spinal astrocytes (rat) in primary culture. Dynorphin A (1-17), dynorphin B, and α-neoendorphin were found to be present in the astrocytes, illustrated by immunofluorescence confocal microscopy, in a discrete punctate pattern of cellular localization. Measurement of astrocyte cellular levels of these dynorphins by radioimmunoassays confirmed the expression of these three dynorphin-related neuropeptides. Notably, BzATP (3'-O-(4-benzoyl)benzoyl adenosine 5'-triphosphate) and KLA (di[3-deoxy-D-manno-octulosonyl]-lipid A) activation of purinergic and toll-like receptors, respectively, resulted in stimulated secretion of dynorphins A and B. However, α-neoendorphin secretion was not affected by BzATP or KLA. These findings suggest that dynorphins A and B undergo regulated secretion from spinal astrocytes. These findings also suggest that spinal astrocytes may provide secreted dynorphins that participate in spinal pain processing.

    Topics: Adenosine Triphosphate; Animals; Astrocytes; Cells, Cultured; Dynorphins; Endorphins; Extracellular Space; Female; Fluorescent Antibody Technique; Glial Fibrillary Acidic Protein; Immunohistochemistry; Microscopy, Confocal; Neuropeptides; Pain; Pregnancy; Protein Precursors; Rats; Rats, Sprague-Dawley; Receptors, Purinergic P2X; Spinal Cord; Toll-Like Receptor 4

2013
Endogenous endomorphin-2 contributes to spinal ĸ-opioid antinociception.
    Pharmacology, 2012, Volume: 89, Issue:3-4

    Multiple opioid receptor (OR) types and endogenous opioid peptides exist in the spinal dorsal horn and there may be interactions among these receptor types that involve opioid peptides. In a previous study we observed that antinociceptive effects of the selective κ-opioid receptor (κOR) agonist, U50,488H, was attenuated in μ-opioid receptor (μOR) knockout mice as compared to wild-type mice when administered spinally. This suggests that an interaction between κORs and μORs exits in the spinal cord. The present study was aimed at investigating whether endogenous opioid peptides were involved in such interaction.. We examined whether the presence of antibodies to endogenous opioid peptides, endomorphin-2, met-enkephalin and dynorphin A affected the antinociceptive effects of spinal U50,488H in rats. The tail-flick test was used to assess pain thresholds.. The increase in tail-flick latency after spinal U50,488H was attenuated when the rats were pretreated intrathecally with antiserum against endomorphin-2. Pretreatments with antisera against met-enkephalin and dynorphin A had no effect on U50,488H antinociception. The antisera alone did not affect pain threshold.. The results suggest that endomorphin-2, an endogenous opioid peptide highly selective to the μOR, plays a role in antinociception induced by κOR activation in the spinal cord.

    Topics: 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer; Analgesics, Non-Narcotic; Animals; Dynorphins; Enkephalin, Methionine; Male; Oligopeptides; Pain; Rats; Rats, Sprague-Dawley; Receptors, Opioid, kappa; Receptors, Opioid, mu; Spinal Cord

2012
Pain inhibition by blocking leukocytic and neuronal opioid peptidases in peripheral inflamed tissue.
    FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 2012, Volume: 26, Issue:12

    Inflammatory pain can be controlled by endogenous opioid peptides. Here we blocked the degradation of opioids in peripheral injured tissue to locally augment this physiological system. In rats with hindpaw inflammation, inhibitors of aminopeptidase N (APN; bestatin) or neutral endopeptidase (NEP; thiorphan), and a dual inhibitor, NH(2)-CH-Ph-P(O)(OH)CH(2)-CH-CH(2)Ph(p-Ph)-CONH-CH-CH(3)-COOH (P8B), were applied to injured paws. Combined bestatin (1.25-5 mg)/thiorphan (0.2-0.8 mg) or P8B (0.0625-1 mg) alone elevated mechanical nociceptive thresholds to 307 and 227% of vehicle-treated controls, respectively. This analgesia was abolished by antibodies to methionine-enkephalin, leucine-enkephalin, and dynorphin A 1-17, by peripherally restricted and by selective μ-, δ-, and κ-opioid receptor antagonists. Flow cytometry and photospectrometry revealed expression and metabolic activity of APN and NEP on macrophages, granulocytes, and sciatic nerves from inflamed tissue. Radioimmunoassays showed that inhibition of leukocytic APN and NEP by bestatin (5-500 μM)/thiorphan (1-100 μM) combinations or by P8B (1-100 μM) prevented the degradation of enkephalins. Blockade of neuronal peptidases by bestatin (0.5-10 mM)/thiorphan (0.1-5 mM) or by P8B (0.1-10 mM) additionally hindered dynorphin A 1-17 catabolism. Thus, leukocytes and peripheral nerves are important sources of APN and NEP in inflamed tissue, and their blockade promotes peripheral opioid analgesia.

    Topics: Alanine; Amino Acid Sequence; Animals; Antibodies; CD13 Antigens; Dose-Response Relationship, Drug; Dynorphins; Enkephalin, Leucine; Enkephalin, Methionine; Enzyme Inhibitors; Flow Cytometry; Hindlimb; Inflammation; Leucine; Leukocytes; Male; Narcotic Antagonists; Neprilysin; Neurons; Opioid Peptides; Pain; Pain Threshold; Phosphinic Acids; Rats; Rats, Wistar; Receptors, Opioid; Thiorphan

2012
Delayed postoperative latent pain sensitization revealed by the systemic administration of opioid antagonists in mice.
    European journal of pharmacology, 2011, Apr-25, Volume: 657, Issue:1-3

    The long-lasting post-surgical changes in nociceptive thresholds in mice, indicative of latent pain sensitization, were studied. The contribution of kappa opioid and N-methyl-d-aspartate (NMDA) receptors was assessed by the administration of nor-binaltorphimine or MK-801; dynorphin levels in the spinal cord were also determined. Animals underwent a plantar incision and/or a subcutaneous infusion of remifentanil (80μg/kg), and mechanical thresholds (von Frey) were evaluated at different times. On day 21, after complete recovery of mechanical thresholds and healing of the wound, one of the following drugs was administered subcutaneously: (-)-naloxone (1mg/kg), (+)-naloxone (1mg/kg), naloxone-methiodide (3mg/kg), or nor-binaltorphimine (5mg/kg). Another group received subcutaneous MK-801 (0.15mg/kg) before nor-binaltorphimine administration. Dynorphin on day 21 was determined in the spinal cord by immunoassay. In mice receiving remifentanil during surgery, the administration of (-)-naloxone or nor-binaltorphimine induced significant hyperalgesia even 5months after manipulation. Nociceptive thresholds remained unaltered after (+)-naloxone or naloxone-methiodide. On day 21 after manipulation, the administration of MK-801 prevented nor-binaltorphimine-induced hyperalgesia. No changes in dynorphin levels were observed before or after opioid antagonist administration. In conclusion, surgery produced latent pain sensitization evidenced by opioid antagonist-precipitated hyperalgesia. The effect was stereospecific, centrally originated, and mediated by kappa opioid receptors. The blockade of nor-binaltorphimine-induced hyperalgesia by MK-801, suggests that NMDA receptors are also involved. Our results show for the first time that surgery induces latent, long-lasting changes in the processing of nociceptive information that can be induced by non-nociceptive stimuli such as the administration of opioid antagonists.

    Topics: Animals; Dizocilpine Maleate; Dynorphins; Hyperalgesia; Male; Mice; Naloxone; Naltrexone; Narcotic Antagonists; Pain; Pain Threshold; Piperidines; Postoperative Complications; Reaction Time; Receptors, N-Methyl-D-Aspartate; Receptors, Opioid, kappa; Remifentanil; Spinal Cord

2011
Oxytocin in the periaqueductal gray participates in pain modulation in the rat by influencing endogenous opiate peptides.
    Peptides, 2011, Volume: 32, Issue:6

    Periaqueductal gray (PAG) plays a very important role in pain modulation through endogenous opiate peptides including leucine-enkephalin (L-Ek), methionine-enkephalin (M-Ek), β-endorphin (β-Ep) and dynorphin A(1-13) (DynA(1-13)). Our pervious study has demonstrated that intra-PAG injection of oxytocin (OXT) increases the pain threshold, and local administration of OXT receptor antagonist decreases the pain threshold, in which the antinociceptive role of OXT can be reversed by pre-PAG administration of OXT receptor antagonist. The experiment was designed to investigate the effect of OXT on endogenous opiate peptides in the rat PAG during the pain process. The results showed that (1) the concentrations of OXT, L-Ek, M-Ek and β-Ep, not DynA(1-13) in the PAG perfusion liquid were increased after the pain stimulation; (2) the concentrations of L-Ek, M-Ek and β-Ep, not DynA(1-13) in the PAG perfusion liquid were decreased by the OXT receptor antagonist; (3) the increased pain threshold induced by the OXT was attenuated by naloxone, an opiate receptor antagonist; and (4) the concentrations of L-Ek, M-Ek and β-Ep, not DynA(1-13) in the PAG perfusion liquid were increased by exogenous OXT administration. The data suggested that OXT in the PAG could influence the L-Ek, M-Ek and β-Ep rather than DynA(1-13) to participate in pain modulation, i.e. OXT in the PAG participate in pain modulation by influencing the L-Ek, M-Ek and β-Ep rather than DynA(1-13).

    Topics: Animals; beta-Endorphin; Catheterization; Dynorphins; Enkephalin, Leucine; Enkephalin, Methionine; Microinjections; Naloxone; Narcotic Antagonists; Oxytocin; Pain; Pain Measurement; Pain Threshold; Peptide Fragments; Periaqueductal Gray; Radioimmunoassay; Rats; Rats, Sprague-Dawley

2011
Determination of specific neuropeptides modulation time course in a rat model of osteoarthritis pain by liquid chromatography ion trap mass spectrometry.
    Neuropeptides, 2011, Volume: 45, Issue:6

    Animal models are useful to evaluate pharmacological therapies to alleviate joint pain. The present study characterized central neuropeptides modulation in the monoiodoacetate (MIA) rat model. Animals receiving a single 3mg MIA injection were euthanized at 3, 7, 14, 21 and 28 days post injection. Spinal cords were analyzed by liquid chromatography ion trap mass spectrometry. Up-regulations of the calcitonin gene-related peptide and substance P were observed starting on days 7 and 28 respectively, whereas big dynorphin(₁₋₃₂) content decreased significantly on day 14 in comparison to control animals (P<0.05). Preclinical drug evaluations using this model should be conducted between 7 and 21 days post injection when the lesions resemble most to human osteoarthritis.

    Topics: Animals; Calcitonin Gene-Related Peptide; Chromatography, Liquid; Disease Models, Animal; Dynorphins; Enzyme Inhibitors; Humans; Iodoacetic Acid; Knee Joint; Male; Neuropeptides; Osteoarthritis; Pain; Rats; Rats, Sprague-Dawley; Spinal Cord; Substance P; Tandem Mass Spectrometry; Time Factors

2011
Direct evidence for the ongoing brain activation by enhanced dynorphinergic system in the spinal cord under inflammatory noxious stimuli.
    Anesthesiology, 2010, Volume: 112, Issue:2

    Dynorphin A in the spinal cord is considered to contribute to nociceptive stimuli. However, it has not yet been determined whether activation of the spinal dynorphinergic system under nociceptive stimuli plays a role in direct acceleration of the ascending nociceptive pathway. In this study, the authors investigated the role of spinal dynorphinergic transmission in ongoing brain activation under noxious stimuli in mice.. The changes in prodynorphin messenger RNA expression and dynorphin A (1-17)-like immunoreactivity in the mouse spinal cord were determined after the intraplantar injection of complete Freund's adjuvant in mice. The signal intensity in different brain regions after the intraplantar injection of complete Freund's adjuvant or intrathecal injection of dynorphin A (1-17) was measured by a pharmacological functional magnetic resonance imaging analysis.. Complete Freund's adjuvant injection produced pain-associated behaviors and induced a dramatic increase in signal intensity in the mouse cingulate cortex, somatosensory cortex, insular cortex, and thalamic nuclei. These effects were not seen in prodynorphin knockout mice. Prodynorphin messenger RNA expression and dynorphin A (1-17)-like immunoreactivity on the ipsilateral side of the spinal cord were markedly increased in complete Freund's adjuvant-injected mice. Furthermore, intrathecal injection of dynorphin A (1-17) at relatively high doses caused pain-associated behaviors and a remarkable increase in the activities of the cingulate cortex, somatosensory cortex, insular cortex, and medial and lateral thalamic nuclei in mice.. These findings indicate that spinally released dynorphin A (1-17) by noxious stimuli leads to the direct activation of ascending pain transmission.

    Topics: Animals; Behavior, Animal; Brain; Brain Chemistry; Dynorphins; Enkephalins; Freund's Adjuvant; Immunohistochemistry; Inflammation; Injections, Spinal; Magnetic Resonance Imaging; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Pain; Protein Precursors; Receptors, N-Methyl-D-Aspartate; Reverse Transcriptase Polymerase Chain Reaction; RNA; Spinal Cord; Synaptic Transmission

2010
Spinal mediators that may contribute selectively to antinociceptive tolerance but not other effects of morphine as revealed by deletion of GluR5.
    Neuroscience, 2010, Aug-11, Volume: 169, Issue:1

    Several groups maintain that morphine tolerance and dependence correlate with increased activity of protein kinases ERK1/2 and P38 MAPK and PKC as well as elevated levels of the neuropeptides dynorphin (DYN), substance P (sP), and calcitonin gene-related peptide (CGRP) in spinal cord dorsal horn (SCDH). They demonstrate that tolerance and dependence can be prevented, and sometimes reversed, by constitutive genetic deletion or pharmacological inhibition of these factors. Recently, we showed that mice with a constitutive deletion of the GluR5 subunit of kainate receptors (GluR5 KO) are not different from wild type (WT) littermates with respect to baseline nociceptive thresholds as well as acute morphine antinociception, morphine physical dependence and conditioned place preference. However, unlike WT, GluR5 KO mice do not develop antinociceptive tolerance following systemic morphine administration. In this report, we examined levels of these mediators in SCDH of WT and GluR5 KO mice following subcutaneous implantation of placebo or morphine pellets. Surprisingly, spinal DYN and CGRP, along with phosphorylated ERK2 (pERK2), P38 (pP38) and PKCgamma (pPKCgamma) are elevated by deletion of GluR5. Additionally, chronic systemic morphine administration increased spinal pERK2, pP38 and pPKCgamma levels in both tolerant WT and non-tolerant GluR5 KO mice. In contrast, while morphine increased spinal DYN and CGRP in WT mice, DYN remained unchanged and CGRP was reduced in GluR5 KO mice. These observations suggest that spinal ERK2, P38 and PKCgamma are likely involved in multiple adaptive responses following systemic morphine administration, whereas DYN and CGRP may contribute selectively to the development of antinociceptive tolerance.

    Topics: Animals; Calcitonin Gene-Related Peptide; Drug Implants; Drug Tolerance; Dynorphins; Enzyme Activation; Enzyme Induction; Exploratory Behavior; Male; Mice; Mice, Knockout; Mitogen-Activated Protein Kinase 1; Morphine; Morphine Dependence; Narcotics; p38 Mitogen-Activated Protein Kinases; Pain; Pain Threshold; Phosphorylation; Posterior Horn Cells; Protein Kinase C; Protein Processing, Post-Translational; Receptors, Kainic Acid

2010
Neuropathic and chronic pain stimuli downregulate central mu-opioid and dopaminergic transmission.
    Trends in pharmacological sciences, 2010, Volume: 31, Issue:7

    Although morphine and other mu-opioid agonists are the main analgesics for severe pain, these compounds have potential for abuse and/or addiction. This has complicated the use of mu-agonists in the treatment of chronic pain. However, clinical studies show that when mu-agonist analgesics are appropriately used to control pain, actual abuse or addiction does not usually occur, although some risk factors that increase vulnerability need to be considered, including genetic variation. We review recent findings on molecular adaptations in sustained pain models, and propose how these adaptations (including sustained release of the endogenous mu-agonist beta-endorphin) can result in decreased abuse potential of mu-agonists in chronic pain states. We also review data on particular gene polymorphisms (e.g. in the mu-receptor gene) that could also influence the relative abuse potential of mu-agonists in clinical pain populations.

    Topics: Analgesics, Opioid; Chronic Disease; Down-Regulation; Dynorphins; Endorphins; Extracellular Signal-Regulated MAP Kinases; Humans; Neuralgia; Opioid-Related Disorders; Pain; Protein Kinase C; Receptors, Dopamine; Receptors, Opioid, mu; Reward; Up-Regulation; Ventral Tegmental Area

2010
Attenuation of persistent experimental pancreatitis pain by a bradykinin b2 receptor antagonist.
    Pancreas, 2010, Volume: 39, Issue:8

    The role of bradykinin (BK) receptors in activating and sensitizing peripheral nociceptors is well known. Recently, we showed that spinal dynorphin was pronociceptive through direct or indirect BK receptor activation. Here, we explored the potential role of BK receptors in pain associated with persistent pancreatitis in rats.. Experimental pancreatitis and abdominal hypersensitivity were induced by intravenous administrations of dibutyltin dichloride (DBTC). [des-Arg-Leu]BK (B1 antagonist) and HOE 140 (B2 antagonist) were given by intraperitoneal or intrathecal injection. Dynorphin antiserum was given intrathecally. Reverse transcription-polymerase chain reaction was used to detect spinal mRNA for BK receptors.. Dibutyltin dichloride-induced pancreatitis upregulated B1 and B2 mRNA in the thoracic dorsal root ganglion and B2, but not B1, in the pancreas. No changes in spinal B1 or B2 mRNA were observed. Intraperitoneal or intrathecal administration of HOE 140 dose dependently abolished DBTC-induced abdominal hypersensitivity, whereas [des-Arg-Leu]BK was without effect by either route of administration. Antiserum to dynorphin (intrathecal) abolished DBTC-induced hypersensitivity.. These results suggest that blockade of peripheral or spinal BK B2 receptors may be an effective approach for diminishing pain associated with pancreatitis. Moreover, it is suggested that spinal dynorphin may maintain pancreatitis pain through direct or indirect activation of BK B2 receptors in the spinal cord.

    Topics: Abdominal Pain; Animals; Anti-Inflammatory Agents, Non-Steroidal; Bradykinin; Bradykinin B1 Receptor Antagonists; Bradykinin B2 Receptor Antagonists; Dynorphins; Ganglia, Spinal; Gene Expression; Immune Sera; Injections, Intraperitoneal; Injections, Intravenous; Injections, Spinal; Male; Organotin Compounds; Pain; Pancreas; Pancreatitis; Rats; Rats, Sprague-Dawley; Receptor, Bradykinin B1; Receptor, Bradykinin B2; Reverse Transcriptase Polymerase Chain Reaction; Spinal Cord

2010
A prolonged nitric oxide-dependent, opioid-mediated antinociceptive effect of hyperbaric oxygen in mice.
    The journal of pain, 2009, Volume: 10, Issue:2

    Hyperbaric oxygen (HBO(2)) therapy is reported to cause pain relief in several conditions of chronic pain. A single 60-minute session of HBO(2) treatment produced a prolonged antinociceptive effect in mice that persisted for 90 minutes after cessation of treatment. The HBO(2)-induced antinociception was significantly attenuated by pretreatment before HBO(2) exposure with the opioid antagonist naltrexone, the nonspecific nitric oxide synthase (NOS)-inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME), and the selective neuronal NOS-inhibitor S-methyl-L-thiocitrulline (SMTC) but not the selective endothelial NOS-inhibitor N(5)-(1-iminoethyl)-L-ornithine (L-NIO). The antinociception was also significantly reduced by central pretreatment with a rabbit antiserum against dynorphin(1-13) but not by rabbit antisera against either beta-endorphin or methionine-enkephalin. The prolonged antinociceptive effect at 90 minutes after HBO(2)-induced treatment was also significantly attenuated by naltrexone but not L-NAME administered 60 minutes after HBO(2) treatment but before nociceptive testing. These findings indicate that the antinociception that persists for 90 minutes after HBO(2) exposure is mediated by nitric oxide (NO) and opioid mechanisms but that the NO involvement is critical during the HBO(2) treatment and not at the time of nociceptive testing. These results are consistent with the concept that HBO(2) may induce an NO-dependent release of opioid peptide to cause a long-acting antinociceptive effect.. This article presents evidence of a persistent antinociceptive effect of hyperbaric oxygen treatment that is mediated by opioid and NO mechanisms. Further elucidation of the underlying mechanism could identify molecular targets to cause a longer-acting activation of endogenous pain-modulating systems.

    Topics: Acetic Acid; Analgesia; Analysis of Variance; Animals; beta-Endorphin; Citrulline; Dynorphins; Enkephalin, Methionine; Enzyme Inhibitors; Hyperbaric Oxygenation; Injections, Intraperitoneal; Male; Mice; Microinjections; Naltrexone; Narcotic Antagonists; Neurotransmitter Agents; NG-Nitroarginine Methyl Ester; Nitric Oxide; Pain; Pain Measurement; Thiourea

2009
Relationship of spinal dynorphin neurons to delta-opioid receptors and estrogen receptor alpha: anatomical basis for ovarian sex steroid opioid antinociception.
    The Journal of pharmacology and experimental therapeutics, 2008, Volume: 326, Issue:3

    Pharmacological and behavioral studies suggest that spinal delta- and kappa-opioid antinociceptive systems are functionally associated with ovarian sex steroids. These interactions can be demonstrated specifically during pregnancy or hormone-simulated pregnancy (HSP). The analgesia associated with both conditions can be abolished by blockade of either spinal kappa-opioid receptors or delta-opioid receptors (DOR). Furthermore, both dynorphin (DYN) release (J Pharmacol Exp Ther 298:1213-1220, 2001) and the processing of the DYN precursor (J Neurochem 65:1374-1380, 1995) are significantly increased in the spinal cord during HSP. We undertook the current study to determine whether DYN, DOR, and estrogen receptor alpha (ERalpha) share anatomical relationships that permit their direct interaction. Coexpression of DOR or ERalpha by DYN neurons was assessed using fluorescence immunohistochemistry and a synaptosomal release assay. Findings show that ERalpha and DYN are coexpressed. Moreover, in the spinal cord of HSP animals, there were significant increases in the number of DYN-immunoreactive (DYN-ir) cells, ERalpha-ir cells, cells double-labeled for DYN-ir and ERalpha-ir and the proportion of DYN-ir cells coexpressing ERalpha. Some varicose fibers in the spinal cord dorsal horn and intermediate gray matter that expressed DYN-ir also expressed DOR-ir. Activation of DORs located on DYN terminals was sufficient to inhibit K(+)-evoked DYN release. These data define, at least in part, the anatomical substrates that may be relevant to the antinociception of gestation and its hormonal simulation. Furthermore, they provide a framework for understanding sex-based nociception and antinociception and suggest novel strategies for treating pain.

    Topics: Analgesics, Opioid; Animals; Dynorphins; Estrogen Receptor alpha; Female; Gonadal Steroid Hormones; Neurons; Ovary; Pain; Pregnancy; Rats; Rats, Sprague-Dawley; Receptors, Opioid, delta; Spinal Cord

2008
Spinal dynorphin and bradykinin receptors maintain inflammatory hyperalgesia.
    The journal of pain, 2008, Volume: 9, Issue:12

    An upregulation of the endogenous opioid, dynorphin A, in the spinal cord is seen in multiple experimental models of chronic pain. Recent findings implicate a direct excitatory action of dynorphin A at bradykinin receptors to promote hyperalgesia in nerve injured rats, and its upregulation may promote, rather than counteract, enhanced nociceptive input due to injury. Here we examined a model of inflammatory pain by unilateral injection of complete Freund's adjuvant (CFA) into the rat hind paw. Rats exhibited tactile hypersensitivity and thermal hyperalgesia in the inflamed paw by 6 hours after CFA injection, whereas a significant elevation of prodynorphin transcripts in the lumbar spinal cord was seen at day 3 but not at 6 hours. Thermal hyperalgesia at day 3, but not at 6 hours, after CFA injection was blocked by intrathecal administration of anti-dynorphin antiserum or by bradykinin receptor antagonists. The antihyperalgesic effect of the latter was not due to de novo production of bradykinin or upregulation of spinal bradykinin receptors. These data suggest that elevated spinal dynorphin on peripheral inflammation mediates chronic inflammatory hyperalgesia. The antihyperalgesic effect of bradykinin receptor antagonists requires the presence of upregulated spinal dynorphin but not of de novo production of bradykinin, supporting our hypothesis that pathological levels of dynorphin may activate spinal bradykinin receptors to mediate inflammatory hyperalgesia.. This study shows that chronic peripheral inflammation induces a significant upregulation of the endogenous opioid peptide dynorphin. Elevated levels of spinal dynorphin and activation of spinal bradykinin receptors are essential to maintain inflammatory hyperalgesia. The results suggest that blockade of spinal bradykinin receptors may have therapeutic potential in chronic inflammatory pain.

    Topics: Adjuvants, Immunologic; Analysis of Variance; Animals; Bradykinin; Dynorphins; Freund's Adjuvant; Hyperalgesia; Inflammation; Injections, Spinal; Kallidin; Kininogens; Male; Pain; Pain Measurement; Pain Threshold; Radioligand Assay; Rats; Rats, Sprague-Dawley; Receptors, Bradykinin; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Spinal Cord; Tritium; Up-Regulation

2008
Involvement of spinal Met-enkephalin in nicotine-induced antinociception in mice.
    Brain research, 2008, Jan-16, Volume: 1189

    Nicotine is neuronal stimulating drug in the central nervous system and elicits various effects through nicotinic acetylcholine receptors. As previously reported, nicotine has an antinociceptive effect through activation of endogenous opioid neurons. However, detailed mechanisms of nicotine-induced antinociception are uncertain. In this study, we focused on spinal cord and investigated the involvement of endogenous opioidergic neurons in nicotine-induced antinociception in mice. In the tail-pinch test, subcutaneously administered nicotine (5 mg/kg) produced maximal antinociception 0.5 h after nicotine administration; this was attenuated by mecamylamine (MEC, 3 mg/kg, s.c.) or naloxone (NLX, 1 mg/kg, s.c.) administration. Intrathecal nicotine (10 mug) produced maximal antinociception at 2 min and this was also attenuated by MEC (3 mg/kg, s.c.) or NLX (1 mg/kg, s.c.) administration. The preproenkephalin (ppENK) mRNA level in spinal cord, but not dorsal root ganglion, was significantly increased 2 h following nicotine administration and recovered to control level 4 h after nicotine (5 mg/kg, s.c.) administration. This increase in ppENK mRNA level was inhibited by MEC (3 mg/kg, s.c.). The mRNA levels of preprodynorphin and preproopiomelanocortin were not increased by nicotine (5 mg/kg, s.c.). In the dorsal horn of the lumbar spinal cord, methionine-enkephalin (Met-ENK)-like IR was remarkably reduced at 0.5 h following nicotine administration and recovered to control levels by 2 h after nicotine (3 mg/kg, s.c.) administration. These results suggest that nicotine has an antinociceptive effect by promoting the release of Met-ENK, but not dynorphins and endorphins, from activated opioidergic neurons in spinal cord.

    Topics: Analgesics; Animals; Dose-Response Relationship, Drug; Down-Regulation; Dynorphins; Enkephalin, Methionine; Enkephalins; Male; Mecamylamine; Mice; Mice, Inbred ICR; Naloxone; Narcotic Antagonists; Nicotine; Nicotinic Agonists; Nicotinic Antagonists; Nociceptors; Pain; Pain Measurement; Pain Threshold; Posterior Horn Cells; Pro-Opiomelanocortin; Protein Precursors; RNA, Messenger

2008
Role of nociceptin/orphanin FQ and the pseudopeptide [Phe1Psi(CH2NH)Gly2]-nociceptin(1-13)-NH2 and their interaction with classic opioids in the modulation of thermonociception in the land snail Helix aspersa.
    European journal of pharmacology, 2008, Feb-26, Volume: 581, Issue:1-2

    The role in nociception of nociceptin/orphanin FQ (N/OFQ) and its receptor, the opioid receptor-like 1 (NOP), remains unclear because this peptide has been implicated in both suppression and enhancement of nociception. The present work characterises the effects of N/OFQ and the NOP receptor antagonist, the pseudopeptide [Phe(1)Psi(CH(2)NH)Gly(2)]-nociceptin(1-13)-NH(2) (Phe(1)Psi), on thermonociception in the snail Helix aspersa using the hot plate assay. Additionally, the possible interaction of each of these compounds with morphine or dynorphin A(1-17) and naloxone was studied. Compounds were administered into the hemocoel cavity of H. aspersa and the latency to the aversive withdrawal behaviour recorded. Dose-response and time course curves were done. N/OFQ and naloxone produced a similar dose-dependent pronociceptive effect; however, N/OFQ reached its peak effect earlier and was 30 times more potent than naloxone. [Phe(1)Psi(CH(2)NH)Gly(2)]-nociceptin(1-13)-NH(2) and the opioid agonists, morphine and dynorphin A(1-17) produced antinociception with a similar efficacy, but [Phe(1)Psi(CH(2)NH)Gly(2)]-nociceptin(1-13)-NH(2) reached its peak effect more rapidly and lasted longer than that of dynorphin A(1-17) and morphine. [Phe(1)Psi(CH(2)NH)Gly(2)]-nociceptin(1-13)-NH(2) was 50 times less potent than dynorphin A(1-17), but 30 times more potent than morphine. N/OFQ significantly reduced morphine and dynorphin A(1-17)-induced antinociception. Combined administration of low doses of [Phe(1)Psi(CH(2)NH)Gly(2)]-nociceptin(1-13)-NH(2) and morphine or dynorphin A(1-17) produced a potent antinociceptive effect. Sub-effective doses of naloxone and N/OFQ also synergised to produce pronociception. Data suggest that these two opioid classes regulate nociception through parallel systems. The H. aspersa model appears as a valuable experimental preparation to continue the study of these opioid receptor systems.

    Topics: Animals; Dose-Response Relationship, Drug; Drug Interactions; Dynorphins; Helix, Snails; Hot Temperature; Morphine; Motor Activity; Naloxone; Narcotic Antagonists; Nociceptin; Nociceptin Receptor; Opioid Peptides; Pain; Peptide Fragments; Reaction Time; Receptors, Opioid

2008
Spinal NK-1 receptor expressing neurons mediate opioid-induced hyperalgesia and antinociceptive tolerance via activation of descending pathways.
    Pain, 2007, Volume: 129, Issue:1-2

    Opioids can induce hyperalgesia in humans and in animals. Mechanisms of opiate-induced hyperalgesia and possibly of spinal antinociceptive tolerance may be linked to pronociceptive adaptations occurring at multiple levels of the nervous system including activation of descending facilitatory influences from the brainstem, spinal neuroplasticity, and changes in primary afferent fibers. Here, the role of NK-1 receptor expressing cells in the spinal dorsal horn in morphine-induced hyperalgesia and spinal antinociceptive tolerance was assessed by ablating these cells with intrathecal injection of SP-saporin (SP-SAP). Ablation of NK-1 receptor expressing cells prevented (a) morphine-induced thermal and mechanical hypersensitivity, (b) increased touch-evoked spinal FOS expression, (c) upregulation of spinal dynorphin content and (d) the rightward displacement of the spinal morphine antinociceptive dose-response curve (i.e., tolerance). Morphine-induced hyperalgesia and antinociceptive tolerance were also blocked by spinal administration of ondansetron, a serotonergic receptor antagonist. Thus, NK-1 receptor expressing neurons play a critical role in sustained morphine-induced neuroplastic changes which underlie spinal excitability reflected as thermal and tactile hypersensitivity to peripheral stimuli, and to reduced antinociceptive actions of spinal morphine (i.e., antinociceptive tolerance). Ablation of these cells likely eliminates the ascending limb of a spinal-bulbospinal loop that engages descending facilitation and elicits subsequent spinal neuroplasticity. The data may provide a basis for understanding mechanisms of prolonged pain which can occur in the absence of tissue injury.

    Topics: Afferent Pathways; Animals; Drug Tolerance; Dynorphins; Hyperalgesia; Male; Morphine; Neurons; Ondansetron; Pain; Pain Measurement; Pain Threshold; Rats; Rats, Sprague-Dawley; Receptors, Neurokinin-1; Ribosome Inactivating Proteins, Type 1; Saporins; Spinal Cord; Substance P

2007
Nociceptin/orphanin FQ blocks the antinociception induced by mu, kappa and delta opioid agonists on the cold water tail-flick test.
    European journal of pharmacology, 2007, Feb-14, Volume: 557, Issue:1

    Nociceptin/orphanin FQ (N/OFQ), a 17-amino-acid peptide, is an endogenous agonist whose receptor is similar in sequence to mu, delta and kappa opioid receptors. It has been reported that N/OFQ can block antinociceptive effects induced by opioid receptor agonists in the radiant heat tail-flick test and warm water tail-withdrawal test. The present study was designed to see the effect of N/OFQ on antinociception induced by opioid receptor agonists in the cold water tail-flick (CWT) test, which measures a different type of pain. In adult male Sprague-Dawley (S-D) rats given subcutaneous (s.c.) injections of saline or morphine (8 mg/kg), intracerebroventricular (i.c.v.) injection of N/OFQ (18 microg) 15 min later produced a significant reversal of morphine antinociception (P<0.01, ANOVA followed by Duncan's test), compared to the corresponding saline control group. Saline (t=+15 min, i.c.v.) had no effect on s.c. morphine antinociception (P>0.01), compared to the corresponding saline control group. When the kappa opioid receptor agonist spiradoline (80 mg/kg, s.c.) was used instead of morphine, similar results were observed. In another series of experiments, it was found that i.c.v. injection of N/OFQ (18 microg) reversed the antinociception induced by i.c.v. injection of the selective mu opioid agonist PL017 (2 microg), delta opioid agonist DPDPE (50 ng) and kappa opioid agonist dynorphin (21.5 microg), respectively. These results indicate that N/OFQ may be an endogenous anti-opioid peptide in the brain of rats in the CWT test.

    Topics: Analgesics, Opioid; Animals; Cold Temperature; Dynorphins; Endorphins; Enkephalin, D-Penicillamine (2,5)-; Male; Morphine; Nociceptin; Opioid Peptides; Pain; Pain Measurement; Pyrrolidines; Rats; Rats, Sprague-Dawley; Receptors, Opioid, delta; Receptors, Opioid, kappa; Receptors, Opioid, mu; Tail; Water

2007
Nerve growth factor sequestering therapy attenuates non-malignant skeletal pain following fracture.
    Pain, 2007, Dec-15, Volume: 133, Issue:1-3

    Current therapies to treat skeletal fracture pain are extremely limited. Some non-steroidal anti-inflammatory drugs have been shown to inhibit bone healing and opiates induce cognitive dysfunction and respiratory depression which are especially problematic in the elderly suffering from osteoporotic fractures. In the present report, we developed a closed femur fracture pain model in the mouse where skeletal pain behaviors such as flinching and guarding of the fractured limb are reversed by 10mg/kg morphine. Using this model we showed that the administration of a monoclonal antibody against nerve growth factor (anti-NGF) reduced fracture-induced pain-related behaviors by over 50%. Treatment with anti-NGF reduced c-Fos and dynorphin up-regulation in the spinal cord at day 2 post-fracture. However, anti-NGF treatment did not reduce p-ERK and c-Fos expression at 20 and 90 min, respectively, following fracture. This suggests NGF is involved in maintenance but not the acute generation of fracture pain. Anti-NGF therapy did not inhibit bone healing as measured by callus formation, bridging of the fracture site or mechanical strength of the bone. As the anti-NGF antibody does not appreciably cross the blood-brain barrier, the present data suggest that the anti-hyperalgesic action of anti-NGF therapy results from blockade of activation and/or sensitization of the CGRP/trkA positive fibers that normally constitute the majority of sensory fibers that innervate the bone. These results demonstrate that NGF plays a significant role in driving fracture pain and that NGF sequestering therapies may be efficacious in attenuating this pain.

    Topics: Animals; Antibodies; Antigens, CD; Antigens, Differentiation, Myelomonocytic; Disease Models, Animal; Dynorphins; Exodeoxyribonucleases; Femoral Fractures; Male; Mice; Mice, Inbred C3H; Nerve Fibers; Nerve Growth Factors; Pain; Proto-Oncogene Proteins c-fos; Radiography; Time Factors; Trans-Activators; Tyrosine 3-Monooxygenase; Up-Regulation

2007
Chronic pain induces anxiety with concomitant changes in opioidergic function in the amygdala.
    Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology, 2006, Volume: 31, Issue:4

    Clinically, it has been reported that chronic pain induces depression, anxiety, and reduced quality of life. The endogenous opioid system has been implicated in nociception, anxiety, and stress. The present study was undertaken to investigate whether chronic pain could induce anxiogenic effects and changes in the opioidergic function in the amygdala in mice. We found that either injection of complete Freund's adjuvant (CFA) or neuropathic pain induced by sciatic nerve ligation produced a significant anxiogenic effect at 4 weeks after the injection or surgery. Under these conditions, the selective mu-opioid receptor agonist [D-Ala2,N-MePhe4,Gly5-ol]-enkephalin (DAMGO)- and the selective delta-opioid receptor agonist (+)-4-[(alphaR)-alpha-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide (SNC80)-stimulated [35S]GTPgammaS binding in membranes of the amygdala was significantly suppressed by CFA injection or nerve ligation. CFA injection was associated with a significant increase in the kappa-opioid receptor agonist 2-(3,4-dichlorophenyl)-N-methyl-N-[(1S)-1-phenyl-2-(1-pyrrolidinyl)ethyl]acetamide hydrochloride (ICI199,441)-stimulated [35S]GTPgammaS binding in membranes of the amygdala. The intracerebroventricular administration and microinjection of a selective mu-opioid receptor antagonist, a selective delta-opioid receptor antagonist, and the endogenous kappa-opioid receptor ligand dynorphin A caused a significant anxiogenic effect in mice. We also found that thermal hyperalgesia induced by sciatic nerve ligation was reversed at 8 weeks after surgery. In the light-dark test, the time spent in the lit compartment was not changed at 8 weeks after surgery. Collectively, the present data constitute the first evidence that chronic pain has an anxiogenic effect in mice. This phenomenon may be associated with changes in opioidergic function in the amygdala.

    Topics: Amygdala; Analgesics, Opioid; Analysis of Variance; Animals; Anxiety; Behavior, Animal; Benzamides; Chronic Disease; Diazepam; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Interactions; Dynorphins; Enkephalin, Ala(2)-MePhe(4)-Gly(5)-; Freund's Adjuvant; Guanosine 5'-O-(3-Thiotriphosphate); Injections, Intraventricular; Male; Maze Learning; Mice; Mice, Inbred C57BL; Naltrexone; Narcotic Antagonists; Narcotics; Pain; Pain Measurement; Piperazines; Protein Binding; Pyrrolidines; Rats; Rats, Sprague-Dawley; Reaction Time; Sciatica; Somatostatin; Sulfur Isotopes; Time Factors; Tranquilizing Agents

2006
Opioid receptor-mediated hyperalgesia and antinociceptive tolerance induced by sustained opiate delivery.
    Neuroscience letters, 2006, Mar-20, Volume: 396, Issue:1

    Opiates are commonly used to treat moderate to severe pain and can be used over prolonged periods in states of chronic pain such as those associated with cancer. In addition, to analgesic actions, studies show that opiate administration can paradoxically induce hyperalgesia. At the pre-clinical level, such hyperalgesia is associated with numerous pronociceptive neuroplastic changes within the primary afferent fibers and the spinal cord. In rodents, sustained opiate administration also induces antinociceptive tolerance. The mechanisms by which prolonged opiate exposure induces hyperalgesia and the relationship of this state to antinociceptive tolerance remain unclear. The present study was aimed at determining whether sustained opiate-induced hyperalgesia, associated neuroplasticity and antinociceptive tolerance are the result of specific opiate interaction at opiate receptors. Enantiomers of oxymorphone, a mu opioid receptor agonist, were administered to rats by spinal infusion across 7 days. Sustained spinal administration of (-)-oxymorphone, but not its inactive enantiomer (+)-oxymorphone or vehicle, upregulated spinal dynorphin content, produced thermal and tactile hypersensitivity, and produced antinociceptive tolerance. These results indicate that these pronociceptive actions of sustained opiate administration require specific interaction with opiate receptors and are unlikely to be the result of accumulation of potentially excitatory metabolic products. While the precise mechanisms, which may account for these pronociceptive changes remain to be unraveled, the present data point to plasticity initiated by opiate receptor interaction.

    Topics: Afferent Pathways; Animals; Central Nervous System; Disease Models, Animal; Drug Administration Schedule; Drug Tolerance; Dynorphins; Hyperalgesia; Isomerism; Male; Narcotics; Neuronal Plasticity; Nociceptors; Oxymorphone; Pain; Pain Measurement; Pain Threshold; Rats; Rats, Sprague-Dawley; Receptors, Opioid; Receptors, Opioid, mu; Spinal Nerve Roots; Up-Regulation

2006
Possible involvement of dynorphin A-(1-17) release via mu1-opioid receptors in spinal antinociception by endomorphin-2.
    The Journal of pharmacology and experimental therapeutics, 2006, Volume: 317, Issue:1

    The antinociception induced by i.t. or i.c.v. administration of endomorphins is mediated via mu-opioid receptors. However, although endomorphins do not have an appreciable affinity for kappa-opioid receptors, pretreatment with the kappa-opioid receptor antagonist norbinaltorphimine markedly reduces the antinociceptive response to i.c.v. or i.t. administered endomorphin-2 but not endomorphin-1. These results suggest that endomorphin-2 initially stimulates mu-opioid receptors, which subsequently induce the release of dynorphins that act on kappa-opioid receptors to produce antinociception. The present study was performed in mice to determine whether the release of dynorphins by i.t. administered endomorphin-2 is mediated through mu-opioid receptors to produce antinociception. Intrathecal pretreatment with an antiserum against dynorphin A-(1-17), but not against dynorphin B-(1-13) or alpha-neoendorphin, dose-dependently prevented the paw-withdrawal inhibition by endomorphin-2. The pretreatments with these antisera did not affect the endomorphin-1- or [D-Ala(2),MePhe(4),Gly(ol)(5)]enkephalin-induced paw-withdrawal inhibition. The attenuation of endomorphin-2-induced antinociception by i.t. pretreatment with an antiserum against dynorphin A-(1-17) or s.c. pretreatment with norbinaltorphimine was blocked dose-dependently by s.c. pretreatment with the mu-opioid receptor antagonist beta-funaltrexamine or the mu(1)-opioid receptor antagonist naloxonazine at ultra-low doses that are ineffective against mu-opioid receptor agonists. These results suggest that the spinal antinociception induced by endomorphin-2 is mediated through the stimulation of a distinct subtype of mu(1)-opioid receptor that induces the release of the endogenous kappa-opioid peptide dynorphin A-(1-17) in the spinal cord.

    Topics: Analgesics, Opioid; Animals; Dose-Response Relationship, Drug; Dynorphins; Hot Temperature; Immune Sera; Injections, Spinal; Male; Mice; Mice, Inbred Strains; Oligopeptides; Pain; Pain Measurement; Physical Stimulation; Receptors, Opioid, mu; Time Factors

2006
Only arginine vasopressin, not oxytocin and endogenous opiate peptides, in hypothalamic paraventricular nucleus play a role in acupuncture analgesia in the rat.
    Brain research bulletin, 2006, Feb-15, Volume: 68, Issue:6

    Our previous study proved that hypothalamic paraventricular nucleus (PVH) plays an important role in acupuncture analgesia. The effect of acupuncture on the concentrations of arginine vasopressin (AVP), oxytocin (OXT), leucine-enkephaline (L-Ek), beta-endorphin (beta-Ep) and dynorphinA(1-13) (DynA(1-13)) was investigated in rat PVH. Electrical acupuncture of "Zusanli" points (St. 36) 30 min increased the AVP, not OXT, L-Ek, beta-Ep and DynA(1-13) concentrations in PVH tissue using micropunch and radioimmunoassay, which showed a negative relationship between the pain threshold and AVP concentrations in PVH tissue. Electrical acupuncture could elevate the AVP concentrations in PVH perfuse liquid during acupuncture, and then reduce the AVP concentrations in PVH perfuse liquid after acupuncture. But no change in OXT, L-Ek, beta-Ep and DynA(1-13) concentrations was detected in PVH perfuse liquid. Electrical acupuncture decreased the number of AVP, not OXT, L-Ek, beta-Ep and DynA(1-13) immunoreactive cells in PVH using immunocytochemistry. The results suggested that only AVP, not OXT and endogenous opiate peptides in PVH involved acupuncture analgesia in the rat.

    Topics: Acupuncture Analgesia; Afferent Pathways; Animals; Arginine Vasopressin; beta-Endorphin; Dynorphins; Electric Stimulation; Enkephalin, Leucine; Extracellular Fluid; Hypothalamo-Hypophyseal System; Immunohistochemistry; Male; Neurons; Opioid Peptides; Oxytocin; Pain; Paraventricular Hypothalamic Nucleus; Radioimmunoassay; Rats; Rats, Sprague-Dawley; Up-Regulation

2006
Big dynorphin as a putative endogenous ligand for the kappa-opioid receptor.
    Journal of neurochemistry, 2006, Volume: 97, Issue:1

    The diversity of peptide ligands for a particular receptor may provide a greater dynamic range of functional responses, while maintaining selectivity in receptor activation. Dynorphin A (Dyn A), and dynorphin B (Dyn B) are endogenous opioid peptides that activate the kappa-opioid receptor (KOR). Here, we characterized interactions of big dynorphin (Big Dyn), a 32-amino acid prodynorphin-derived peptide consisting of Dyn A and Dyn B, with human KOR, mu- (hMOR) and delta- (hDOR) opioid receptors and opioid receptor-like receptor 1 (hORL1) expressed in cells transfected with respective cDNA. Big Dyn and Dyn A demonstrated roughly similar affinity for binding to hKOR that was higher than that of Dyn B. Dyn A was more selective for hKOR over hMOR, hDOR and hORL1 than Big Dyn, while Dyn B demonstrated low selectivity. In contrast, Big Dyn activated G proteins through KOR with much greater potency, efficacy and selectivity than other dynorphins. There was no correlation between the rank order of the potency for the KOR-mediated activation of G proteins and the binding affinity of dynorphins for KOR. The rank of the selectivity for the activation of G proteins through hKOR and of the binding to this receptor also differed. Immunoreactive Big Dyn was detected using the combination of radioimmunoassay (RIA) and HPLC in the human nucleus accumbens, caudate nucleus, hippocampus and cerebrospinal fluid (CSF) with the ratio of Big Dyn and Dyn B being approximately 1:3. The presence in the brain implies that Big Dyn, along with other dynorphins, is processed from prodynorphin and secreted from neurons. Collectively, the high potency and efficacy and the relative abundance suggest that Big Dyn may play a role in the KOR-mediated activation of G proteins.

    Topics: Animals; Binding, Competitive; Central Nervous System; Cerebrospinal Fluid; Dynorphins; Endorphins; Guanosine 5'-O-(3-Thiotriphosphate); Humans; Ligands; Mice; Mice, Knockout; Neural Pathways; Neurons; Nociceptin Receptor; Pain; Radioimmunoassay; Radioligand Assay; Receptors, Opioid; Receptors, Opioid, delta; Receptors, Opioid, kappa; Receptors, Opioid, mu

2006
Opioid, cheating on its receptors, exacerbates pain.
    Nature neuroscience, 2006, Volume: 9, Issue:12

    Opioids are commonly thought of as compounds that alleviate pain. A new study finds that elevated levels of the opioid dynorphin can unexpectedly activate bradykinin receptors, contributing to the maintenance of neuropathic pain.

    Topics: Animals; Dynorphins; Ganglia, Spinal; Humans; Pain; Pain Threshold; Receptors, Bradykinin; Signal Transduction

2006
Direct evidence for the involvement of the mesolimbic kappa-opioid system in the morphine-induced rewarding effect under an inflammatory pain-like state.
    Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology, 2005, Volume: 30, Issue:1

    Recent clinical studies have demonstrated that when morphine is used to control pain in cancer patients, psychological dependence is not a major concern. The present study was undertaken to ascertain the modulation of psychological dependence on morphine under a chronic pain-like state in rats. The prototypical mu-opioid receptor agonist morphine (8 mg/kg, i.p.) induced a dose-dependent place preference. In the present study, we found that an inflammatory pain-like state following formalin injection significantly suppressed the morphine-induced rewarding effect. This effect was almost reversed by s.c. pretreatment with the kappa-opioid receptor antagonist nor-binaltorphimine (nor-BNI, 5 mg/kg). Furthermore, the morphine-induced increase in dopamine (DA) turnover in the limbic forebrain was significantly inhibited by treatment with formalin. This inhibition was also suppressed by pretreatment with nor-BNI. In addition, in vivo microdialysis studies clearly showed that the morphine-induced increase in the extracellular levels of DA and its metabolites, 3,4-dihydroxyphenylacetic acid and homovanillic acid, in the nucleus accumbens (N.Acc.) was significantly decreased in rats that had been pretreated with formalin. This effect was in turn reversed by the microinjection of a specific dynorphin A antibody into the N.Acc. These findings suggest that the inflammatory pain-like state induced by formalin injection may have caused a sustained activation of the kappa-opioidergic system within the N.Acc., resulting in suppression of the morphine-induced rewarding effect in rats. The present study provides further evidence of the clinical usefulness of morphine in patients suffering from severe pain.

    Topics: Animals; Brain Chemistry; Conditioning, Operant; Dopamine; Dynorphins; Edema; Foot; Formaldehyde; Inflammation; Limbic System; Male; Microdialysis; Microinjections; Morphine; Naltrexone; Narcotic Antagonists; Narcotics; Pain; Pain Measurement; Rats; Rats, Sprague-Dawley; Receptors, Opioid, kappa; Reward

2005
Effects of synchronous or asynchronous electroacupuncture stimulation with low versus high frequency on spinal opioid release and tail flick nociception.
    Experimental neurology, 2005, Volume: 192, Issue:1

    Electroacupuncture stimulation (EAS) is known to change brain neurotransmitter release. In the present study, we investigated the effects of synchronous or asynchronous electroacupuncture stimulation with low versus high frequency on spinal opioid release and tail flick nociception. Rats were given "2/100 Hz" EAS, which stands for an asynchronous mode of stimulation, in which 2 Hz was alternated with 100 Hz, each lasting for 3 s, or "(2 + 100) Hz" EAS, a mode of stimulation in which 2 Hz stimulation was applied to the left hind leg simultaneously with 100 Hz stimulation on the right hind leg. The rats were subjected to the same total number of electrical stimulations in these two modes. Results were as follows: (1) 2/100 Hz EAS was 40% more potent than (2 + 100) Hz EAS (P < 0.01) in producing an anti-nociceptive effect. (2) Intrathecal (i.t.) injection of the mu-opioid receptor antagonist D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr amide (CTOP) blocked in a dose-dependent manner the anti-nociceptive effect produced by 2/100 Hz EAS but not by (2 + 100) Hz EAS, whereas i.t. injection of the kappa-opioid receptor antagonist norbinaltorphimide (Nor-BNI) blocked the anti-nociceptive effect induced by both modes of EAS. (3) I.t. injection of endomorphin-2 antiserum blocked in a dose-dependent manner the anti-nociceptive effect of 2/100 Hz EAS but not that of (2 + 100) Hz EAS, whereas i.t. injection of dynorphin antiserum blocked the anti-nociceptive effect induced by both modes of stimulation. (4) 2/100 Hz EAS increased the release of both endomorphin-2 and dynorphin, whereas (2 + 100) Hz EAS increased the release of dynorphin but not of endmorphin-2. We conclude that the more potent anti-nociceptive effect induced by 2/100 Hz EAS, as compared with that of (2 + 100) Hz EAS, was due, at least partly, to the synergistic interaction of endomorphin-2 and dynorphin in rat spinal cord.

    Topics: Afferent Pathways; Animals; beta-Endorphin; Dose-Response Relationship, Drug; Drug Synergism; Dynorphins; Electroacupuncture; Female; Narcotic Antagonists; Narcotics; Neurons, Afferent; Nociceptors; Pain; Pain Management; Pain Measurement; Rats; Rats, Wistar; Receptors, Opioid, kappa; Receptors, Opioid, mu; Reflex; Spinal Cord

2005
c-Fos antisense oligodeoxynucleotide offsets behavioral nociceptive responses and both up-regulations of c-Fos protein and dynorphin a (1-8) in dorsal horn: a study using the formalin test in rats.
    The International journal of neuroscience, 2005, Volume: 115, Issue:7

    The formalin test was used to elicit acute and chronic pain in rats, and antisense oligodeoxynucleotide (AS-ODN) was used as a tool to modulate the expression of nociceptive behavioral and neurochemical responses. AS-ODN complementary to c-Fos mRNA was administered intrathecally (i.t.) 4 h before formalin injection in the experimental group. Normal saline or reverse AS-ODN was pre-administered i.t. at the same time in two control groups (saline and reverse AS-ODN). The results showed that the acute phase of nociceptive behavior showed no change by AS-ODN administration, whereas the tonic phase of nociceptive licking and biting behavior was significantly suppressed by AS-ODN as compared with the saline or the reverse AS-ODN group, respectively (p < .05 and p < .01). At the same time, both Fos-like immunoreactive (FLI) neurons and density of dynorphin-like immunoreactivities (DLI) were decreased significantly (p < .05 and p < .01) in the AS-ODN group as compared with that in two control groups. The results indicate that the long-lasting nociceptive responses elicited by sustained noxious inputs are based on the up-regulation of c-Fos gene expression, which in turn induces the upregulation of Dyn A production. It is proposed that intensified Dyn A production in the dorsal horn may be pivotal for the appearance of chronic pain.

    Topics: Animals; Behavior, Animal; Dynorphins; Female; Formaldehyde; Injections, Spinal; Male; Oligodeoxyribonucleotides, Antisense; Pain; Pain Threshold; Peptide Fragments; Posterior Horn Cells; Proto-Oncogene Proteins c-fos; Rats; Rats, Wistar; RNA, Messenger; Up-Regulation

2005
Development of highly potent and selective dynorphin A analogues as new medicines.
    The journal of peptide research : official journal of the American Peptide Society, 2005, Volume: 66, Issue:5

    Dynorphin A (Dyn A), a 17 amino acid peptide H-Tyr-Gly-Gly-Phe-Leu-Arg-Arg-Ile-Arg-Pro-Lys-Leu-Lys-Trp-Asp-Asn-Gln-OH, is a potent opioid peptide which interacts preferentially with kappa-opioid receptors. Research in the development of selective and potent opioid peptide ligands for the kappa-receptor is important in mediating analgesia. Several cyclic disulphide bridge-containing peptide analogues of Dyn A, which were conformationally constrained in the putative message or address segment of the opioid ligand, were designed, synthesized and assayed. To further investigate the conformational and topographical requirements for the residues in positions 5 and 11 of these analogues, a systematic series of Dyn A(1-11)-NH2 cyclic analogues incorporating the sulphydryl-containing amino acids L- and D-Cys and L- and D-Pen in positions 5 and 11 were synthesized and assayed. Cyclic lactam peptide analogues were also synthesized and assayed. Several of these cyclic analogues, retained the same affinity and selectivity (vs. the mu- and delta-receptors) as the parent Dyn A(1-11)-NH2 peptide in the guinea-pig brain (GPB), but exhibited a much lower activity in the guinea-pig ileum (GPI), thus leading to centrally vs. peripherally selective peptides. Studies of the structure-activity relationship of Dyn A peptide provide new insights into the importance of each amino acid residue (and their configurations) in Dyn A analogues for high potency and good selectivity at kappa-opioid receptors. We report herein the progress towards the development of Dyn A peptide ligands, which can act as agonists or antagonists at cell surface receptors that modulate cell function and animal behaviour using various approaches to rational peptide ligand-based drug design.

    Topics: Animals; Biological Assay; Brain; Capsaicin; Drug Design; Dynorphins; Guinea Pigs; Ileum; Ligands; Macaca mulatta; Mice; Muscle, Smooth; Opioid Peptides; Pain; Peptides; Protein Conformation; Receptors, Cell Surface; Species Specificity

2005
Nonopioidergic mechanism mediating morphine-induced antianalgesia in the mouse spinal cord.
    The Journal of pharmacology and experimental therapeutics, 2004, Volume: 310, Issue:1

    Intrathecal (i.t.) pretreatment with a low dose (0.3 nmol) of morphine causes an attenuation of i.t. morphine-produced analgesia; the phenomenon has been defined as morphine-induced antianalgesia. The opioid-produced analgesia was measured with the tail-flick (TF) test in male CD-1 mice. Intrathecal pretreatment with low dose (0.3 nmol) of morphine time dependently attenuated i.t. morphine-produced (3.0 nmol) TF inhibition and reached a maximal effect at 45 min. Intrathecal pretreatment with morphine (0.009-0.3 nmol) for 45 min also dose dependently attenuated morphine-produced TF inhibition. The i.t. morphine-induced antianalgesia was dose dependently blocked by the nonselective mu-opioid receptor antagonist (-)-naloxone and by its nonopioid enantiomer (+)-naloxone, but not by endomorphin-2-sensitive mu-opioid receptor antagonist 3-methoxynaltrexone. Blockade of delta-opioid receptors, kappa-opioid receptors, and N-methyl-D-aspartate (NMDA) receptors by i.t. pretreatment with naltrindole, nor-binaltorphimine, and (-)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine maleate (MK-801), respectively, did not affect the i.t. morphine-induced antianalgesia. Intrathecal pretreatment with antiserum against dynorphin A(1-17), [Leu]-enkephalin, [Met]-enkephalin, beta-endorphin, cholecystokinin, or substance P also did not affect the i.t. morphine-induced antianalgesia. The i.t. morphine pretreatment also attenuated the TF inhibition produced by opioid muagonist [D-Ala2, N-Me-Phe4,Gly-ol5]-enkephalin, delta-agonist deltorphin II, and kappa-agonist U50,488H. It is concluded that low doses (0.009-0.3 nmol) of morphine given i.t. activate an antianalgesic system to attenuate opioid mu-, delta-, and kappa-agonist-produced analgesia. The morphine-induced antianalgesia is not mediated by the stimulation of opioid mu-, delta-, or kappa-receptors or NMDA receptors. Neuropeptides such as dynorphin A(1-17), [Leu]-enkephalin, [Met]-enkephalin, beta-endorphin, cholecystokinin, and substance P are not involved in this low-dose morphine-induced antianalgesia.

    Topics: Analgesia; Animals; beta-Endorphin; Dizocilpine Maleate; Drug Interactions; Drug Tolerance; Dynorphins; Enkephalins; Male; Mice; Morphine; Naloxone; Naltrexone; Oligopeptides; Pain; Pain Measurement; Receptors, Opioid, delta; Receptors, Opioid, kappa; Receptors, Opioid, mu; Spinal Cord; Substance P

2004
The antinociceptive effect of Delta9-tetrahydrocannabinol in the arthritic rat.
    European journal of pharmacology, 2004, Jun-16, Volume: 493, Issue:1-3

    Our study addressed the hypothesis that spinal release of endogenous opioids underlies Delta9-tetrahydrocannabinol (Delta9-THC)-induced antinociception in Freund's adjuvant-induced arthritic and nonarthritic rats. The paw-pressure test was used to assess the antinociceptive effects of Delta9-THC versus those of morphine, and opioid and cannabinoid receptor-selective antagonists were used to characterize the involved receptors. Cerebrospinal fluid was collected after Delta9-THC injection (i.p.) for the measurement of endogenous opioid peptides. Our results indicate that morphine or Delta9-THC is equally potent and efficacious in both nonarthritic and arthritic rats. Delta9-THC-induced antinociception is attenuated by the kappa opioid receptor antagonist, nor-binaltorphimine, in arthritic rats only. Delta9-THC induces increased immunoreactive dynorphin A (idyn A) levels in nonarthritic rats while decreasing idyn A in arthritic rats. We hypothesize that the elevated idyn A level in arthritic rats contributes to hyperalgesia by interaction with N-methyl-D-aspartate receptors, and that Delta9-THC induces antinociception by decreasing idyn A release.

    Topics: Animals; Arthritis, Experimental; Cannabinoid Receptor Antagonists; Dose-Response Relationship, Drug; Dronabinol; Dynorphins; Enkephalin, Leucine; Enkephalin, Methionine; Freund's Adjuvant; Injections, Intradermal; Injections, Intraperitoneal; Male; Morphine; Mycobacterium; Naloxone; Naltrexone; Narcotic Antagonists; Pain; Pain Measurement; Piperidines; Pyrazoles; Rats; Rats, Sprague-Dawley; Receptor, Cannabinoid, CB1; Receptors, Opioid; Rimonabant

2004
Endothelin and the tumorigenic component of bone cancer pain.
    Neuroscience, 2004, Volume: 126, Issue:4

    Tumors including sarcomas and breast, prostate, and lung carcinomas frequently grow in or metastasize to the skeleton where they can induce significant bone remodeling and cancer pain. To define products that are released from tumors that are involved in the generation and maintenance of bone cancer pain, we focus here on endothelin-1 (ET-1) and endothelin receptors as several tumors including human prostate and breast have been shown to express high levels of ETs and the application of ETs to peripheral nerves can induce pain. Here we show that in a murine osteolytic 2472 sarcoma model of bone cancer pain, the 2472 sarcoma cells express high levels of ET-1, but express low or undetectable levels of endothelin A (ETAR) or B (ETBR) receptors whereas a subpopulation of sensory neurons express the ETAR and non-myelinating Schwann cells express the ETBR. Acute (10 mg/kg, i.p.) or chronic (10 mg/kg/day, p.o.) administration of the ETAR selective antagonist ABT-627 significantly attenuated ongoing and movement-evoked bone cancer pain and chronic administration of ABT-627 reduced several neurochemical indices of peripheral and central sensitization without influencing tumor growth or bone destruction. In contrast, acute treatment (30 mg/kg, i.p.) with the ETBR selective antagonist, A-192621 increased several measures of ongoing and movement evoked pain. As tumor expression and release of ET-1 has been shown to be regulated by the local environment, location specific expression and release of ET-1 by tumor cells may provide insight into the mechanisms that underlie the heterogeneity of bone cancer pain that is frequently observed in humans with multiple skeletal metastases.

    Topics: Analysis of Variance; Animals; Atrasentan; Behavior, Animal; Bone Neoplasms; Calcitonin Gene-Related Peptide; Disease Models, Animal; Dynorphins; Endothelin Receptor Antagonists; Endothelin-1; Ganglia, Spinal; Gene Expression Regulation, Neoplastic; Glial Fibrillary Acidic Protein; Immunohistochemistry; Male; Mice; Mice, Inbred Strains; Pain; Pain Measurement; Pyrrolidines; Receptors, Endothelin; Sarcoma; Sciatic Nerve; Time Factors

2004
Delta-opioid agonist SNC80 can attenuate the development of dynorphin A-induced tactile allodynia in rats.
    Anesthesiology, 2004, Volume: 101, Issue:2

    Topics: Animals; Benzamides; Catheterization; Dizocilpine Maleate; Dose-Response Relationship, Drug; Dynorphins; Excitatory Amino Acid Antagonists; Injections, Spinal; Male; Nociceptors; Pain; Physical Stimulation; Piperazines; Rats; Rats, Sprague-Dawley; Receptors, Opioid, delta

2004
[MK-801 suppresses dynorphin A (1-17)-induced facilitation of nociceptive responses to formalin in rats].
    Sheng li xue bao : [Acta physiologica Sinica], 2003, Feb-25, Volume: 55, Issue:1

    To explore the facilitation of nociceptive response by dynorphin (Dyn ) A in a model of formalin test in rats, the effects of single intrathecal injection (i.t.) of normal saline (NS), MK-801 (antagonist of NMDA receptor), naloxone (antagonist of opioid receptor), or Dyn A (1-17) were observed, and the effects of i.t. MK-801 or naloxone followed by i.t. Dyn A (1-17) were observed as well. The nociceptive licking and biting induced by injection of formalin exhibited two phases. The first phase lasted for a relatively short period of 3-9 min, and the second phase lasted for a relatively longer period after a 3 to 6- min quietness. The results showed that there were no differences in the first phase in all groups; however, there were differences in the second phase as follows: (1) the duration of nociceptive response was significantly increased in Dyn A (1-17) group (489.5+/-22.5 s) as compared to that of NS group (344.7+/-12.9 s), MK-801 group (331.4+/-20.7 s) or naloxone group (352.5+/-18.4 s) (P<0.01 in three cases); (2) the duration of nociceptive response was significantly shortened in MK-801 plus Dyn A (1-17) group (285.7+/-19.4 s) as compared to that of Dyn A (1-17) group (P<0.01), but there were no significant differences as compared to that of MK-801 group; and (3) there was no significant difference in the second phase between naloxone plus Dyn A (1-17) group (473.8+/-17.8 s) and Dyn A (1-17) group, but the duration of nociceptive response was longer than that of NS group or naloxone group (P<0.01 in both). The results obtained suggest: (1) at the spinal cord, Dyn A (1-17) facilitates nociceptive responses; (2) NMDA receptors, but not opioid receptors, are possibly involved in the nociception by Dyn A (1-17).

    Topics: Animals; Dizocilpine Maleate; Dynorphins; Formaldehyde; Injections, Spinal; Naloxone; Nociceptors; Pain; Rats; Receptors, N-Methyl-D-Aspartate

2003
Topical capsaicin-induced allodynia in unanesthetized primates: pharmacological modulation.
    The Journal of pharmacology and experimental therapeutics, 2003, Volume: 306, Issue:3

    Topically administered capsaicin produces thermal allodynia, and this effect has been used to investigate pain transduction and its pharmacological modulation. This study investigated the parameters of topical capsaicin-induced thermal allodynia in unanesthetized rhesus monkeys and its pharmacological modulation by centrally acting compounds [a kappa-opioid agonist: (5alpha,7alpha,8beta)-(+)-N-methyl-N-(7-[1-pyrrolidinyl]-1-oxaspiro [4.5]dec-8-yl)-benzeneacetamide (U69,593); and noncompetitive N-methyl-d-aspartate (NMDA) antagonists: ketamine and MK-801 (dizocilpine)]. Rhesus monkeys (n = 4) were studied within the warm water tail withdrawal assay (20-s maximum latency), using thermal stimuli that are normally not noxious (38 and 42 degrees C). Capsaicin was applied topically on the tail (0.0013 and 0.004 M capsaicin solution on a 1-cm2 patch; 15-min contact). Topical capsaicin produced concentration-dependent thermal allodynia in both temperatures, robustly detected 15 to 90 min after topical capsaicin removal. A similar allodynic profile was observed with topical administration of the "endovanilloid" N-arachidonoyl-dopamine. The kappa-agonist U69,593 (0.01-0.1 mg/kg, s.c.) dose dependently prevented capsaicin (0.004 M)-induced allodynia in 38 and 42 degrees C, and the largest U69,593 dose also reversed ongoing allodynia within this model. Two NMDA antagonists, ketamine and MK-801 (0.32-1.8 and 0.032-0.056 mg/kg, respectively), also prevented capsaicin-induced allodynia in 38 degrees C, but only variably in 42 degrees C, at doses that did not cause robust thermal antinociceptive effects. At the largest doses studied, ketamine but not MK-801 also briefly reversed ongoing capsaicin-induced allodynia. The present model of topical capsaicin administration may be used to study antiallodynic effects of opioid and nonopioid compounds, as well as their ability to prevent and reverse allodynia, in unanesthetized nonhuman primates in the absence of tissue disruption.

    Topics: Aminobutyrates; Analgesics; Animals; Benzeneacetamides; Capsaicin; Disease Models, Animal; Dizocilpine Maleate; Dynorphins; Female; Ketamine; Macaca mulatta; Pain; Pain Measurement; Peptide Fragments; Pyrrolidines

2003
Different mechanisms of intrinsic pain inhibition in early and late inflammation.
    Journal of neuroimmunology, 2003, Volume: 141, Issue:1-2

    Neuroimmune interactions control pain through activation of opioid receptors on sensory nerves by immune-derived opioid peptides. Here we evaluate mechanisms of intrinsic pain inhibition at different stages of Freund's adjuvant-induced inflammation of the rat paw. We use immunohistochemistry and paw pressure testing. Our data show that in early (6 h) inflammation leukocyte-derived beta-endorphin, met-enkephalin and dynorphin A activate peripheral mu-, delta- and kappa-receptors to inhibit nociception. In addition, central opioid mechanisms seem to contribute significantly to this effect. At later stages (4 days), antinociception is exclusively produced by leukocyte-derived beta-endorphin acting at peripheral mu and delta receptors. Corticotropin-releasing hormone (CRH) is an endogenous trigger of these effects at both stages. These findings indicate that peripheral opioid mechanisms of pain inhibition gain functional relevance with the chronicity of inflammation.

    Topics: Animals; Corticotropin-Releasing Hormone; Dynorphins; Edema; Endorphins; Enkephalin, Methionine; Freund's Adjuvant; Hindlimb; Inflammation; Injections, Subcutaneous; Leukocytes; Male; Naloxone; Pain; Pain Threshold; Rats; Rats, Wistar; Stress, Physiological; Time Factors

2003
Multiple actions of systemic artemin in experimental neuropathy.
    Nature medicine, 2003, Volume: 9, Issue:11

    The clinical management of neuropathic pain is particularly challenging. Current therapies for neuropathic pain modulate nerve impulse propagation or synaptic transmission; these therapies are of limited benefit and have undesirable side effects. Injuries to peripheral nerves result in a host of pathophysiological changes associated with the sustained expression of abnormal pain. Here we show that systemic, intermittent administration of artemin produces dose- and time-related reversal of nerve injury-induced pain behavior, together with partial to complete normalization of multiple morphological and neurochemical features of the injury state. These effects of artemin were sustained for at least 28 days. Higher doses of artemin than those completely reversing experimental neuropathic pain did not elicit sensory or motor abnormalities. Our results indicate that the behavioral symptoms of neuropathic pain states can be treated successfully, and that partial to complete reversal of associated morphological and neurochemical changes is achievable with artemin.

    Topics: Animals; Biomarkers; Calcitonin Gene-Related Peptide; Dynorphins; Male; Nerve Tissue Proteins; Pain; Rats; Spinal Nerves

2003
Breaking the pain barrier.
    Nature medicine, 2003, Volume: 9, Issue:11

    Topics: Animals; Calcitonin Gene-Related Peptide; Dynorphins; Nerve Tissue Proteins; Pain; Rats; Spinal Nerves

2003
Comparison of antiepileptic drugs tiagabine, lamotrigine, and gabapentin in mouse models of acute, prolonged, and chronic nociception.
    The Journal of pharmacology and experimental therapeutics, 2002, Volume: 302, Issue:3

    Some antiepileptic drugs have been shown to be clinically effective in the treatment of neuropathic pain. This study determined whether the new antiepileptic drug tiagabine, a GABA uptake inhibitor, is efficacious in mice in a broad range of nociceptive tests (hot-plate, formalin, and dynorphin-induced chronic allodynia) and compared tiagabine's potency with two other antiepileptic drugs, gabapentin and lamotrigine. Intraperitoneally administered tiagabine, but not lamotrigine, gabapentin, or i.t. tiagabine, produced dose-dependent antinoception in the hot-plate test. A 5-min pretreatment with tiagabine (2-29 nmol i.t.) dose-dependently inhibited both the acute and late phase formalin behaviors; pretreatment with lamotrigine (4-265 nmol i.t.) inhibited only the late phase. In the formalin assay the GABA(A) antagonist bicuculline reversed the acute phase antinociception, whereas the GABA(B) antagonist saclofen reversed both the acute and late phase tiagabine-induced antinociception. Tiagabine administered i.p. but not i.t. dose-dependently reduced dynorphin-induced chronic allodynia for 120 min. Gabapentin and lamotrigine produced antinociception administered either i.t. or i.p. in a dose-dependent manner. Thus, we have shown that gabapentin and lamotrigine produced antinociception in two mouse models of pain, whereas tiagabine produced antinociception in all three mouse models of pain.

    Topics: Acetates; Acute Disease; Adrenergic alpha-Agonists; Amines; Analgesics, Non-Narcotic; Analgesics, Opioid; Animals; Anticonvulsants; Chronic Disease; Cyclohexanecarboxylic Acids; Dose-Response Relationship, Drug; Dynorphins; Formaldehyde; GABA Antagonists; Gabapentin; gamma-Aminobutyric Acid; Hot Temperature; Injections, Intraperitoneal; Injections, Spinal; Lamotrigine; Male; Mice; Mice, Inbred ICR; Nipecotic Acids; Pain; Pain Measurement; Reaction Time; Tiagabine; Triazines

2002
Lack of the nociceptin receptor does not affect acute or chronic nociception in mice.
    Peptides, 2002, Volume: 23, Issue:9

    The peptide nociceptin/orphanin FQ (N/OFQ) and its receptor ORL-1, also designated opioid receptor 4 (OP(4)) are involved in the modulation of nociception. Using OP(4)-knockout mice, we have studied their response following opioid receptor stimulation and under neuropathic conditions.In vas deferens from wild-type and OP(4)-knockout mice, DAMGO (mu/OP(3) agonist), deltorphine II (delta/OP(1) agonist) and (-)-U-50488 (kappa/OP(2) agonist) induced similar concentration-dependent inhibition of electrically-evoked contractions. Naloxone and naltrindole (delta/OP(1) antagonists) shifted the curves of DAMGO (pA(2)=8.6) and deltorphine II (pA(2)=10.2) to the right, in each group. In the hot-plate assay, N/OFQ (10 nmol per mouse, i.t.) increased baseline latencies two-fold in wild-type mice while morphine (10mg/kg, s.c.), deltorphine II (10 nmol per mouse, i.c.v.) and dynorphin A (20 nmol per mouse, i.c.v.) increased hot-plate latencies by about four- to five-fold with no difference observed between wild-type and knockout mice. Furthermore, no change was evident in the development of the neuropathic condition due to chronic constriction injury (CCI) of the sciatic nerve, after both thermal and mechanical stimulation. Altogether these results suggest that the presence of OP(4) receptor is not crucial for (1) the development of either acute or neuropathic nociceptive responses, and for (2) the regulation of full receptor-mediated responses to opioid agonists, even though compensatory mechanisms could not be excluded.

    Topics: 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer; Analgesics, Non-Narcotic; Analgesics, Opioid; Animals; Dose-Response Relationship, Drug; Dynorphins; Enkephalin, Ala(2)-MePhe(4)-Gly(5)-; Female; Male; Mice; Mice, Knockout; Morphine; Naloxone; Naltrexone; Narcotic Antagonists; Nociceptin; Nociceptin Receptor; Oligopeptides; Opioid Peptides; Pain; Receptors, Opioid; Time Factors; Vas Deferens

2002
'Knock-down' of spinal CB1 receptors produces abnormal pain and elevates spinal dynorphin content in mice.
    Pain, 2002, Volume: 100, Issue:1-2

    Recent studies demonstrate the possible existence of tonic modulatory control of nociceptive input mediated by spinal cannabinoid receptors (CB1). Accordingly, it is predicted that a reduction in the spinal CB1 receptors may enhance sensitivity to sensory stimuli and a decrease in spinal antinociceptive potency to cannabinoid agonists. An antisense oligodeoxynucleotide (ODN) specific to the CB1 receptor was used to 'knock-down' CB1 receptors in the lumbar spinal cord and dorsal root ganglia by the local, repeated intrathecal (i.th.) administration of the ODN. This treatment resulted in a decrease in lumbar spinal CB1 receptor expression accompanied by a decrease in the response thresholds to both innocuous tactile and noxious thermal stimuli. The antinociceptive action of the CB1 agonist, WIN 55,212-2, by i.th. administration was also significantly attenuated after treatment with the antisense ODN. Similar treatment using a mismatch control ODN had no effect on receptor protein or on sensory thresholds. The effects of the antisense ODN treatment on sensory thresholds were fully reversed after discontinuation of the ODN injection. The antisense ODN treated rats also showed a significant increase in lumbar spinal dynorphin A. Acute i.th. injection of MK-801 or an antidynorphin antiserum blocked the antisense ODN-induced tactile and thermal hypersensitivity. These data support the possibility of endogenous inhibitory cannabinoid tone to limit spinal afferent input of thermal and tactile stimuli. Lifting of this inhibitory tone through a 'knock-down' of spinal CB1 receptors apparently lowers the thresholds for sensory input, as reflected by the actions of MK-801 to block tactile and thermal hypersensitivity. The increased spinal dynorphin may act to further promote afferent outflow and abnormal pain because sequestration of spinal dynorphin with antiserum also reverses the manifestations of abnormal pain following knock-down of CB1 receptors.

    Topics: Analgesics; Animals; Antibodies; Benzoxazines; Cyclohexanols; Dizocilpine Maleate; Dynorphins; Excitatory Amino Acid Antagonists; Male; Mice; Mice, Inbred ICR; Morpholines; Naphthalenes; Oligodeoxyribonucleotides, Antisense; Pain; Receptors, Cannabinoid; Receptors, Drug; Spinal Cord; Tritium

2002
Dynorphin-independent spinal cannabinoid antinociception.
    Pain, 2002, Volume: 100, Issue:3

    Spinal antinociception produced by delta 9-tetrahydro-cannabinol (Delta(9)-THC) and other cannabinoid agonists has been suggested to be mediated by the release of dynorphin acting at the kappa opioid receptor. Alternatively, as cannabinoid receptors are distributed appropriately in the pain transmission pathway, cannabinoid agonists might act directly at the spinal level to inhibit nociception, without requiring dynorphin release. Here, these possibilities were explored using mice with a deletion of the gene encoding prodynorphin. Antinociceptive dose-response curves were constructed for spinal Delta(9)-THC and WIN 55,212-2 in prodynorphin knock-out mice and in wild-type littermates. WIN 55,212-2 and Delta(9)-THC were equipotent in the wild-type and prodynorphin knock-out mice. Spinal pretreatment with a kappa opioid receptor antagonist, nor-binaltorphimine (nor-BNI), did not alter the dose-response curves for either WIN 55,212-2 or Delta(9)-THC in prodynorphin knock-out and wild-type mice. However, the same dose of nor-BNI used blocked U50,488H-induced antinociception in both wild-type and prodynorphin knock-out mice, confirming kappa opioid receptor activity. Pretreatment with SR141716A, a cannabinoid receptor antagonist blocked the antinociceptive actions of both WIN 55,212-2 and Delta(9)-THC. These data support the conclusion that antinociception produced by spinal cannabinoids are likely to be mediated directly through activation of cannabinoid receptors without the requirement for dynorphin release or activation of kappa opioid receptors.

    Topics: Analgesics; Analgesics, Non-Narcotic; Animals; Benzoxazines; Dose-Response Relationship, Drug; Dronabinol; Dynorphins; Injections, Spinal; Male; Mice; Mice, Knockout; Morpholines; Naltrexone; Naphthalenes; Pain; Pain Measurement; Reference Values; Reproducibility of Results; Sensitivity and Specificity; Single-Blind Method; Species Specificity; Spinal Cord

2002
Alteration in endogenous opioid systems due to chronic inflammatory pain conditions.
    European journal of pharmacology, 2002, Jan-25, Volume: 435, Issue:2-3

    The influence of chronic arthritic pain on two endogenous opioid peptides, dynorphin B and [Met5]enkephalin-Arg6-Phe7, and multiple opioid receptors in discrete brain, lumbar spinal cord and pituitary pools was investigated. Using radioimmunoassay and receptor binding assay, we examined the changes in regional opioid peptide levels and opioid receptor activity due to chronic inflammation in adjuvant arthritic rats. At 4 weeks post-inoculation, increased levels of immunoreactive dynorphin B and [Met5]enkephalin-Arg6-Phe7 were measured in tissues of arthritic rats compared with controls. No significant changes in mu-, delta- or kappa-opioid receptors were seen after chronic inflammation. Taken together, these results indicate that in chronic arthritis, opioid receptor changes do not follow the peptide alterations of pro-dynorphin and pro-enkephalin systems. Thus, dynamic modification and modulation of nociceptive information takes place during chronic inflammation. This supports the key role of the central nervous system in chronic inflammatory pain conditions.

    Topics: Animals; Brain; Chronic Disease; Disease Models, Animal; Dynorphins; Endorphins; Enkephalin, Methionine; Female; Inflammation; Opioid Peptides; Pain; Pain Measurement; Rats; Rats, Inbred Lew; Receptors, Opioid; Spinal Cord

2002
Heritability of nociception. III. Genetic relationships among commonly used assays of nociception and hypersensitivity.
    Pain, 2002, Volume: 97, Issue:1-2

    We and others have previously demonstrated that nociception in the mouse is heritable. A genetic correlation analysis of 12 common measures of nociception among a common set of inbred strains revealed three major clusters (or 'types') of nociception in this species. In the present study, we re-evaluated the major types of nociception and their interrelatedness using ten additional assays of nociception and hypersensitivity, including: three thermal assays (tail withdrawal from 47.5 degrees C water or -15 degrees C ethanol; tail flick from radiant heat), two chemical assays of spontaneous nociception (bee venom test; capsaicin test) and their subsequent thermal hypersensitivity states (including contralateral hypersensitivity in the bee venom test), a mechanical nociceptive assay (tail-clip test), and a mechanical hypersensitivity assay (intrathecal dynorphin). Confirming our earlier findings, the results demonstrate distinct thermal and chemical nociceptive types. It is now clear that mechanical hypersensitivity and thermal hypersensitivity are genetically dissociable phenomena. Furthermore, we now see at least two distinct types of thermal hypersensitivity: afferent-dependent, featuring a preceding significant period of spontaneous nociceptive behavior associated with afferent neural activity, and non-afferent-dependent. In conclusion, our latest analysis suggests that there are at least five fundamental types of nociception and hypersensitivity: (1) baseline thermal nociception; (2) spontaneous responses to noxious chemical stimuli; (3) thermal hypersensitivity; (4) mechanical hypersensitivity; and (5) afferent input-dependent hypersensitivity.

    Topics: Animals; Bee Venoms; Capsaicin; Carrageenan; Dynorphins; Hyperalgesia; Injections, Spinal; Male; Mice; Mice, Inbred AKR; Mice, Inbred BALB C; Mice, Inbred C3H; Mice, Inbred C57BL; Mice, Inbred CBA; Mice, Inbred DBA; Multivariate Analysis; Neurogenic Inflammation; Nociceptors; Pain; Pain Measurement; Species Specificity; Tail

2002
Time-dependent descending facilitation from the rostral ventromedial medulla maintains, but does not initiate, neuropathic pain.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2002, Jun-15, Volume: 22, Issue:12

    Although injury-induced afferent discharge declines significantly over time, experimental neuropathic pain persists unchanged for long periods. These observations suggest that processes that initiate experimental neuropathic pain may differ from those that maintain such pain. Here, the role of descending facilitation arising from developing plasticity in the rostral ventromedial medulla (RVM) in the initiation and maintenance of experimental neuropathic pain was explored. Tactile and thermal hypersensitivity were induced in rats by spinal nerve ligation (SNL). RVM lidocaine blocked SNL-induced tactile and thermal hypersensitivity on post-SNL days 6-12 but not on post-SNL day 3. Lesion of RVM cells expressing mu-opioid receptors with dermorphin-saporin did not prevent the onset of SNL-induced tactile and thermal hypersensitivity, but these signs reversed to baseline levels beginning on post-SNL day 4. Similarly, lesions of the dorsolateral funiculus (DLF) did not prevent the onset of SNL-induced tactile and thermal hypersensitivity, but these signs reversed to baseline levels beginning on post-SNL day 4. Lesions of the DLF also blocked the SNL-induced increase in spinal dynorphin content, which has been suggested to promote neuropathic pain. These data distinguish mechanisms that initiate the neuropathic state as independent of descending supraspinal influences and additional mechanism(s) that require supraspinal facilitation to maintain such pain. In addition, the data indicate that these time-dependent descending influences can underlie some of the SNL-induced plasticity at the spinal level. Such time-dependent descending influences driving associated spinal changes, such as the upregulation of dynorphin, are key elements in the maintenance, but not initiation, of neuropathic states.

    Topics: Animals; Behavior, Animal; Denervation; Dynorphins; Immunotoxins; Kinetics; Lidocaine; Ligation; Male; Medulla Oblongata; Microinjections; N-Glycosyl Hydrolases; Oligopeptides; Opioid Peptides; Pain; Plant Proteins; Rats; Rats, Sprague-Dawley; Ribosome Inactivating Proteins, Type 1; Saporins; Spinal Cord; Spinal Nerves

2002
Pronociceptive effects of spinal dynorphin promote cannabinoid-induced pain and antinociceptive tolerance.
    Pain, 2002, Volume: 98, Issue:1-2

    Recent studies indicate that sustained opioid administration produces increased expression of spinal dynorphin, which promotes enhanced sensitivity to non-noxious and noxious stimuli. Such increased "pain" may manifest behaviorally as a decrease in spinal antinociceptive potency. Here, the possibility of similar mechanisms in the antinociception of spinal cannabinoids was explored. Response thresholds to non-noxious mechanical and noxious thermal stimuli were assessed. Antinociception was determined using the 52 degrees C tail-flick test. Mice received repeated WIN 55,212-2, its inactive enantiomer, WIN 55,212-3 or vehicle (i.th., bid, 5 days). WIN 55,212-2, but not WIN 55,212-3 or vehicle, produced a time-related increased sensitivity to non-noxious and noxious stimuli. WIN 55,212-2, but not WIN 55,212-3 or vehicle, elicited a significant increase in lumbar spinal dynorphin content at treatment day 5. Increased sensitivity to mechanical and thermal stimuli produced by WIN 55,212-2 was reversed to baseline levels by i.th. MK-801 or dynorphin antiserum; control serum had no effect. WIN 55,212-2, but not WIN 55,212-3 or vehicle, produced dose-related antinociception and repeated administration resulted in antinociceptive tolerance. While MK-801 and dynorphin antiserum did not alter acute antinociception produced by WIN 55,212-2, these substances significantly blocked antinociceptive tolerance when given immediately prior to WIN 55,212-2 challenge on day 5. Daily MK-801 pretreatments, prior to WIN 55,212-2 injection, also produced a significant block of antinociceptive tolerance. These data suggest that like opioids, repeated spinal administration of a cannabinoid CB1 agonist elicits abnormal pain, which results in increased expression of spinal dynorphin. Manipulations that block cannabinoid-induced pain also block the behavioral manifestation of cannabinoid tolerance.

    Topics: Analgesics; Animals; Antibodies; Benzoxazines; Cannabinoids; Cross Reactions; Drug Synergism; Drug Tolerance; Dynorphins; Hot Temperature; Immunoenzyme Techniques; Injections, Spinal; Isomerism; Male; Mice; Mice, Inbred ICR; Morpholines; Naphthalenes; Nociceptors; Pain; Pain Threshold; Physical Stimulation; Spinal Cord

2002
Antinociceptive effect produced by intracerebroventricularly administered dynorphin A is potentiated by p-hydroxymercuribenzoate or phosphoramidon in the mouse formalin test.
    Brain research, 2001, Feb-09, Volume: 891, Issue:1-2

    The antinociceptive effects of intracerebroventricularly (i.c.v.) administered dynorphin A, an endogenous agonist for kappa-opioid receptors, in combination with various protease inhibitors were examined using the mouse formalin test in order to clarify the nature of the proteases involved in the degradation of dynorphin A in the mouse brain. When administered i.c.v. 15 min before the injection of 2% formalin solution into the dorsal surface of a hindpaw, 1-4 nmol dynorphin A produced a dose-dependent reduction of the nociceptive behavioral response consisting of licking and biting of the injected paw during both the first (0-5 min) and second (10-30 min) phases. When co-administered with p-hydroxymercuribenzoate (PHMB), a cysteine protease inhibitor, dynorphin A at the subthreshold dose of 0.5 nmol significantly produced an antinociceptive effect during the second phase. This effect was significantly antagonized by nor-binaltorphimine, a selective kappa-opioid receptor antagonist, but not by naltrindole, a selective delta-opioid receptor antagonist. At the same dose of 0.5 nmol, dynorphin A in combination with phosphoramidon, an endopeptidase 24.11 inhibitor, produced a significant antinociceptive effect during both phases. The antinociceptive effect was significantly antagonized by naltrindole, but not by nor-binaltorphimine. Phenylmethanesulfonyl fluoride (PMSF), a serine protease inhibitor, bestatin, a general aminopeptidase inhibitor, and captopril, an angiotensin-converting enzyme inhibitor, were all inactive. The degradation of dynorphin A by mouse brain extracts in vitro was significantly inhibited only by the cysteine protease inhibitors PHMB and N-ethylmaleimide, but not by PMSF, phosphoramidon, bestatin or captopril. The present results indicate that cysteine proteases as well as endopeptidase 24.11 are involved in two steps in the degradation of dynorphin A in the mouse brain, and that phosphoramidon inhibits the degradation of intermediary delta-opioid receptor active fragments enkephalins which are formed from dynorphin A.

    Topics: Animals; Brain; Cell Extracts; Drug Interactions; Dynorphins; Glycopeptides; Hydroxymercuribenzoates; Injections, Intraventricular; Mice; Naltrexone; Narcotic Antagonists; Nociceptors; Pain; Pain Measurement; Protease Inhibitors; Rats

2001
Spinal and supraspinal changes in opioid mRNA expression are related to the onset of pain behaviors following excitotoxic spinal cord injury.
    Pain, 2001, Feb-01, Volume: 90, Issue:1-2

    Excitotoxic spinal cord injury (SCI) causes anatomic, physiologic and molecular changes within the spinal cord and brain. Intraspinal injection of quisqualic acid (QUIS) produces an excitotoxic injury that leads to the onset of behavioral syndromes, believed to be related to the clinical condition of chronic pain. The opioid system, classically involved in the suppression of pain transmission, has been associated with the onset of pain-related behaviors and changes in spinal opioid peptide expression have been demonstrated in various models of SCI and chronic pain. Recently, changes in opioid peptide expression have been demonstrated in both spinal and supraspinal areas following excitotoxic SCI. Therefore, the purpose of this study was to examine changes in opioid peptide gene expression as they relate to the onset of pain behaviors following excitotoxic SCI. Male, Long-Evans rats were given an intraspinal injection of 1.2 microl of 125 mM QUIS and allowed to survive for 10 days, a duration sufficient for the development of pain-related behaviors. Animals were assessed daily for the presence of excessive grooming behavior, i.e. self-directed biting and scratching resulting in damage to superficial and deeper layers of the skin. Animals were also tested for thermal hypersensitivity using a cold plate apparatus on days 5, 7, and 10 following QUIS injection. After sacrifice, quantitative in situ hybridization was performed on regions of the spinal cord surrounding the lesion site as well as whole brain sections through various levels of the thalamus and cortex. Spinal preproenkephalin (PPE) and preprodynorphin (PPD) expression was significantly increased in animals that developed excessive grooming behaviors vs. those that did not. For PPE, this difference was seen bilaterally, in areas of cord caudal to the site of injury. For PPD, this difference was seen only ipsilateral to the site of injection, rostral to the site of injury. In addition, PPE expression in the anterior cingulate cortex and PPD expression in the contralateral parietal cortex were significantly higher in grooming vs. non-grooming animals. These results support previous conclusions that both spinal and supraspinal regulation of endogenous opioid peptide expression plays a role in the response to or onset of post-SCI pain. These results also suggest that the opioid peptides are regulated independently and serve different functions in response to SCI.

    Topics: Animals; Brain; Dynorphins; Enkephalins; Excitatory Amino Acid Agonists; Gene Expression; Grooming; Male; Opioid Peptides; Pain; Protein Precursors; Quisqualic Acid; Rats; Rats, Long-Evans; RNA, Messenger; Spinal Cord; Spinal Cord Injuries

2001
Absence of spinal response to extracorporeal shock waves on the endogenous opioid systems in the rat.
    Ultrasound in medicine & biology, 2001, Volume: 27, Issue:2

    Extracorporeal shock wave therapy (ESWT) seems to be a new therapeutic strategy for chronic pain due to tendopathies. Neurophysiological mechanisms of action for pain relief following ESWT are still unknown. The aim of this study was to investigate if the analgesic effect of ESWT is caused by modulation of the endogenous spinal opioid system. Rats were treated with two different energy flux densities (0.04 and 0.11mJ/mm(2)) and immunohistochemical analysis of met-enkephalin (MRGL) and dynorphin (Dyn) was performed at 4 or 72 h after ESWT. ESWT had no modulatory influence on the expression of the spinal opioid systems. Different energy doses or repetitive treatment did not alter MRGL or Dyn immunoreactivity in the spinal cord. Furthermore, a delayed effect of ESWT at 72 h after treatment was not detectable. We conclude from these findings that the analgesic effects of ESWT treatment are not supported by endogenous opioids.

    Topics: Animals; Chi-Square Distribution; Dynorphins; Enkephalin, Methionine; Immunohistochemistry; Lithotripsy; Pain; Rats; Rats, Wistar; Spinal Cord

2001
The effect of a paracetamol and morphine combination on dynorphin A levels in the rat brain.
    Biochemical pharmacology, 2001, Jun-01, Volume: 61, Issue:11

    The purpose of this study was to find out whether the combination of inactive doses of paracetamol (PARA) and morphine was able to change dynorphin (DYN) A levels, evaluated by radioimmunoassay, and whether naloxone or [(-)-2-(3 furylmethyl)-normetazocine] (MR 2266), a kappa-opioid antagonist, modifies or prevents the activity of this combination on nociception and on DYN levels. The work was suggested by our previous findings which demonstrated that inactive doses of PARA and morphine, when given in combination, share an antinociceptive effect, and that PARA, at antinociceptive doses, decreases DYN levels in the frontal cortex, thus indicating a selective action within the CNS. Our present results demonstrate that the combination of inactive doses of PARA (100 mg/kg) and morphine (3 mg/kg) is just as effective in decreasing the levels of DYN A as full antinociceptive doses of PARA or morphine alone in the frontal cortex of the rat. The values, expressed in pmol/g tissue, were: control = 2.83 +/- 0.20; paracetamol (100) = 2.60 +/- 0.23; morphine (3) = 2.73 +/- 0.24; paracetamol + morphine = 1.34 + 0.16 (P < 0.05). The decrease was partially antagonised by MR 2266, but not by naloxone, suggesting that the activity of PARA and morphine in combination on DYN A levels could be mediated, at least in part, through kappa-receptors, although other systems may be involved. On the other hand, both naloxone and MR 2266 prevented the antinociceptive effect of the combination in the hot plate test. All our experimental data suggest that PARA and morphine in combination exert their antinociceptive effect through the opioidergic system, which in turn may cause a decrease in DYN levels in the CNS of the rat.

    Topics: Acetaminophen; Analgesics; Analgesics, Non-Narcotic; Analgesics, Opioid; Animals; Brain; Drug Combinations; Dynorphins; Male; Morphine; Pain; Pain Measurement; Rats; Rats, Wistar

2001
Antisera against endogenous opioids increase the nocifensive response to formalin: demonstration of inhibitory beta-endorphinergic control.
    European journal of pharmacology, 2001, Jun-01, Volume: 421, Issue:1

    The roles of endogenous opioid peptides in the brain in the modulation of nocifensive responses to formalin in ICR mice were studied. Mice were pretreated intracerebroventricularly (i.c.v.) with rabbit antiserum against beta-endorphin, [Leu5]enkephalin, [Met5]enkephalin or dynorphin A-(1-17) 1 h prior to intraplantar injection of formalin (0.5%, 25 microl) and the nocifensive licking responses were then observed. Pretreatment of mice with antiserum against beta-endorphin enhanced the second phase, but not the first phase of the nocifensive responses to formalin. Pretreatment with antiserum against [Leu5]enkephalin also caused a small but statistically significant enhancement of the second phase, but not the first phase of nocifensive responses to formalin. On the other hand, pretreatment with antiserum against [Met5]enkephalin or dynorphin A-(1-17) did not affect the nocifensive response to formalin. Our results indicate that beta-endorphinergic, and to a lesser extent, [Leu5]enkephalinergic systems are activated at the supraspinal sites to attenuate the nocifensive responses to formalin stimulation.

    Topics: Animals; beta-Endorphin; Dynorphins; Enkephalin, Leucine; Enkephalin, Methionine; Formaldehyde; Immune Sera; Injections, Intraventricular; Male; Mice; Mice, Inbred ICR; Nociceptors; Opioid Peptides; Pain

2001
Expression of c-fos mRNA is increased and related to dynorphin mRNA expression following excitotoxic spinal cord injury in the rat.
    Neuroscience letters, 2001, Jul-20, Volume: 307, Issue:3

    Previous studies have demonstrated that excitotoxic spinal cord injury (SCI) created by the intraspinal injection of quisqualic acid (QUIS) is capable of inducing opioid peptide gene expression within the spinal cord and cortex. The opioids are classically involved in the suppression of pain transmission but specifically, dynorphin, has been implicated in the secondary pathophysiologic response to SCI. Activation of the immediate early gene, c-fos, has been implicated in the induction of preprodynorphin (PPD) gene expression and therefore, may be an important intermediate step in the generation of the opioid response to SCI. The purpose of this study was to investigate whether intraspinal QUIS injection induces c-fos expression within the spinal cord. Male, Long-Evans, adult rats (n=5) received an intraspinal injection of 1.2 microl of 125 mM QUIS directed at spinal segments T12-L2. Four hours post-injection brain and spinal cord tissues were removed and processed for in situ hybridization. Integrated density of c-fos and PPD mRNA expression was increased in the spinal dorsal horn following QUIS injection as compared to sham-injected animals. This indicates that SCI rapidly induces c-fos and PPD expression and suggests that c-fos plays a role in the induction of PPD expression.

    Topics: Animals; Dynorphins; Excitatory Amino Acid Agonists; Gene Expression Regulation; Male; Neurotoxins; Nociceptors; Pain; Posterior Horn Cells; Protein Precursors; Proto-Oncogene Proteins c-fos; Quisqualic Acid; Rats; Rats, Long-Evans; RNA, Messenger; Spinal Cord Injuries; Up-Regulation

2001
The role of kainic acid/AMPA and metabotropic glutamate receptors in the regulation of opioid mRNA expression and the onset of pain-related behavior following excitotoxic spinal cord injury.
    Neuroscience, 2001, Volume: 104, Issue:3

    Intraspinal injection of quisqualic acid, a mixed kainic acid/2-amino-3(3-hydroxy-5-methylisoxazol-4-yl)propionic acid and metabotropic glutamate receptor agonist, produces an excitotoxic injury that leads to the onset of both spontaneous and evoked pain behavior as well as changes in spinal and cortical expression of opioid peptide mRNA, preprodynorphin and preproenkephalin. What characteristics of the quisqualic acid-induced injury are attributable to activation of each receptor subtype is unknown. This study attempted to define the role of activation of the kainic acid/2-amino-3(3-hydroxy-5-methylisoxazol-4-yl)propionic acid (AMPA) and metabotropic glutamate receptor subtypes in the regulation of opioid peptide expression and the onset of spontaneous and evoked pain-related behavior following excitotoxic spinal cord injury by comparing quisqualic acid-induced changes with those created by co-injection of quisqualic acid and the kainic acid/AMPA antagonist, 2,3-dihydroxy-6-nitro-7-sulfamoylbenzo[f]quinoxaline, (NBQX) or the metabotropic antagonist, (RS)-1-aminoindan-1,5-dicarboxylic acid (AIDA). Therefore, 42 male Long-Evans adult rats were divided into seven treatment groups and received intraspinal microinjections of saline (sham), 0.5% dimethylsulphoxide (sham), quisqualic acid (1.2 microl, 125 mM), NBQX (1.2 microl, 60 microM), AIDA (1.2 microl, 250 microM), quisqualic acid/NBQX (1.2 microl, 125 mM/60 microM), or quisqualic acid/AIDA (1.2 microl, 125 mM/250 microM) directed at spinal levels thoracic 12-lumbar 2. Behavioral observations of spontaneous and evoked pain responses were completed following surgery. After a 10-day survival period, animals were killed and brain and spinal cord tissues were removed and processed for histologic analysis and in situ hybridization. Both AIDA and NBQX affected the quisqualic acid-induced total lesion volume but only AIDA caused a decrease in the percent tissue damage at the lesion epicenter. Preprodynorphin and preproenkephalin expression is increased in both spinal and cortical areas in quisqualic acid-injected animals versus sham-, NBQX or AIDA-injected animals. NBQX did not affect quisqualic acid-induced spinal or cortical expression of preprodynorphin or preproenkephalin except for a significant decrease in preproenkephalin expression in the spinal cord. In contrast, AIDA significantly decreases quisqualic acid-induced preprodynorphin and preproenkephalin expression within the spinal cord and cortex. AIDA, b

    Topics: Animals; Behavior, Animal; Dynorphins; Enkephalins; Excitatory Amino Acid Agonists; Excitatory Amino Acid Antagonists; Grooming; Indans; Male; Nerve Degeneration; Neurons; Neurotoxins; Opioid Peptides; Pain; Pain Measurement; Protein Precursors; Quinoxalines; Rats; Rats, Long-Evans; Receptors, AMPA; Receptors, Glutamate; Receptors, Kainic Acid; Receptors, Metabotropic Glutamate; RNA, Messenger; Spinal Cord; Spinal Cord Injuries

2001
Methionine-enkephalin-and Dynorphin A-release from immune cells and control of inflammatory pain.
    Pain, 2001, Volume: 93, Issue:3

    We have previously shown that beta-endorphin (END) is contained and released from memory-type T-cells within inflamed tissue and that it is capable to control pain (J Clin Invest 100(1) (1997) 142). Methionine-enkephalin (MET) and Dynorphin-A (DYN) are endogenous opioids with preference for delta- and kappa-opioid receptors, respectively. Both MET and DYN are produced and contained within immune cells. The goal of this study was to determine the release characteristics of MET and DYN in a rat model of localized hindpaw inflammation and to examine the antinociceptive role of MET and DYN in a Freund's adjuvant induced model of inflammatory pain. We found that corticotropin-releasing factor (CRF) can stimulate the release of both MET and DYN from lymphocytes. This release is dose-dependent and reversible by the selective CRF antagonist alpha-helical-CRF. Furthermore, CRF (1.5 ng) produces analgesia when injected into the inflamed paw, which is reversible by direct co-administration of antibodies to MET. Lymphocyte content of MET was 7.0+/-1.4 ng/million cells, whilst DYN content was ~30-fold lower. Both END and DYN, but not MET, were released by IL-1. Consistently, IL-1 produced peripheral analgesic effects which were not reversed by antibodies to MET. These results indicate that both MET and DYN play a role in peripheral analgesia but have different characteristics of release. These studies further support a role of the immune system in the control of inflammatory pain. This may be particularly important in patients suffering from compromised immune systems as with cancer and AIDS.

    Topics: Animals; Antibodies; Corticotropin-Releasing Hormone; Dose-Response Relationship, Drug; Dynorphins; Enkephalin, Methionine; Humans; Inflammation; Interleukin-2; Lymphocytes; Male; Pain; Rats; Rats, Wistar

2001
Differential antinociceptive effects induced by intrathecally administered endomorphin-1 and endomorphin-2 in the mouse.
    European journal of pharmacology, 2001, Sep-21, Volume: 427, Issue:3

    Two highly selective mu-opioid receptor agonists, endomorphin-1 and endomorphin-2, have been identified and postulated to be endogenous ligands for mu-opioid receptors. Intrathecal (i.t.) administration of endomorphin-1 and endomorphin-2 at doses from 0.039 to 5 nmol dose-dependently produced antinociception with the paw-withdrawal test. The paw-withdrawal inhibition rapidly reached its peak at 1 min, rapidly declined and returned to the pre-injection levels in 20 min. The inhibition of the paw-withdrawal responses to endomorphin-1 and endomorphin-2 at a dose of 5 nmol observed at 1 and 5 min after injection was blocked by pretreatment with a non-selective opioid receptor antagonist naloxone (1 mg/kg, s.c.). The antinociceptive effect of endomorphin-2 was more sensitive to the mu (1)-opioid receptor antagonist, naloxonazine than that of endomorphin-1. The endomorphin-2-induced paw-withdrawal inhibition at both 1 and 5 min after injection was blocked by pretreatment with kappa-opioid receptor antagonist nor-binaltorphimine (10 mg/kg, s.c.) or the delta(2)-opioid receptor antagonist naltriben (0.6 mg/kg, s.c.) but not the delta(1)-opioid receptor antagonist 7-benzylidine naltrexone (BNTX) (0.6 mg/kg s.c.). In contrast, the paw-withdrawal inhibition induced by endomorphin-1 observed at both 1 and 5 min after injection was not blocked by naloxonazine (35 mg/kg, s.c.), nor-binaltorphimine (10 mg/kg, s.c.), naltriben (0.6 mg/kg, s.c.) or BNTX (0.6 mg/kg s.c.). The endomorphin-2-induced paw-withdrawal inhibition was blocked by the pretreatment with an antiserum against dynorphin A-(1-17) or [Met(5)]enkephalin, but not by antiserum against dynorphin B-(1-13). Pretreatment with these antisera did not affect the endomorphin-1-induced paw-withdrawal inhibition. Our results indicate that endomorphin-2 given i.t. produces its antinociceptive effects via the stimulation of mu (1)-opioid receptors (naloxonazine-sensitive site) in the spinal cord. The antinociception induced by endomophin-2 contains additional components, which are mediated by the release of dynorphin A-(1-17) and [Met(5)]enkephalin which subsequently act on kappa-opioid receptors and delta(2)-opioid receptors to produce antinociception.

    Topics: Analgesics; Animals; Benzylidene Compounds; Dose-Response Relationship, Drug; Dynorphins; Enkephalin, Ala(2)-MePhe(4)-Gly(5)-; Enkephalin, Leucine; Enkephalin, Methionine; Immune Sera; Injections, Spinal; Injections, Subcutaneous; Male; Mice; Naloxone; Naltrexone; Narcotic Antagonists; Oligopeptides; Pain; Pain Measurement; Pain Threshold; Peptide Fragments; Time Factors

2001
Central changes in nociceptin dynorphin B and Met-enkephalin-Arg-Phe in different models of nociception.
    Brain research, 2000, Feb-28, Volume: 857, Issue:1-2

    The newly identified neuropeptide nociceptin/orphanin FQ (NOC) was measured in different rat brain areas related to the descending anti-nociceptive pathways and compared to two opioid peptides, dynorphin B (DYN B) and Met-enkephalinArgPhe (MEAP). Two experimental models of chronic nociception, one neurogenic and one inflammatory, used in this study, reveal how different pathological conditions may influence these endogenous systems. Nerve injury is induced by ligation of the sciatic nerve and inflammation by a carrageenan injection in the gluteal muscle, 2 weeks prior to decapitation. Selected brain areas were dissected out and frozen. NOC-, DYN B- and MEAP-like immunoreactivity (LI) is determined by radioimmunoassay. Nerve injury increased the NOC-LI levels in the cortex cinguli, DYN B-LI levels in the dorsal and the ventral part of the spinal cord, whereas a decrease in the MEAP-LI levels is seen in the dorsal part of the periaqueductal grey (PAG). After inflammation, the NOC-LI levels increased in cortex cinguli, hypothalamus and in the dorsal spinal cord, whereas DYN B-LI levels increased in the dorsal part of the PAG. A general increase in MEAP-LI levels is found after inflammation in all analyzed brain areas except in hippocampus. In conclusion, increased levels of NOC-LI were found in cortex cinguli in both treatment groups and in hypothalamus and spinal cord following carrageenan treatment. The changes in the NOC-LI concentrations were not parallelled by changes in DYN B-LI and MEAP-LI, suggesting that NOC and opioid peptides elicit different reactions in the systems of nociception/antinociception.

    Topics: Animals; Brain; Dynorphins; Efferent Pathways; Endorphins; Enkephalin, Methionine; Inflammation; Male; Nerve Crush; Nociceptin; Nociceptors; Opioid Peptides; Pain; Rats; Rats, Sprague-Dawley

2000
Dynorphin B and spinal analgesia: induction of antinociception by the cannabinoids CP55,940, Delta(9)-THC and anandamide.
    Brain research, 2000, Feb-28, Volume: 857, Issue:1-2

    The endogenous opioid dynorphin B was evaluated for its role in cannabinoid-induced antinociception. Previous work in our laboratory has shown that the synthetic, bicyclic cannabinoid, CP55,940, induces the release of dynorphin B whilst the naturally occurring cannabinoid, Delta(9)-tetrahydrocannabinol (Delta(9)-THC), releases dynorphin A. The dynorphins contribute in part to the antinociceptive effects of both cannabinoids at the level of the spinal cord. The present study compares dynorphin B released from perfused rat spinal cord in response to acute administration of anandamide (AEA), Delta(9)-THC and CP55,940 at two time points, 10 min and 30 min post administration, and attempts to correlate such release with antinociceptive effects of the drugs. Dynorphin B was collected from spinal perfusates of rats pretreated with Delta(9)-THC, CP55,940 or AEA. The supernatant was lyophilized and the concentrations of dynorphin B were measured via radioimmunoassay. At a peak time of antinociception (10 min), CP55,940 and Delta(9)-THC induced significant two-fold increases in the release of dynorphin B. AEA did not significantly release dynorphin B. Upon a 30-min pretreatment with the drugs, no significant dynorphin B release was observed, although antinociceptive effects persisted for CP55,940 and Delta(9)-THC. Previous work indicates that Delta(9)-THC releases dynorphin A while AEA releases no dynorphin A. This study confirms that although all three test drugs produced significant antinociception at 10 min, the endocannabinoid, AEA, does not induce antinociception via dynorphin release. Thus, our data indicate a distinct mechanism which underlies AEA-induced antinociception.

    Topics: Analgesia; Analgesics; Analgesics, Non-Narcotic; Animals; Arachidonic Acids; Calcium Channel Blockers; Cannabinoid Receptor Modulators; Cannabinoids; Cyclohexanols; Dronabinol; Dynorphins; Endocannabinoids; Endorphins; Male; Pain; Polyunsaturated Alkamides; Rats; Rats, Sprague-Dawley; Spinal Cord

2000
Effects of peptidase inhibitors on anti-nociceptive action of dynorphin-(1-8) in rats.
    Naunyn-Schmiedeberg's archives of pharmacology, 2000, Volume: 361, Issue:3

    Previous in vitro studies showed that the degradation of dynorphin-(1-8) [dyn-(1-8)] by cerebral membrane preparations is almost completely prevented by a mixture of three peptidase inhibitors (PIs), amastatin, captopril and phosphoramidon. In the present investigations, effects of the three PIs on the anti-nociception induced by the intra-third-ventricular (i.t.v.) administration of dyn-(1-8) were examined. The inhibitory effect of dyn-(1-8) on the tail-flick response was increased more than 100-fold by the i.t.v. pretreatment of rats with the three PIs. The inhibition produced by dyn-(1-8) in rats pretreated with any combination of two PIs was significantly smaller than that in rats pretreated with three PIs, indicating that any residual single peptidase could inactivate significant amounts of dyn-(1-8). The antagonistic effectiveness of naloxone, a relatively selective mu-opioid antagonist, indicates that dyn-(1-8)-induced inhibition of tail-flick response in rats pretreated with three PIs is mediated by mu-opioid receptors. Furthermore, mu-receptor-mediated inhibition induced by dyn-(1-8) was significantly greater than that produced by [Met5]-enkephalin in rats pretreated with three PIs. The data obtained in the present investigations together with those obtained in previous studies strongly indicate that dyn-(1-8) not only has well-known kappa-agonist activity but also has high mu-agonist activity.

    Topics: Analgesics, Opioid; Analysis of Variance; Angiotensin-Converting Enzyme Inhibitors; Animals; Anti-Bacterial Agents; Captopril; Drug Interactions; Dynorphins; Enkephalin, Ala(2)-MePhe(4)-Gly(5)-; Glycopeptides; Injections, Intraventricular; Male; Naloxone; Narcotic Antagonists; Pain; Pain Measurement; Peptide Fragments; Peptides; Protease Inhibitors; Rats; Rats, Wistar; Receptors, Opioid, mu

2000
Extraterritorial neuropathic pain correlates with multisegmental elevation of spinal dynorphin in nerve-injured rats.
    Pain, 2000, Volume: 86, Issue:1-2

    Neuropathic pain is often associated with the appearance of pain in regions not related to the injured nerve. One mechanism that may underlie neuropathic pain is abnormal, spontaneous afferent drive which may contribute to NMDA-mediated central sensitization by the actions of glutamate and by the non-opioid actions of spinal dynorphin. In the present study, injuries to lumbar or sacral spinal nerves elicited elevation in spinal dynorphin content which correlated temporally and spatially with signs of neuropathic pain. The increase in spinal dynorphin content was coincident with the onset of tactile allodynia and thermal hyperalgesia. Injury to the lumbar (L(5)/L(6)) spinal nerves produced elevated spinal dynorphin content in the ipsilateral dorsal spinal quadrant at the L(5) and L(6) spinal segments and in the segments immediately adjacent. Lumbar nerve injury elicited ipsilateral tactile allodynia and thermal hyperalgesia of the hindpaw. In contrast, S(2) spinal nerve ligation elicited elevated dynorphin content in sacral spinal segments and bilaterally in the caudal lumbar spinal cord. The behavioral consequences of S(2) spinal nerve ligation were also bilateral, with tactile allodynia and thermal hyperalgesia seen in both hindpaws. Application of lidocaine to the site of S(2) ligation blocked thermal hyperalgesia and tactile allodynia of the hindpaws suggesting that afferent drive was critical to maintenance of the pain state. Spinal injection of antiserum to dynorphin A((1-17)) and of MK-801 both blocked thermal hyperalgesia, but not tactile allodynia, of the hindpaw after S(2) ligation. These data suggest that the elevated spinal dynorphin content consequent to peripheral nerve injury may drive sensitization of the spinal cord, in part through dynorphin acting directly or indirectly on the NMDA receptor complex. Furthermore, extrasegmental increases in spinal dynorphin content may partly underlie the development of extraterritorial neuropathic pain.

    Topics: Anesthetics, Local; Animals; Antibodies, Blocking; Dizocilpine Maleate; Dynorphins; Excitatory Amino Acid Antagonists; Hot Temperature; Hyperalgesia; Immunoassay; Lidocaine; Ligation; Male; Pain; Pain Measurement; Pain Threshold; Rats; Rats, Sprague-Dawley; Receptors, N-Methyl-D-Aspartate; Spinal Cord; Spinal Nerves

2000
Murine models of inflammatory, neuropathic and cancer pain each generates a unique set of neurochemical changes in the spinal cord and sensory neurons.
    Neuroscience, 2000, Volume: 98, Issue:3

    The aim of this investigation was to determine whether murine models of inflammatory, neuropathic and cancer pain are each characterized by a unique set of neurochemical changes in the spinal cord and sensory neurons. All models were generated in C3H/HeJ mice and hyperalgesia and allodynia behaviorally characterized. A variety of neurochemical markers that have been implicated in the generation and maintenance of chronic pain were then examined in spinal cord and primary afferent neurons.Three days after injection of complete Freund's adjuvant into the hindpaw (a model of persistent inflammatory pain) increases in substance P, calcitonin gene-related peptide, protein kinase C gamma, and substance P receptor were observed in the spinal cord. Following sciatic nerve transection or L5 spinal nerve ligation (a model of persistent neuropathic pain) significant decreases in substance P and calcitonin gene-related peptide and increases in galanin and neuropeptide Y were observed in both primary afferent neurons and the spinal cord. In contrast, in a model of cancer pain induced by injection of osteolytic sarcoma cells into the femur, there were no detectable changes in any of these markers in either primary afferent neurons or the spinal cord. However, in this cancer-pain model, changes including massive astrocyte hypertrophy without neuronal loss, increase in the neuronal expression of c-Fos, and increase in the number of dynorphin-immunoreactive neurons were observed in the spinal cord, ipsilateral to the limb with cancer. These results indicate that a unique set of neurochemical changes occur with inflammatory, neuropathic and cancer pain in C3H/HeJ mice and further suggest that cancer induces a unique persistent pain state. Determining whether these neurochemical changes are involved in the generation and maintenance of each type of persistent pain may provide insight into the mechanisms that underlie each of these pain states.

    Topics: Animals; Astrocytes; Axotomy; Behavior, Animal; Disease Models, Animal; Dynorphins; Fluorescent Antibody Technique; Freund's Adjuvant; Ganglia, Spinal; Male; Mice; Mice, Inbred C3H; Neoplasm Transplantation; Neuralgia; Neuritis; Neurons, Afferent; Osteolysis; Pain; Palpation; Physical Stimulation; Proto-Oncogene Proteins c-fos; Receptors, Neurokinin-1; Sarcoma, Experimental; Sciatic Nerve; Spinal Cord; Spinal Nerves; Tumor Cells, Cultured

2000
Local inhibitory effects of dynorphin A-(1-17) on capsaicin-induced thermal allodynia in rhesus monkeys.
    European journal of pharmacology, 2000, Aug-18, Volume: 402, Issue:1-2

    Although dynorphin A-(1-17) has been characterized in vitro as a high efficacy kappa-opioid receptor agonist, functional studies of dynorphin A-(1-17) following central or systemic administration indicate the involvement of both opioid and non-opioid components. The aim of this study was to investigate whether local administration of dynorphin-related analogs can attenuate capsaicin (8-methyl-N-vanillyl-6-nonenamide)-induced nociception and what type of opioid receptor mediates the local action of dynorphin A-(1-17) in monkeys. Capsaicin (100 microg) was used to evoke a nociceptive response, thermal allodynia, which was manifested as a reduced tail-withdrawal latency in normally innocuous 46 degrees C warm water. Co-administration of dynorphin A-(1-17) (0.3-10 microg) with capsaicin in the tail dose-dependently inhibited thermal allodynia; however, both non-opioid fragments dynorphin A-(2-17) (10-300 microg) and dynorphin A-(2-13) (10-300 microg) were ineffective. Local antiallodynia of dynorphin A-(1-17) was antagonized by a small dose (100 microg) of an opioid receptor antagonist, quadazocine, applied s.c. in the tail. Pretreatment with a selective kappa-opioid receptor antagonist, nor-binaltorphimine (nor-BNI), s.c. 320 microg in the tail also reversed local antiallodynia of dynorphin A-(1-17). Both locally effective doses of antagonists, when applied s.c. in the back, did not antagonize local dynorphin A-(1-17), indicating that peripheral kappa-opioid receptors selectively mediated the local action of dynorphin A-(1-17) in the tail. In addition, a much larger dose of dynorphin A-(1-17) (1000 microg), when administered s. c. in the back or i.m. in the thigh, did not cause sedative or diuretic effects. These results suggest that in vivo opioid actions of dynorphin-related peptides can be differentiated locally in this procedure. They also indicate that local application of peptidic ligands may be a useful medication for localized pain.

    Topics: 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer; Animals; Capsaicin; Diuresis; Dynorphins; Female; Injections, Intramuscular; Injections, Subcutaneous; Macaca mulatta; Male; Pain; Pain Measurement; Reaction Time; Receptors, Opioid, kappa

2000
Involvement of dynorphin in immobilization stress-induced antinociception in the mouse.
    European neuropsychopharmacology : the journal of the European College of Neuropsychopharmacology, 2000, Volume: 10, Issue:5

    The effect of antiserum against [Met(5)]-enkephalin, [Leu(5)]-enkephalin, beta-endorphin, or dynorphin A-(1-13) administered intracerebroventricularly (i.c.v.) or intrathecally (i. t.) on immobilization-induced antinociception was studied in ICR mice. Antinociception was assessed by the tail-flick assay. Immobilization of the mouse increased inhibition of the tail-flick response at least 1 h. The i.c.v. or i.t. injection with antiserum against dynorphin A-(1-13) at the dose of 200 microg significantly attenuated immobilization-induced inhibition of the tail-flick response. However, antiserum against [Met(5)]-enkephalin, [Leu(5)]-enkephalin, or beta-endorphin did not affect the immobilization stress-induced antinociception. Furthermore, i.c.v. or i.t. injection with nor-binaltorphimine (Nor-BNI; from 1 to 20 microg) effectively inhibited immobilization stress-induced inhibition of the tail-flick response in a dose-dependent manner. However, beta-FNA (from 0.5 to 2 microg) or naltrindole (from 1 to 20 microg) administered i.c.v. or i.t. did not affect immobilization stress-induced antinociception. Our results suggest that supraspinally and spinally located dynorphin appears to be involved in the production of immobilization stress-induced antinociception via stimulating kappa-opioid receptors.

    Topics: Animals; beta-Endorphin; Cerebral Ventricles; Dynorphins; Enkephalin, Leucine; Enkephalin, Methionine; Immune Sera; Injections, Intraventricular; Injections, Spinal; Male; Mice; Mice, Inbred ICR; Pain; Pain Measurement; Reaction Time; Restraint, Physical; Stress, Psychological

2000
Arginine-rich peptides are blockers of VR-1 channels with analgesic activity.
    FEBS letters, 2000, Sep-15, Volume: 481, Issue:2

    Vanilloid receptors (VRs) play a fundamental role in the transduction of peripheral tissue injury and/or inflammation responses. Molecules that antagonize VR channel activity may act as selective and potent analgesics. We report that synthetic arginine-rich hexapeptides block heterologously expressed VR-1 channels with submicromolar efficacy in a weak voltage-dependent manner, consistent with a binding site located near/at the entryway of the aqueous pore. Dynorphins, natural arginine-rich peptides, also blocked VR-1 activity with micromolar affinity. Notably, synthetic and natural arginine-rich peptides attenuated the ocular irritation produced by topical capsaicin application onto the eyes of experimental animals. Taken together, our results imply that arginine-rich peptides are VR-1 channel blockers with analgesic activity. These findings may expand the development of novel analgesics by targeting receptor sites distinct from the capsaicin binding site.

    Topics: Amino Acid Sequence; Analgesics; Animals; Arginine; Capsaicin; Dynorphins; Electric Conductivity; Eye; Inhibitory Concentration 50; Mice; Oocytes; Pain; Peptides; Receptors, Drug; TRPV Cation Channels; Xenopus laevis

2000
Redox manipulation of NMDA receptors in vivo: alteration of acute pain transmission and dynorphin-induced allodynia.
    Pain, 1999, Volume: 80, Issue:1-2

    The redox modulatory site of the N-methyl-D-aspartate (NMDA) receptor directly regulates NMDA receptor function. Sulfhydryl reducing agents, such as dithiothreitol (DTT), potentiate NMDA receptor-evoked currents in vitro, whereas oxidizing agents, such as 5,5'-dithio-bis-(2-nitrobenzoic acid) (DTNB), attenuate these currents. In this study, we examined the effect of this redox manipulations on nociceptive spinal cord signaling in mice. Intrathecal (i.t.) administration of DTT (0.1-30 nmol), presumably reducing the NMDA receptor, dose-dependently enhanced NMDA-induced nociceptive behaviors, and this enhancement was blocked by the oxidizing agent, DTNB. Pretreatment with DTT (10 nmol, i.t.) enhanced NMDA-induced tail-flick thermal hyperalgesia and intraplantar formalin-induced nociceptive behaviors. Finally, DTT pretreatment enhanced the long lasting allodynia induced by i.t. administration of dynorphin, whereas post-treatment with DTNB reduced the permanent allodynia induced by dynorphin for 5 days. Potentiation of all four of these NMDA-dependent nociceptive behaviors by DTT suggests that the reduction of the NMDA receptor by endogenous reducing agents may contribute to augmented pain transmission in response to activation by endogenous glutamate. Moreover, blockade of in vivo NMDA receptor reducing agents or oxidation of the NMDA receptor redox site may prove therapeutically useful in the treatment of chronic pain.

    Topics: Acute Disease; Animals; Dithionitrobenzoic Acid; Dithiothreitol; Dynorphins; Excitatory Amino Acid Agonists; Hyperalgesia; Injections, Spinal; Male; Mice; Mice, Inbred ICR; N-Methylaspartate; Oxidation-Reduction; Pain; Pain Measurement; Receptors, N-Methyl-D-Aspartate; Reducing Agents; Signal Transduction; Spinal Cord; Sulfhydryl Reagents; Synaptic Transmission

1999
Analgesia-producing mechanism of processed Aconiti tuber: role of dynorphin, an endogenous kappa-opioid ligand, in the rodent spinal cord.
    Japanese journal of pharmacology, 1999, Volume: 79, Issue:3

    The analgesia-producing mechanism of processed Aconiti tuber was examined using rodents whose nociceptive threshold was decreased by loading repeated cold stress (RCS). The antinociceptive effect of processed Aconiti tuber (0.3 g/kg, p.o.) in RCS-loaded mice was antagonized by pretreatment with a kappa-opioid antagonist, nor-binaltorphimine (10 mg/kg, s.c.), and was abolished by an intrathecal injection of anti-dynorphin antiserum (5 microg). The Aconiti tuber-induced antinociception was inhibited by both dexamethasone (0.4 mg/kg, i.p.) and a dopamine D2 antagonist, sulpiride (10 mg/kg, i.p.), in RCS-loaded mice, and it was eliminated by both an electric lesion of the hypothalamic arcuate nucleus (HARN) and a highly selective dopamine D2 antagonist, eticlopride (0.05 microg), administered into the HARN in RCS-loaded rats. These results suggest that the analgesic effect of processed Aconiti tuber was produced via the stimulation of kappa-opioid receptors by dynorphin released in the spinal cord. It was also shown that dopamine D2 receptors in the HARN were involved in the expression of the analgesic activity of processed Aconiti tuber.

    Topics: 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer; Administration, Topical; Analgesics; Animals; Arcuate Nucleus of Hypothalamus; Cold Temperature; Dexamethasone; Dopamine Antagonists; Drugs, Chinese Herbal; Dynorphins; Glucocorticoids; Hypothalamus; Immune Sera; Ligands; Male; Mice; Naltrexone; Narcotic Antagonists; Nociceptors; Pain; Pain Threshold; Rats; Rats, Sprague-Dawley; Receptors, Opioid, delta; Receptors, Opioid, kappa; Receptors, Opioid, mu; Salicylamides; Spinal Cord; Sulpiride

1999
Preprodynorphin-like immunoreactivity in medullary dorsal horn neurons projecting to the thalamic regions in the rat.
    Neuroscience letters, 1999, Apr-02, Volume: 264, Issue:1-3

    Preprodynorphin (PPD)-like immunoreactive (-LI) neuronal cell bodies in the trigeminal sensory nuclear complex of the rat were found in laminae I and II of the medullary dorsal horn (MDH; caudal spinal trigeminal nucleus) and the paratrigeminal nucleus. A PPD immunofluorescence histochemistry combined with a fluorescence retrograde tract-tracing method revealed that some of the PPD-LI neurons in the MDH and paratrigeminal nucleus projected to the thalamic regions. Nociceptive nature of the PPD-LI MDH neurons projecting to the thalamic regions was also demonstrated by a triple labeling method, using the technique of the noxious stimulus-evoked expression of the immediate-early gene, c-fos. In the rats which were subcutaneously injected with formalin into the upper and lower lips, c-fos protein (Fos) was found in PPD-LI neurons which were labeled with a retrograde tracer injected into the thalamic regions.

    Topics: Animals; Dynorphins; Formaldehyde; Injections, Subcutaneous; Lip; Male; Neurons; Nociceptors; Pain; Protein Precursors; Proto-Oncogene Proteins c-fos; Rats; Rats, Wistar; Spinal Cord; Synaptic Transmission; Thalamus; Trigeminal Nuclei; Trigeminal Nucleus, Spinal

1999
Cannabinoid modulation of dynorphin A: correlation to cannabinoid-induced antinociception.
    European journal of pharmacology, 1999, Aug-13, Volume: 378, Issue:3

    Intrathecal administration of anandamide, delta9-tetrahydrocannabinol (THC) and (-)-3-[2-hydroxy-4-(1,1-dimethyheptyl)ptyl)phenyl]-4-(3-hydr oxypropyl)-cicloexan-1-ol (CP55,940) induced spinal antinociception accompanied by differential kappa-opioid receptor involvement and dynorphin A peptide release. Antinociception using the tail-flick test was induced by the classical cannabinoid THC and was blocked totally by 17,17'-bis(cyclopropylmethyl)-6',6,7,7'-tetrahydro-4,5,4'5'-diepoxy++ +-6,6'-(imino)[7,7'-bimorphinan]-3,3',14,14'-tetrol (norbinaltorphimine) indicating a significant and critical kappa-opioid receptor component. The endogenous cannabinoid, anandamide and the non-classical bicyclic cannabinoid, CP55,940, induced non-nor-BNI-sensitive effects. The N-piperidino-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-3-pyrazo le-carboxamide (SR141716A)-mediated attenuation of spinal antinociception imparted by the various cannabinoids indicates cannabinoid CB1 receptor involvement. THC-induced an enhancement of immunoreactive dynorphin A release which coincided with the onset, but not duration antinociception. The release of dynorphin A was also attenuated by SR141716A suggesting it is cannabinoid CB1 receptor-mediated. These data indicate a critical role for dynorphin A release in the initiation of the antinociceptive effects of the cannabinoids at the spinal level.

    Topics: Analgesics; Animals; Cannabinoids; Cyclohexanols; Dimethyl Sulfoxide; Dronabinol; Dynorphins; Injections, Spinal; Male; Naltrexone; Narcotic Antagonists; Nociceptors; Pain; Pain Measurement; Piperidines; Pyrazoles; Rats; Rats, Sprague-Dawley; Rimonabant

1999
Dynorphin-(1-8) inhibits the release of substance P-like immunoreactivity in the spinal cord of rats following a noxious mechanical stimulus.
    European journal of pharmacology, 1997, Apr-04, Volume: 323, Issue:2-3

    This study was conducted to determine the effect of the opioid peptide dynorphin-(1-8) on the release of substance P-like immunoreactivity in the dorsal horn during mechanical activation of peripheral nociceptors. A push-pull cannula was used to perfuse the dorsal horn of decerebrate/spinal transected rats before, during and following the application of a noxious mechanical stimulus to the ipsilateral hindpaw and lower limb. The collected perfusates were assayed for substance P-like immunoreactivity using radioimmunoassay. Dynorphin-(1-8) applied to the spinal cord at a concentration of 1 microM reduced the basal release of substance P-like immunoreactivity by 28 +/- 11% and prevented the mechanically evoked release of substance P-like immunoreactivity. This effect of dynorphin-(1-8) was reversed by 2 microM of the selective kappa-opioid receptor antagonist nor-binaltorphimine. Moreover, blockade of the kappa-opioid receptors by nor-binaltorphimine resulted in a 33 +/- 5% increase in the basal release of substance P-like immunoreactivity. These data show that activation of nor-binaltorphimine-sensitive sites by dynorphin-(1-8) results in inhibition of the release of substance P-like immunoreactivity in the dorsal horn of the rat.

    Topics: Animals; Decerebrate State; Dynorphins; Hindlimb; Male; Naltrexone; Pain; Peptide Fragments; Physical Stimulation; Radioimmunoassay; Rats; Rats, Wistar; Receptors, Opioid, kappa; Spinal Cord; Substance P

1997
[Met5]enkephalin and delta2-opioid receptors in the spinal cord are involved in the cold water swimming-induced antinociception in the mouse.
    Life sciences, 1997, Volume: 61, Issue:7

    Mice made cold water swimming (CWS: 4 degrees C, 3 min) produced an opioid-mediated antinociception. Experiments were designed to determine what types of opioid receptors and endogenous opioid peptides in the spinal cord are involved in the CWS-induced antinociception in male ICR mice. Antinociception was measured by the tail-flick test. CWS-induced antinociception was blocked by intrathecal (i.t.) pretreatment with antiserum to [Met5]enkephalin (100 microg, 1 hr), but not by antiserum (100 microg, 1 hr) to [Leu5]enkephalin, beta-endorphin or dynorphin A (1-17). Moreover, i.t. pretreatment with delta2-opioid receptor antagonist naltriben (NTB: 10 microg, 10 min) blocked the antinociception induced by CWS or i.t.-administered [Met5]enkephalin (10 microg). However, the antinociception induced by CWS or i.t.-administered [Met5]enkephalin was not blocked by i.t. pretreatment with delta1-opioid receptor antagonist 7-benzylidene naltrexone (BNTX: 1 microg, 10 min), mu-opioid receptor antagonist D-Phe-Cys-Try-D-Try-Om-Thr-Phe-Thr-NH2 (CTOP: 50 ng, 10 min), or kappa-opioid receptor antagonist norbinaltorphimine (norBNI: 5 microg, 24 hr). These data indicate that [Met5]enkephalin and delta2-opioid receptor in the spinal cord are involved in antinociception induced by CWS.

    Topics: Animals; beta-Endorphin; Cold Temperature; Dynorphins; Enkephalin, Leucine; Enkephalin, Methionine; Immune Sera; Injections, Spinal; Male; Mice; Mice, Inbred ICR; Pain; Receptors, Opioid, delta; Receptors, Opioid, kappa; Receptors, Opioid, mu; Spinal Cord

1997
Spinally administered dynorphin A produces long-lasting allodynia: involvement of NMDA but not opioid receptors.
    Pain, 1997, Volume: 72, Issue:1-2

    The endogenous opioid peptide dynorphin A has non-opioid effects that can damage the spinal cord when given in high doses. Dynorphin has been shown to increase the receptive field size of spinal cord neurons and facilitate C-fiber-evoked reflexes. Furthermore, endogenous dynorphin levels increase following damage to the spinal cord, injury to peripheral nerves, or inflammation. In this study, sensory processing was characterized following a single, intrathecal injection of dynorphin A (1-17) in mice. A single intrathecal injection of dynorphin A (1-17) (3 nmol, i.t.) induced mechanical allodynia (hind paw, von Frey filaments) lasting 70 days, tactile allodynia (paint brush applied to flank) lasting 14 days, and cold allodynia (acetone applied to the dorsal hind paw) lasting 7 days. Similarly, dynorphin A (2-17) (3 nmol, i.t.), a non-opioid peptide, induced cold and tactile allodynia analogous to that induced by dynorphin A (1-17), indicating the importance of non-opioid receptors. Pretreatment with the NMDA antagonists, MK-801 and LY235959, but not the opioid antagonist, naloxone, blocked the induction of allodynia. Post-treatment with MK-801 only transiently blocked the dynorphin-induced allodynia, suggesting the NMDA receptors may be involved in the maintenance of allodynia as well as its induction. We have induced a long-lasting state of allodynia and hyperalgesia by a single intrathecal injection of dynorphin A (1-17) in mice. The allodynia induced by dynorphin required NMDA receptors rather than opioid receptors. This result is consistent with results in rats and with signs of clinically observed neuropathic pain. This effect of exogenously administered dynorphin raises the possibility that increased levels of endogenous dynorphins associated with spinal cord injuries may participate in the genesis and maintenance of neuropathic pain.

    Topics: Animals; Dizocilpine Maleate; Dynorphins; Excitatory Amino Acid Antagonists; Injections, Spinal; Isoquinolines; Male; Mice; Mice, Inbred ICR; Narcotic Antagonists; Pain; Receptors, N-Methyl-D-Aspartate; Receptors, Opioid

1997
Temporal changes in spinal cord expression of mRNA for substance P, dynorphin and enkephalin in a model of chronic pain.
    Acta physiologica Scandinavica, 1997, Volume: 161, Issue:4

    We have used a partial sciatic nerve ligation model to examine the time course for changes in the expression of mRNA for three peptides related to pain transmission at spinal sites (dynorphin, enkephalin and substance P), during the development of allodynia. Enhanced expression of mRNA for dynorphin and substance P was observed in the dorsal horn on the same side as the partial nerve ligation. Increased expression of dynorphin mRNA was biphasic. The initial increases in expression of dynorphin mRNA occurred at 3 h, and a secondary peak was observed 1-3 days after surgery. The secondary increases coincided roughly with increased substance P mRNA expression. However, both dynorphin and substance P mRNA returned to control values after 1 week despite continuing allodynia. No significant changes in expression of mRNA for enkephalin were observed. The elevation of substance P mRNA in intrinsic spinal cord neurons may be secondary to changes in immediate early genes c-fos and jun-B, whereas the expression of dynorphin and enkephalin mRNA is differently regulated. The results also suggest that changes in the expression of the three neuropeptides are not critically involved in the development and maintenance of chronic pain or allodynia.

    Topics: Animals; Behavior, Animal; Chronic Disease; Disease Models, Animal; Dynorphins; Enkephalins; Gene Expression; Male; Neuropeptides; Pain; Rats; Rats, Sprague-Dawley; RNA, Messenger; Spinal Cord; Substance P; Time Factors

1997
Kappa-opioid receptor modulation of the release of substance P in the dorsal horn.
    Brain research, 1996, Jan-08, Volume: 706, Issue:1

    Substance P (SP), a member of the tachykinin peptide family, has been found in high concentrations in the superficial laminae of the dorsal horn and it is thought to play a major role in the transmission of nociceptive information. Dynorphin(1-8), an opioid peptide with high selectivity for the kappa-opioid receptor subtype, is also found in the dorsal horn of the spinal cord. The aim of this study was to determine the effect of dynorphin(1-8) on the release of SP-like-immunoreactivity (SPLI) in the dorsal horn before and during the activation of peripheral nociceptors by a thermal stimulus. A push-pull canula was used to perfuse the dorsal horn of non-anesthetized decerebrate/spinal transected rats and the collected perfusates were assayed for SPLI by using radioimmunoassay. Dynorphin(1-8) applied to the spinal cord at a concentration of 1 microM elicited a 27 +/- 8% decrease in the basal release of SPLI and prevented the increase in the release of SPLI evoked by the application of a noxious thermal stimulus to the ipsilateral hind paw and lower limb. The effect of dynorphin(1-8) was reversed by 2 microM of nor-binaltorphimine (nor-BNI), a selective kappa opioid receptor antagonist. Application of nor-BNI alone to the perfusate resulted in a 62 +/- 23% increase in the basal release of SPLI. In conclusion, dynorphin(1-8) reduces the basal release of SPLI and prevents the increase in the release of SPLI elicited by the application of a noxious cutaneous thermal stimulus. This effect is mediated through the kappa-opioid receptor, which appears to tonically regulate the release of SPLI in the dorsal horn.

    Topics: Animals; Basal Metabolism; Dynorphins; Hindlimb; Hot Temperature; Intercellular Signaling Peptides and Proteins; Male; Pain; Peptide Fragments; Peptides; Radioimmunoassay; Rats; Rats, Sprague-Dawley; Rats, Wistar; Receptors, Opioid, kappa; Spinal Cord; Substance P

1996
Single intrathecal injections of dynorphin A or des-Tyr-dynorphins produce long-lasting allodynia in rats: blockade by MK-801 but not naloxone.
    Pain, 1996, Volume: 68, Issue:2-3

    Neuropathic pain states are accompanied by increased sensitivity to both noxious and non-noxious sensory stimuli, characterized as hyperalgesia and allodynia, respectively. In animal models of neuropathic pain, the presence of hyperalgesia and allodynia are accompanied by neuroplastic changes including increased spinal levels of substance P, cholecystokinin (CCK), and dynorphin. N-Methyl-D-aspartate (NMDA) receptors appear to be involved in maintaining the central sensitivity which contributes to neuropathic pain. In addition to its opioid activities, dynorphin has been suggested to act at the NMDA receptor complex. In an attempt to mimic the increased levels of spinal dynorphin seen in animal models of neuropathic pain, rats received a single intrathecal (i.t.) injection of dynorphin A(1-17), dynorphin A(1-13), dynorphin A(2-17) or dynorphin A(2-13) through indwelling catheters. Tactile allodynia was determined by measuring response threshold to probing with von Frey filaments. Dynorphin A(1-17) administration evoked significant and long-lasting tactile allodynia (i.e. > 60 days). Likewise, the i.t. administration of dynorphin A(1-13) or dynorphin A(2-17) or dynorphin A(2-13) also produced long-lasting tactile allodynia. Intrathecal pretreatment, but not post-treatment, with MK-801 prevented dynorphin A(1-17)-induced development of allodynia; i.t. administration of MK-801 alone had no effect on responses to tactile stimuli. In contrast, i.t. pretreatment with naloxone did not affect the development of tactile allodynia induced by dynorphin A(1-17) or alter sensory threshold when given alone. These results demonstrate that a single dose of dynorphin A, or its des-Tyr fragments, produces long-lasting allodynia which may be irreversible in the rat. Further, this effect appears to be mediated through activation of NMDA, rather than opioid, receptors. While the precise mechanisms underlying the development and maintenance of the allodynia is unclear, it seems possible that dynorphin may produce changes in the spinal cord, which may contribute to the development of signs reminiscent of a "neuropathic' state. Given that levels of dynorphin are elevated following nerve injury, it seems reasonable to speculate that dynorphin may have a pathologically relevant role in neuropathic pain states.

    Topics: Animals; Chronic Disease; Dizocilpine Maleate; Dynorphins; Excitatory Amino Acid Antagonists; Injections, Spinal; Male; Naloxone; Narcotic Antagonists; Pain; Peptide Fragments; Peripheral Nervous System Diseases; Rats; Rats, Sprague-Dawley; Sensory Thresholds

1996
Cerebrospinal alterations of immunoreactive dynorphin A after unilateral dorsal rhizotomy in the rat.
    Brain research, 1995, Jan-30, Volume: 670, Issue:2

    Possible alterations of immunoreactive dynorphin A (ir-dyn A) were investigated at different levels of the spinal cord and in discrete brain regions of male rats 10, 30 and 60 days after unilateral dorsal rhizotomy, i.e., during the development of deafferentation pain and autotomy behavior that follows afferent nerve interruption. Dorsal rhizotomy caused an increase of spinal ir-dyn A at 10 days in the cervical segment; subsequent assays showed a progressive increase in other spinal regions too. At the last observation, 60 days after rhizotomy, neuropeptide levels were still significantly higher than in sham-lesioned animals in the cervical, thoracic and lumbosacral spinal cord. The spinal ir-dyn A changes were both ipsi- and contralateral to the lesion. No alterations were found in the brainstem and midbrain and a not significant decrease was observed in the hypothalamus. In the striatum and cortex, however, there was a bilateral significant increase 30 days after surgery and a constant and significant elevation was detected in the hippocampus at all three intervals. These data cast additional light on the neurochemical changes caused by the interruption of afferent nerves, followed by development of the deafferentation pain syndrome in laboratory animals and human beings. They also support the concept of central neuroplasticity in pathological pain and indicate that the opioid neuropeptide dynorphin is involved.

    Topics: Analysis of Variance; Animals; Brain; Denervation; Dynorphins; Male; Pain; Radioimmunoassay; Rats; Rats, Sprague-Dawley; Self Mutilation; Spinal Cord; Spinal Nerve Roots

1995
[Expression of preprodynorphin mRNA in the spinal cord after inflammatory abdominal stimulation in rats].
    [Hokkaido igaku zasshi] The Hokkaido journal of medical science, 1994, Volume: 69, Issue:1

    Dynorphin, an opioid peptide, is thought to play an important role in the modulation of nociceptive neural networks at the level of the spinal cord. Fos protein is involved in the transcriptional regulation of the dynorphin gene. Although several studies have been carried out on dynorphin gene expression by noxious somatic stimuli, few have evaluated the effect of noxious visceral stimuli on the expression of dynorphin gene. In the present studies we analysed the expression of the dynorphin gene mediated by a noxious visceral stimulus in a rat model by exposure of abdominal tissue to carrageenan. Expression of preprodynorphin and c-fos mRNAs in the spinal cord neuron was examined using ribonuclease protection assays. After inflammation, a rapid increase in the levels of c-fos mRNA in the thoracic spinal cord was observed. c-fos mRNA levels rose within 30 minutes after injection, and remained elevated for 1 hour, subsequently falling to control levels. In contrast, preprodynorphin mRNA began to increase from 30 minutes after injection and remained elevated for at least 2 days. In situ hybridization with alpha 35S-labeled cRNA probe demonstrated that in the lower thoracic spinal cord preprodynorphin mRNA was expressed in dorsal horn neurons. In celiac ganglia, both preprodynorphin and c-fos mRNAs were not detected. In the peripancreatic abdominal tissue, there was acute severe inflammation consisting of necrosis and marked polymorphonuclear leucocytic infiltration. These data demonstrate that after abdominal tissue inflammation, activation of dynorphin biosynthesis occurred in thoracic spinal cord.(ABSTRACT TRUNCATED AT 250 WORDS)

    Topics: Abdomen; Animals; Carrageenan; Dynorphins; Gene Expression; Genes, fos; In Situ Hybridization; Inflammation; Male; Pain; Protein Precursors; Proto-Oncogene Proteins c-fos; Rats; Rats, Inbred Strains; RNA, Messenger; Spinal Cord; Stimulation, Chemical

1994
Interleukin 1 beta and corticotropin-releasing factor inhibit pain by releasing opioids from immune cells in inflamed tissue.
    Proceedings of the National Academy of Sciences of the United States of America, 1994, May-10, Volume: 91, Issue:10

    Local analgesic effects of exogenous opioid agonists are particularly prominent in painful inflammatory conditions and are mediated by opioid receptors on peripheral sensory nerves. The endogenous ligands of these receptors, opioid peptides, have been demonstrated in resident immune cells within inflamed tissue of animals and humans. Here we examine in vivo and in vitro whether interleukin 1 beta (IL-1) or corticotropin-releasing factor (CRF) is capable of releasing these endogenous opioids and inhibiting pain. When injected into inflamed rat paws (but not intravenously), IL-1 and CRF produce antinociception, which is reversible by IL-1 receptor antagonist and alpha-helical CRF, respectively, and by the immunosuppressant cyclosporine A. In vivo administration of antibodies against opioid peptides indicates that the effects of IL-1 and CRF are mediated by beta-endorphin and, in addition, by dynorphin A and [Met]enkephalin, respectively. Correspondingly, IL-1 effects are inhibited by mu-, delta-, and kappa-opioid antagonists, whereas CRF effects are attenuated by all except a kappa-antagonist. Finally, IL-1 and CRF produce acute release of immunoreactive beta-endorphin in cell suspensions freshly prepared from inflamed lymph nodes. This effect is reversible by IL-1 receptor antagonist and alpha-helical CRF, respectively. These findings suggest that IL-1 and CRF activate their receptors on immune cells to release opioids that subsequently occupy multiple opioid receptors on sensory nerves and result in antinociception. beta-Endorphin, mu- and delta-opioid receptors play a major role, but IL-1 and CRF appear to differentially release additional opioid peptides.

    Topics: Analysis of Variance; Animals; Antibodies; beta-Endorphin; Corticotropin-Releasing Hormone; Cyclosporine; Dose-Response Relationship, Drug; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalin, Methionine; Humans; Inflammation; Injections; Interleukin-1; Male; Naloxone; Pain; Rats; Rats, Wistar; Recombinant Proteins; Regression Analysis; Somatostatin

1994
Infrared laser diode irradiation has no behavioral or biochemical effect on pain in the sciatic nerve ligation-induced mononeuropathy in rat.
    Anesthesia progress, 1994, Volume: 41, Issue:4

    The aim of this study was to evaluate the effect of acute and repeated (5 days) treatment with various types of infrared (IR) diode lasers and probes (single- vs cluster-beam) on the pain response in rats with peripheral mononeuropathy produced by sciatic nerve ligation. Male Sprague-Dawley rats were anesthetized with sodium pentobarbital, and the mid-thigh was surgically exposed to reveal the sciatic nerve, around which four ligatures were loosely tied. On postoperative day 5, the skin over the sciatic nerve lesion was subjected to a 30-min daily local exposure from a 904-nm IR diode laser (700 Hz, average output power 10 mW) with a single-beam probe, a 830-nm IR diode laser (700 Hz) with either a single-beam (average output power 50 mW) or cluster-beam probe (average output power 15 mW), or placebo for 5 consecutive days. Two pain responses (foot-withdrawal time and the hind-paw elevation time) were measured on both sides using the radiant heat method on days 5 and 9. In addition, cold allodynia was measured on day 9 of treatment by placing the rats on a chilled metal plate (4 degrees C) and measuring the duration of elevation of either of the hind paws. On day 9, the animals were sacrificed for collection of the samples of brain and lumbar spinal cord for the determination of the tissue concentrations of dynorphin A1-8-like immunoactivity (DYN) using specific radioimmunoassay (RIA). The hind-paw withdrawal and elevation times on the right side in all groups subjected to the various methods of IR laser irradiation did not differ significantly as compared with the placebo-treated group when measured on days 5 and 9 after surgery. No statistically significant differences in withdrawal response and elevation time of the unaffected left hind paw were noted either. The measurement of cold allodynia similarly failed to reveal any effect in laser-treated groups versus placebo. The RIA analysis found that tissue concentrations of DYN were significantly elevated in the spinal cord ipsilaterally to the ligation side, as compared with the contralateral side, in all rats with sciatic nerve ligation. All modalities of IR diode laser treatment did not produce any significant difference in the brain and spinal cord level of DYN on postoperative day 9 in all treatment groups. It is concluded that repeated IR diode laser treatment did not reduce hyperalgesia induced by sciatic nerve ligation in rats.

    Topics: Animals; Dynorphins; Infrared Rays; Laser Therapy; Ligation; Male; Pain; Rats; Rats, Sprague-Dawley; Sciatic Nerve

1994
Spinal dynorphin immunoreactivity increases bilaterally in a neuropathic pain model.
    Brain research, 1993, Dec-03, Volume: 629, Issue:2

    Increased spinal levels of dynorphin, an endogenous opioid kappa agonist, are seen in models of both chronic and acute hyperalgesia. This study determined the extent and localization of spinal immunoreactive dynorphin following sciatic cryoneurolysis (SCN), a neuropathic pain model produced by a peripheral nerve freeze lesion. SCN results in behaviors associated with neuropathic pain such as autotomy (the gnawing and scratching of the affected limb), touch-evoked and mechanical allodynia, and spontaneous nociceptive behavior. Following SCN, 4 rats that displayed autotomy and 3 rats that did not were randomly chosen for immunohistochemical staining of dynorphin-like immunoreactivity (DLIR). The area of DLIR above a standardized threshold level was quantified in both dorsal horns of each spinal cord section using a computer-assisted image analyzer to express DLIR in pixels. DLIR was observed both ipsilateral and contralateral to the injured peripheral nerve. In addition, the area of DLIR was significantly greater (P = 0.05) in rats that showed autotomy behavior (mean = 52.6 x 10(3) +/- 25.6) compared to rats with no autotomy (mean = 13.8 x 10(3) +/- 4.78). In sharp contrast to the ipsilateral dynorphin increases observed in other neuropathic pain models, we observe a bilateral increase at 21 days following SCN.

    Topics: Animals; Behavior, Animal; Dynorphins; Freezing; Image Processing, Computer-Assisted; Immunohistochemistry; Male; Pain; Rats; Rats, Sprague-Dawley; Sciatic Nerve; Self Mutilation; Spinal Cord

1993
Pain: persistence and disease.
    Journal of the American Dental Association (1939), 1993, Volume: 124, Issue:2

    Topics: Animals; Dynorphins; Nociceptors; Pain; Rats; Synaptic Transmission

1993
Cyclic AMP mediates mu and delta, but not kappa opioid analgesia in the spinal cord of the rat.
    Life sciences, 1993, Volume: 52, Issue:24

    Intrathecal (i.t.) injection of the membrane-permeable cyclic AMP (cAMP) analogue dibutyryl-cyclic AMP (Bt2cAMP) in 3 successive doses of 12.5, 12.5 and 25 ug in rats was very effective in reversing the antinociceptive effects produced by mu agonist morphine or delta agonist DPDPE, but not that by kappa agonist dynorphin A-(1-13). cAMP content of the spinal cord was significantly decreased by morphine, but not by dynorphin A-(1-13). The results imply that a decrease in spinal cAMP content may be important for the antinociceptive effect elicited by mu and delta, but not kappa opioid receptor agonists.

    Topics: Analgesia; Analgesics; Animals; Bucladesine; Cyclic AMP; Dynorphins; Enkephalin, D-Penicillamine (2,5)-; Enkephalins; Injections, Spinal; Male; Morphine; Pain; Peptide Fragments; Radioimmunoassay; Rats; Rats, Wistar; Receptors, Opioid, delta; Receptors, Opioid, kappa; Receptors, Opioid, mu; Spinal Cord; Time Factors

1993
Dynorphin, a preferential ligand for kappa-opioid receptors, is present in nerve fibers and immune cells within inflamed tissue of the rat.
    Neuroscience letters, 1992, Jun-08, Volume: 140, Issue:1

    Exogenous kappa-opioid agonists have been shown to produce peripheral antinociceptive effects in inflamed tissue. This study sought to determine whether endogenous kappa-receptor ligands are present at the site of inflammation. In Freund's adjuvant-induced hindpaw inflammation in the rat, we show, by immunohistochemistry, that dynorphin is detectable within inflammatory cells and in the cutaneous nerves in a similar distribution as calcitonin gene-related peptide, a specific marker for sensory neurons. These findings extend our previous observations in that not only beta-endorphin and Met-enkephalin (mu- and delta-receptor ligands), but also a preferential kappa-ligand is present within inflamed subcutaneous tissue.

    Topics: Animals; Calcitonin Gene-Related Peptide; Dynorphins; Freund's Adjuvant; Inflammation; Male; Nerve Fibers; Pain; Rats; Rats, Wistar; Receptors, Opioid, kappa; Skin

1992
Analgesic actions of dynorphin A(1-13) antiserum in the rat brain stem.
    Brain research bulletin, 1992, Volume: 29, Issue:5

    This study was performed to evaluate the effects of dynorphin A(1-3) antiserum when microinjected into an active hyperalgesic region within the rat brain stem. When administered within the dorsal posterior mesencephalic tegmentum (DPMT) of intact conscious rats, dynorphin A(1-13) antiserum produced rapid onset and persistent prolongation of a low intensity thermally evoked tail avoidance response (LITETAR). These analgesic actions of the dynorphin A(1-13) antiserum appeared to be dose dependent. These studies support previous hypotheses about the existence of tonically active brain stem opioid hyperalgesic process. Further, the results provide indirect evidence for a potential role of brain stem dynorphin(s) in facilitating pain.

    Topics: Analgesics; Animals; Antibodies; Brain Stem; Dose-Response Relationship, Drug; Dynorphins; Evoked Potentials; Female; Microinjections; Pain; Peptide Fragments; Rats; Rats, Sprague-Dawley; Tegmentum Mesencephali

1992
[Endogenous opioid systems as biochemical basis of pain control].
    Minerva anestesiologica, 1992, Volume: 58, Issue:10

    Topics: Amino Acid Sequence; Dynorphins; Endorphins; Enkephalins; Humans; Molecular Sequence Data; Pain

1992
Demonstration of dynorphin A 1-8 immunoreactive axons contacting spinal cord projection neurons in a rat model of peripheral inflammation and hyperalgesia.
    Pain, 1992, Volume: 51, Issue:2

    Using a double-labeling technique, we evaluated the input of afferents immunoreactive for dynorphin peptide onto a population of lumbar spinal neurons contributing to the spinoparabrachial tract in rats with 1 inflamed hind paw. We found that the frequency and distribution with which dynorphin immunoreactive varicosities were in apposition to projection neurons varied according to neuron location. In particular, neurons in the superficial dorsal horn and neck of the dorsal horn receive a high degree of dynorphin input. We also determine that unilateral peripheral inflammation is associated with both an increase in the number of projection neurons receiving detectable DYN input and in the frequency of this input onto a given neuron, with the largest increase seen in the superficial dorsal horn. Since almost all superficial dorsal horn neurons contributing to the spinoparabrachial tract respond either exclusively or maximally to noxious stimulation, our data supports dynorphin's involvement in nociception.

    Topics: Animals; Axons; Dendrites; Dynorphins; Immunohistochemistry; Inflammation; Male; Nerve Endings; Neurons; Pain; Peptide Fragments; Peripheral Nerves; Rats; Rats, Sprague-Dawley; Receptors, Opioid, kappa; Spinal Cord

1992
Involvement of spinal kappa opioid receptors in the antagonistic effect of dynorphins on morphine antinociception.
    Life sciences, 1991, Volume: 48, Issue:15

    The modulatory effects of intrathecally (i.t.) administered dynorphin A(1-17) and dynorphin A(1-13) on morphine antinociception have been studied previously in rats by other investigators. However, both potentiating and attenuating effects have been reported. In this study, the modulatory effects of i.t. administered dynorphin A(1-17) as well as the smaller fragment, dynorphin A(1-8), were studied in mice. In addition, nor-binaltorphimine (nor-BNI), a highly selective kappa opioid receptor antagonist, and naltrindole (NTI), a highly selective delta opioid receptor antagonist, were used to characterize the possible involvement of spinal kappa and delta opioid receptors in the modulatory effects of the dynorphins. Dynorphin A(1-17) and dynorphin A(1-8) administered i.t. at doses that did not alter tail-flick latencies, were both able to antagonize in a dose-dependent manner, the antinociceptive action of s.c. administered morphine sulfate. The antinociceptive ED50 of morphine sulfate was increased 3.9- and 5.3-fold by 0.4 nmol/mouse of dynorphin A(1-17) and dynorphin A(1-8), respectively. Injections of 0.4 and 0.8 nmol/mouse of nor-BNI i.t., but not its inactive enantiomer (+)-1-nor-BNI, inhibited dose-dependently the antagonistic effects of the dynorphins. These doses of nor-BNI alone did not affect the antinociceptive action of morphine sulfate. Intrathecal administration of 5 nmol/mouse of NTI also did not affect the modulatory effects of dynorphins. These observations that dynorphins exert their antagonistic effects on morphine-induced antinociception stereoselectively through spinal kappa opioid receptors may suggest a coupling between spinal kappa and mu opioid receptors.

    Topics: Animals; Dynorphins; Indoles; Male; Mice; Morphinans; Morphine; Naltrexone; Pain; Receptors, Opioid; Receptors, Opioid, delta; Receptors, Opioid, kappa; Spine; Stereoisomerism

1991
Prolonged inflammatory pain modifies corticotropin-releasing factor-induced opioid peptide release in the hypothalamus.
    Brain research, 1991, Nov-01, Volume: 563, Issue:1-2

    The influence of prolonged pain upon hypothalamic opioid peptide release in vitro was examined in rats subjected to Freund's adjuvant (FA)-induced unilateral inflammation of the hindlimb. Basal release of enkephalin (ENK) but not beta-endorphin (END) or dynorphin (DYN) was increased 10 days following FA treatment. Superfusion of corticotropin-releasing factor (CRF; 10(-8) M) stimulated the release of opioid peptides in control hypothalami. CRF, however, failed to modify beta-END and DYN release in hypothalami of FA-treated rats, whereas ENK release was markedly reduced. In contrast, KCl-stimulated opioid peptide release did not differ between FA and control hypothalami. These data demonstrate that prolonged inflammatory pain alters the responsiveness of hypothalamic opioid systems to CRF. It is suggested that this effect is mediated at the level of the CRF neuron or its receptor.

    Topics: Animals; Arthritis, Experimental; Behavior, Animal; beta-Endorphin; Corticotropin-Releasing Hormone; Dynorphins; Electroshock; Endorphins; Enkephalin, Methionine; Hypothalamus; In Vitro Techniques; Inflammation; Male; Pain; Potassium Chloride; Radioimmunoassay; Rats; Rats, Inbred Strains; Restraint, Physical; Stress, Psychological

1991
Differential release of substance P and somatostatin in the rat spinal cord in response to noxious cold and heat; effect of dynorphin A(1-17).
    The Journal of pharmacology and experimental therapeutics, 1990, Volume: 252, Issue:2

    Dynorphin A(1-17), the proposed endogenous ligand for the kappa receptor, has been reported to demonstrate no antinociceptive activity when tested in analgesic assays involving noxious (heat (e.g., tail-flick and hot-plate assays). By using a rat tail-flick analgesic assay that utilizes extreme cold as its noxious stimulus (an ethylene glycol-water mixture maintained at -10 degrees C), we have recently reported a dose-related and naloxone-reversible antinociceptive effect for i.c.v. administered dynorphin A(1-17). To elucidate the biochemical mechanism of this antinociception, we designed a push-pull perfusion system which would allow us to measure changes in neuropeptide release in the spinal cord during exposure to noxious heat or cold. Male Sprague-Dawley rats were implanted surgically with two lengths of PE-10 tubing inserted into the spinal subarachnoid space via the cisterna magna, with the push cannula at the level of T-1, and the pull cannula at the rostral edge of the lumbar enlargement. At the time of testing, samples of cerebrospinal fluid were collected both in the presence and absence of a noxious stimulus. Substance P (SP) and somatostatin (SST) levels were measured by radioimmunoassay. Exposing the animal's tail to the noxious cold (30 sec/min for 20 min) resulted in a significant elevation in SP release (69% above base-line levels), but no change in the level of SST release. Conversely, exposure to noxious heat (50 degrees C, 20 sec/min for 20 min) produced a significant increase in SST release (56% above base line), but no change in the level of SP release.(ABSTRACT TRUNCATED AT 250 WORDS)

    Topics: 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer; Animals; Cold Temperature; Dynorphins; Hot Temperature; Male; Naloxone; Pain; Pyrrolidines; Rats; Rats, Inbred Strains; Somatostatin; Spinal Cord; Substance P

1990
Intrathecal somatostatin, somatostatin analogs, substance P analog and dynorphin A cause comparable neurotoxicity in rats.
    Neuroscience, 1990, Volume: 39, Issue:3

    Rats chronically implanted with intrathecal catheters received intrathecal injections (10 microliters followed by 10 microliters saline flush) of either saline (n = 5), somatostatin (100 micrograms, n = 10), the somatostatin analog BIM 23003 (100 micrograms, n = 5), the somatostatin analog SMS 201-995 (100 micrograms, n = 5), the substance P analog [D-Pro2, D-Trp7,9] SP (10 micrograms, n = 10), or dynorphin A (1-17) (20 nmol, n = 8). These doses (somatostatin, substance P and dynorphin A) were selected based on previous studies in which they caused significant motor deficits. Effects on thermal cutaneous nociception, behavior, motor function and spinal cord histopathology were evaluated. All peptides caused severe neurotoxicity, evidenced by flaccid hind leg paralysis and lumbar spinal neuronal degeneration, which was accompanied by an inflammatory reaction in meninges and spinal gray matter. Histopathological changes had developed within 24 h after injection of somatostatin, substance P analog and dynorphin A, showing mild to severe neuronal degeneration and mild inflammatory responses in spinal cord and meninges. Significant antinociceptive effects, due to severe neurotoxic effects, were only observed following intrathecal injection of SMS 201-995 and the substance P analog. Potential neurotoxic mechanisms of the different peptides are discussed.

    Topics: Animals; Dynorphins; Injections, Spinal; Male; Motor Activity; Neurotoxins; Octreotide; Pain; Peptide Fragments; Rats; Rats, Inbred Strains; Reference Values; Seizures; Somatostatin; Spinal Cord; Stereotyped Behavior; Substance P; Time Factors

1990
Opioids from immunocytes interact with receptors on sensory nerves to inhibit nociception in inflammation.
    Proceedings of the National Academy of Sciences of the United States of America, 1990, Volume: 87, Issue:15

    Exogenous opioids can produce localized opioid receptor-mediated antinociception in peripheral inflamed tissue. Previous studies show that activation of endogenous opioids by a cold water swim in rats with hind paw inflammation results in a similar local antinociceptive effect but suggest that pituitary-adrenal opioid pools are not directly involved in producing this effect. Here we show increased amounts of opioid peptides in immune cells infiltrating the inflamed tissue. Furthermore, we demonstrate immunoreactive opioid receptors on peripheral terminals of sensory neurons. The local administration of antibodies against opioid peptides or receptors or systemic pretreatment with the immunosuppressant cyclosporine blocks cold water swim-induced antinociception. These findings suggest that antinociception in inflammation can be brought about by endogenous opioids from immune cells interacting with opioid receptors on peripheral sensory nerves.

    Topics: Animals; Antibodies, Monoclonal; beta-Endorphin; Dynorphins; Enkephalin, Methionine; Freund's Adjuvant; Immunoenzyme Techniques; Inflammation; Male; Neurons, Afferent; Pain; Physical Exertion; Radioimmunoassay; Rats; Rats, Inbred Strains; Receptors, Opioid; Reference Values; Skin

1990
Cholecystokinin octapeptide antagonized opioid analgesia mediated by mu- and kappa- but not delta-receptors in the spinal cord of the rat.
    Brain research, 1990, Jul-16, Volume: 523, Issue:1

    Intrathecal (ith) injection of cholecystokinin octapeptide (CCK-8) to the rat with single dose of 4 or 40 ng, or successive doses from 0.1 to 1 microgram at 10 min intervals produced neither analgesia nor hyperalgesia. However, the analgesia produced by ith injection of PL017, a specific mu-receptor agonist or 66A-078, a specific kappa-receptor agonist could be markedly antagonized by CCK-8 at a dose as small as 4 ng. In contrast, analgesia produced by ith injection of delta-agonist DPDPE could not be blocked by CCK-8 even at a dose as high as 40 ng. Since the effect of CCK-8 could be totally reversed by the CCK receptor antagonist proglumide, this effect is most probably mediated by CCK receptors.

    Topics: Animals; Dose-Response Relationship, Drug; Dynorphins; Endorphins; Enkephalin, D-Penicillamine (2,5)-; Enkephalins; Male; Narcotic Antagonists; Pain; Pain Measurement; Peptide Fragments; Proglumide; Rats; Rats, Inbred Strains; Reaction Time; Receptors, Opioid; Receptors, Opioid, delta; Receptors, Opioid, kappa; Receptors, Opioid, mu; Sincalide; Spinal Cord

1990
[Influence of microinjection of dynorphin antibody into periaqueductal gray (PAG) on analgesia induced by electroacupuncture of different frequencies in rats].
    Zhen ci yan jiu = Acupuncture research, 1990, Volume: 15, Issue:2

    Previous studies have shown that 2 Hz but not 100 Hz electroacupuncture (EA) stimulation released beta-endorphin in PAG of the rat to induce analgesia. The present study was undertaken to see whether dynorphin plays a role in PAG in mediating analgesia induced by low and high frequency EA. Injection of affinity purified dynorphin antibody (1:350,000 in RIA titer) 1 microliter into PAG blocked 2 Hz EA analgesia almost completely, and 15 Hz EA analgesia partly, leaving 100 Hz EA analgesia intact. This blocking effect was totally disappeared when the antibody was diluted to 1/10 of its original concentration (1:35,000). Besides, injection of dynorphin into PAG through chronically implanted cannula showed no analgesic effect. The results suggest that the blockade of 2 Hz EA analgesia by high titer dynorphin antibody injected into PAG may have been the result of cross reactivity of the antibody to other opioid peptides (such as beta-endorphin, enkephalin, etc) released in the PAG area. The data also stress the importance of using antibodies of proper titer and concentration to exclude false positive or false negative conclusions in adopting the antibody microinjection techniques, as was repeatedly shown in the immunohistochemical studies.

    Topics: Acupuncture Analgesia; Animals; Antibodies; Dynorphins; Electroacupuncture; Female; Microinjections; Pain; Periaqueductal Gray; Rats; Sensory Thresholds

1990
[Cholecystokinin octapeptide (CCK-8) antagonized analgesia mediated by mu and kappa opioid receptors].
    Sheng li xue bao : [Acta physiologica Sinica], 1990, Volume: 42, Issue:3

    CCK-8 has been shown to antagonize the analgesia produced by morphine or endogenous opioid peptides. The present study was performed to clarify the interaction between CCK-8 and different opioid ligands. Analgesia produced by intrathecal (I.T.) injection of the specific mu receptor agonist PL017 10 ng or kappa receptor agonist NDAP 500 ng can be antagonized by I.T. injection CCK-8 at a dose as small as 4 ng. In contrast, analgesia produced by I.T. injection of the delta receptor agonist DPDPE (6.5, 13 and 26 micrograms) was not blocked by CCK-8 (4 ng or 40 ng, I.T.). The antagonistic effect of CCK-8 on PL017 and NDAP could be completely reversed by proglumide (3 micrograms, I.T.). I.T. injection of CCK-8 (4 or 40 ng single dose or cumulative dose of 0.1, 0.2, 0.5 and 1.0 microgram at 10 min intervals) produced neither analgesia nor hyperalgesia. In conclusion, CCK-8 preferentially antagonizes opioid analgesia mediated by mu and kappa receptors, and this antagonistic effect is mediated by CCK receptors.

    Topics: Analgesics; Animals; Dynorphins; Endorphins; Injections, Spinal; Male; Pain; Peptide Fragments; Proglumide; Rats; Rats, Inbred Strains; Receptors, Opioid; Receptors, Opioid, kappa; Receptors, Opioid, mu; Sensory Thresholds; Sincalide; Spinal Cord

1990
Localization of dynorphin-induced neurotoxicity in rat spinal cord.
    Life sciences, 1989, Volume: 44, Issue:20

    Intrathecally injected dynorphin A (1-13) in rats results in a reversible hindlimb paralysis and an irreversible loss of the tail-flick reflex. Histologic examination of the spinal cords of dynorphin treated rats demonstrated dead and/or dying neurons predominately localized in the central area which approximates Rexed lamina VII and X. In this area a maximum effect of the dynorphin-induced neurotoxicity is evident. Thus, the dynorphin-induced neuron death is suggestive of an anatomical selectivity.

    Topics: Animals; Cell Survival; Dose-Response Relationship, Drug; Dynorphins; Male; Neurons; Pain; Paralysis; Peptide Fragments; Rats; Rats, Inbred Strains; Reflex; Spinal Cord Diseases

1989
Induction of the gene encoding pro-dynorphin by experimentally induced arthritis enhances staining for dynorphin in the spinal cord of rats.
    Neuroscience, 1989, Volume: 31, Issue:1

    The response of dynorphinergic neurons in the lumbosacral spinal cord of the rat to chronic arthritic inflammation was studied by the combined use of biochemical and immunohistochemical procedures. In polyarthritic rats, in which all four limbs showed a swelling, inflammation and hyperalgesia, a pronounced elevation was seen in the level of messenger ribonucleic acid encoding prodynorphin (pro-enkephalin B) in the lumbosacral spinal cord. In addition, the levels of immunoreactive dynorphin A1-17, a primary gene product of this precursor, were greatly increased. This activation was reflected in a striking intensification of the immunohistochemical staining of both dynorphin and alpha/beta-neo-endorphin, a further major product of pro-dynorphin. In control animals perikarya were stained exceedingly rarely and encountered only in laminae I and II. Stained fibres and varicosities were seen throughout the dorsal and ventral gray matter, being most concentrated in laminae I, II, IV and V of the dorsal horn and dorsolateral to the central canal. In polyarthritic rats, fibres and varicosities were much more intensely stained throughout the cord, particularly in laminae I/II, IV and V and dorsolateral to the central canal. Many strongly-stained perikarya could be seen: these comprised many small diameter cells in laminae I and II, and some large diameter marginal neurons and large diameter cells, heterogenous in appearance, in the deeper laminae IV and V. Monolaterally inflamed rats injected in the right hind-paw showed pathological changes only in this limb. Correspondingly, in unilateral inflammation, an elevation in immunoreactive dynorphin was seen exclusively in the right dorsal horn and the above-described intensification of staining for dynorphin and neo-endorphin was seen only in this quadrant. This reveals the neuroanatomical specificity of the response. Thus, in the lumbosacral cord of the rat, pro-dynorphin neurons are most preponderant in laminae I, II, IV and V. A pronounced intensification of the immunohistochemical staining of these neurons is seen in chronic arthritis. Furthermore, there is a parallel elevation in the levels of messenger ribonucleic acid encoding pro-dynorphin and of its primary products dynorphin and neo-endorphin. These findings demonstrate an enhancement in the functional activity of spinal cord localized dynorphin neurons in the response to chronic arthritic inflammation.

    Topics: Animals; Arthritis; Dynorphins; Enkephalins; Gene Expression Regulation; Immunohistochemistry; Male; Pain; Protein Precursors; Rats; Rats, Inbred Strains; RNA, Messenger; Spinal Cord

1989
Opioid-like activity in the cerebrospinal fluid of pain patients treated by electroacupuncture.
    Neuropharmacology, 1989, Volume: 28, Issue:9

    Thirteen patients with pain from various causes were treated by electroacupuncture for 30 min. Cerebrospinal fluid (CSF) was obtained before and after treatment. Opioid-like substances in the CSF were fractionated by high pressure liquid chromatography and assayed by competitive receptor binding using a mu-specific radioligand, [D-ala2, MePhe4, gly-ol5]-enkephalin (DAGO). Opioid activity, associated with a fraction, eluted at 18-20% acetonitrile, consistently showed an increase in level after acupuncture. Two other fractions eluted at larger concentrations of acetonitrile also increased significantly after acupuncture; however the increase was not consistently observed in every patient. Measurements of beta-endorphin and dynorphin by radioimmunoassay indicated that 80 and 60% of the patients, respectively, had a higher level of these peptides after acupuncture. The nature of the opioid activity, eluted at 18-20% acetonitrile is unknown; however a small amount of it could be found in various parts of the brain of rat.

    Topics: beta-Endorphin; Chromatography, High Pressure Liquid; Chronic Disease; Dynorphins; Electroacupuncture; Enkephalin, Ala(2)-MePhe(4)-Gly(5)-; Enkephalins; Humans; Pain; Pain Management; Radioimmunoassay

1989
Lidocaine treatment of painful diabetic neuropathy and endogenous opioid peptides in plasma.
    The Clinical journal of pain, 1989, Volume: 5, Issue:3

    Intravenous infusion of lidocaine has a pain-relieving effect in patients with painful diabetic neuropathy. We measured plasma beta-endorphin (beta-EP), dynorphin immunoreactivity (DYN), and met-enkephalin (MET) before and after lidocaine infusion in 8 patients with painful diabetic neuropathy and in 10 controls. The pretreatment level of beta-EP and DYN was identical in the two groups. After lidocaine, beta-EP increased in diabetic patients from 3.4 to 5.5 pmol/L (median) (p less than 0.02) and in controls from 3.4 to 5.0 pmol/L (p less than 0.02). The concentration of DYN was stable, and MET was undetectable before and after lidocaine. Lidocaine reduced symptoms and pain score in diabetic patients was uncorrelated with the changes in beta-EP. Intravenous lidocaine increased plasma beta-EP and diminished complaints in patients with painful diabetic neuropathy.

    Topics: Adult; beta-Endorphin; Diabetic Neuropathies; Dynorphins; Endorphins; Female; Humans; Lidocaine; Male; Middle Aged; Pain; Radioimmunoassay

1989
Enkephalins interact with substance P-induced aversive behaviour in mice.
    Brain research, 1988, Feb-23, Volume: 442, Issue:1

    The effect of the endogenous opioid peptides, methionine-enkephalin (Met-ENK), beta-endorphin (beta-END) and dynorphin-(1-17) (DYN) on the aversive behavior produced by intrathecal (i.t.) administration of substance P (SP) was studied in mice. A low dose of i.t. administered Met-ENK gave a marked reduction of the SP-induced response. In the tail-flick assay, such doses of Met-ENK were ineffective in producing antinociception. At much higher doses, however, Met-ENK obtained antinociceptive activity. In contrast, beta-END and DYN had about the same potency in inhibiting the SP-induced behavioural response and in the tail-flick test, respectively. These results suggest that opioid peptides, particularly enkephalin neurons in the spinal cord influence SP-induced aversive behaviour.

    Topics: Animals; beta-Endorphin; Dose-Response Relationship, Drug; Dynorphins; Enkephalin, Methionine; Injections, Spinal; Mice; Nociceptors; Pain; Reaction Time; Substance P

1988
A novel interaction between dynorphin(1-13) and an N-methyl-D-aspartate site.
    Brain research, 1988, Mar-08, Volume: 443, Issue:1-2

    Dynorphin injected intrathecally in the rat results in a neurotoxicity behaviorally expressed as an irreversible loss of the thermally evoked tail-flick reflex. The excitatory amino acid antagonists DL-2-amino-5-phosphonovalerate (APV) and gamma-D-glutamylglycine (DGG) blocked the loss of the tail-flick reflex. The order of potency (APV greater than DGG) suggests that the N-methyl-D-aspartate (NMDA) subclass of excitatory amino acid receptors participate in the neurotoxicity. Additionally, intrathecal injection of APV results in a reversible loss of the tail-flick reflex, whereas with DGG doses which block the tail-flick reflex also result in hindlimb paralysis. These data suggest that neurotransmission in the tail-flick reflex pathway is, in part, mediated by NMDA receptors. From these and previous findings it was concluded that dynorphin neurotoxicity results from enhanced, excitotoxic, transmission across these synapses utilizing NMDA receptors.

    Topics: 2-Amino-5-phosphonovalerate; Analgesics; Animals; Aspartic Acid; Atropine; Binding, Competitive; Dipeptides; Dynorphins; Hot Temperature; Kinetics; Male; N-Methylaspartate; Pain; Peptide Fragments; Rats; Rats, Inbred Strains; Receptors, N-Methyl-D-Aspartate; Receptors, Neurotransmitter; Valine

1988
Investigations on dynorphin structurally-related opioid peptides. I. Impact on nociceptive transmission. II. Regulation of pituitary content.
    Annali dell'Istituto superiore di sanita, 1988, Volume: 24, Issue:3

    Topics: Animals; Benzomorphans; Cyclazocine; Dynorphins; Estradiol; Ethylketocyclazocine; Female; Hypothalamo-Hypophyseal System; Male; Ovary; Pain; Pain Measurement; Rats; Rats, Inbred Strains; Structure-Activity Relationship

1988
[Frequency of electroacupuncture as a cardinal factor determining the potency of analgesia and its vulnerability to naloxone blockade in rabbits].
    Zhen ci yan jiu = Acupuncture research, 1988, Volume: 13, Issue:1

    Topics: Acupuncture Therapy; Animals; Dynorphins; Electric Stimulation Therapy; Male; Naloxone; Nociceptors; Pain; Rabbits; Sensory Thresholds; Transcutaneous Electric Nerve Stimulation

1988
Distinguishable effects of intrathecal dynorphins, somatostatin, neurotensin and s-calcitonin on nociception and motor function in the rat.
    Pain, 1988, Volume: 35, Issue:1

    We determined the effects on nociceptive threshold and motor function of dynorphin-gene products, dynorphin A-(1-32) (DYN A-(1-32), DYN A-(1-8), DYN B and DYN B-29 and the non-opioid peptides somatostatin, neurotensin and salmon calcitonin (s-CT) after intrathecal administration in the rat. DYN A-(1-32) (25 nmol) produced maximal elevation of tail-flick latency accompanied by severe hind limb paralysis and tail flaccidity lasting 6 h and still present at 24 h in several animals. Antinociception evaluated by the vocalization test wore off within 2 h. A lower dose of the peptide (6.25 nmol) did not alter the tail-flick reflex and motor function but significantly elevated the vocalization threshold. The other dynorphins showed weaker, short-lasting activity on the nociceptive threshold, the order of potency being as follows: DYN B-29 greater than DYN B greater than DYN A-(1-8). On the other hand, at the high doses DYN B (100 nmol) and DYN B-29 (50 and 100 nmol) caused moderately severe hind limb paralysis whereas DYN A-(1-8) did not cause any motor impairment up to the dose of 100 nmol. MR 1452, a relatively preferential antagonist of the kappa opioid receptor, prevented both the antinociceptive and motor effects of dynorphins. Intrathecal somatostatin (25 nmol) had a profile of activity superimposable on that of DYN A-(1-32): long-lasting (up to 24 h) elevation of tail-flick latency with hind limb paralysis, and a shorter (4 h) elevation of the vocalization threshold. MR 1452 did not modify these effects. Intrathecal neurotensin (25 nmol) and s-CT (0.5 nmol) did not alter tail-flick latency or vocalization threshold. However, adopting the hot plate as the analgesimetric test, both peptides elevated the time of hind paw licking, taken as an index of nociception. No signs of motor dysfunction were observed at the doses employed.

    Topics: Animals; Calcitonin; Dynorphins; Injections, Spinal; Male; Movement; Neurotensin; Nociceptors; Pain; Rats; Rats, Inbred Strains; Somatostatin; Time Factors

1988
Inflammation of the hind limb as a model of unilateral, localized pain: influence on multiple opioid systems in the spinal cord of the rat.
    Pain, 1988, Volume: 35, Issue:3

    Inoculation of the right hind paw with Mycobacterium butyricum rapidly led to swelling and inflammation. The afflicted limb showed an enhanced sensitivity to noxious pressure (hyperalgesia) and a reduced sensitivity to noxious heat 24 h following treatment. Both naloxone and MR 2266 (which has greater activity at kappa-opioid receptors) further increased the sensitivity to pressure (that is, potentiated the hyperalgesia) but did not affect the response to heat. They did not affect the response of the uninflamed paw. At 1 week, only MR 2266 was effective. At both 24 h and 1 week, the inflamed paw showed pronounced supersensitivity to the antinociceptive action of morphine against noxious pressure. At both 24 h and (to a greater extent) 1 week, a rise in levels of immunoreactive (ir)-dynorphin (DYN) was seen in the ipsilateral dorsal horn of the lumbar spinal cord. There was no alteration in the contralateral dorsal horn or in either ventral horn. Furthermore, levels of ir-met-enkephalin (ME) and ir-leu-enkephalin (LE) were unaffected. There was no difference in the density of mu-, delta- or kappa-binding sites in any part of the lumbar cord, at either 24 h or 1 week, between ipsilateral and contralateral tissue. By 3 and 5 weeks postinoculation, the symptoms had spread to the contralateral hind limb and ir-DYN was elevated in the contralateral dorsal horn and the ipsilateral ventral horn. At 5 weeks, levels of ir-ME and ir-LE also were increased in the ipsilateral and contralateral dorsal horns, but not in the contralateral ventral horn. Furthermore, levels of ir-DYN were increased in the cervico-thoracic spinal cord, and rats displayed adrenal hypertrophy and a rise in plasma levels of ir-beta-endorphin (beta-EP). These data indicate: (1) Peripheral inflammation localized to a single limb selectively modifies levels of ir-DYN in ipsilateral dorsal horn. The effect is specific to DYN as compared to ME and LE. The density of mu-, delta-, or kappa-receptors in the lumbar spinal cord is unmodified. (2) The altered response to opioid agonists and antagonists shown by rats with an inflamed limb may be selective to the injured tissue. (3) Alterations in opioid systems associated with unilateral hind limb inflammation may not be exclusively chronic in nature: they appear very rapidly (within 24 h) of the induction of pain. With time, the contralateral limb becomes affected and, eventually, the effects resemble those seen with generalized polyarthritis.

    Topics: Animals; Benzomorphans; Disease Models, Animal; Dynorphins; Endorphins; Hindlimb; Inflammation; Male; Naloxone; Pain; Rats; Rats, Inbred Strains; Receptors, Opioid; Spinal Cord

1988
Spinal antinociceptive action of three representative opioid peptides in frogs.
    Brain research, 1987, Jan-27, Volume: 402, Issue:1

    Spinal administration of low doses of dynorphin, beta-endorphin or Met-enkephalin produces a potent, dose-dependent increase in the nociceptive threshold in the unanesthetized frog, Rana pipiens. Nociceptive thresholds were determined by using the acetic acid test, previously shown to be a sensitive indicator of antinociception in this amphibian species. Of particular interest, spinally administered dynorphin produces a potent antinociception in frogs without any signs of motor dysfunction seen after spinally administered dynorphin in mammalian species.

    Topics: Analgesics; Animals; beta-Endorphin; Dynorphins; Endorphins; Enkephalin, Methionine; Pain; Rana pipiens; Sensory Thresholds; Spinal Cord

1987
Time-dependent neurobiological effects of colchicine administered directly into the hippocampus of rats.
    Brain research, 1987, Apr-07, Volume: 408, Issue:1-2

    Rats were given bilateral injections of colchicine into the dorsal and ventral hippocampus. Behavioral, neurochemical and histopathological measurements were taken, up to 12 weeks after surgery. Colchicine produced a consistent increase in spontaneous motor activity, enhanced acoustic startle reactivity, and accelerated acquisition of two-way shuttle box avoidance, but did not affect reactivity to a noxious thermal stimulus. Measurement of dynorphin in the hippocampus indicated that colchicine rapidly depleted this neuropeptide, which is thought to be contained preferentially in the mossy fibers of granule cells of the hippocampus. Colchicine also decreased Met-enkephalin in the hippocampus, but the magnitude of the change (22%) was less than that (89% depletion) observed for hippocampal dynorphin. Examination of hippocampal morphology using light microscopic techniques indicated that colchicine caused approximately 60% degeneration of granule cells in the hippocampus. Although the length of the pyramidal cells was decreased (12-16%), the width of the CA1 and CA3 region of the hippocampus was not affected. These data underscore the importance of the granule cells in the mediation of behavioral processes such as motor activity, startle reactivity and performance of shuttle box avoidance.

    Topics: Acoustic Stimulation; Animals; Avoidance Learning; Behavior, Animal; Colchicine; Dynorphins; Enkephalin, Methionine; Hippocampus; Male; Motor Activity; Pain; Peptide Fragments; Rats; Rats, Inbred F344; Reaction Time; Reflex, Startle; Time Factors

1987
Bilateral intranigral microinjection of morphine and opioid peptides produces antinociception in rats.
    Brain research, 1987, May-12, Volume: 411, Issue:1

    Bilateral intranigral microinjection of morphine produced dose-related and naloxone-reversible antinociceptive effects on the tail-flick and hot-plate tests. Intranigral injection of enkephalin had antinociceptive effects on both tests, and dynorphin had an antinociceptive effect on the hot-plate test. This is the first report of evidence that nigral opiate receptors may mediate antinociception.

    Topics: Animals; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalin, Leucine-2-Alanine; Enkephalins; Male; Microinjections; Morphine; Naloxone; Pain; Pain Measurement; Rats; Rats, Inbred Strains; Reaction Time; Substantia Nigra

1987
Effects of selective and non-selective kappa-opioid receptor agonists on cutaneous C-fibre-evoked responses of rat dorsal horn neurones.
    Brain research, 1987, Jul-07, Volume: 415, Issue:1

    We have studied the effects of 3 putative kappa-opioid receptor agonists, U50488H, ethylketocyclazocine (EKC) and dynorphin A1-13 (DYN) on the processing of nociceptive information in the dorsal horn of the rat under halothane anaesthesia. Extracellular single unit recordings were made from convergent or multireceptive lumbar dorsal horn neurones, which could be excited by impulses in A beta and C fibre afferents following transcutaneous electrical stimulation of their ipsilateral hind paw receptive fields and also by noxious and innocuous natural stimuli. Agonists were applied directly onto the surface of the spinal cord. DYN and U50488H consistently produced both a facilitation and inhibition of the C-fibre evoked nociceptive responses of individual cells, these dual effects being relatively insensitive to naloxone antagonism and cancelled each other for the whole population of cells. A beta fibre-evoked responses were little altered. In contrast, EKC consistently depressed C-fibre transmission in a dose-dependent, naloxone reversible manner, analogous to, but considerably less potent than intrathecal morphine under identical experimental conditions. Agonist-induced effects on neuronal responses to natural stimulation (noxious pinch and innocuous prod) were consistent with the changes observed with the electrically evoked responses. The present results therefore indicate that EKC probably exerts its spinal antinociceptive activity in the rat spinal cord in a manner akin to mu-receptor activation. Results with U50488H and DYN indicate that -opioids can excite and inhibit individual neurones but produce no overall change on the whole population, so differing from effects mediated by the other opiate receptors.

    Topics: 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer; Action Potentials; Animals; Cyclazocine; Dose-Response Relationship, Drug; Dynorphins; Electric Stimulation; Ethylketocyclazocine; Injections, Spinal; Male; Naloxone; Nerve Fibers; Pain; Peptide Fragments; Pyrrolidines; Rats; Rats, Inbred Strains; Receptors, Opioid; Receptors, Opioid, kappa; Skin; Spinal Cord; Time Factors

1987
Intrathecal dynorphin(1-13) results in an irreversible loss of the tail-flick reflex in rats.
    Brain research, 1987, Dec-01, Volume: 435, Issue:1-2

    Intrathecal injection of dynorphin produced a loss of the tail-flick reflex that lasted throughout the 14-day experimental period whereas, the inclined plane test of motor function and tail-shock vocalization recovered within an hour. An important aspect of the loss of the tail-flick reflex was that it was an all-or-none event. At any dose of tail-flick latency either remained unchanged when compared with pre-injection latencies or the latency was elevated to the cut off time of 14 s. The ED50's +/- S.E.M. for tail-flick, inclined plane and tail-shock vocalization were 65.4 +/- 5.0, 67.7 +/- 5.0 and 68.0 +/- 3.9 nmol respectively. Results from the hot-plate test revealed no statistical difference between saline and dynorphin injected animals one day following the injection. Animals injected with morphine sulphate s.c. lost the tail-flick reflex but completely recovered by 24 h. Histology of the spinal cord of animals treated with dynorphin 24 h prior to sacrifice revealed dead neurons primarily in the ventral horn with little or no damage in the dorsal horn. These data demonstrate that dynorphin(1-13) injected intrathecally results in a rather specific neurotoxic action in the spinal cord.

    Topics: Animals; Dynorphins; Injections, Spinal; Male; Neurons; Pain; Peptide Fragments; Rats; Rats, Inbred Strains; Reflex; Spinal Cord; Vocalization, Animal

1987
A model of chronic pain in the rat: response of multiple opioid systems to adjuvant-induced arthritis.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 1986, Volume: 6, Issue:4

    Chronic arthritic pain was induced by intradermally inoculating rats at the tail-base with Mycobacterium butyricum, which results in swelling, inflammation, and hyperalgesia of the joints. These symptoms peak at 3 weeks after inoculation and disappear by 10 weeks. The following changes were seen at 3 weeks. Immunoreactive dynorphin (ir-Dyn) and ir-alpha-neo-endorphin (alpha-NE) manifested comparable patterns of change. Their levels were increased in the anterior, but not neurointermediate, pituitary. The thalamus showed a rise in ir-Dyn and ir-alpha-NE, but no alterations were seen in other brain regions. In each case, cervical, thoracic, and lumbosacral sections of the spinal cord showed a rise in ir-Dyn and ir-alpha-NE: This was most pronounced in the lumbosacral region, where the magnitude of these shifts correlated with the intensity of arthritic symptoms. In addition, a moderate elevation in ir-methionine-enkephalin (ME) was seen in lumbosacral spinal cord. In brain, ir was not changed. The level of ir-beta-endorphin (beta-EP) was elevated both in the plasma and the anterior, but not the neurointermediate, pituitary. In addition, the content of messenger RNA encoding the beta-EP precursor, proopiomelanocortin (POMC), was enhanced in the anterior lobe. Thus, there was a selective activation of synthesis of beta-EP in, and its secretion from, the anterior lobe. In no brain tissue did levels of ir-beta-EP change. At 10 weeks postinoculation, the above changes were no longer apparent, indicating their reversibility.(ABSTRACT TRUNCATED AT 250 WORDS)

    Topics: Animals; Arthritis; Arthritis, Experimental; beta-Endorphin; Brain; Brain Chemistry; Chronic Disease; Diprenorphine; Disease Models, Animal; Dynorphins; Endorphins; Hypothalamus; Male; Mesencephalon; Pain; Rats; Rats, Inbred Strains; Receptors, Opioid; Spinal Cord; Thalamus

1986
[Peripheral analgesic actions of opioid peptides and morphine analogues].
    Nihon yakurigaku zasshi. Folia pharmacologica Japonica, 1986, Volume: 88, Issue:2

    Opiates and opioid peptides were administered in the order of 10(-9)-10(-6) mol peripherally, and their action on pain sensitivity was investigated by the modified formalin test which has two characteristic pain responses (the first and the second phase) in the mouse hindpaw. Opioid peptides (20-500 pmol) had dose-dependent analgesia against both first and second phases, and their action ranked dynorphin greater than [D-Ala2, Met5]-enkephalinamide greater than [Met5]-enkephalin. EKC and morphine (0.4-2.5 nmol) inhibited pain response of the first phase, but produced hyperalgesia in the second phase dose-dependently. Lidocaine hydrochloride had peripheral analgesic action, but was about 500-10000 times weaker than these substances. So, these peripheral analgesic actions have a different mechanism from that of local anesthetic action. N-methyl levallorphan which is thought to be a peripherally selective narcotic antagonist reversed these peripheral analgesic actions at the first and second phases and also prevented the hyperalgesic effects of EKC and morphine at the second phase. Naloxone reversed analgesia at only the first phase. These results suggest that an analgesic mechanism by opioids may exist at the peripheral site as well. Furthermore, it is estimated that a receptor exists which is antagonized by N-methyl levallorphan but not by naloxone and that there is a system of hyperalgesia by EKC and morphine in pain modulation.

    Topics: Analgesics, Opioid; Animals; Cyclazocine; Dynorphins; Enkephalin, Methionine; Ethylketocyclazocine; Levallorphan; Lidocaine; Male; Mice; Morphine; Naloxone; Pain; Peptide Fragments

1986
[Effects of opiate and opioid peptides administered intrathecally on the pain threshold and micturition reflex in rats].
    Masui. The Japanese journal of anesthesiology, 1986, Volume: 35, Issue:6

    Topics: Animals; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalin, Leucine-2-Alanine; Female; Injections, Spinal; Morphine; Pain; Rats; Rats, Inbred Strains; Reflex; Sensory Thresholds; Urination

1986
Behavioral effects of centrally administered dynorphin and [D-ala2-D-leu] enkephalin (DADLE) in rats.
    Neuropeptides, 1986, Volume: 8, Issue:3

    Dynorphin and [D-ala2-D-leu]enkephalin (DADLE) were administered directly into the cerebrolateral ventricles of rats and effects on various indices of sensorimotor function and retention of a passive avoidance task were measured. Dynorphin markedly suppressed exploratory motor activity and decreased responsiveness to an acoustic stimulus. Although increases in latency to respond to a noxious thermal stimulus were seen in rats after dynorphin, these changes were always associated with alterations in motor capacity. Injection of dynorphin immediately after a passive avoidance training trial had no significant effect on retention 1 week later. The physiological effects of DADLE were clearly different than those of dynorphin. DADLE produced a biphasic decrease followed by an increase in motor activity and an increased acoustic startle reactivity. DADLE had no effect on reactivity to a noxious thermal stimulus. Posttrial administration of DADLE significantly impaired retention of a step-through passive avoidance task 1 week after training. These data indicate different neurobiological roles for kappa and delta opiate receptors in the central nervous system.

    Topics: Acoustic Stimulation; Animals; Avoidance Learning; Cerebral Ventricles; Dynorphins; Enkephalin, Leucine; Enkephalin, Leucine-2-Alanine; Hot Temperature; Injections, Intraventricular; Male; Motor Activity; Pain; Rats; Rats, Inbred F344; Reflex, Startle

1986
[Dynorphin as an analgesic for the dental pulp].
    Practica odontologica, 1986, Volume: 7, Issue:8

    Topics: Dental Pulp Diseases; Dynorphins; Humans; Pain

1986
[Frequency as the cardinal determinant for electroacupuncture analgesia to be reversed by opioid antagonists].
    Sheng li xue bao : [Acta physiologica Sinica], 1986, Volume: 38, Issue:5

    Topics: Acupuncture Therapy; Animals; beta-Endorphin; Dynorphins; Electric Stimulation Therapy; Endorphins; Enkephalins; Naloxone; Naltrexone; Pain; Rats; Sensory Thresholds; Transcutaneous Electric Nerve Stimulation

1986
Modulators of pain in the spinal cord.
    NIDA research monograph, 1986, Volume: 75

    Intrathecal injection of subanalgesic doses of morphine (7.5 nmol) and dynorphin-A-(1-13) (1.25 nmol) in combination resulted in a marked analgesic effect as assessed by tail flick latency in the rat. The analgesic effect of the composite of dynorphin/morphine was dose-dependent in serial dilutions and not accompanied by any signs of motor dysfunction. Synergism between dynorphin and morphine can also be demonstrated without motor dysfunction, when dynorphin is injected together intrathecally with a serial increasing dosages of morphine. Metenkephalin showed an analogous synergistic effect with dynorphin and also morphine in the spinal cord, although dynorphin (and also metenkephalin) antagonized morphine analgesia by the intraventricular route. The underlying mechanism of the interactions among different classes of opioid ligands at different levels of the central nervous system deserves further study.

    Topics: Analgesia; Animals; Cerebral Ventricles; Dynorphins; Morphine; Pain; Peptide Fragments; Rats; Spinal Cord

1986
[Lars Terenius about neuropeptides and pain: Endorphines alleviate stress--a curative effect? A new spinal-active analgesic insight. Interview by Birgit Lindell].
    Nordisk medicin, 1985, Volume: 100, Issue:3

    Topics: Analgesics; Dynorphins; Endorphins; Humans; Pain; Receptors, Opioid; Receptors, Opioid, kappa; Substance P

1985
Chronic pain in the rat: selective alterations in CNS and pituitary pools of dynorphin as compared to vasopressin.
    Neuropeptides, 1985, Volume: 5, Issue:4-6

    Inoculation of rats at the tail-base with Mycobacterium led to arthritic swelling and inflammation of all four limbs. Immunoreactive (ir)-dynorphin (DYN) increased in anterior but not neurointermediate pituitary. In the brain, only thalamus showed a rise and, in spinal cord, a large elevation was seen. Ir-vasopressin (VP) was not affected in these tissues but increased in midbrain. These effects might reflect a role of DYN in the control of chronic pain. In addition, they support a differential modulation of DYN as compared to VP extrinsic to the hypothalamic-neurohypophyseal axis.

    Topics: Animals; Central Nervous System; Chronic Disease; Dynorphins; Mesencephalon; Pain; Pituitary Gland, Anterior; Rats; Spinal Cord; Thalamus; Vasopressins

1985
Possible involvement of dynorphinergic system in nociceptive transmission at spinal level.
    Neuropeptides, 1985, Volume: 5, Issue:4-6

    The opioid peptide dynorphin1-32 (DYN1-32, 25 nmol) intrathecally administered causes, in the rat, an elevation of nociceptive threshold of longer duration than that of DYN A, as ascertained by vocalization test. Comparative findings obtained with tail flick test allow to differentiate antinociception from motor dysfunction. The breakdown of DYN A at spinal level is very rapid. The electrical stimulation of the tail associated to a restraint condition of the rat produces a significant increase of immunoreactive DYN in cervical, thoracic and lumbar segments of spinal cord, therefore indicating a correlative, if not causal, relationship between the spinal dynorphinergic system and aversive stimuli.

    Topics: Analgesics; Animals; Dynorphins; Male; Pain; Rats; Rats, Inbred Strains; Sensory Thresholds; Spinal Cord; Synaptic Transmission

1985
Characterization of dynorphin A-induced antinociception at spinal level.
    European journal of pharmacology, 1985, Mar-26, Volume: 110, Issue:1

    Dynorphin A (DYN A) injected intrathecally in the rat produced a significant elevation of the nociceptive threshold, measured by the tail flick test. The highest dose of DYN A (25 nmol) produced maximal elevation of tail flick latency to radiant heat together with hindlimb paralysis and tail flaccidity lasting several hours, thus confirming several previous reports. A lower dose of DYN A (12.5 nmol) produced only a smaller, not constant, short-lasting change in the nociceptive threshold. The vocalization test (electrical stimulation of the tail) gave a different result: the time course curve showed that the antinociceptive effect had worn off 60 min after DYN A 25 nmol. Thus it can be assumed that the prolonged depression of the tail flick reflex was related to motor dysfunction and did not completely reflect the animal's response to painful stimuli. Tolerance to the antinociceptive and motor effects developed after the chronic intrathecal infusion of DYN A with osmotic minipumps. Intrathecal MR 1452 (30 nmol), a purported kappa-receptor blocker, fully prevented the effects of DYN A but not morphine-induced antinociception. Naloxone antagonized DYN A only at a 4 fold higher dose. MR 1452 (90 nmol) administered after DYN A reversed the elevation of the vocalization threshold while tail flick latency remained unmodified. Analysis by high performance liquid chromatography of intrathecally injected radiolabelled DYN A revealed that DYN A was largely broken down about 10 min after its administration. Our results seem to indicate that DYN A in the spinal cord causes alterations in nociception and motor function, clearly distinguishable in time and both mediated by an opioid receptor, probably of the kappa type. However, different mechanism(s), possibly non-opioid in nature, may contribute to the prolonged depression of the tail flick.

    Topics: Analgesia; Animals; Benzomorphans; Drug Tolerance; Dynorphins; Male; Naloxone; Pain; Rats; Rats, Inbred Strains; Sensory Thresholds; Spinal Cord

1985
Spinal cord dynorphin may modulate nociception via a kappa-opioid receptor in chronic arthritic rats.
    Brain research, 1985, Aug-05, Volume: 340, Issue:1

    Inoculation of rats with Mycobacterium butyricum produced an arthritis of the limbs which revealed an enhanced sensitivity to noxious mechanical pressure (hyperalgesia). Arthritic rats displayed a pronounced rise in immunoreactive dynorphin in lumbo-sacral spinal cord which correlated both with the intensity and time-course of this hyperalgesia. MR-2266, a relatively preferential antagonist at the chi-opioid receptor (at which dynorphin is considered to act) potentiated this hyperalgesia. In contrast, MR 2267 (its inactive stereo-isomer) was ineffective. Further, naloxone (a weak chi-antagonist), and ICI 154,129 (a preferential delta-antagonist) were, in each case, inactive. The data demonstrate a pronounced response of spinal dynorphin to chronic arthritic pain in the rat. In addition, they raise the possibility of a function of spinal DYN, via a chi-receptor, in the modulation of chronic arthritic pain.

    Topics: Animals; Arthritis; Chronic Disease; Conditioning, Classical; Dynorphins; Enkephalin, Methionine; Mycobacterium Infections; Naloxone; Nociceptors; Pain; Radioimmunoassay; Rats; Receptors, Opioid; Receptors, Opioid, kappa; Spinal Cord

1985
Naloxone-reversible and non-reversible shock-induced analgesia during development.
    Neuroscience letters, 1985, Oct-10, Volume: 60, Issue:3

    We evaluated the development of naloxone-reversible and naloxone-non-reversible analgesia induced by footshock in rats of different ages and correlated it with the concentrations of beta-endorphin and dynorphin in brain areas and the spinal cord. We observed that naloxone-non-reversible shock-induced analgesia appeared first and its appearance might be related to the early presence of high dynorphin concentrations in the spinal cord. Naloxone-reversible analgesia appeared later together with the reaching of adult concentrations of cerebral beta-endorphin.

    Topics: Analgesia; Animals; beta-Endorphin; Central Nervous System; Dynorphins; Electroshock; Endorphins; Enkephalin, Methionine; Male; Naloxone; Pain; Rats; Rats, Inbred Strains

1985
Antinociceptive profiles of opioid peptide agonists in a rat tooth pulp stimulation procedure.
    Peptides, 1985, Volume: 6 Suppl 2

    The analgesic activity of the prototypic opioid peptides for the mu (D-Ala2-Me-Phen4-Gly-ol5-enkephalin [DAGO]) kappa (Dynorphin 1-13), delta (D-Ala2-D-Leu5-enkephalin [DADLE]), or epsilon (beta-endorphin) receptor was assessed in a rat tooth pulp stimulation procedure. All opioid peptides tested and the opioid alkaloid U50, 488H (kappa receptor agonist) significantly elevated response thresholds. The rank order of potency based on the Minimum Effective Dose values was beta-endorphin greater than DAGO = dynorphin A (1-13) amide greater than DADLE greater than dynorphin A (1-13) greater than U50,488H. Based on absolute magnitude, the rank order of dose response slopes was DAGO greater than U50,488H greater than dynorphin A (1-13) amide greater than beta-endorphin greater than DADLE. Dynorphin A (1-13) produced the shallowest dose response slope and the magnitude of response threshold was the lowest for all compounds tested. Finally, the general conclusion that mu agonists are effective against noxious stimuli derived from thermal, chemical, and mechanical is extended by our data to include electrical sources derived from tooth pulp stimulation; kappa agonists are effective against noxious stimuli derived from chemical, mechanical, and electrical sources (tooth pulp stimulation) and delta agonists are effective analgesics against thermal, chemical and electrical stimuli (tooth pulp stimulation).

    Topics: 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer; Analgesics; Animals; beta-Endorphin; Dental Pulp; Dynorphins; Electric Stimulation; Endorphins; Enkephalin, Ala(2)-MePhe(4)-Gly(5)-; Enkephalin, Leucine; Enkephalin, Leucine-2-Alanine; Enkephalins; Male; Pain; Peptide Fragments; Pyrrolidines; Rats; Rats, Inbred Strains

1985
Multiple opioid peptides and the modulation of pain: immunohistochemical analysis of dynorphin and enkephalin in the trigeminal nucleus caudalis and spinal cord of the cat.
    The Journal of comparative neurology, 1985, Oct-22, Volume: 240, Issue:4

    Using immunocytochemistry, we have identified important differences in the distribution of immunoreactive dynorphin and enkephalin cells and terminals in the trigeminal nucleus caudalis and in the spinal dorsal horn of the cat. Dynorphin immunoreactive processes are more closely associated with those regions of cord that process nociceptive information, specifically laminae I and V. Enkephalin neurons and terminals are more widespread. Based on the staining pattern with an antiserum to the octapeptide-metenkephalin-arg-gly-leu, we suggest that the dense enkephalin terminal immunoreactivity in the inner part of the substantia gelatinosa derives from cells in lamina III. There are also significant differences in the anatomical relationship of the two opioid peptides with the organization of parasympathetic autonomic preganglionic neurons. The functional significance of these observations must await physiological analysis; nevertheless, it is almost certain that differences will be found and that these will be important in understanding the mechanisms through which exogenous opiates and a variety of descending control systems exert their effects on spinal cord neurons.

    Topics: Animals; Cats; Dynorphins; Enkephalins; Immunohistochemistry; Nerve Endings; Pain; Spinal Cord; Trigeminal Nucleus, Spinal

1985
Dynorphin: potent analgesic effect in spinal cord of the rat.
    Scientia Sinica. Series B, Chemical, biological, agricultural, medical & earth sciences, 1984, Volume: 27, Issue:2

    Evidences are presented to show a strong and long-lasting analgesic effect after injection of dynorphin into the subarachnoid space of the spinal cord in the rat. Taking the amplitude and time course of the increase of tail flick latency as the indices of analgesia, dynorphin elicited dose-dependent analgesic effect in the range of 2.3-18.6 nmol. Calculating on a molar basis dynorphin was 6-10 times more potent than morphine and 65-100 times more potent than morphiceptin, another mu opiate receptor agonist. Dynorphin analgesia was completely reversed by intrathecal injection of anti-dynorphin IgG and partially reversed by naloxone. Acute tolerance to morphine analgesia did not affect the occurrence of dynorphin analgesia, indicating the absence of cross tolerance between morphine and dynorphin. Evidence from different lines of approach suggests that dynorphin may bind with kappa opiate receptors in the spinal cord to exert its analgesic effect.

    Topics: Analgesia; Analgesics; Animals; Drug Tolerance; Dynorphins; Endorphins; Female; Injections, Intraventricular; Injections, Spinal; Morphine; Naloxone; Pain; Peptide Fragments; Rats; Sensory Thresholds; Spinal Cord

1984
[Dynorphin: analgesic effect via kappa receptors in spinal cord of the spinal cords of rats].
    Zhongguo yao li xue bao = Acta pharmacologica Sinica, 1984, Volume: 5, Issue:4

    Topics: Analgesics; Animals; Cyclazocine; Dynorphins; Ethylketocyclazocine; Female; Morphine; Naloxone; Pain; Rats; Receptors, Opioid; Receptors, Opioid, kappa; Sensory Thresholds; Spinal Cord

1984
Peptides and nociception.
    International review of neurobiology, 1984, Volume: 25

    Topics: Adrenocorticotropic Hormone; Animals; beta-Endorphin; Bombesin; Bradykinin; Cholecystokinin; Dynorphins; Endorphins; Enkephalins; Melanocyte-Stimulating Hormones; Mice; Nervous System; Neurotensin; Pain; Peptide Fragments; Peptides; Rats; Somatostatin; Substance P; Thyrotropin-Releasing Hormone; Vasopressins

1984
Evidence for a role of the ventro-medial posterior hypothalamus in nociceptive processes in the rat.
    Pharmacology, biochemistry, and behavior, 1983, Volume: 18, Issue:6

    Bilateral, radio-frequency destruction of the ventro-medial posterior hypothalamus (VMPH) resulted, as compared to sham-operated and control rats and evaluated in the tail-flick and vocalization tests, in a significant decrease in basal nociceptive threshold on day 4 post-surgery. By day 12, however, no significant difference between sham and lesioned rats was seen. At this time the antinociception elicited by either acute foot-shock or cold-water-immersion stress was profoundly attenuated. The antinociceptive response to various doses of morphine was not, in contrast, diminished. As established by use of radioimmunoassay, these lesions did not significantly alter hypothalamic levels of beta-endorphin, met-enkephalin, dynorphin or alpha-neo-endorphin. They did, however, produce a pronounced and significant fall in the hypothalamic content of substance P. These data are indicative that the VMPH may, via a mechanism not involving endorphins, be of importance in the determination of basal nociceptive threshold and in the generation of stress-, but not morphine-, evoked antinociception. The relationship of these findings to the interconnections of the VMPH, and to the possible significance of substance P and the pituitary in nociceptive processes, is discussed.

    Topics: Animals; beta-Endorphin; Dynorphins; Electroshock; Endorphins; Enkephalin, Methionine; Hypothalamus; Hypothalamus, Posterior; Male; Morphine; Naltrexone; Nociceptors; Pain; Protein Precursors; Rats; Rats, Inbred Strains; Stereotaxic Techniques

1983
Possible medullary kappa hyperalgesic mechanism. I. A new potential role for endogenous opioid peptides in pain perception.
    Life sciences, 1983, Oct-31, Volume: 33, Issue:18

    The kappa-agonist, ethylketazocine, produces hyperalgesia in the acutely decerebrated dog as indicated by a shortening of the skin twitch reflex latency whereas fentanyl is inactive. Naloxone produces analgesia and antagonizes the hyperalgesic effect of ethylketazocine. Spinal cord transection decreases the latency of the skin twitch reflex and allowed the analgesic effect of fentanyl and ethylketazocine on this nociceptive reflex to become manifest. These observations indicate that there is a non-opioid analgesic and kappa hyperalgesic mechanism present in the pontine-medullary region of the dog brainstem. It suggests that the hyperalgesic mechanism is mediated by an endogenous kappa-opioid peptide and that the analgesic effect of naloxone is in part related to antagonism of the activity of this hyperalgesia producing opioid peptide.

    Topics: Animals; Cyclazocine; Dogs; Dynorphins; Endorphins; Ethylketocyclazocine; Medulla Oblongata; Naloxone; Pain; Receptors, Opioid; Reflex

1983
Sites of analgesic action of dynorphin.
    Life sciences, 1983, Volume: 33 Suppl 1

    Analgesic effects of dynorphin and dynorphin-(1-13) injected into various regions of the CNS of rats were investigated using the tail pinch method. Dynorphin and dynorphin-(1-13) produced a dose-dependent analgesic effect when injected into the third ventricle (3rd vent.) or the nuclei reticularis gigantocellularis and paragigantocellularis (NRGC-NRPG) and the analgesic activity of dynorphin was 3 to 5 times more potent than that of dynorphin-(1-13). These peptides in higher doses elicited transient "barrel-rolling" and rigidity in behavior. On the other hand, when injected intrathecally, dynorphin exerted an analgesia accompanied by paralysis of hindlimbs which were long lasting. But its analgesic activity was less effective as compared with the 3rd vent. and NRGC-NRPG injections.

    Topics: Analgesia; Analgesics; Animals; Dynorphins; Endorphins; Injections, Intraventricular; Male; Pain; Peptide Fragments; Rats; Rats, Inbred Strains

1983
In vivo evidence for multiple opiate receptors mediating analgesia in the rat spinal cord.
    Brain research, 1982, Sep-09, Volume: 247, Issue:1

    In rats implanted with chronic catheters in the spinal subarachnoid space, intrathecal injections of SKF 10047 and dynorphin did not produce any elevation of the nociceptive threshold as defined by hot-plate and tail-flick tests. In contrast, intrathecal ethylketocyclazocine (EKC) and (D-Ala2,D-Leu5)-enkephalin (DADL) administration resulted in a dose-dependent antinociceptive effect which was reversible with intraperitoneal naloxone. Calculation of the Schild dose-ratio plots for the data derived from systemically administered naloxone reveals a slope of--1 and a calculated pA2 value of 6.8 for EKC and 6.2 for DADL. Also, animals made tolerant to systemic morphine showed a diminished analgesic response to intrathecal morphine and EKC when compared to naive animals. There was, however, no significant change in the dose response curve of intrathecal DADL. Thus, these experiments suggest that in addition to mu receptors a separate subpopulation of delta but not kappa or sigma receptors are involved with spinally mediated analgesia.

    Topics: Animals; Cyclazocine; Dose-Response Relationship, Drug; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalin, Leucine-2-Alanine; Ethylketocyclazocine; Injections, Spinal; Male; Naloxone; Pain; Phenazocine; Rats; Rats, Inbred Strains; Receptors, Opioid; Spinal Cord

1982
Nonopiate effects of dynorphin and des-Tyr-dynorphin.
    Science (New York, N.Y.), 1982, Dec-10, Volume: 218, Issue:4577

    Intracerebroventricular administration of dynorphin produced potent and long-lasting effects on motor function and the electroencephalogram in rats. In addition, local iontophoretic or pressure ejection of dynorphin consistently inhibited hippocampal unit activity. None of these effects were significantly affected by naloxone even at high doses. Moreover, a fragment of dynorphin that failed to displace any of a number of tritiated narcotics from rat brain homogenates produced similar effects on these physiological measures in vivo. On the basis of a variety of criteria for "opiate action," the results suggest that a second biologically active site within the dynorphin sequence is capable of quite potent but nonopiate effects.

    Topics: Action Potentials; Amino Acid Sequence; Animals; Dynorphins; Endorphins; Hippocampus; Male; Pain; Rats; Structure-Activity Relationship

1982
Immunoreactive dynorphin in mammalian spinal cord and dorsal root ganglia.
    Proceedings of the National Academy of Sciences of the United States of America, 1981, Volume: 78, Issue:12

    The distribution of immunoreactive dynorphin (ir-dynorphin) has been determined in dorsal and ventral aspects of spinal cord and in dorsal root ganglia of rabbit and rat. Concentrations are highest in dorsal root, with intermediate levels in ventral cord and low levels in dorsal root ganglia of both species. Levels of ir-dynorphin are relatively uniform over examined segments (vertebrae C2-S3) of rabbit spinal cord and dorsal root ganglia. Gel permeation chromatography of extracts from rabbit dorsal and ventral spinal cord and dorsal root ganglia revealed at least three immunoreactive components of differing molecular size in all three structures. Multiple unilateral or bilateral dorsal rhizotomy (vertebrae C5-T1) in rat did not affect levels of ir-dynorphin in spinal cord. As reported [Goldstein, A. & Ghazarossian, V. E. (1980) Proc. Natl. Acad. Sci. USA 77, 6207-6210], midthoracic spinal transection was without effect. Within the spinal cord, the neuropeptide appears, most probably, to be contained in short-axoned neurons. We surmise that this potent opioid peptide may participate in the processing of sensory information in spinal cord.

    Topics: Animals; Dynorphins; Endorphins; Ganglia, Spinal; Neurotransmitter Agents; Pain; Rabbits; Radioimmunoassay; Spinal Cord

1981
Behavioral effects of dynorphin 1-13 in the mouse and rat: initial observations.
    Peptides, 1980,Winter, Volume: 1, Issue:4

    Dynorphin is a recently identified, pharmacologically potent endogenous opioid peptide. Heretofore it has not been characterized for its behavioral effects. The effects of centrally infused dynorphin upon a variety of behaviors were therefore examined in mice and rats. The present findings point to a specific profile of behavioral activity. The peptide was active in facilitating feeding and grooming, but was inactive in modifying pain sensitivity and rearing behavior. Naloxone was generally ineffective in reversing behavioral effects. Dynorphin thus appears to have some opiate-like effects upon exogenous administration but may be rapidly broken down into a behaviorally potent non-opiate peptide fragment.

    Topics: Animals; Behavior, Animal; Dynorphins; Endorphins; Feeding Behavior; Grooming; Male; Mice; Naloxone; Pain; Peptide Fragments; Rats; Rats, Inbred Strains

1980