116 research outputs found

    Involvement of S-nitrosylation of actin in inhibition of neurotransmitter release by nitric oxide

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>The role of the diffusible messenger nitric oxide (NO) in the regulation of pain transmission is still a debate of matter, pro-nociceptive and/or anti-nociceptive. <it>S</it>-Nitrosylation, the reversible post-translational modification of selective cysteine residues in proteins, has emerged as an important mechanism by which NO acts as a signaling molecule. The occurrence of <it>S</it>-nitrosylation in the spinal cord and its targets that may modulate pain transmission remain unclarified. The "biotin-switch" method and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry were employed for identifying <it>S</it>-nitrosylated proteins.</p> <p>Results</p> <p>Here we show that actin was a major protein <it>S</it>-nitrosylated in the spinal cord by the NO donor, <it>S</it>-nitroso-<it>N</it>-acetyl-DL-penicillamine (SNAP). Interestingly, actin was <it>S</it>-nitrosylated, more in the S2 fraction than in the P2 fraction of the spinal homogenate. Treatment of PC12 cells with SNAP caused rapid <it>S</it>-nitrosylation of actin and inhibited dopamine release from the cells. Just like cytochalasin B, which depolymerizes actin, SNAP decreased the amount of filamentous actin cytoskeleton just beneath the membrane. The inhibition of dopamine release was not attenuated by inhibitors of soluble guanylyl cyclase and cGMP-dependent protein kinase.</p> <p>Conclusion</p> <p>The present study demonstrates that actin is a major <it>S</it>-nitrosylated protein in the spinal cord and suggests that NO directly regulates neurotransmitter release by <it>S</it>-nitrosylation in addition to the well-known phosphorylation by cGMP-dependent protein kinase.</p

    Localization of orphanin FQ (nociceptin) peptide and messenger RNA in the central nervous system of the rat

    Full text link
    Orphanin FQ (OFQ) is the endogenous agonist of the opioid receptor-like receptor (ORL-1). It and its precursor, prepro-OFQ, exhibit structural features suggestive of the opioid peptides. A cDNA encoding the OFQ precursor sequence in the rat recently has been cloned, and the authors recently generated a polyclonal antibody directed against the OFQ peptide. In the present study, the authors used in situ hybridization and immunohistochemistry to examine the distribution of OFQ peptide and mRNA in the central nervous system of the adult rat. OFQ immunoreactivity and prepro-OFQ mRNA expression correlated virtually in all brain areas studied. In the forebrain, OFQ peptide and mRNA were prominent in the neocortex endopiriform nucleus, claustrum, lateral septum, ventral forebrain, hypothalamus, mammillary bodies, central and medial nuclei of the amygdala, hippocampal formation, paratenial and reticular nuclei of the thalamus, medial habenula, and zona incerta. No OFQ was observed in the pineal or pituitary glands. In the brainstem, OFQ was prominent in the ventral tegmental area, substantia nigra, nucleus of the posterior commissure, central gray, nucleus of Darkschewitsch, peripeduncular nucleus, interpeduncular nucleus, tegmental nuclei, locus coeruleus, raphe complex, lateral parabrachial nucleus, inferior olivary complex, vestibular nuclear complex, prepositus hypoglossus, solitary nucleus, nucleus ambiguous, caudal spinal trigeminal nucleus, and reticular formation. In the spinal cord, OFQ was observed throughout the dorsal and ventral horns. The wide distribution of this peptide provides support for its role in a multitude of functions, including not only nociception but also motor and balance control, special sensory processing, and various autonomic and physiologic processes. J. Comp. Neurol. 406:503–547, 1999. © 1999 Wiley-Liss, Inc.Peer Reviewedhttp://deepblue.lib.umich.edu/bitstream/2027.42/34452/1/7_ftp.pd

    Anti-nociceptive responses produced by human putative counterpart of nocistatin

    No full text
    b-nocistatin is a heptadecapeptide produced from bovine prepronociceptin and blocks the induction of hyperalgesia and touch-evoked pain (allodynia) by intrathecal administration of nociceptin or prostaglandin E(2) (PGE(2)). Human prepronociceptin may generate a 30-amino acid peptide different in length from b-nocistatin. Here, we examine whether the human putative counterpart of nocistatin (h-nocistatin) possessed the same biological activities as b-nocistatin. Simultaneous intrathecal injection of h-nocistatin in mice blocked the induction of allodynia by nociceptin and PGE(2) in a dose-dependent manner with ID(50) values of 329 pg kg(−1) and 16.6 ng kg(−1), respectively. h-nocistatin was about 10 times less potent than b-nocistatin. h-nocistatin also attenuated the nociceptin- and PGE(2)-induced hyperalgesia. These results demonstrate that h-nocistatin is biologically active and may be involved in the processing of pain at the spinal level in humans

    Towards a Receptor for Nocistatin?

    No full text
    Nocistatin is a peptide derived from the pronociceptin precursor, the source of nociceptin, the endogenous ligand for the nociceptin (NOP or ORL1). Despite nocistatin showing activity in a wide range of assays for nociception and other CNS activities, there is a dearth of information regarding the cellular actions of this peptide in the brain, and no receptor for nocistatin has been identified. In a study published in this issue of the British Journal of Pharmacology, Fantin and colleagues demonstrate that nocistatin inhibits 5-HT release from cortical synaptosomes in a concentration-dependent and Pertussis toxin-sensitive manner. The actions of nocistatin are independent of activity at NOP receptors. This study represents the first unambiguous demonstration of nocistatin agonist actions in brain and, taken together with previous work in the spinal cord, provides strong evidence that there is an as yet unidentified G protein-coupled receptor for nocistatin

    The D Prostanoid Receptor Agonist BW245C [(4 S

    No full text

    Central injections of nocistatin or its C-terminal hexapeptide exert anxiogenic-like effect on behaviour of mice in the plus-maze test

    No full text
    1. Nocistatin (NST) antagonizes several actions of nociceptin/orphanin FQ (N/OFQ), but acts on distinct receptors. As N/OFQ exerts anxiolytic-like actions in various tests, its behavioural actions in the elevated plus-maze (EPM) test were compared with those of bovine NST. 2. Five minutes after i.c.v. treatment, mice were placed on the EPM for 5 min and entries into and time spent on open and closed arms were recorded alongside other parameters. 3. NST (0.1 – 3 pmol) reduced percentages of entries into (control 39.6±3.1%, peak effect at 1 pmol NST 8.5±2.9%) and time spent on open arms (control 30.8±2.3%, NST 2.7±1.5%). The C-terminal hexapeptide of NST (NST-C6; 0.01 – 10 pmol) closely mimicked these actions of NST, with peak effects at 0.1 pmol. 4. N/OFQ (1 – 100 pmol) increased percentages of entries into (control 38.5±3.4%; peak effect at 10 pmol N/OFQ 67.9±4.9%) and time spent on open arms (control 32.0±3.8%; N/OFQ 74.9±5.8%). Closed arm entries, an index of locomotor activity, were unchanged by all peptides. 5. Effects of NST or NST-C6, but not N/OFQ, were still detectable 15 min after injection. Behaviour of animals co-injected with NST (1 pmol) or NST-C6 (0.1 pmol) plus N/OFQ (10 pmol) was indistinguishable from that of controls. 6. These results reveal potent anxiogenic-like actions of NST and NST-C6, and confirm the anxiolytic-like properties of N/OFQ. As NST and N/OFQ both derive from preproN/OF, anxiety may be modulated in opposing directions depending on how this precursor is processed
    corecore