dynorphins has been researched along with rimorphin* in 177 studies
2 review(s) available for dynorphins and rimorphin
Article | Year |
---|---|
Combinatorial chemistry--applications of light-directed chemical synthesis.
Combinatorial methods in biology and chemistry are proving to be powerful methods for generating molecular diversity. One approach, light-directed chemical synthesis, combines semiconductor-based photolithography technologies with solid-phase organic chemistry to synthesize large arrays of molecules with potential biological activity. This novel technology has the potential to provide libraries of both natural and synthetic molecules that might be screened rapidly for biological activity. Topics: Amino Acid Sequence; Base Sequence; Biotechnology; Carbamates; DNA; Dynorphins; Endorphins; Molecular Sequence Data; Oligonucleotides; Peptides; Photochemistry | 1994 |
Pain, nociception and spinal opioid receptors.
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 |
2 trial(s) available for dynorphins and rimorphin
Article | Year |
---|---|
Opioid peptides in response to mental stress in asymptomatic dilated cardiomyopathy.
Fourteen asymptomatic dilated cardiomyopathy patients showing normal plasma levels of beta-endorphin, Met-enkephalin, dynorphin B, norepinephrine and endothelin-1 but elevated atrial natriuretic factor (ANF) levels underwent two Mental Arithmetic Tests (MAT), with placebo and naloxone hydrochloride infusion, respectively. MAT significantly (p < 0.01) increased blood pressure, heart rate, opioid peptides, norepinephrine, ANF, but not endothelin-1. Naloxone infusion significantly (p < 0.05) attenuated the increments produced by MAT in all measured parameters during placebo infusion. These results indicate that in asymptomatic dilated cardiomyopathy the endogenous opioid system, activated by stress-induced sympathoadrenergic hyperactivity, may further increase the sympathetic tone in a positive feedback that is interrupted by naloxone. Topics: Atrial Natriuretic Factor; beta-Endorphin; Blood Pressure; Cardiomyopathy, Dilated; Dynorphins; Endorphins; Endothelin-1; Enkephalin, Methionine; Female; Heart Rate; Humans; Intelligence Tests; Male; Mathematics; Middle Aged; Naloxone; Narcotic Antagonists; Norepinephrine; Opioid Peptides; Stress, Psychological | 1998 |
Endogenous opioid system and atrial natriuretic factor in normotensive offspring of hypertensive parents at rest and during exercise test.
To investigate the effects of the endogenous opioid system on plasma atrial natriuretic factor (ANF) levels during sympathetic hyperactivity.. We studied the young normotensive offspring of parents who both had essential hypertension, who are characterized by a hyperactive sympathetic nervous system.. We assessed plasma beta-endorphin, met-enkephalin, dynorphin B, ANF and noradrenaline levels, blood pressure and heart rate values in eight normotensive offspring and in 10 young normotensive subjects with no family history of hypertension (controls) at rest and during two exercise tests: the first test performed with the infusion of placebo (1.5 ml/min saline) and the second test with the infusion of an opioid antagonist (9.5 micrograms/kg per min naloxone hydrochloride). ANF and opioids were radioimmunoassayed after chromatographic pre-extraction.. At rest plasma met-enkephalin, dynorphin B, ANF and noradrenaline values in the normotensive offspring were significantly higher than in the controls. Exercise with placebo significantly raised all hormonal and haemodynamic parameters in the two groups. This increase was significantly higher in the normotensive offspring than in the controls. Naloxone did not modify any parameter in either group at rest, but it enhanced further the rise in plasma noradrenaline levels induced by exercise in both groups. A similar effect of naloxone during exercise was observed for plasma ANF levels in the normotensive offspring.. Our findings show that plasma met-enkephalin, dynorphin B, ANF and noradrenaline levels at rest and during exercise are higher in normotensive offspring than in controls. The effects of naloxone indicate that in normotensive offspring at rest the opioid system does not affect ANF release, whereas during exercise it attenuates ANF hypersecretion, possibly by reducing noradrenaline release. Topics: Adult; Atrial Natriuretic Factor; beta-Endorphin; Disease Susceptibility; Dynorphins; Endorphins; Enkephalin, Methionine; Exercise Test; Female; Hemodynamics; Humans; Hypertension; Male; Naloxone; Norepinephrine; Opioid Peptides; Physical Exertion; Radioimmunoassay | 1994 |
173 other study(ies) available for dynorphins and rimorphin
Article | Year |
---|---|
The endomorphin-1/2 and dynorphin-B peptides display biased agonism at the mu opioid receptor.
Opioid agonist activation at the mu opioid receptor (MOR) can lead to a wide variety of physiological responses. Many opioid agonists share the ability to selectively and preferentially activate specific signaling pathways, a term called biased agonism. Biased opioid ligands can theoretically induce specific physiological responses and might enable the generation of drugs with improved side effect profiles.. Dynorphins, enkephalins, and endomorphins are endogenous opioid agonist peptides that may possess distinct bias profiles; biased agonism of endogenous peptides could explain the selective roles of these ligands in vivo. Our purpose in the present study was to investigate biased signaling and potential underlying molecular mechanisms of bias using. We found that endomorphin-1/2 preferentially activated cAMP signaling, while dynorphin-B preferentially activated. We found that endomorphin-1/2 and dynorphin-B displayed contrasting bias profiles at the MOR, and ruled out potential AC6 and RGS4 mechanisms in this bias. This identified signaling bias could be involved in specifying endogenous peptide roles in vivo, where these peptides have low selectivity between opioid receptor family members. Topics: Adenylyl Cyclases; Animals; Cell Culture Techniques; Cell Line; CHO Cells; Cricetulus; Cyclic AMP; Dynorphins; Endorphins; Gene Knockdown Techniques; Guanosine 5'-O-(3-Thiotriphosphate); Humans; Oligopeptides; Receptors, Opioid, mu; RGS Proteins; Signal Transduction | 2020 |
Alanine scan of the opioid peptide dynorphin B amide.
To date structure-activity relationship (SAR) studies of the dynorphins (Dyn), endogenous peptides for kappa opioid receptors (KOR), have focused almost exclusively on Dyn A with minimal studies on Dyn B. While both Dyn A and Dyn B have identical N-terminal sequences, their C-terminal sequences differ, which could result in differences in pharmacological activity. We performed an alanine scan of the non-glycine residues up through residue 11 of Dyn B amide to explore the roles of these side chains in the activity of Dyn B. The analogs were synthesized by fluorenylmethyloxycarbonyl (Fmoc)-based solid phase peptide synthesis and evaluated for their opioid receptor affinities and opioid potency and efficacy at KOR. Similar to Dyn A the N-terminal Tyr Topics: Alanine; Amides; Amino Acid Sequence; Animals; CHO Cells; Cricetinae; Cricetulus; Dynorphins; Endorphins; Mutagenesis; Opioid Peptides; Protein Binding; Receptors, Opioid, kappa; Solid-Phase Synthesis Techniques; Structure-Activity Relationship | 2017 |
Region-specific bioconversion of dynorphin neuropeptide detected by in situ histochemistry and MALDI imaging mass spectrometry.
Brain region-specific expression of proteolytic enzymes can control the biological activity of endogenous neuropeptides and has recently been targeted for the development of novel drugs, for neuropathic pain, cancer, and Parkinson's disease. Rapid and sensitive analytical methods to profile modulators of enzymatic activity are important for finding effective inhibitors with high therapeutic value. Combination of in situ enzyme histochemistry with MALDI imaging mass spectrometry allowed developing a highly sensitive method for analysis of brain-area specific neuropeptide conversion of synthetic and endogenous neuropeptides, and for selection of peptidase inhibitors that differentially target conversion enzymes at specific anatomical sites. Conversion and degradation products of Dynorphin B as model neuropeptide and effects of peptidase inhibitors applied to native brain tissue sections were analyzed at different brain locations. Synthetic dynorphin B (2pmol) was found to be converted to the N-terminal fragments on brain sections whereas fewer C-terminal fragments were detected. N-ethylmaleimide (NEM), a non-selective inhibitor of cysteine peptidases, almost completely blocked the conversion of dynorphin B to dynorphin B(1-6; Leu-Enk-Arg), (1-9), (2-13), and (7-13). Proteinase inhibitor cocktail, and also incubation with acetic acid displayed similar results. Bioconversion of synthetic dynorphin B was region-specific producing dynorphin B(1-7) in the cortex and dynorphin B (2-13) in the striatum. Enzyme inhibitors showed region- and enzyme-specific inhibition of dynorphin bioconversion. Both phosphoramidon (inhibitor of the known dynorphin converting enzyme neprilysin) and opiorphin (inhibitor of neprilysin and aminopeptidase N) blocked cortical bioconversion to dynorphin B(1-7), wheras only opiorphin blocked striatal bioconversion to dynorphin B(2-13). This method may impact the development of novel therapies with aim to strengthen the effects of endogenous neuropeptides under pathological conditions such as chronic pain. Combining histochemistry and MALDI imaging MS is a powerful and sensitive tool for the study of inhibition of enzyme activity directly in native tissue sections. Topics: Animals; Brain; Cysteine Endopeptidases; Dynorphins; Endorphins; Glycopeptides; Humans; Neuropeptides; Oligopeptides; Parkinson Disease; Protease Inhibitors; Rats; Salivary Proteins and Peptides; Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization | 2017 |
Single housing during early adolescence causes time-, area- and peptide-specific alterations in endogenous opioids of rat brain.
A number of experimental procedures require single housing to assess individual behaviour and physiological responses to pharmacological treatments. The endogenous opioids are closely linked to social interaction, especially early in life, and disturbance in the social environment may affect opioid peptides and thereby confound experimental outcome. The aim of the present study was to examine time-dependent effects of single housing on opioid peptides in rats.. Early adolescent Sprague Dawley rats (post-natal day 22) were subjected to either prolonged (7 days) or short (30 min) single housing. Several brain regions were dissected and immunoreactive levels of Met-enkephalin-Arg(6) Phe(7) (MEAP), dynorphin B and nociception/orphanin FQ, as well as serum corticosterone were measured using RIA.. Prolonged single housing reduced immunoreactive MEAP in hypothalamus, cortical regions, amygdala, substantia nigra and periaqueductal grey. Short single housing resulted in an acute stress response as indicated by high levels of corticosterone, accompanied by elevated immunoreactive nociceptin/orphanin FQ in medial prefrontal cortex, nucleus accumbens and amygdala. Neither short nor prolonged single housing affected dynorphin B.. Disruption in social environmental conditions of rats, through single housing during early adolescence, resulted in time-, area- and peptide-specific alterations in endogenous opioids in the brain. These results provide further evidence for an association between early life social environment and opioids. Furthermore, the results have implications for experimental design; in any pharmacological study involving opioid peptides, it is important to distinguish between effects induced by housing and treatment.. This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2. Topics: Aging; Animals; Brain; Dynorphins; Endorphins; Enkephalin, Methionine; Housing, Animal; Male; Nociceptin; Opioid Peptides; Rats, Sprague-Dawley; Social Environment | 2015 |
Activation of DREAM (downstream regulatory element antagonistic modulator), a calcium-binding protein, reduces L-DOPA-induced dyskinesias in mice.
Previous studies have implicated the cyclic adenosine monophosphate/protein kinase A pathway as well as FosB and dynorphin-B expression mediated by dopamine D1 receptor stimulation in the development of 3,4-dihydroxyphenyl-L-alanine (L-DOPA)-induced dyskinesia. The magnitude of these molecular changes correlates with the intensity of dyskinesias. The calcium-binding protein downstream regulatory element antagonistic modulator (DREAM) binds to regulatory element sites called DRE in the DNA and represses transcription of target genes such as c-fos, fos-related antigen-2 (fra-2), and prodynorphin. This repression is released by calcium and protein kinase A activation. Dominant-active DREAM transgenic mice (daDREAM) and DREAM knockout mice (DREAM(-/-)) were used to define the involvement of DREAM in dyskinesias.. Dyskinesias were evaluated twice a week in mice with 6-hydroxydopamine lesions during long-term L-DOPA (25 mg/kg) treatment. The impact of DREAM on L-DOPA efficacy was evaluated using the rotarod and the cylinder test after the establishment of dyskinesia and the molecular changes by immunohistochemistry and Western blot.. In daDREAM mice, L-DOPA-induced dyskinesia was decreased throughout the entire treatment. In correlation with these behavioral results, daDREAM mice showed a decrease in FosB, phosphoacetylated histone H3, dynorphin-B, and phosphorylated glutamate receptor subunit, type 1 expression. Conversely, genetic inactivation of DREAM potentiated the intensity of dyskinesia, and DREAM(-/-) mice exhibited an increase in expression of molecular markers associated with dyskinesias. The DREAM modifications did not affect the kinetic profile or antiparkinsonian efficacy of L-DOPA therapy.. The protein DREAM decreases development of L-DOPA-induced dyskinesia in mice and reduces L-DOPA-induced expression of FosB, phosphoacetylated histone H3, and dynorphin-B in the striatum. These data suggest that therapeutic approaches that activate DREAM may be useful to alleviate L-DOPA-induced dyskinesia without interfering with the therapeutic motor effects of L-DOPA. Topics: Acetylation; Animals; Antiparkinson Agents; Blotting, Western; Corpus Striatum; Dynorphins; Dyskinesia, Drug-Induced; Endorphins; Histones; Immunohistochemistry; Kv Channel-Interacting Proteins; Levodopa; Mice, Knockout; Motor Activity; Oxidopamine; Parkinsonian Disorders; Phosphorylation; Proto-Oncogene Proteins c-fos; Receptors, AMPA; Repressor Proteins; Rotarod Performance Test | 2015 |
Membrane interactions in small fast-tumbling bicelles as studied by 31P NMR.
Small fast-tumbling bicelles are ideal for studies of membrane interactions at molecular level; they allow analysis of lipid properties using solution-state NMR. In the present study we used 31P NMR relaxation to obtain detailed information on lipid head-group dynamics. We explored the effect of two topologically different membrane-interacting peptides on bicelles containing either dimyristoylphosphocholine (DMPC), or a mixture of DMPC and dimyristoylphosphoglycerol (DMPG), and dihexanoylphosphocholine (DHPC). KALP21 is a model transmembrane peptide, designed to span a DMPC bilayer and dynorphin B is a membrane surface active neuropeptide. KALP21 causes significant increase in bicelle size, as evidenced by both dynamic light scattering and 31P T2 relaxation measurements. The effect of dynorphin B on bicelle size is more modest, although significant effects on T2 relaxation are observed at higher temperatures. A comparison of 31P T1 values for the lipids with and without the peptides showed that dynorphin B has a greater effect on lipid head-group dynamics than KALP21, especially at elevated temperatures. From the field-dependence of T1 relaxation data, a correlation time describing the overall lipid motion was derived. Results indicate that the positively charged dynorphin B decreases the mobility of the lipid molecules--in particular for the negatively charged DMPG--while KALP21 has a more modest influence. Our results demonstrate that while a transmembrane peptide has severe effects on overall bilayer properties, the surface bound peptide has a more dramatic effect in reducing lipid head-group mobility. These observations may be of general importance for understanding peptide-membrane interactions. Topics: Amino Acid Sequence; Anisotropy; Dimyristoylphosphatidylcholine; Dynorphins; Endorphins; Kinetics; Lasers; Light; Lipid Bilayers; Magnetic Resonance Spectroscopy; Membrane Lipids; Membrane Proteins; Molecular Sequence Data; Motion; Peptides; Phosphatidylglycerols; Phospholipid Ethers; Phosphorus Isotopes; Protein Binding; Scattering, Radiation | 2015 |
Plasma membrane poration by opioid neuropeptides: a possible mechanism of pathological signal transduction.
Neuropeptides induce signal transduction across the plasma membrane by acting through cell-surface receptors. The dynorphins, endogenous ligands for opioid receptors, are an exception; they also produce non-receptor-mediated effects causing pain and neurodegeneration. To understand non-receptor mechanism(s), we examined interactions of dynorphins with plasma membrane. Using fluorescence correlation spectroscopy and patch-clamp electrophysiology, we demonstrate that dynorphins accumulate in the membrane and induce a continuum of transient increases in ionic conductance. This phenomenon is consistent with stochastic formation of giant (~2.7 nm estimated diameter) unstructured non-ion-selective membrane pores. The potency of dynorphins to porate the plasma membrane correlates with their pathogenic effects in cellular and animal models. Membrane poration by dynorphins may represent a mechanism of pathological signal transduction. Persistent neuronal excitation by this mechanism may lead to profound neuropathological alterations, including neurodegeneration and cell death. Topics: Analgesics, Opioid; Animals; Cell Membrane; Dynorphins; Endorphins; Enkephalins; Humans; Ligands; Microscopy, Confocal; Neuropeptides; Opioid Peptides; PC12 Cells; Protein Precursors; Rats; Signal Transduction | 2015 |
The opioid peptide dynorphin A induces leukocyte responses via integrin Mac-1 (αMβ2, CD11b/CD18).
Opioid peptides, including dynorphin A, besides their analgesic action in the nervous system, exert a broad spectrum of effects on cells of the immune system, including leukocyte migration, degranulation and cytokine production. The mechanisms whereby opioid peptides induce leukocyte responses are poorly understood. The integrin Mac-1 (αMβ2, CD11b/CD18) is a multiligand receptor which mediates numerous reactions of neutrophils and monocyte/macrophages during the immune-inflammatory response. Our recent elucidation of the ligand recognition specificity of Mac-1 suggested that dynorphin A and dynorphin B contain Mac-1 recognition motifs and can potentially interact with this receptor.. In this study, we have synthesized the peptide library spanning the sequence of dynorphin AB, containing dynorphin A and B, and showed that the peptides bound recombinant αMI-domain, the ligand binding region of Mac-1. In addition, immobilized dynorphins A and B supported adhesion of the Mac-1-expressing cells. In binding to dynorphins A and B, Mac-1 cooperated with cell surface proteoglycans since both anti-Mac-1 function-blocking reagents and heparin were required to block adhesion. Further focusing on dynorphin A, we showed that its interaction with the αMI-domain was activation independent as both the α7 helix-truncated (active conformation) and helix-extended (nonactive conformation) αMI-domains efficiently bound dynorphin A. Dynorphin A induced a potent migratory response of Mac-1-expressing, but not Mac-1-deficient leukocytes, and enhanced Mac-1-mediated phagocytosis of latex beads by murine IC-21 macrophages.. Together, the results identify dynorphins A and B as novel ligands for Mac-1 and suggest a role for the Dynorphin A-Mac-1 interactions in the induction of nonopiod receptor-dependent effects in leukocytes. Topics: Amino Acid Sequence; Cell Adhesion; Chemotaxis; Dynorphins; Endorphins; Fibrinogen; HEK293 Cells; Humans; Leukocytes; Ligands; Macrophage-1 Antigen; Microspheres; Molecular Sequence Data; Peptide Library; Phagocytosis; Protein Binding; Protein Structure, Tertiary; Solubility; U937 Cells | 2015 |
A ghrelin receptor (GHS-R1A) antagonist attenuates the rewarding properties of morphine and increases opioid peptide levels in reward areas in mice.
Gut-brain hormones such as ghrelin have recently been suggested to have a role in reward regulation. Ghrelin was traditionally known to regulate food intake and body weight homoeostasis. In addition, recent work has pin-pointed that this peptide has a novel role in drug-induced reward, including morphine-induced increase in the extracellular levels of accumbal dopamine in rats. Herein the effect of the ghrelin receptor (GHS-R1A) antagonist, JMV2959, on morphine-induced activation of the mesolimbic dopamine system was investigated in mice. In addition, the effects of JMV2959 administration on opioid peptide levels in reward related areas were investigated. In the present series of experiment we showed that peripheral JMV2959 administration, at a dose with no effect per se, attenuates the ability of morphine to cause locomotor stimulation, increase the extracellular levels of accumbal dopamine and to condition a place preference in mice. JMV2959 administration significantly increased tissue levels of Met-enkephalin-Arg(6)Phe(7) in the ventral tegmental area, dynorphin B in hippocampus and Leu-enkephalin-Arg(6) in striatum. We therefore hypothesise that JMV2959 prevents morphine-induced reward via stimulation of delta receptor active peptides in striatum and ventral tegmental areas. In addition, hippocampal peptides that activate kappa receptor may be involved in JMV2959׳s ability to regulate memory formation of reward. Given that development of drug addiction depends, at least in part, of the effects of addictive drugs on the mesolimbic dopamine system the present data suggest that GHS-R1A antagonists deserve to be elucidated as novel treatment strategies of opioid addiction. Topics: Analgesics, Opioid; Animals; Brain; Conditioning, Operant; Dopamine; Dynorphins; Endorphins; Extracellular Fluid; Glycine; Male; Mice; Microdialysis; Morphine; Motor Activity; Opioid Peptides; Receptors, Ghrelin; Reward; Triazoles | 2015 |
Development and characterization of functionalized niosomes for brain targeting of dynorphin-B.
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 |
Effects of rearing conditions on behaviour and endogenous opioids in rats with alcohol access during adolescence.
Causal links between early-life stress, genes and later psychiatric diagnoses are not possible to fully address in human studies. Animal models therefore provide an important complement in which conditions can be well controlled and are here used to study and distinguish effects of early-life stress and alcohol exposure. The objective of this study was to investigate the impact of rearing conditions on behaviour in young rats and if these changes could be followed over time and to examine interaction effects between early-life environment and adolescent alcohol drinking on behaviour and immunoreactive levels of the opioid peptides dynorphin B, met-enkephalin-Arg(6)Phe(7) and beta-endorphin. We employed a rodent model, maternal separation, to study the impact of rearing conditions on behaviour, voluntary alcohol consumption and alcohol-induced effects. The consequences of short, 15 min (MS 15), and long, 360 min (MS 360), maternal separation in combination with adolescent voluntary alcohol consumption on behaviour and peptides were examined. A difference in the development of risk taking behaviour was found between the MS15 and MS360 while the development of general activity was found to differ between intake groups. Beta-endorphin levels in the pituitary and the periaqueductal gray area was found to be higher in the MS15 than the MS360. Adolescent drinking resulted in higher dynorphin B levels in the hippocampus and higher met-enkephalin-Arg(6)Phe(7) levels in the amygdala. Amygdala and hippocampus are involved in addiction processes and changes in these brain areas after adolescent alcohol drinking may have consequences for cognitive function and drug consumption behaviour in adulthood. The study shows that individual behavioural profiling over time in combination with neurobiological investigations provides means for studies of causality between early-life stress, behaviour and vulnerability to psychiatric disorders. Topics: Age Factors; Alcohol Drinking; Amygdala; Animals; Animals, Newborn; Behavior, Animal; beta-Endorphin; Dynorphins; Endorphins; Enkephalin, Methionine; Female; Hippocampus; Male; Maternal Deprivation; Periaqueductal Gray; Pituitary Gland; Rats; Rats, Wistar; Risk-Taking; Stress, Physiological | 2013 |
Spinal astrocytes produce and secrete dynorphin neuropeptides.
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 |
Differences in basal and ethanol-induced levels of opioid peptides in Wistar rats from five different suppliers.
One major cause for discrepancies in results from animal experimental studies is the use of different animal strains and suppliers. We have previously reported that Wistar rats from five different suppliers display profound differences in ethanol intake and behavior. One of the neurobiological processes that could be underlying these differences is the endogenous opioid system, which has been implicated in the rewarding and reinforcing effects of alcohol. We therefore hypothesized that the differences between the supplier groups would also be evident in the endogenous opioid system. Radioimmunoassay was used to determine the levels of the opioid peptides Met-enkephalin-Arg(6)Phe(7) and dynorphin B in several brain areas of ethanol-drinking and ethanol naïve Wistar rats from five different suppliers. In the ethanol naïve animals, differences between the supplier groups were found in the pituitary gland, hypothalamus, frontal cortex, dorsal striatum and hippocampus. In the ethanol-drinking rats, differences were found in the same structures, with the addition of medial prefrontal cortex and substantia nigra. Correlations between ethanol intake and peptide levels were also found in several of the areas examined. The structures in which differences were found have all been implicated in the transition from drug use to addiction and these differences may lead to different propensities and vulnerability to this transition. Because the endogenous opioids have been suggested to be involved in a number of neurobiological disorders the results do not only have implications for research on alcohol or drug addiction, but many other fields as well. Topics: Alcohol Drinking; Animals; Animals, Outbred Strains; Brain; Choice Behavior; Dynorphins; Endorphins; Enkephalin, Methionine; Ethanol; Male; Opioid Peptides; Organ Specificity; Pituitary Gland; Rats; Rats, Wistar; Species Specificity; Statistics, Nonparametric | 2012 |
Mixed dimers of insulin-degrading enzyme reveal a cis activation mechanism.
Insulin-degrading enzyme (IDE) exists primarily as a dimer being unique among the zinc metalloproteases in that it exhibits allosteric kinetics with small synthetic peptide substrates. In addition the IDE reaction rate is increased by small peptides that bind to a distal site within the substrate binding site. We have generated mixed dimers of IDE in which one or both subunits contain mutations that affect activity. The mutation Y609F in the distal part of the substrate binding site of the active subunit blocks allosteric activation regardless of the activity of the other subunit. This effect shows that substrate or small peptide activation occurs through a cis effect. A mixed dimer composed of one wild-type subunit and the other subunit containing a mutation that neither permits substrate binding nor catalysis (H112Q) exhibits the same turnover number per active subunit as wild-type IDE. In contrast, a mixed dimer in which one subunit contains the wild-type sequence and the other contains a mutation that permits substrate binding, but not catalysis (E111F), exhibits a decrease in turnover number. This indicates a negative trans effect of substrate binding at the active site. On the other hand, activation in trans is observed with extended substrates that occupy both the active and distal sites. Comparison of the binding of an amyloid β peptide analog to wild-type IDE and to the Y609F mutant showed no difference in affinity, indicating that Y609 does not play a significant role in substrate binding at the distal site. Topics: Amyloid beta-Peptides; Animals; Bradykinin; Catalysis; Catalytic Domain; Circular Dichroism; Dimerization; Dynorphins; Endorphins; Humans; Insulin; Kinetics; Molecular Weight; Mutation; Peptides; Substrate Specificity | 2011 |
Substance P N-terminal fragment SP(1-7) attenuates chronic morphine tolerance and affects dynorphin B and nociceptin in rats.
The N-terminal substance P fragment SP(1-7) is known to modulate hyperalgesia and opioid withdrawal in animal models. This study examined the effects of intraperitoneal (i.p.) injections of SP(1-7) on chronic morphine tolerance and on the levels of dynorphin B (DYN B) and nociceptin/orphanin FQ (N/OFQ) in various brain areas of male Sprague-Dawley rats. Morphine tolerance was induced by subcutaneous injections of the opioid (10mg/kg) twice daily for 7 days. SP(1-7) injected i.p. (185 nmol/kg) 30 min prior to morphine reduced the development of morphine tolerance. Immunoreactive (ir) DYN B and N/OFQ peptide levels were measured in several areas of the central nervous system. Levels of ir DYN B in rats treated with SP(1-7) and morphine were decreased in the nucleus accumbens, substantia nigra and ventral tegmental area and increased in the frontal cortex. The ir N/OFQ levels were increased in the periaqueductal gray and decreased in the nucleus accumbens. Since the concentration profiles of the two peptides were altered by SP(1-7) in the areas that are implicated in the modulation of opioid tolerance and analgesia, it is suggested that DYN B and N/OFQ systems may be involved in the effects of SP(1-7) on opioid tolerance. Topics: Analgesics, Opioid; Animals; Drug Tolerance; Dynorphins; Endorphins; Male; Morphine; Nociceptin; Opioid Peptides; Pain Measurement; Peptide Fragments; Rats; Rats, Sprague-Dawley; Substance P | 2011 |
A morphologically distinct granule cell type in the dentate gyrus of the red fox correlates with adult hippocampal neurogenesis.
Wild red foxes, proverbially cunning carnivores, are investigated for adult hippocampal neurogenesis and morphological characteristics of the dentate gyrus. Adult red foxes harbor almost 15-times more young, doublecortin-positive neurons in their dentate gyrus than domesticated dogs. The number of doublecortin-positive cells corresponds to 4.4% of the total granule cell number, whereas dividing cells amount to only 0.06%. Compared to laboratory mice, proliferating (Ki67-positive) and dying cells are rare, but the percentage of new neurons is quite similar. The numbers of proliferating cells, young cells of neuronal lineage and dying cells correlate. Resident granule cells can be divided into two types with strikingly different morphologies, staining patterns and distinct septotemporal distributions. Small sized granule cells with a nuclear diameter of 7.3 microm account for approximately 83% of all granule cells. The remaining granule cells are significantly larger with a nuclear diameter of 9.4 microm diameter and stain heavily for NeuN. Septally and mid-septotemporally, densely packed small cells dominate. Here, only few large granule cells are scattered throughout the layer. Temporally, granule cells become more loosely packed and most of the cells are of the large type. High rates of neurogenesis are observed in foxes with high numbers of large granule cells, whereas the number of small granule cells does not correlate with any of the neurogenesis-related cell counts. Staining for parvalbumin, glutamate receptor 2/3, GAP-43 and dynorphin shows an anatomical context that is a composite of features common also to other mammalian species. In summary, we report a morphologically distinct granule cell type which correlates with adult hippocampal neurogenesis in the fox. Furthermore, the maturation phase of the young neurons may be prolonged as in other long living species such as primates. Topics: Adaptation, Physiological; Aging; Animals; Basic Helix-Loop-Helix Transcription Factors; Biomarkers; Cell Count; Cell Differentiation; Cell Proliferation; Cell Shape; Dentate Gyrus; Doublecortin Domain Proteins; Dynorphins; Endorphins; Female; Foxes; GAP-43 Protein; Immunohistochemistry; Intelligence; Ki-67 Antigen; Learning; Male; Microtubule-Associated Proteins; Nerve Tissue Proteins; Neural Cell Adhesion Molecule L1; Neurogenesis; Neuronal Plasticity; Neurons; Neuropeptides; Parvalbumins; Proto-Oncogene Proteins c-fos; Receptors, Glutamate; Sex Characteristics; Sialic Acids; Species Specificity | 2010 |
The contribution of serotonin 1A receptors to kappa opioid immunosuppression.
Activation of kappa opioid receptors (kappa-OR) with the selective agonist rimorphin (0.1 mg/kg) produced marked suppression of the immune response in CBA mice. This effect was not seen on administration of rimorphin on the background of a reduction in the activity of the serotoninergic (5-HTergic) system resulting from stimulation of presynaptic (8-OH-DPAT, 0.1 mg/kg) or blockade of postsynaptic (WAY-100635, 1.0 mg/kg) 5-HT1A receptors. These data led to the conclusion that 5-HTergic mechanisms involving preand postsynaptic 5-HT1A receptors have a role in kappa-opioid-mediated immunosuppression. Topics: 8-Hydroxy-2-(di-n-propylamino)tetralin; Animals; Dynorphins; Endorphins; Immunosuppression Therapy; Male; Mice; Mice, Inbred CBA; Neuroimmunomodulation; Piperazines; Pyridines; Receptor, Serotonin, 5-HT1A; Receptors, Opioid, kappa; Receptors, Presynaptic; Serotonin 5-HT1 Receptor Agonists; Serotonin 5-HT1 Receptor Antagonists | 2009 |
Immune response of submissive and aggressive mice under conditions of opioid receptor activation.
Experiments on the model of paired sensory contact demonstrated that stimulation of immune response in aggressive CBA and C57Bl/6J mice with 10- and 20-day experience of victories, respectively, was prevented by selective δ(2)- and -opioid receptor agonists DSLET and κ-opioid receptor agonists rimorphin in a dose of 100 microg/kg. In C57Bl/6J mice with depression-like behavior, normalization (but not suppression) of the immune response under conditions of μ-opioid receptor stimulation (100 microg/kg) was observed. Selective modulation of activity of a certain type of opioid receptors can normalize the immune function modified during the formation of a certain behavioral type. Topics: Aggression; Animals; Dynorphins; Endorphins; Enkephalin, Leucine; Immunomodulation; Male; Mice; Mice, Inbred C57BL; Mice, Inbred CBA; Receptors, Opioid, delta; Receptors, Opioid, kappa; Receptors, Opioid, mu | 2009 |
Possible involvement of dynorphin A release via mu1-opioid receptor on supraspinal antinociception of endomorphin-2.
It has been demonstrated that the antinociception induced by i.t. or i.c.v. administration of endomorphins is mediated through mu-opioid receptors. Moreover, though endomorphins do not have appreciable affinity for kappa-opioid receptors, pretreatment with the kappa-opioid receptor antagonist nor-binaltorphimine markedly blocks the antinociception induced by i.c.v.- or i.t.-injected endomorphin-2, but not endomorphin-1. These evidences propose the hypothesis that endomorphin-2 may initially stimulate the mu-opioid receptors, which subsequently induces the release of dynorphins acting on kappa-opioid receptors to produce antinociception. The present study was performed to determine whether the release of dynorphins by i.c.v.-administered endomorphin-2 is mediated through mu-opioid receptors for producing antinociception. Intracerebroventricular pretreatment with an antiserum against dynorphin A, but not dynorphin B or alpha-neo-endorphin, and s.c. pretreatment with kappa-opioid receptor antagonist nor-binaltorphimine dose-dependently attenuated the antinociception induced by i.c.v.-administered endomorphin-2, but not endomorphin-1 and DAMGO. The attenuation of endomorphin-2-induced antinociception by pretreatment with antiserum against dynorphin A or nor-binaltorphimine was dose-dependently eliminated by additional s.c. pretreatment with a selective mu-opioid receptor antagonist beta-funaltrexamine or a selective mu1-opioid receptor antagonist naloxonazine at ultra low doses, which are inactive against micro-opioid receptor agonists in antinociception, suggesting that endomorphin-2 stimulates distinct subclass of micro1-opioid receptor that induces the release of dynorphin A acting on kappa-opioid receptors in the brain. It concludes that the antinociception induced by supraspinally administered endomorphin-2 is in part mediated through the release of endogenous kappa-opioid peptide dynorphin A, which is caused by the stimulation of distinct subclass of micro1-opioid receptor. Topics: Analgesics; Animals; Dynorphins; Endorphins; Enkephalin, Ala(2)-MePhe(4)-Gly(5)-; Immune Sera; Injections, Intraventricular; Male; Mice; Naloxone; Naltrexone; Oligopeptides; Protein Precursors; Receptors, Opioid, kappa; Receptors, Opioid, mu | 2008 |
[Impact of serotonin 1A receptors in the kappa-opioid immunosuppression].
The data obtained indicate that rimorphin (0.1 mg/kg), a specific kappa-agonist, evoked a significant inhibition of the immune response in CBA mice. Pretreatment of the animals with 8-OH-DPAT (0.1 mg/kg), a selective serotonin (5-HT) agonist, activating presynaptic 5-HT(1A) receptors, or WAY-100635 (1.0 mg/kg), a selective 5-HT(1A) receptors blocker of postsynaptic 5-HTIA receptors, prevents kappa-opioid effect. The present data indicate that kappa-opioid-induced immunosuppression is due to the involvement of the 5-HT-ergic mechanisms that are modulated via pre- and postsynaptic 5-HT(1A) receptors. Topics: 8-Hydroxy-2-(di-n-propylamino)tetralin; Animals; Dynorphins; Endorphins; Immunosuppression Therapy; Male; Mice; Mice, Inbred CBA; Neuroimmunomodulation; Piperazines; Pyridines; Receptor, Serotonin, 5-HT1A; Receptors, Opioid, kappa; Receptors, Presynaptic; Serotonin 5-HT1 Receptor Agonists; Serotonin 5-HT1 Receptor Antagonists | 2008 |
The impact of postnatal environment on opioid peptides in young and adult male Wistar rats.
Early environmental influences can change the neuronal development and thereby affect behavior in adult life. The aim in the present study was to thoroughly examine the impact of early environmental factors on endogenous opioids by using a rodent maternal separation (MS) model. The endogenous opioid peptide system is not fully developed at birth, and short- and/or long-term alterations may occur in these neural networks in animals exposed to manipulation of the postnatal environment. Rat pups were subjected to one of five rearing conditions; 15 min (MS15) litter (l) or individual (i), 360 min (MS360) l or i daily MS, or housed under normal animal facility rearing (AFR) conditions during postnatal days 1-21. Measurements of immunoreactive (ir) Met-enkephalin-Arg6Phe7 (MEAP) and dynorphin B (DYNB) peptide levels in the pituitary gland and in a number of brain areas, were performed at three and 10 weeks of age, respectively. MS-induced changes were more pronounced in ir MEAP levels, especially in individually separated rats at three weeks of age and in litter-separated rats at 10 weeks of age. The enkephalin and dynorphin systems have different developmental patterns, dynorphin appearing earlier, which may point at a more sensitive enkephalin system during the early postnatal weeks. The results provide evidence that opioid peptides are sensitive for early environmental factors and show that the separation conditions are critical and also result in changes manifesting at different time points. MS-induced effects were observed in areas related to stress, drug reward and dependence mechanisms. By describing effects on opioid peptides, the study addresses the possible role of a deranged endogenous opioid system in the previously described behavioral consequences of MS. Topics: Age Factors; Animals; Animals, Newborn; Body Weight; Dynorphins; Endorphins; Enkephalin, Methionine; Environment; Female; Male; Maternal Behavior; Maternal Deprivation; Opioid Peptides; Pituitary Gland; Pregnancy; Rats; Rats, Wistar; Social Isolation; Stress, Psychological | 2008 |
Dysregulation of dynorphins in Alzheimer disease.
The opioid peptides dynorphins may be involved in pathogenesis of Alzheimer disease (AD) by inducing neurodegeneration or cognitive impairment. To test this hypothesis, the dynorphin system was analyzed in postmortem samples from AD and control subjects, and subjects with Parkinson or cerebro-vascular diseases for comparison. Dynorphin A, dynorphin B and related neuropeptide nociceptin were determined in the Brodmann area 7 by radioimmunoassay. The precursor protein prodynorphin, processing convertase PC2 and the neuroendocrine pro7B2 and 7B2 proteins required for PC2 maturation were analyzed by Western blot. AD subjects displayed robustly elevated levels of dynorphin A and no differences in dynorphin B and nociceptin compared to controls. Subjects with Parkinson or cerebro-vascular diseases did not differ from controls with respect to any of the three peptides. PC2 levels were also increased, whereas, those of prodynorphin and pro7B2/7B2 were not changed in AD. Dynorphin A levels correlated with the neuritic plaque density. These results along with the known non-opioid ability of dynorphin A to induce neurodegeneration suggest a role for this neuropeptide in AD neuropathology. Topics: Aged; Aged, 80 and over; Alzheimer Disease; Brain; Dynorphins; Endorphins; Female; Humans; Male; Nerve Degeneration; Nociceptin; Opioid Peptides; Up-Regulation | 2007 |
Dynorphin peptides differentially regulate the human kappa opioid receptor.
Dynorphins, endogenous peptides for the kappa opioid receptor, play important roles in many physiological and pathological functions. Here, we examined how prolonged treatment with three major prodynorphin peptides, dynorphin A (1-17) (Dyn A), dynorphin B (1-13) (Dyn B) and alpha-neoendorphin (alpha-Neo), regulated the human kappa opioid receptor (hKOR) stably expressed in Chinese hamster ovary (CHO) cells. Results from receptor binding and [(35)S]GTPgammaS binding assays showed that these peptides were potent full agonists of the hKOR with comparable receptor reserve and intrinsic efficacy to stimulate G proteins. A 4-h incubation with alpha-Neo at a concentration of approximately 600xEC(50) value (from [(35)S]GTPgammaS binding) resulted in receptor down-regulation to a much lower extent than the incubation with Dyn A and Dyn B at comparable concentrations ( approximately 10% vs. approximately 65%). Extending incubation period and increasing concentrations did not significantly affect the difference. The plateau level of alpha-Neo-mediated receptor internalization (30 min) was significantly less than those of Dyn A and Dyn B. Omission of the serum from the incubation medium or addition of peptidase inhibitors into the serum-containing medium enhanced alpha-Neo-, but not Dyn A- or Dyn B-, mediated receptor down-regulation and internalization; however, the degrees of alpha-Neo-induced adaptations were still significantly less than those of Dyn A and Dyn B. Thus, these endogenous peptides differentially regulate KOR after activating the receptor with similar receptor occupancy and intrinsic efficacy. Both stability in the presence of serum and intrinsic capacity to promote receptor adaptation play roles in the observed discrepancy among the dynorphin peptides. Topics: Animals; Binding, Competitive; Blotting, Western; CHO Cells; Cricetinae; Cricetulus; Down-Regulation; Dynorphins; Electrophoresis, Polyacrylamide Gel; Endorphins; Enkephalins; Flow Cytometry; Guanosine 5'-O-(3-Thiotriphosphate); Humans; Ligands; Peptides; Protease Inhibitors; Protein Precursors; Receptors, Opioid, kappa | 2007 |
Ethanol-induced effects on opioid peptides in adult male Wistar rats are dependent on early environmental factors.
The vulnerability to develop alcoholism is dependent on both genetic and environmental factors. The neurobiological mechanisms underlying these factors are not fully understood but individual divergence in the endogenous opioid peptide system may contribute. We have previously reported that early-life experiences can affect endogenous opioids and also adult voluntary ethanol intake. In the present study, this line of research was continued and the effects of long-term voluntary ethanol drinking on the opioid system are described in animals reared in different environmental settings. Rat pups were subjected to 15 min (MS15) or 360 min (MS360) of daily maternal separation during postnatal days 1-21. At 10 weeks of age, male rats were exposed to voluntary ethanol drinking in a four-bottle paradigm with 5%, 10% and 20% ethanol solution in addition to water for 2 months. Age-matched controls received water during the same period. Immunoreactive (ir) Met-enkephalin-Arg6Phe7 (MEAP) and dynorphin B (DYNB) peptide levels were thereafter measured in the pituitary gland and several brain areas. In water-drinking animals, lower ir MEAP levels were observed in the MS360 rats in the hypothalamus, medial prefrontal cortex, striatum and the periaqueductal gray, whereas no differences were seen in ir DYNB levels. Long-term ethanol drinking induced lower ir MEAP levels in MS15 rats in the medial prefrontal cortex and the periaqueductal gray, whereas higher levels were detected in MS360 rats in the hypothalamus, striatum and the substantia nigra. Chronic voluntary drinking affected ir DYNB levels in the pituitary gland, hypothalamus and the substantia nigra, with minor differences between MS15 and MS360. In conclusion, manipulation of the early environment caused changes in the opioid system and a subsequent altered response to ethanol. The altered sensitivity of the opioid peptides to ethanol may contribute to the previously reported differences in ethanol intake between MS15 and MS360 rats. Topics: Alcohol Drinking; Animals; Animals, Newborn; Body Weight; Central Nervous System Depressants; Dose-Response Relationship, Drug; Dynorphins; Endorphins; Enkephalin, Methionine; Environment; Ethanol; Female; Male; Maternal Deprivation; Opioid Peptides; Pregnancy; Radioimmunoassay; Rats; Rats, Wistar; Risk; Stress, Psychological | 2007 |
Big dynorphin as a putative endogenous ligand for the kappa-opioid receptor.
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 |
Membrane leakage induced by dynorphins.
Dynorphins, endogeneous opioid peptides, function as ligands to the opioid kappa receptors and induce non-opioid excitotoxic effects. Here we show that big dynorphin and dynorphin A, but not dynorphin B, cause leakage effects in large unilamellar phospholipid vesicles (LUVs). The effects parallel the previously studied potency of dynorphins to translocate through biological membranes. Calcein leakage caused by dynorphin A from LUVs with varying POPG/POPC molar ratios was promoted by higher phospholipid headgroup charges, suggesting that electrostatic interactions are important for the effects. A possibility that dynorphins generate non-opioid excitatory effects by inducing perturbations in the lipid bilayer of the plasma membrane is discussed. Topics: Amino Acid Sequence; Dynorphins; Endorphins; Fluoresceins; Liposomes; Membranes, Artificial; Molecular Sequence Data; Phosphatidylcholines; Phosphatidylglycerols; Phospholipids | 2006 |
Contribution of spinal mu(1)-opioid receptors and dynorphin B to the antinociception induced by Tyr-d-Arg-Phe-Sar.
The antinociceptive effect of Tyr-d-Arg-Phe-Sar (TAPS) at the spinal level was characterized with the mouse tail-flick test. Intrathecal (i.t.) administration of TAPS produced a dose-dependent antinociception. The antinociception induced by TAPS was completely blocked by i.t. pretreatment with the mu-opioid receptor antagonist beta-funaltrexamine, the mu(1)-opioid receptor antagonist naloxonazine or the kappa-opioid receptor antagonist nor-binaltorphimine, but not with the delta-opioid receptor antagonist naltrindole. Moreover, TAPS-induced antinociception was dose-dependently attenuated by i.t. pretreatment with an antiserum against dynorphin B, but not against dynorphin A, alpha-neo-endorphin, [Met(5)]enkephalin, or [Leu(5)]enkephalin. In mice lacking prodynorphin, TAPS-induced antinociception was significantly reduced compared to that in wild-type mice. These results suggest that TAPS mainly stimulates mu(1)-opioid receptors, which subsequently induce the release of dynorphin B, which then acts on kappa-opioid receptors to produce antinociception. Topics: Analgesics, Opioid; Animals; Dynorphins; Endorphins; Enkephalin, Ala(2)-MePhe(4)-Gly(5)-; Male; Mice; Mice, Inbred C57BL; Narcotic Antagonists; Nociceptors; Oligopeptides; Receptors, Opioid, mu; Spinal Cord | 2006 |
REM sleep changes in rats induced by siRNA-mediated orexin knockdown.
Short interfering RNAs (siRNA) targeting prepro-orexin mRNA were microinjected into the rat perifornical hypothalamus. Prepro-orexin siRNA-treated rats had a significant (59%) reduction in prepro-orexin mRNA compared to scrambled siRNA-treated rats 2 days postinjection, whereas prodynorphin mRNA was unaffected. The number of orexin-A-positive neurons on the siRNA-treated side decreased significantly (23%) as compared to the contralateral control (scrambled siRNA-treated) side. Neither the colocalized dynorphin nor the neighbouring melanin-concentrating hormone neurons were affected. The number of orexin-A-positive neurons on the siRNA-treated side did not differ from the number on the control side 4 or 6 days postinjection. Behaviourally, there was a persistent (approximately 60%) increase in the amount of time spent in rapid eye movement (REM) sleep during the dark (active) period for 4 nights postinjection, in rats treated with prepro-orexin siRNA bilaterally. This increase occurred mainly because of an increased number of REM episodes and decrease in REM-to-REM interval. Cataplexy-like episodes were also observed in some of these animals. Wakefulness and NREM sleep were unaffected. The siRNA-induced increase in REM sleep during the dark cycle reverted to control values on the 5th day postinjection. In contrast, the scrambled siRNA-treated animals only had a transient increase in REM sleep for the first postinjection night. Our results indicate that siRNA can be usefully employed in behavioural studies to complement other loss-of-function approaches. Moreover, these data suggest that the orexin system plays a role in the diurnal gating of REM sleep. Topics: Animals; Behavior, Animal; Brain; Cell Count; Dynorphins; Endorphins; Functional Laterality; Immunohistochemistry; Intracellular Signaling Peptides and Proteins; Male; Neuropeptides; Orexins; Rats; Rats, Sprague-Dawley; RNA, Small Interfering; Sleep, REM; Time Factors; Wakefulness | 2006 |
Membrane interactions of dynorphins.
The dynorphins are primarily endogenous ligands to the kappa-opioid receptor, but a variety of non-opioid effects have also been observed, including direct effects on membranes. The peptides are rich in Arg residues, a characteristic feature of the cell-penetrating peptides. In this investigation, we have examined the interaction of the two peptides dynorphin A and dynorphin B with model membranes. A variety of NMR methods, as well as CD and fluorescence spectroscopy, have been used to characterize the structure of the two peptides and, more importantly, the position of the peptides in phospholipid bicelles. Both peptides interact to a large extent with both zwitterionic and partly negatively charged bicelles but are only marginally structured in either solvent. Dynorphin A was found to insert its N-terminus into the bilayer of the bicelle, while dynorphin B was found to reside on the surface of the bilayer. Despite the high degree of similarity in the sequence of the two peptides, it has previously been observed that dynorphin A has membrane perturbing effects and causes leakage of calcein from large unilamellar phospholipid vesicles while dynorphin B has no such effects. Our results provide a possible explanation for the difference in membrane perturbation. Topics: Amino Acid Sequence; Circular Dichroism; Deuterium Exchange Measurement; Dynorphins; Endorphins; Lipid Bilayers; Magnetic Resonance Spectroscopy; Micelles; Molecular Sequence Data; Phospholipids; Protein Structure, Secondary; Spectrometry, Fluorescence | 2006 |
Progesterone-receptive beta-endorphin and dynorphin B neurons in the arcuate nucleus project to regions of high gonadotropin-releasing hormone neuron density in the ovine preoptic area.
Progesterone inhibits gonadotropin-releasing hormone (GnRH) secretion through interneuronal systems located in the mediobasal hypothalamus in ewes. Endogenous opioid peptides are implicated in this inhibition of GnRH secretion. The distributions of endogenous opioid peptides are known to overlap with progesterone receptors (PR) in the arcuate nucleus. We investigated whether PR is expressed by beta-endorphin and dynorphin B neurons in the arcuate nucleus and if a subset of double-labeled cells projects to the preoptic area where most GnRH neurons are detected. Injection of a retrograde tracer, Fluorogold, into the rostral preoptic area was performed in ovariectomized ewes pretreated with estrogen and progesterone. Brain sections were processed using double immunocytochemistry. Only brains of ewes with an injection site encompassing at least 80 GnRH neurons were processed for PR and then either beta-endorphin or dynorphin B immunocytochemistry. Antigen retrieval is essential for PR detection but causes Fluorogold to fade. Thus, quantitative analysis was performed on photographs taken before and after antigen retrieval. We found that 25-30% of PR-containing neurons, 20% of beta-endorphin cells and 22% of dynorphin B neurons in the arcuate nucleus project toward the preoptic area. From the PR/beta-endorphin double-labeled cells that represent 25 and 36% of PR and beta-endorphin cells, respectively, 35% were labeled with Fluorogold. From the PR/dynorphin B double-labeled cells that account for 39 and 62% of PR and dynorphin B neurons, respectively, 26% contained Fluorogold. These data strongly support the hypothesis that progesterone acts in the arcuate nucleus through beta-endorphin and dynorphin B neurons to affect preoptic area GnRH neurons. Topics: Animals; Arcuate Nucleus of Hypothalamus; beta-Endorphin; Dynorphins; Endorphins; Female; Gonadotropin-Releasing Hormone; Neural Pathways; Neurons; Preoptic Area; Progesterone; Receptors, Opioid; Receptors, Progesterone; Sheep; Staining and Labeling; Tissue Distribution | 2005 |
Differential effects of N-peptidyl-O-acyl hydroxylamines on dynorphin-induced antinociception in the mouse capsaicin test.
In the capsaicin test, intrathecal (i.t.) dynorphins are antinociceptive. Cysteine protease inhibitors such as p-hydroxymercuribenzoate (PHMB) given i.t. augment and prolong their activity. The effect of two novel cysteine protease inhibitors, N-peptidyl-O-acyl hydroxylamines, on the antinociception induced by i.t. administered dynorphin A or dynorphin B has been investigated. When administered i.t. 5 min before the injection of capsaicin (800 ng) into the plantar surface of the hindpaw, dynorphin A (62.5-1000 pmol) or dynorphin B (0.5-4 nmol) produced a dose-dependent and significant antinociceptive effect. The effect of dynorphin A (1 nmol) and dynorphin B (4 nmol) disappeared completely within 180 and 60 min, respectively. PHMB (2 nmol) and Boc-Tyr-Gly-NHO-Bz (BYG-Bz) (2 nmol) co-administered with dynorphin A or dynorphin B significantly prolonged antinociception induced by both. On the other hand, Z-Phe-Phe-NHO-Bz (ZFF-Bz) (1 and 2 nmol) only prolonged antinociception induced by dynorphin A. The results suggest that Z-Phe-Phe-NHO-Bz is an inhibitor of cysteine proteases preferring cleavage of dynorphin A, with less specificity towards dynorphin B in the mouse spinal cord. Topics: Analgesics; Animals; Capsaicin; Dynorphins; Endorphins; Humans; Hydroxylamines; Hydroxymercuribenzoates; Injections, Spinal; Male; Mice; Pain Measurement; Protease Inhibitors | 2005 |
Relative contribution of endogenous opioids to myocardial ischemic tolerance.
Opioid preconditioning by exogenous opioids experimentally protects the myocardium against ischemia/reflow injury. Additionally, endogenous opioid peptides released during ischemia also enhance ischemic tolerance. Promiscuous opioid receptor agonists conceal the differential contribution of the mu, delta, and kappa opioid subtypes. This study compared the impact of selective delta and kappa opioid receptor antagonists on postischemic functional and metabolic recovery. Also measured were changing levels of peptides dynorphin B and met-enkephalin during ischemia/reflow injury.. Using the rabbit Langendorff model, the functional recovery of control hearts (following 2 h of global ischemia) was compared to hearts pretreated with delta antagonist NTB (1 microM) or kappa antagonist, nor-BNI (1 microM). Measures included percentage of return of isovolumetric developed pressure (LVDP), myocardial oxygen consumption (MVO(2)) and coronary flow (CF). In additional studies, untreated hearts were harvested at baseline, following ischemia, or following 5 or 45 min of reflow. Tissue concentrations of met-enkephalin and dynorphin B were measured by RIA.. After 45 min of reflow, hearts pretreated with either NTB or nor-BNI showed impaired functional recovery by a decrease in LVDP (P < 0.05); however, MVO(2) or CF were unaffected. RIA data shows that baseline levels of both peptides are similar and increase significantly during ischemia, but reflow dynorphin levels drop far below baseline, while met-enkephalin returns to baseline.. Antagonism of both delta and kappa opioid receptor subtypes equally contributes to impaired left ventricular function, independent of altered perfusion or metabolic rate. Endogenous kappa-receptor agonists may contribute primarily during ischemia or early reflow, since low late reflow dynorphin content did not correlate with altered functional recovery. Topics: Animals; Dynorphins; Endorphins; Enkephalin, Methionine; In Vitro Techniques; Ischemic Preconditioning, Myocardial; Naltrexone; Narcotic Antagonists; Rabbits; Receptors, Opioid, delta; Receptors, Opioid, kappa; Recovery of Function; Ventricular Function, Left | 2004 |
The influence of opioid peptides on steroidogenesis in porcine granulosa cells.
The present studies were undertaken to examine the influence of mu (beta-endorphin, DAMGO, FK 33-824), delta (met-enkephalin, leu-enkephalin, DPLPE) and kappa opioid receptor agonists (dynorphin A, dynorphin B, U 50488) used at different doses (1-1000 nM) alone and in combination with LH (100 ng/ml) on steroidogenesis in porcine granulosa cells derived from large follicles. The effects of mu, delta and kappa receptor agonists on both basal and LH-induced progesterone (P4) secretion were negligible. Agonists of mu opioid receptors reduced basal androstenedione (A4), testosterone (T) and oestradiol (E2) release. Co-treatment with LH entirely abolished the inhibitory effect of these agonists on A4 and E2 secretion and resulted in an increase in T release. The addition of delta receptor agonists was followed by a decrease in basal A4, T and E2 secretion. The cells incubated in the presence of LH increased the androgen production and abrogated the inhibitory effect of delta agonists on E2 output. Basal A4, T and E2 release was also suppressed by kappa receptor agonists. The presence of LH in culture media extended the inhibitory effect of these opioids on E2 output and caused either abolition of the inhibitory influence of kappa agonists or even augmentation of both androgen release in response to the opioids. In conclusion, these data support the involvement of three major types of opioid receptors in the regulation of porcine granulosa cell steroidogenesis. Topics: 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer; Animals; beta-Endorphin; D-Ala(2),MePhe(4),Met(0)-ol-enkephalin; Dose-Response Relationship, Drug; Dynorphins; Endorphins; Enkephalin, Ala(2)-MePhe(4)-Gly(5)-; Enkephalin, D-Penicillamine (2,5)-; Enkephalins; Female; Granulosa Cells; Luteinizing Hormone; Opioid Peptides; Ovarian Follicle; Swine | 2004 |
Stress increases dynorphin immunoreactivity in limbic brain regions and dynorphin antagonism produces antidepressant-like effects.
Rats exposed to learned helplessness (LH), an animal model of depression, showed a recovery following an intracerebroventricular injection of nor-binaltorphimine dihydrochloride (norBNI; a kappa-opioid antagonist). To investigate the potential role of dynorphin A and dynorphin B, we examined the effects of different stress/depression models on dynorphin A and dynorphin B immunoreactivity in hippocampus and nucleus accumbens (NAc). Immobilization stress (3 h) caused an increase in levels of dynorphin A and dynorphin B immunoreactivity in the hippocampus and the NAc. Forced swim stress also temporally increased dynorphin A levels in the hippocampus. Furthermore, exposure to LH produced a similar increase in dynorphin A and dynorphin B in the hippocampus and NAc. Infusions of norBNI into the dentate gyrus or CA3 regions of hippocampus and into the shell or core regions of NAc produced antidepressant-like effects in the LH paradigm. The degrees of norBNI's effects were stronger in the CA3 region and NAc shell and less effective in the dentate gyrus of hippocampus and NAc core. These results indicate that both dynorphin A and dynorphin B contribute to the effects of stress, and suggest that blockade of kappa-opioid receptors may have therapeutic potential for the treatment of depression. Topics: Animals; Behavior, Animal; Cell Count; Disease Models, Animal; Dose-Response Relationship, Drug; Dynorphins; Endorphins; Escape Reaction; Helplessness, Learned; Immobilization; Immunohistochemistry; Limbic System; Male; Naltrexone; Rats; Rats, Sprague-Dawley; Stress, Physiological | 2004 |
Maternal separation alters maternal care, but has minor effects on behavior and brain opioid peptides in adult offspring.
The aim of the study was to investigate the effect of repeated maternal separation (MS; 4 hr per day) during postnatal Days 1 to 15 on emotionality and voluntary ethanol intake in adult male and female Wistar rat offspring relative to controls exposed to a brief (5-min) daily handling procedure. Brain immunoreactive opioid peptide levels and plasma levels of corticosterone also were measured. There were mainly no alterations in any of the tested behaviors (i.e., fleeing and freezing responses, exploratory behavior, spontaneous and amphetamine-induced locomotor activity and competitive behavior), ethanol intake, or immunoreactive opioid peptide levels in MS offspring, either in males or females, compared to their respective controls nor were there any differences in plasma corticosterone between groups. In addition, the dams' retrieval behavior of the pups also was studied, showing that MS dams spent more time in the nest with the pups after the 4-hr separation period compared to control dams. With respect to the used protocol of the MS procedure in the present study, our results do not provide support for the suggestion that this procedure is a relevant model for studying development of psychopathology and vulnerability to drug abuse. Topics: Animals; Brain; Corticosterone; Dynorphins; Emotions; Endorphins; Enkephalin, Methionine; Female; Male; Maternal Behavior; Maternal Deprivation; Motor Activity; Nesting Behavior; Rats; Reference Values; Social Behavior | 2004 |
Protein kinase C signaling transduces endorphin-primed cardiogenesis in GTR1 embryonic stem cells.
The prodynorphin gene and its product, dynorphin B, have been found to promote cardiogenesis in embryonic cells by inducing the expression of GATA-4 and Nkx-2.5, two transcription factor-encoding genes essential for cardiogenesis. The molecular mechanism(s) underlying endorphin-induced cardiogenesis remain unknown. In the present study, we found that GTR1 embryonic stem (ES) cells expressed cell surface kappa opioid receptors, as well as protein kinase C (PKC)-alpha, -beta1, -beta2, -delta, -epsilon, and -zeta. Cardiac differentiation was associated with a marked increase in the Bmax value for a selective opioid receptor ligand and complex subcellular redistribution of selected PKC isozymes. PKC-alpha, -beta1, -beta2, -delta, and -epsilon all increased in the nucleus of ES-derived cardiac myocytes, compared with nuclei from undifferentiated cells. In both groups of cells, PKC-delta and -epsilon were mainly expressed at the nuclear level. The nuclear increase of PKC-alpha, -beta1, and -beta2 was due to a translocation from the cytosolic compartment. In contrast, the increase of both PKC-delta and PKC-epsilon in the nucleus of ES-derived cardiomyocytes occurred independently of enzyme translocation, suggesting changes in isozyme turnover and/or gene expression during cardiogenesis. No change in PKC-zeta expression was observed during cardiac differentiation. Opioid receptor antagonists prevented the nuclear increase of PKC-alpha, PKC-beta1, and PKC-beta2 and reduced cardiomyocyte yield but failed to affect the nuclear increase in PKC-delta and -epsilon. PKC inhibitors prevented the expression of cardiogenic genes and dynorphin B in ES cells and abolished their development into beating cardiomyocytes. Topics: Animals; Cell Differentiation; Cell Line; Cell Nucleus; Dynorphins; Embryo, Mammalian; Embryo, Nonmammalian; Endorphins; Enkephalins; Enzyme Inhibitors; Gene Expression Regulation, Developmental; Heart; Isoenzymes; Myocytes, Cardiac; Myosin Heavy Chains; Narcotic Antagonists; Protein Kinase C; Protein Precursors; Receptors, Opioid, kappa; RNA, Messenger; Signal Transduction; Stem Cells | 2003 |
Dynorphin B is an agonist of nuclear opioid receptors coupling nuclear protein kinase C activation to the transcription of cardiogenic genes in GTR1 embryonic stem cells.
The cardiac differentiation of embryonic stem (ES) cells was found to involve prodynorphin gene and dynorphin B expression and was associated with the interaction of secreted dynorphin B with cell surface opioid receptors coupled with protein kinase C (PKC) signaling and complex subcellular redistribution patterning of selected PKC isozymes. Here, confocal microscopy revealed the presence of immunoreactive dynorphin B-like material in GTR1 ES cells, suggesting that dynorphin peptides may also act intracellularly. Opioid binding sites were identified in ES cell nuclei, with a single dissociation constant in the low nanomolar range. A significant increase in Bmax for a kappa opioid receptor ligand was observed in nuclei isolated from ES-derived cardiomyocytes compared with nuclei from undifferentiated cells. Direct exposure of nuclei isolated from undifferentiated ES cells to dynorphin B or U-50,488H, a synthetic kappa opioid receptor agonist, time- and dose-dependently activated the transcription of GATA-4 and Nkx-2.5, 2 cardiac lineage-promoting genes. Nuclear exposure to dynorphin B also enhanced the rate of prodynorphin gene transcription. These responses were abolished in a stereospecific fashion by the incubation of isolated nuclei with selective opioid receptor antagonists. Nuclei isolated from undifferentiated cells were able to phosphorylate the acrylodan-labeled MARCKS peptide, a high-affinity fluorescent PKC substrate. Exposure of isolated nuclei to dynorphin B induced a remarkable increase in nuclear PKC activity, which was suppressed by opioid receptor antagonists. Nuclear treatment with PKC inhibitors abolished the capability of dynorphin B to prime the transcription of cardiogenic genes. Topics: Animals; Cell Differentiation; Cell Line; Cell Lineage; Cell Nucleus; DNA-Binding Proteins; Dynorphins; Embryo, Mammalian; Embryo, Nonmammalian; Endorphins; Enkephalins; Enzyme Activation; GATA4 Transcription Factor; Gene Expression Regulation, Developmental; Heart; Homeobox Protein Nkx-2.5; Homeodomain Proteins; Myocytes, Cardiac; Protein Kinase C; Protein Precursors; Receptors, Opioid; RNA, Messenger; Signal Transduction; Stem Cells; Transcription Factors; Transcriptional Activation; Xenopus Proteins | 2003 |
Purification and quantification of opioid peptides in bone and joint tissues--a methodological study in the rat.
The occurrence of methionine-enkephalin-Arg(6)-Phe(7) (MEAP) and dynorphin B (DYNB) representing two main precursors of opioids was analyzed in specimens from rat cortical bone, periosteum, bone marrow and joint tissue by radioimmunoassay (RIA). MEAP and DYNB were extracted in a solution of 4% EDTA in 2 M acetic acid previously proven suitable for extraction of sensory and autonomic neuropeptides in bone and joints. In crude extracts of cortical bone, the immunoreactive (ir) levels of both opioids were under the detection limit of RIA. As for DYNB this also applied to crude extracts of joints and periosteum. Therefore, two purification methods were tested and compared, i.e. reverse phase C 18 and ion exchange chromatography. RIA of the elution fraction disclosed a significant difference between the two methods in terms of recovery, i.e. <5% and 50%, respectively. Thus, purification by ion exchange chromatography prior to RIA appeared to be the most suitable by providing measurable levels of both MEAP and DYNB in all tissues analyzed (highest in bone marrow, lowest in cortical bone). The described method offers a means of quantifying opioid peptides in bone and joints, which may be utilized in the analysis of regulatory mechanisms of nociception, growth and immune responses in different conditions. Topics: Animals; Bone and Bones; Chromatography, Ion Exchange; Dynorphins; Endorphins; Enkephalin, Methionine; Joints; Male; Radioimmunoassay; Rats; Rats, Inbred Lew; Reproducibility of Results | 2003 |
Activation of delta- and kappa-opioid receptors by opioid peptides protects cardiomyocytes via KATP channels.
To examine the receptor specificity and the mechanism of opioid peptide-induced protection, we examined freshly isolated adult rabbit cardiomyocytes subjected to simulated ischemia. Cell death as a function of time was assessed by trypan blue permeability. Dynorphin B (DynB) and Met5-enkephalin (ME) limitation of cell death (expressed as area under the curve) was sensitive to blockade by naltrindole (NTI, a delta-selective antagonist) and 5'-guanidinyl-17-(cyclopropylmethyl)-6,7-dehydro-4,5alpha-epoxy-3,14-dihydroxy-6,7-2',3'-indolomorphinan (GNTI dihydrochloride, a kappa-selective antagonist): 85.7 +/- 2.7 and 142.9 +/- 2.7 with DynB and DynB + NTI, respectively (P < 0.001), 94.1 +/- 4.2 and 164.5 +/- 7.3 with DynB and DynB + GNTI, respectively (P < 0.001), 111.9 +/- 7.0 and 192.1 +/- 6.4 with ME and ME + NTI, respectively (P < 0.001), and 120.2 +/- 4.3 and 170.0 +/- 3.3 with ME and ME + GNTI, respectively (P < 0.001). Blockade of ATP-sensitive K+ channels eliminated DynB- and ME-induced protection: 189.6 +/- 5.4 and 139.0 +/- 5.4 for control and ME, respectively (P < 0.001), and 210 +/- 5.9 and 195 +/- 6.1 for 5-HD and ME + 5-HD, respectively (P < 0.001); 136.0 +/- 5.7 and 63.4 +/- 5.4 for control and ME, respectively (P < 0.001), and 144.6 +/- 4.5 and 114.6 +/- 7.7 for HMR-1098 and ME + HMR-1098, respectively (P < 0.01); 189.6 +/- 5.4 and 139.0 +/- 5.4 for control and ME, respectively (P < 0.001), and 210 +/- 5.9 and 195 +/- 6.1 for 5-HD and ME + 5-HD, respectively (P < 0.001); and 136.0 +/- 5.7 and 63.4 +/- 5.4 for control and ME, respectively (P < 0.001), and 144.6 +/- 4.5 and 114.6 +/- 7.7 for HMR-1098 and ME + HMR-1098, respectively (P < 0.01). We conclude that opioid peptide-induced cardioprotection is mediated by delta- and kappa-receptors and involves sarcolemmal and mitochondrial ATP-sensitive K+ channels. Topics: Adenosine Triphosphate; Animals; Benzamides; Cardiotonic Agents; Dose-Response Relationship, Drug; Dynorphins; Endorphins; Enkephalin, Methionine; Guanidines; Ischemic Preconditioning, Myocardial; Male; Mitochondria; Morphinans; Myocardial Ischemia; Myocytes, Cardiac; Naltrexone; Narcotic Antagonists; Potassium Channels; Rabbits; Receptors, Opioid, delta; Receptors, Opioid, kappa; Sarcolemma | 2003 |
The regulation of steroidogenesis by opioid peptides in porcine theca cells.
The present study was designed to investigate basal and LH-induced steroidogenesis in porcine theca cells from large follicles in response to various concentrations (1-1000 nM) of mu opioid receptor agonists (beta-endorphin, DAMGO, FK 33-824), delta receptor agonists (met-enkephalin, leu-enkephalin, DPLPE) and kappa receptor agonists (dynorphin A, dynorphin B, U 50488). Agonists of mu opioid receptors suppressed basal androstenedione (A4), testosterone (T) and oestradiol-17beta (E2) secretion and enhanced LH-induced A4 and T release by theca cells. The inhibitory effect of the agonists on E2 secretion was abolished in the presence of LH. All delta receptor agonists depressed basal progesterone (P4) output. However, the influence of these agents on LH-treated cells was negligible. Among delta receptor agonist used only leu-enkephalin and DPLPE at the lowest concentrations inhibited basal A4 release. The presence of LH in culture media changed the influence of these opioids from inhibitory to stimulatory. Similarly, DPLPE reduced T secretion by non-stimulated theca cells and enhanced T secretion of stimulated cells. All of delta agonists inhibited basal E2 secretion and unaffected its release from LH-treated theca cells. Agonists of kappa receptors inhibited basal, non-stimulated, P4 secretion and two of them (dynorphin B, U 50488) potentiated LH-induced P4 output. Basal A4 and T release remained unaffected by kappa agonist treatment, but the cells cultured in the presence of LH generally increased both androgen production in response to these opioids. Basal secretion of E2 was also suppressed by kappa agonists. This inhibitory effect was not observed when the cells were additionally treated with LH. In view of these findings we suggest that opioid peptides derived from three major opioid precursors may directly participate in the regulation of porcine theca cell steroidogenesis. Topics: 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer; Animals; beta-Endorphin; Cells, Cultured; D-Ala(2),MePhe(4),Met(0)-ol-enkephalin; Dynorphins; Endorphins; Enkephalin, Ala(2)-MePhe(4)-Gly(5)-; Enkephalin, D-Penicillamine (2,5)-; Enkephalin, Leucine; Enkephalin, Methionine; Estradiol; Female; Opioid Peptides; Progesterone; Receptors, Opioid, delta; Receptors, Opioid, kappa; Receptors, Opioid, mu; Steroids; Swine; Testosterone; Theca Cells | 2003 |
Long-term effects of short and long periods of maternal separation on brain opioid peptide levels in male Wistar rats.
Environmental manipulations early in life may induce persistent alterations in adult behaviour and physiology. The underlying neural mechanisms of these responses are not yet clear. We have previously reported long-term changes in brain opioid peptide levels in male and female Sprague-Dawley rats after short periods (15 min, known as neonatal handling) of maternal separation (MS) until weaning. To study this further, we investigated behavioural and neurochemical effects of repeated MS in male Wistar rats. The rat pups were separated from their dams in litters for either 360 min (MS360) or 15 min (MS15) daily from postnatal day 1 to 21 or exposed to normal animal facility rearing. Behavioural analysis showed that MS360 rats had increased ultrasonic calls on postnatal day 5 compared to MS15 rats, but not on postnatal day 6. Moreover, the MS360 rats had more animals with higher frequency of calls at day 5 than 6 than the MS15 rats. Analysis of the opioid peptides dynorphin B and Met-enkephalin-Arg(6)Phe(7) with radioimmunoassay 7 weeks after the MS procedure, revealed long-term neurochemical changes in several brain areas and in the pituitary gland. Immunoreactive dynorphin B and Met-enkephalin-Arg(6)Phe(7) levels were affected in the hypothalamus and dynorphin B levels in the neurointermediate pituitary lobe, amygdala, substantia nigra and the periaqueductal gray. Together, these findings show that repeated periods of MS early in life in male Wistar rats affect the development of the ultrasonic call response and induce long-lasting and possibly permanent alterations in the opioid peptide systems. Topics: Animals; Animals, Newborn; Behavior, Animal; Body Weight; Brain; Corticosterone; Dynorphins; Endorphins; Enkephalin, Methionine; Enkephalins; Female; Male; Maternal Deprivation; Opioid Peptides; Pituitary Gland; Protein Precursors; Radioimmunoassay; Rats; Rats, Wistar; Time Factors; Ultrasonics | 2003 |
Substrate activation of insulin-degrading enzyme (insulysin). A potential target for drug development.
The rate of the insulin-degrading enzyme (IDE)-catalyzed hydrolysis of the fluorogenic substrate 2-aminobenzoyl-GGFLRKHGQ-ethylenediamine-2,4-dinitrophenyl is increased 2-7-fold by other peptide substrates but not by peptide non-substrates. This increased rate is attributed to a decrease in Km with little effect on Vmax. An approximately 2.5-fold increase in the rate of amyloid beta peptide hydrolysis is produced by dynorphin B-9. However, with insulin as substrate, dynorphin B-9 is inhibitory. Immunoprecipitation of differentially tagged IDE and gel filtration analysis were used to show that IDE exists as a mixture of dimers and tetramers. The equilibrium between dimer and tetramer is concentration-dependent, with the dimer the more active form. Bradykinin shifted the equilibrium toward dimer. Activation of substrate hydrolysis is not seen with a mixed dimer of IDE containing one active subunit and one subunit that is catalytically inactive and deficient in substrate binding. On the other hand, a mixed dimer containing one active subunit and one subunit that is catalytically inactive but binds substrate with normal affinity is activated by peptides. These findings suggest that peptides bind to one subunit of IDE and induce a conformational change that shifts the equilibrium to the more active dimer as well as activates the adjacent subunit. The selective activation of IDE toward amyloid beta peptide relative to insulin suggests the potential for development of compounds that increase IDE activity toward amyloid beta peptide as a therapeutic intervention for the treatment of Alzheimer's disease. Topics: Animals; Binding Sites; Chromatography, Gel; COS Cells; Dimerization; Dose-Response Relationship, Drug; Drug Design; Dynorphins; Endorphins; Enzyme Activation; Hydrolysis; Insecta; Insulin; Insulysin; Kinetics; Metalloendopeptidases; Peptides; Precipitin Tests; Protein Conformation; Rats; Recombinant Proteins; Substrate Specificity; Time Factors; Trypsin | 2003 |
The alpha9/alpha10-containing nicotinic ACh receptor is directly modulated by opioid peptides, endomorphin-1, and dynorphin B, proposed efferent cotransmitters in the inner ear.
Opioid peptides have been detected in the auditory and vestibular efferent neurons where they colocalize with the major neurotransmitter, acetylcholine. We investigated the function of opioids to modulate neurotransmission mediated by hair cell's alpha9/alpha10-containing nicotinic acetylcholine receptors (alpha9/alpha10nAChRs). The endogenous opioid peptides, endomorphin-1 (mu agonist) and dynorphin B (kappa agonist), but not a delta agonist [D-Pen2,D-Pen-5]enkephalin, inhibited the acetylcholine-evoked currents in frog saccular hair cells and rat inner hair cells. This inhibition was noncompetitive, voltage-independent, and was accompanied by an acceleration of the rate of current decay. Selective mu- and kappa-opioid receptor antagonists did not block the inhibition, although partial reduction by naloxone was observed. All opioid antagonists tested also reduced the acetylcholine response. Endomorphin-1 and dynorphin B inhibited the acetylcholine-evoked currents in alpha9/alpha10-expressing Xenopus oocytes. Because oocytes lack opioid receptors, it provides strong evidence for the direct interaction of opioid peptides with alpha9/alpha10nAChR.. alpha9/alpha10nAChR is a target for modulation by endomorphin-1 and dynorphin B, efferent cotransmitters in the inner ear. Topics: Acetylcholine; Animals; Anura; Cochlea; Dynorphins; Ear, Inner; Electric Conductivity; Endorphins; Enkephalin, D-Penicillamine (2,5)-; Hair Cells, Auditory; In Vitro Techniques; Narcotic Antagonists; Neurotransmitter Agents; Oligopeptides; Oocytes; Protein Isoforms; Rats; Rats, Sprague-Dawley; Receptors, Nicotinic; Saccule and Utricle; Synapses; Xenopus laevis | 2002 |
Intrathecally administered big dynorphin, a prodynorphin-derived peptide, produces nociceptive behavior through an N-methyl-D-aspartate receptor mechanism.
Intrathecal (i.t.) administration of big dynorphin (1-10 fmol), a prodynorphin-derived peptide consisting of dynorphin A and dynorphin B, to mice produced a characteristic behavioral response, the biting and/or licking of the hindpaw and the tail along with slight hindlimb scratching directed toward the flank, which peaked at 5-15 min after an injection. Dynorphin A produced a similar response, though the doses required were higher (0.1-30 pmol) whereas dynorphin B was practically inactive even at 1000 pmol. The behavior induced by big dynorphin (3 fmol) was dose-dependently inhibited by intraperitoneal injection of morphine (0.125-2 mg/kg) and also dose-dependently, by i.t. co-administration of D(-)-2-amino-5-phosphonovaleric acid (D-APV) (1-4 nmol), a competitive N-methyl-D-aspartate (NMDA) receptor antagonist, MK-801 (0.25-4 nmol), an NMDA ion-channel blocker, and ifenprodil (2-8 pmol), an inhibitor of the NMDA receptor ion-channel complex interacting with the NR2B subunit and the polyamine recognition site. On the other hand, naloxone, an opioid receptor antagonist, 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX), a non-NMDA glutamate receptor antagonist, 7-chlorokynurenic acid, a competitive antagonist of the glycine recognition site on the NMDA receptor ion-channel complex, [D-Phe(7),D-His(9)]-substance P(6-11), a specific antagonist for substance P (NK1) receptors, and MEN-10376, a tachykinin NK2 receptor antagonist, had no effect. These results suggest that big dynorphin-induced nociceptive behavior is mediated through the activation of the NMDA receptor ion-channel complex by acting on the NR2B subunit and/or the polyamine recognition site but not on the glycine recognition site, and does not involve opioid, non-NMDA glutamate receptor mechanisms or tachykinin receptors in the mouse spinal cord. Topics: 2-Amino-5-phosphonovalerate; 6-Cyano-7-nitroquinoxaline-2,3-dione; Analgesics, Opioid; Animals; Behavior, Animal; Dizocilpine Maleate; Dynorphins; Endorphins; Enkephalins; Excitatory Amino Acid Antagonists; Injections, Spinal; Kynurenic Acid; Male; Mice; Morphine; Neurokinin A; Nociceptors; Peptide Fragments; Piperidines; Protein Precursors; Receptors, N-Methyl-D-Aspartate; Receptors, Tachykinin; Substance P | 2002 |
Effect of single and repeated methamphetamine treatment on neurotransmitter release in substantia nigra and neostriatum of the rat.
The main purpose of this study was to characterize the initial neurotransmission cascade elicited by methamphetamine, analysing simultaneously with in vivo microdialysis monoamine, amino acid and neuropeptide release in substantia nigra and neostriatum of the rat. The main effect of a single systemic dose of methamphetamine (15 mg/kg, subcutaneously) was an increase in dopamine levels, both in substantia nigra ( approximately 10-fold) and neostriatum ( approximately 40-fold), accompanied by a significant, but lesser, increase in dynorphin B ( approximately two-fold, in both regions), and a decrease in monoamine metabolites. A similar effect was also observed after local administration of methamphetamine (100 microm) via the microdialysis probes, but restricted to the treated region. In other experiments, rats were repeatedly treated with methamphetamine or saline, with the last dose administered 12 h before microdialysis. Dopamine K+-stimulated release was decreased following repeated methamphetamine administration compared with that following saline, both in the substantia nigra (by approximately 65%) and neostriatum (by approximately 20%). In contrast, the effect of K+-depolarization on glutamate, aspartate and GABA levels was increased following repeated administration of methamphetamine. In conclusion, apart from an impairment of monoamine neurotransmission, repeated methamphetamine produces changes in amino acid homeostasis, probably leading to NMDA-receptor overstimulation. Topics: Amino Acids; Animals; Aspartic Acid; Biogenic Monoamines; Central Nervous System Stimulants; Dopamine; Drug Administration Routes; Drug Administration Schedule; Dynorphins; Endorphins; Extracellular Space; gamma-Aminobutyric Acid; Glutamic Acid; Male; Methamphetamine; Microdialysis; Neostriatum; Neurotransmitter Agents; Potassium; Rats; Rats, Sprague-Dawley; Substantia Nigra; Synaptic Transmission | 2002 |
Alteration in endogenous opioid systems due to chronic inflammatory pain conditions.
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 |
Orexin/hypocretin neurons: chemical phenotype and possible interactions with melanin-concentrating hormone neurons.
We showed earlier that a specific neuron population of the rat lateral hypothalamus, differing from the codistributed melanin-concentrating hormone (MCH) neurons, express both dynorphin (DYN) and secretogranin II (SgII) genes. We demonstrated later that this population corresponds in fact to the newly identified orexin/hypocretin (OX/Hcrt) neurons. In the present study, by revisiting the chemical phenotype of these neurons, we confirm that all of them contain DYN B- and SgII-immunoreactive materials. The roles played by these peptide/protein in OX/Hcrt neurons are still unclear. Double immunocytochemical stainings highlight putative somasomatic, axosomatic and axodendritic contacts between OX/Hcrt and MCH neurons. Adding OX/Hcrt to the culture medium of hypothalamic slices from 8-day-old rats results either in a significant increase of MCH mRNA after 24 h survival or a strong fall after 10 days culture. These results taken together suggest that OX/Hcrt can directly and/or indirectly affect MCH expression, and that both OX/Hcrt and MCH neuron populations interact to respond in a coordinated manner to central and peripheral signals. Topics: Animals; Carrier Proteins; Cell Communication; Chromogranins; Dynorphins; Endorphins; Hypothalamic Area, Lateral; Hypothalamic Hormones; Immunohistochemistry; Intracellular Signaling Peptides and Proteins; Male; Melanins; Neurons; Neuropeptides; Orexins; Pituitary Hormones; Protein Biosynthesis; Proteins; Rats; Rats, Sprague-Dawley; Tissue Distribution | 2002 |
Central changes in nociceptin dynorphin B and Met-enkephalin-Arg-Phe in different models of nociception.
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.
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 |
The effect on opioid peptides in the rat brain, after chronic treatment with the anabolic androgenic steroid, nandrolone decanoate.
In recent years, an increase in abuse of anabolic androgenic steroids (AAS) has been seen among individuals not directly connected to sports. Clinical evidence suggests that abuse of these steroids may result in profound changes in personality, expressed by depressive symptoms, irritability and increased aggression. It is still unknown whether these alterations are related to changes in any particular transmitter system or whether they are persistent or reversible. In this study we focused on AAS effect on the endogenous dynorphin and enkephalin system in the brain. Male rats were given intramuscular injections of the AAS nandrolone decanoate (15 mg/kg), once daily for 2 weeks. The levels of the opioid peptide immunoreactivities (ir) were assessed by radioimmunoassay in two groups immediately after the treatment and in two other groups after additional 3 weeks without any drug treatment (recovery period). The result indicates that chronic AAS treatment increased the activity in the dynorphin B- and Met-enkephalin-Arg(6)Phe(7)-ir in the hypothalamus, striatum and periaqueductal gray (PAG) compared to controls. In addition, the steroid induced an imbalance between the dynorphin and the enkephalin opioid system in the nucleus accumbens, hypothalamus and PAG. This imbalance remained after the recovery period. Since increased peptide activity was found in brain regions regulating emotions, dependence, defensive reactions and aggression, it was suggested that the actual endogenous opioid systems are involved in previously reported AAS-induced changes in these behaviours. Topics: Anabolic Agents; Animals; Brain; Corpus Striatum; Dynorphins; Endorphins; Enkephalin, Methionine; Hypothalamus; Male; Nandrolone; Opioid Peptides; Periaqueductal Gray; Radioimmunoassay; Rats; Rats, Sprague-Dawley; Receptors, Opioid, delta; Receptors, Opioid, kappa; Receptors, Opioid, mu | 2000 |
Elf-pulsed magnetic fields modulate opioid peptide gene expression in myocardial cells.
Magnetic fields have been shown to affect cell proliferation and growth factor expression in cultured cells. Although the activation of endorphin systems is a recurring motif among the biological events elicited by magnetic fields, compelling evidence indicating that magnetic fields may modulate opioid gene expression is still lacking. We therefore investigated whether extremely low frequency (ELF) pulsed magnetic fields (PMF) may affect opioid peptide gene expression and the signaling pathways controlling opioid peptide gene transcription in the adult ventricular myocyte, a cell type behaving both as a target and as a source for opioid peptides.. Prodynorphin gene expression was investigated in adult rat myocytes exposed to PMF by the aid of RNase protection and nuclear run-off transcription assays. In PMF-exposed nuclei, nuclear protein kinase C (PKC) activity was followed by measuring the phosphorylation rate of the acrylodan-labeled MARCKS peptide. The effect of PMF on the subcellular distribution of different PKC isozymes was assessed by immunoblotting. A radioimmunoassay procedure coupled to reversed-phase high performance liquid chromatography was used to monitor the expression of dynorphin B.. Here, we show that PMF enhanced myocardial opioid gene expression and that a direct exposure of isolated myocyte nuclei to PMF markedly enhanced prodynorphin gene transcription, as in the intact cell. The PMF action was mediated by nuclear PKC activation but occurred independently from changes in PKC isozyme expression and enzyme translocation. PMF also led to a marked increase in the synthesis and secretion of dynorphin B.. The present findings demonstrate that an opioid gene is activated by myocyte exposure to PMF and that the cell nucleus and nuclear embedded PKC are a crucial target for the PMF action. Due to the wide ranging importance of opioid peptides in myocardial cell homeostasis, the current data may suggest consideration for potential biological effects of PMF in the cardiovascular system. Topics: Analysis of Variance; Animals; Cell Nucleus; Cells, Cultured; Dynorphins; Electromagnetic Fields; Endorphins; Gene Expression Regulation; Immunoblotting; Male; Myocardium; Opioid Peptides; Protein Kinase C; Rats; Rats, Wistar; RNA, Messenger | 2000 |
Opioid peptide gene expression primes cardiogenesis in embryonal pluripotent stem cells.
Zinc finger-containing transcription factor GATA-4 and homeodomain Nkx-2.5 govern crucial developmental fates and have been found to promote cardiogenesis in embryonic cells exposed to the differentiating agent DMSO. Nevertheless, intracellular activators of these transcription factors are largely unknown. In this study, pluripotent P19 cells expressed the prodynorphin gene, an opioid gene encoding for the dynorphin family of opioid peptides. P19 cells were also able to synthesize and secrete dynorphin B, a biologically active end product of the prodynorphin gene. DMSO-primed GATA-4 and Nkx-2.5 gene expression was preceded by a marked increase in prodynorphin gene expression and dynorphin B synthesis and secretion. The DMSO effect occurred at the transcriptional level. In the absence of DMSO, dynorphin B triggered GATA-4 and Nkx-2.5 gene expression and led to the appearance of both alpha-myosin heavy chain and myosin light chain-2V transcripts, two markers of cardiac differentiation. Moreover, dynorphin B-exposed cells were positively stained in the presence of MF 20, a mouse monoclonal antibody raised against the alpha-myosin heavy chain. Opioid receptor antagonism and inhibition of opioid gene expression by a prodynorphin antisense phosphorothioate oligonucleotide blocked DMSO-induced cardiogenesis, suggesting an autocrine role of an opioid gene in developmental decisions. Topics: Animals; Autocrine Communication; Cell Differentiation; Dimethyl Sulfoxide; DNA-Binding Proteins; Dynorphins; Endorphins; Enkephalins; Fetal Heart; Fetal Proteins; GATA4 Transcription Factor; Gene Expression Regulation, Developmental; Homeobox Protein Nkx-2.5; Homeodomain Proteins; Mice; Oligonucleotides, Antisense; Opioid Peptides; Protein Precursors; Recombinant Fusion Proteins; RNA, Messenger; Stem Cells; Thionucleotides; Transcription Factors; Transfection; Xenopus Proteins; Zinc Fingers | 2000 |
Basal levels and alcohol-induced changes in nociceptin/orphanin FQ, dynorphin, and enkephalin levels in C57BL/6J mice.
In order to investigate the involvement of the opioid and nociceptin/orphanin FQ (N/OFQ) system in alcohol drinking behaviour, N/OFQ and the opioid peptides dynorphin B (DYNB) and Met-enkephalin-Arg(6) Phe(7) (MEAP) were examined in the alcohol-preferring C57BL/6J mice. Basal peptide levels were compared in the brain and the pituitary gland with basal levels in the alcohol-avoiding DBA/2J mice. Furthermore, the effects of chronic alcohol self-administration on peptides were studied in the C57BL/6J mice. Compared to the DBA/2J mice, C57BL/6J mice had low immunoreactive (ir) levels of DYNB and MEAP in the nucleus accumbens, the hippocampus, and the substantia nigra, low ir-DYNB levels in the striatum and low ir-MEAP levels in the frontal cortex. Higher ir-DYNB levels in the pituitary gland and in the periaqueductal gray (PAG) and higher ir-N/OFQ levels in the frontal cortex and in the hippocampus were detected in C57BL/6J mice compared to the DBA/2J mice. After 4 weeks of voluntary alcohol consumption, only minor changes in steady-state peptide levels were identified. However, 5 days after the alcohol-drinking period, lower levels of all peptides were detected in the ventral tegmental area and ir-DYNB levels were also lower in the amygdala and in the substantia nigra. Twenty-one days after cessation of alcohol self-administration, the opioid peptides in alcohol-consuming C57BL/6J mice were lower in the PAG, the N/OFQ was lower in the frontal cortex and DYNB was higher in the amygdala and substantia nigra as compared to control C57BL/6J mice. This study demonstrates strain differences between C57BL/6J mice and DBA/2J mice that could contribute to divergent drug-taking behaviour, and it also demonstrates time- and structure-specific changes in neuropeptide levels after alcohol self-administration in the C57BL/6J mice. Topics: Alcohol Drinking; Alcohol-Induced Disorders, Nervous System; Animals; Brain; Brain Chemistry; Drug Administration Schedule; Dynorphins; Endorphins; Enkephalin, Methionine; Enkephalins; Ethanol; Male; Mice; Mice, Inbred C57BL; Mice, Inbred DBA; Nociceptin; Opioid Peptides; Pituitary Gland; Self Administration | 2000 |
Repeated ethanol administration induces short- and long-term changes in enkephalin and dynorphin tissue concentrations in rat brain.
Recently, we have shown that rats repeatedly treated with ethanol and/or cocaine have decreased kappa-opioid receptor mRNA levels in the mesolimbic system. The aim of the present study was to investigate the short- and long-term effects of repeated ethanol administration on opioid peptide concentrations in brain tissue of male Sprague-Dawley rats. Dynorphin B (1-13) (Dyn B) and Met-enkephalinArg(6)Phe(7) (MEAP), endogenous ligands to kappa- and delta-opioid receptors, respectively, were measured using radioimmunoassays. The rats were given either ethanol [intraperitoneal (ip), twice daily, 2 g/kg bw/dose] or saline for 13 consecutive days. Thirty minutes after the last ethanol dose on Day 13, the Dyn B tissue concentration was significantly decreased in the cingulate cortex. The MEAP tissue concentration was decreased in the hippocampus 5 days after the last ethanol injection as compared to saline-treated controls. Furthermore, the Dyn B and the MEAP concentrations were increased in the periaqueductal grey area (PAG) at this time point. Of particular interest were the significant increases in Dyn B tissue concentrations found in the nucleus accumbens (NAcc) at 30 min and at 21 days after the last ethanol dose. The results suggest that repeated ethanol administration induces both short- and long-term changes in the tissue concentrations of opioids in certain brain regions associated with motivation and reward. Topics: Animals; Brain; Brain Chemistry; Dynorphins; Endorphins; Enkephalin, Methionine; Ethanol; Hippocampus; Kinetics; Male; Nucleus Accumbens; Periaqueductal Gray; Rats; Rats, Sprague-Dawley | 2000 |
Antisense mapping KOR-1: evidence for multiple kappa analgesic mechanisms.
In binding assays, both dynorphin B and alpha-neoendorphin are relatively selective for the kappa1b site, unlike U50,488H which has high affinity for both kappa1a and kappa1b sites. In vivo, U50,488H, dynorphin B and alpha-neoendorphin analgesia are reversed by the kappa1-selective antagonist, nor-binaltorphimine (norBNI). Antisense mapping the three exons of KOR-1 revealed that probes targeting all three exons blocked U50,488H analgesia, as expected. However, the selectivity profile of dynorphin B and alpha-neoendorphin analgesia towards the various antisense oligodeoxynucleotides differed markedly from U50,488H, implying a different receptor mechanism of action. Topics: 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer; Analgesics, Non-Narcotic; Animals; Antisense Elements (Genetics); Binding Sites; Chromosome Mapping; Dynorphins; Endorphins; Ion Channel Gating; Male; Mice; Mice, Inbred ICR; Protein Precursors; Receptors, Opioid, kappa | 1999 |
Morphometry of a peptidergic transmitter system: dynorphin B-like immunoreactivity in the rat hippocampal mossy fiber pathway before and after seizures.
While the morphometry of classical transmitter systems has been extensively studied, relatively little quantitative information is available on the subcellular distribution of peptidergic dense core vesicles (DCVs) within axonal arbors and terminals, and how distribution patterns change in response to neural activity. This study used correlated quantitative light and electron microscopic immunohistochemistry to examine dynorphin B-like immunoreactivity (dyn B-LI) in the rat hippocampal mossy fiber pathway before and after seizures. Forty-eight hours after seizures induced by two pentylenetetrazol injections, light microscopic dyn B-LI was decreased dorsally and increased ventrally. Ultrastructural examination indicated that, in the hilus of the dentate gyrus, these alterations resulted from changes that were almost entirely restricted to the profiles of the large mossy-like terminals formed by mossy fiber collaterals (which primarily contact spines), compared to the profiles of the smaller, less-convoluted terminals found on the same collaterals (which primarily contact aspiny dendritic shafts). Dorsally, mossy terminal profile labeled DCV (/DCV) density dropped substantially, while ventrally, both mossy terminal profile perimeter and /DCV density increased. In all terminal profile examined, /DCVs also were closely associated with the plasma membrane. Following seizures, there was a reorientation of /DCVs along the inner surface of mossy terminal profile membranes, in relation to the types of profiles adjacent to the membrane: in both the dorsal and ventral hilus, significantly fewer /DCVs were observed at sites apposed to dendrites, and significantly more were observed at sites apposed to spines. Thus, after seizures, changes specific to: (1) the dorsoventral level of the hippocampal formation, (2) the type of terminal, and (3) the type of profile in apposition to the portion of the terminal membrane examined were all observed. An explanation of these complex, interdependent alterations will probably require evoking multiple interrelated mechanisms, including selective prodynorphin synthesis, transport, and release. Topics: Animals; Disease Models, Animal; Dynorphins; Endorphins; Immunohistochemistry; Linear Models; Male; Microscopy, Electron; Mossy Fibers, Hippocampal; Neuropeptides; Neurotransmitter Agents; Rats; Rats, Sprague-Dawley; Seizures | 1999 |
Variation in hippocampal dynorphin b-immunoreactive mossy fiber terminal fields of apomorphine-(un)susceptible rats.
The size of distinct hippocampal sub-fields were measured in the apomorphine-susceptible and apomorphine-unsusceptible rat lines. Mossy fiber terminal fields were delineated using dynorphin B immunoreactivity and area measurements were taken from (1) the supra-pyramidal mossy fiber terminal field; (2) the intra- and infra-pyramidal mossy fiber terminal field; (3) the hilus of the fascia dentata (4) the non dynorphin B immunoreactive area of the regio inferior and fascia dentata and (5) the total area of regio inferior and fascia dentata. The data indicate that statistically significant differences in the morphometry of the hippocampal subfields of the apomorphine susceptible and unsusceptible rats are confined to the intra- and infra terminal field: the relative size of the left and right intra- and infra terminal field of apomorphine unsusceptible rats are significantly larger than those of the apomorphine susceptible rats. These data explain at least in part the differential response of these rats to novelty. Topics: Animals; Apomorphine; Dentate Gyrus; Dopamine Agonists; Dynorphins; Endorphins; Hippocampus; Male; Mossy Fibers, Hippocampal; Rats; Rats, Wistar | 1999 |
Neonatal handling in rats induces long-term effects on dynorphin peptides.
The effects of neonatal handling on the opioid dynorphin peptides in the brain and pituitary gland of Sprague-Dawley rats were investigated. Ten weeks after the neonatal handling, handled rats had higher tissue levels of dynorphin A and B in the hypothalamus, pituitary gland and striatum and slightly higher dynorphin B levels in the hippocampus, medulla oblongata and midbrain as compared with non-handled controls. The results indicate a persistent upregulation of the dynorphin system in certain brain areas after neonatal handling, which could contribute to the behavioural changes in these rats observed later in life. Observation in the open field and the elevated plus-maze tests confirmed behavioural effects of neonatal handling, i.e. showing that handled rats exhibit attenuated fearfulness in novel environments as compared with non-handled rats. Topics: Animals; Animals, Newborn; Behavior, Animal; Brain Chemistry; Corpus Striatum; Dynorphins; Endorphins; Female; Handling, Psychological; Hippocampus; Hypothalamus; Maze Learning; Medulla Oblongata; Mesencephalon; Physical Stimulation; Pituitary Gland; Pregnancy; Rats; Rats, Sprague-Dawley; Substance-Related Disorders | 1999 |
Hippocampal dynorphin B injections impair spatial learning in rats: a kappa-opioid receptor-mediated effect.
The hippocampus plays a central role in the acquisition and storage of information. Long-term potentiation in the mossy fibre pathway to the CA3 region in the hippocampus, an animal model of memory acquisition, is modulated by dynorphin peptides. This study investigated the possible role of hippocampal dynorphin in spatial learning. Male rats were trained in the Morris Water Task after microinjection with different doses of dynorphin B (1, 3.3 or 10 nmol/rat) or artificial cerebrospinal fluid (as control) into the CA3 region of the dorsal hippocampus. Dynorphin B was found to impair spatial learning at all tested doses. The synthetic kappa1-selective opiate receptor antagonist nor-binaltorphimine (2 nmol) also given into the hippocampus fully blocked the acquisition impairment caused by dynorphin B (10 nmol), while nor-binaltorphimine alone did not affect learning performance. These findings suggest that dynorphin peptides could play a modulatory role in hippocampal plasticity by acting on hippocampal kappa-receptors and thereby impair spatial learning. Topics: Animals; Behavior, Animal; Dose-Response Relationship, Drug; Dynorphins; Endorphins; Hippocampus; Male; Maze Learning; Microinjections; Naltrexone; Rats; Rats, Sprague-Dawley; Receptors, Opioid, kappa; Spatial Behavior | 1998 |
[Effect of opioid peptides on immunomodulation].
Activation of the opioid receptors by delta-agonist DSLET and by kappa-agonist rimorphin led to a significant inhibition of plaque-forming and rosette-forming cells in the CBA mice. On the other hand, mu-agonist DAGO stimulated the immune response on the 4th and 5th days after immunization with SRBC (5 x 10(8)). Lesion of the hypothalamo-hypophyseal connection prevented immuno-stimulating as well as immuno-depressive effects. The latter seems to be due to an interaction with the serotoninergic system, whereas immuno-stimulating effect involves the dopaminergic system. Topics: Animals; Cyproheptadine; Dopamine D2 Receptor Antagonists; Dynorphins; Endorphins; Enkephalin, Ala(2)-MePhe(4)-Gly(5)-; Enkephalin, Leucine; Enkephalins; Haloperidol; Male; Mice; Mice, Inbred CBA; Neuroimmunomodulation; Pituitary Gland; Receptor, Serotonin, 5-HT2A; Receptors, Dopamine D2; Receptors, Opioid; Receptors, Opioid, delta; Receptors, Opioid, kappa; Receptors, Opioid, mu; Receptors, Serotonin; Spleen | 1998 |
Characterization of immunoreactive dynorphin B and beta-endorphin in human plasma.
Dynorphins and beta-endorphin in human plasma were characterized and studied quantitatively using radioimmunoassay, high-performance liquid chromatography (HPLC), and mass spectrometry. Most immunoreactive (ir) dynorphin B and beta-endorphin in human plasma coeluted with authentic peptides in analysis. Dynorphin A was not detected. Added to human plasma it was rapidly converted into Leu-enkephalin-Arg6 followed by elimination of the C-terminal arginine after prolonged incubation. The rate of dynorphin A conversion was estimated at 40 pmol/min/microl plasma. This process was inhibited by the thiol protease inhibitor, PHMB and by EDTA. Dynorphin B, alpha-neoendorphin and big dynorphin were virtually not metabolized by plasma proteases under the same conditions. beta-endorphin was processed into beta-endorphin(1-19) and the corresponding C-terminal counterpart beta-endorphin(20-31) at a rate of about 25 pmol/min/microl of plasma. Based on the above data, a reliable strategy was established to measure dynorphin B- and beta-endorphin-ir in human plasma samples. The basal levels in a male control group were 0.99 +/- 0.11 (n = 11) and 16.3 +/- 1.5 (n = 11) fmol/ml plasma, respectively. Topics: Adult; beta-Endorphin; Chromatography, High Pressure Liquid; Dynorphins; Endopeptidases; Endorphins; Humans; Male; Mass Spectrometry; Neuropeptides; Peptide Fragments; Protease Inhibitors; Radioimmunoassay | 1998 |
The hippocampal opioidergic system: a comparative immunocytochemical study in four rodents.
The occurrence and distribution of four opioid peptides (methionine-enkephalin /met-enk/, leucine-enkephalin /leu-enk/, dynorphin-A /dyn-A/, dynorphin-B /dyn-B/) were studied and compared in the subregions and layers of the hippocampi of four rodent species (rat, mouse, hamster and guinea-pig) by means of pre-embedding immunocytochemistry in order to reveal species-specific morphological characteristics. Concerning the mossy fibre system, in the rat met-enk and dyn-A, in the mouse met-enk, in the the hamster dyn-A, in the guinea-pig dyn-B was found to be the dominating immunocytochemically detectable hippocampal opioid peptide. Outside the mossy fibre system, in other hippocampal areas met-enk-immunopositive varicose nerve fibres were most frequently detected in the rat and mouse, whereas dyn-B was the most abundant in the guinea-pig. In the hamster hippocampus all the four studied opioid peptides occurred at least in few varicose fibres. In the rat and mouse enkephalinergic boutons formed pericellular baskets around non-principal cells, whereas dynorphin-immunopositive boutons were visualized in similar arrangement around principal cells (both granule and pyramidal cells) in three of the studied species, except the rat. Among other species-specific differences, the presence of leu-enk-immunopositive perikarya in the golden hamster hippocampus, and a highly ordered leu-enk-immunoreactive fibre system exclusively detected in the CA1 area of the guinea-pig hippocampus are noteworthy. The results confirm the high degree of species-specific heterogeneity characterizing the distribution of the opioid peptides in the hippocampal formation. Topics: Animals; Cricetinae; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalin, Methionine; Female; Guinea Pigs; Hippocampus; Immunohistochemistry; Male; Mesocricetus; Mice; Nerve Fibers; Neurons; Pyramidal Cells; Rats; Species Specificity | 1998 |
Pressor effects of endogenous opioid system during acute episodes of blood pressure increases in hypertensive patients.
To investigate the involvement of endogenous opioids in acute increases in blood pressure and their functional relationship with atrial natriuretic factor and endothelin-1, we assessed plasma levels of beta-endorphin, met-enkephalin, dynorphin B, catecholamines, atrial natriuretic factor, and endothelin-1 before and after administration of the opioid antagonist naloxone hydrochloride (8 mg i.v.) in 28 hypertensive patients with a stress-induced acute increase in blood pressure. Ten patients with established mild or moderate essential hypertension and 10 normotensive subjects served as control groups. Opioids, atrial natriuretic factor, and endothelin-I were radioimmunoassayed after chromatographic preextraction; catecholamines were determined by high-performance liquid chromatography with electrochemical detection. Patients with an acute increase in blood pressure (systolic, 203.2 +/- 2.2 mm Hg; diastolic, 108.4 +/- 1.3) had plasma opioid, catecholamine, and atrial natriuretic factor levels significantly (P < .01) higher than hypertensive control patients (systolic pressure, 176.4 +/- 1.0 mm Hg; diastolic, 100.0 +/- 1.4), who had a hormonal pattern similar to that of normotensive subjects (systolic pressure, 123.2 +/- 1.5 mm Hg; diastolic, 75.0 +/- 2.0). Endothelin-1 did not differ in any group. In patients with an acute increase in blood pressure, naloxone significantly (P < .01) reduced blood pressure, heart rate, opioids, catecholamines, and atrial natriuretic factor 10 minutes after administration. Naloxone effects on blood pressure, heart rate, opioids, and catecholamines wore off within 20 minutes. In control groups, naloxone failed to modify any of the considered parameters. Our findings suggest that pressor effects of opioid peptides mediated by the autonomic nervous system during stress-induced acute episodes of blood pressure increase in hypertensive patients. Topics: Atrial Natriuretic Factor; beta-Endorphin; Blood Pressure; Dynorphins; Endorphins; Endothelin-1; Enkephalin, Methionine; Female; Heart Rate; Humans; Hypertension; Infusions, Intravenous; Male; Middle Aged; Naloxone; Narcotic Antagonists; Norepinephrine; Opioid Peptides; Stress, Physiological; Stress, Psychological | 1997 |
Opioid peptide gene expression in the primary hereditary cardiomyopathy of the Syrian hamster. III. Autocrine stimulation of prodynorphin gene expression by dynorphin B.
Prodynorphin mRNA and dynorphin B expression have been previously shown to be greatly increased in cardiac myocytes of BIO 14.6 cardiomyopathic hamsters. Here we report that exogenous dynorphin B induced a dose-dependent increase in prodynorphin mRNA levels and stimulated prodynorphin gene transcription in normal hamster myocytes. Similar responses were elicited by the synthetic selective kappa opioid receptor agonist U-50,488H. These effects were counteracted by the kappa opioid receptor antagonist Mr-1452 and were not observed in the presence of chelerythrine or calphostin C, two specific protein kinase C (PKC) inhibitors. Treatment of cardiomyopathic cells with Mr-1452 significantly decreased both prodynorphin mRNA levels and prodynorphin gene transcription. In control myocytes, dynorphin B induced the translocation of PKC-alpha to the nucleus and increased nuclear PKC activity without affecting the expression of PKC-delta, -epsilon, or -zeta. Acute release of either U-50,488H or dyn B over single normal or cardiomyopathic cells transiently increased the cytosolic Ca2+ concentration. A sustained treatment with each opioid agonist increased the cytosolic Ca2+ level for a more prolonged period in cardiomyopathic than in control myocytes and led to a depletion of Ca2+ from the sarcoplasmic reticulum in both groups of cells. The possibility that prodynorphin gene expression may affect the function of the cardiomyopathic cell through an autocrine mechanism is discussed. Topics: 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer; Animals; Calcium; Cardiomyopathy, Hypertrophic; Cell Compartmentation; Cell Nucleus; Cricetinae; Cytosol; Dynorphins; Endorphins; Enkephalins; Enzyme Inhibitors; Gene Expression; Mesocricetus; Myocardium; Protein Kinase C; Protein Precursors; Pyrrolidines; Receptors, Opioid, kappa; Ryanodine; Sarcoplasmic Reticulum | 1997 |
The Leu-enkephalin-encoding sequence DNA-binding factor (LEF) is the transcription factor YY1.
The Leu-enkephalin-encoding sequence DNA-binding factor (LEF) with high affinity for the Leu-enkephalin-encoding sequences in the prodynorphin and proenkephalin genes has earlier been identified. This factor is composed of three subunits of about 60, 70 (the major DNA-binding subunit), and 95 kDa, respectively. Estimated molecular mass, sequence specificity of DNA-binding, and supershift/inhibition with specific antibodies in a band shift assay showed that the DNA-binding subunit of LEF is identical to the multifunctional transcription factor YY1. However, an antibody against the C-terminus of YY1 distinguished the YY1 complexes with a Leu-enkephalin-encoding sequence and canonical YY1 binding site oligonucleotides, suggesting different protein conformations in complexes with these two DNA fragments. Topics: Animals; Binding Sites; Cell Nucleus; DNA; DNA-Binding Proteins; Dynorphins; Endorphins; Enkephalin, Leucine; Erythroid-Specific DNA-Binding Factors; Molecular Weight; Oligodeoxyribonucleotides; Protein Conformation; Rats; Rats, Sprague-Dawley; Transcription Factors; YY1 Transcription Factor | 1997 |
Opioid peptide modulation of circulatory and endocrine response to mental stress in humans.
Healthy subjects were classified according to their percent increase in systolic blood pressure (SBP) after mental arithmetic test (MAT) as low (delta SBP 9.3-15.1%, n = 15) and high (delta SBP 35.1-45.4%, n = 15) responders. During MAT, low responders showed significantly (p < 0.01) increased plasma levels of beta-endorphin, cortisol, catecholamines, and atrial natriuretic factor (ANF) and decreased levels of endothelin-1, whereas high responders showed increased (p < 0.01) levels of Metenkephalin, dynorphin B, and catecholamines. Pretreatment with naloxone hydrochloride enhanced (p < 0.01) SBP, heart rate, noradrenaline, cortisol, and endothelin-1 levels, and reduced (p < 0.01) ANF in low responders in response to MAT, whereas it decreased (p < 0.01) hemodynamic parameters, noradrenaline, and endothelin-1 in high responders. The individual differences in hemodynamic and endocrine responses to MAT may depend on a different activation of the endogenous opioid system. Topics: Adult; Analysis of Variance; Atrial Natriuretic Factor; beta-Endorphin; Blood Pressure; Catecholamines; Dynorphins; Endorphins; Endothelin-1; Enkephalin, Methionine; Female; Heart Rate; Hormones; Humans; Hydrocortisone; Male; Mathematics; Naloxone; Norepinephrine; Opioid Peptides; Stress, Psychological | 1997 |
Cholecystokinin-8S increases dynorphin B, aspartate and glutamate release in the fronto-parietal cortex of the rat via different receptor subtypes.
The effect of sulphated cholecystokinin-8 (CCK-8S) on extracellular dynorphin B, aspartate, glutamate and GABA levels in the rat fronto-parietal cortex was investigated with in vivo microdialysis. The peptide was infused through the microdialysis probe trying to mimic local CCK-8S release. Basal levels of dynorphin B were around 20 pM, aspartate 100 nM, glutamate 600 nM and GABA 30 nM. CCK-8S (10 microM) induced a approximately 3-fold increase in extracellular dynorphin B, aspartate and glutamate levels, while GABA levels were only slightly increased. The effect of CCK-8S was restricted to the stimulated neocortex. Systemic pretreatment with the CCKB antagonist, L-365, 260, but not with the CCKA antagonist, L-364, 718, significantly antagonised the effect of CCK-8S on cortical dynorphin B and aspartate release. However, both CCKA and CCKB antagonists inhibited the increase in cortical glutamate levels. Thus, the present results indicate that cortical CCK release exerts a stimulatory modulation on cortical dynorphin B and aspartate release via the CCKB receptor subtype, and on glutamate release via both CCKA and CCKB receptor subtypes. Considering electrophysiological evidence that CCK increases neuronal firing rates in many brain regions, it may be suggested that CCK represents a stimulatory system modulating the function of the neocortex. Topics: Animals; Aspartic Acid; Benzodiazepinones; Devazepide; Dynorphins; Endorphins; Frontal Lobe; gamma-Aminobutyric Acid; Glutamic Acid; Hormone Antagonists; Male; Microdialysis; Neurons; Nootropic Agents; Parietal Lobe; Phenylurea Compounds; Rats; Rats, Sprague-Dawley; Receptor, Cholecystokinin A; Receptor, Cholecystokinin B; Receptors, Cholecystokinin; Sincalide | 1997 |
Involvement of dynorphin B in the antinociceptive effects of the cannabinoid CP55,940 in the spinal cord.
Intrathecal administration of delta 9-tetrahydrocannabinol (delta 9-THC) but not the cannabinoid agonist CP55,940 enhances the antinociception produced by morphine. In addition, CP55,940- and delta 9-THC-induced antinociception is blocked by the kappa opioid antagonist norbinaltorphimine, and both cannabinoids are cross-tolerant to kappa agonists but do not act directly at the kappa receptor. Previous work in our laboratory has implicated dynorphins in the antinociceptive effects of delta 9-THC and its enhancement of morphine-induced antinociception. The goal of the present study was to evaluate the role of dynorphins in the antinociceptive effects of CP55,940 at the spinal level. Pretreatment of mice with antisera to dynorphin A(1-17), dynorphin A(1-8) or alpha-neoendorphin, all of which have been shown to retain specificity for blockade of their respective peptide in vivo, blocked the antinociceptive effects of delta 9-THC but not CP55,940. Dynorphin B produced antinociceptive effects on intrathecal administration to mice. Like CP55,940, dynorphin B failed to enhance the antinociceptive effects of morphine, whereas dynorphin A(1-17) and alpha-neoendorphin enhanced the antinociceptive effects of morphine. Using spinal catheterization of the rat, CP55,940 administration was shown to produce a significant release of dynorphin B concurrent with the production of antinociception. Our data suggest that CP55,940 induces a release of spinal dynorphin B that contributes at least in part to its antinociceptive effects in the spinal cord. Topics: Analgesics; Animals; Cannabinoids; Cyclohexanols; Dynorphins; Endorphins; Male; Mice; Mice, Inbred ICR; Rats; Spinal Cord | 1997 |
Levels of dynorphin peptides in the central nervous system and pituitary gland of the spontaneously hypertensive rat.
The levels of dynorphin A-like immunoreactivity (Dyn A-LI) and dynorphin B-like immunoreactivity (Dyn B-LI) were determined in various regions of brain, spinal cord and pituitary gland in spontaneously hypertensive rats (SHRs) as compared with the normotensive Wistar-Kyoto rats (WKYs). SHRs had significantly lower levels of Dyn A-LI and Dyn B-LI in the neurointermediate pituitary lobe and in the hippocampus. Conversely, the levels of Dyn A-LI and Dyn B-LI were higher in the hypothalamus, striatum and periaqueductal gray of the SHRs. Topics: Animals; Biomarkers; Brain; Dynorphins; Endorphins; Enkephalins; Hypertension; Immunohistochemistry; Lumbosacral Region; Male; Pituitary Gland; Protein Precursors; Rats; Rats, Inbred SHR; Rats, Inbred WKY; Spinal Cord | 1997 |
A comparison between microwave irradiation and decapitation: basal levels of dynorphin and enkephalin and the effect of chronic morphine treatment on dynorphin peptides.
Opioid peptides were analysed in tissue extracts of various brain structures and the pituitary gland from rats sacrificed by microwave irradiation, and compared with peptide levels in tissue extracts from decapitated rats. Dynorphin A, dynorphin B and Leu-enkephalinArg6, derived from prodynorphin, and Met-enkephalinArg6Phe7 from proenkephalin, were measured. Basal immunoreactive levels of dynorphin A and B were consistently higher in extracts from microwave-irradiated rats, whereas in these extracts immunoreactive levels of Leu-enkephalinArg6, an endogenous metabolite of dynorphin peptides, were either lower than, the same as or higher than in decapitated rats. Immunoreactive levels of Met-enkephalinArg6Phe7 were higher in microwave-irradiated rats. Effects of morphine treatment on prodynorphin peptide levels were evaluated and compared with previous findings in decapitated rats. Dynorphin immunoreactive levels were higher in the nucleus accumbens and striatum of morphine-tolerant rats than in corresponding areas in saline-treated rats. These results indicate tissue-specific metabolism of prodynorphin peptides and show that metabolism of opioid peptides occurs during the dissection procedure after decapitation of the rat even though precautions are taken to minimize degradation. Topics: Animals; Brain Chemistry; Decerebrate State; Dynorphins; Endorphins; Enkephalins; Injections, Subcutaneous; Male; Microwaves; Morphine; Opioid Peptides; Rats; Rats, Sprague-Dawley; Substance Withdrawal Syndrome | 1997 |
Differential metabolism of dynorphins in substantia nigra, striatum, and hippocampus.
To map the proteolytic enzymes metabolizing dynorphins in brain structures, size-exclusion chromatography linked to electrospray ionization mass spectrometry was used. Enzymes extracted from rat hippocampus, striatum, and substantia nigra were tested for their capability of converting dynorphin-related peptides. Dynorphin A was the most resistant to proteolytic conversion, whereas Big dynorphin and dynorphin B-29 were slowly converted to dynorphin A and dynorphins A and B, respectively. Dynorphin B and alpha-neoendorphin were the least resistant. Dynorphin B was rapidly converted to Leu-enkephalin in the striatum and hippocampus but to Leu-enkephalin-Arg6 in the substantia nigra. alpha-Neoendorphin was converted to Leu-enkephalin in all tissues investigated. Topics: Amino Acid Sequence; Animals; Chromatography, High Pressure Liquid; Corpus Striatum; Dynorphins; Endorphins; Enkephalin, Leucine; Hippocampus; Male; Mass Spectrometry; Protein Precursors; Rats; Substantia Nigra; Tissue Distribution | 1997 |
Endogenous opioid peptides in parasympathetic, sympathetic and sensory nerves in the guinea-pig heart.
Research has suggested that exogenous opioid substances can have direct effects on cardiac muscle or influence neurotransmitter release via presynaptic modulation of neuronal inputs to the heart. In the present study, multiple-labelling immunohistochemistry was employed to determine the distribution of endogenous opioid peptides within the guinea-pig heart. Approximately 40% of cardiac ganglion cells contained immunoreactivity for dynorphin A (1-8), dynorphin A (1-17) and dynorphin B whilst 20% displayed leu-enkephalin immunoreactivity. Different populations of opioid-containing ganglion cells were identified according to the co-existence of opioid immunoreactivity with immunoreactivity for somatostatin and neuropeptide Y. Immunoreactivity for prodynorphin-derived peptides was observed in many sympathetic axons in the heart and was also observed, though to a lesser extent, in sensory axons. Leu-enkephalin immunoreactivity was observed in occasional sympathetic and sensory axons. No immunoreactivity was observed for met-enkephalin-arg-gly-leu or for beta-endorphin. These results demonstrate that prodynorphin-derived peptides are present in parasympathetic, sympathetic and sensory nerves within the heart, but suggest that only the prodynorphin gene is expressed in guinea-pig cardiac nerves. This study has shown that endogenous opioid peptides are well placed to regulate cardiac function via both autonomic and sensory pathways. Topics: Adrenergic Fibers; Animals; Antibody Specificity; Axons; beta-Endorphin; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalin, Methionine; Enkephalins; Female; Ganglia, Autonomic; Guinea Pigs; Heart; Immunohistochemistry; Male; Neurons, Afferent; Opioid Peptides; Parasympathetic Fibers, Postganglionic; Protein Precursors | 1996 |
Endogenous opioids in frontal cortex of patients with Down syndrome.
The main purpose of this study was to investigate differences regarding endogenous opioids in post-mortem frontal cortex of adult patients with Down syndrome (DS), patients with Alzheimer disease (AD) and neurologically healthy persons, respectively, using specific radioimmunoassays. The results of this study show that there is an increase in the levels of leu-enkephalin and dynorphin A in the frontal cortex of patients with DS as compared to the control group. An almost identical increase was also observed when comparing patients with AD to controls. In conclusion, the results of this study suggest a relationship between elevated tissue levels of leuenkephalin and dynorphin A in cerebral cortex and cognitive impairments in patients with DS and AD. Topics: Aged; Alzheimer Disease; Down Syndrome; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalin, Methionine; Enkephalins; Female; Frontal Lobe; Gene Expression Regulation; Humans; Male; Middle Aged; Protein Precursors; Radioimmunoassay | 1996 |
The PC12 rat pheochromocytoma cell line expresses the prodynorphin gene and secretes the 8 kDa dynorphin product.
Most adrenal chromaffin cells synthesize opioids derived from proenkephalin but not from prodynorphin. However, human pheochromocytomas and the PC12 rat pheochromocytoma cell line synthesize dynorphins. The aim of this study was to confirm the presence of the authentic prodynorphin transcript and its dynorphin product in PC12 cells. We have found that the sequence of a 458 bp cDNA fragment derived from RT-PCR amplification of total PC12 RNA was in complete accordance with the published sequence of the equivalent region of the prodynorphin gene. It encodes the potent endogenous kappa opioid agonists alpha-neo-endorphin, dynorphin A and dynorphin B. Furthermore, immunoaffinity-purified PC12 cell extracts were subjected to RP-HPLC. Most of its IR-dynorphin eluted on a peak exhibiting the retention time of similarly treated rat anterior pituitary. The expression of the prodynorphin gene in pheochromocytomas can be explained as either the result of (a) the process of dedifferentiation of chromaffin cells to pheochromocytoma which may thus cause the expression of a previously unexpressed prodynorphin or that (b) those pheochromocytomas expressing the prodynorphin gene derive from the few, centrally located chromaffin cells, which express this gene even under normal conditions. Topics: Adrenal Medulla; Animals; Chromatography, Affinity; Chromatography, High Pressure Liquid; Dynorphins; Endorphins; Enkephalins; Gene Expression; PC12 Cells; Pheochromocytoma; Pituitary Gland; Polymerase Chain Reaction; Protein Precursors; Rats; Sequence Analysis | 1996 |
Modulation of neurotransmitter release by cholecystokinin in the neostriatum and substantia nigra of the rat: regional and receptor specificity.
The effect of cholecystokinin peptides on the release of dynorphin B, aspartate, glutamate, dopamine and GABA in the neostriatum and substantia nigra of the rat was investigated using in vivo microdialysis. Sulphated cholecystokinin-8S in the dialysis perfusate (1-100 microM) induced a concentration-dependent increase in extracellular dynorphin B and aspartate levels, both in the neostriatum and substantia nigra. Striatal dopamine levels were only increased by 100 microM of cholecystokinin-8S, while in the substantia nigra they were increased by 10-100 microM of cholecystokinin-8S. Extracellular GABA and glutamate levels were increased following 100 microM of cholecystokinin-8S only. Striatal cholecystokinin-8S administration also produced a significant increase in nigral dynorphin B levels. Local cholecystokinin-4 (100 microM) produced a moderate, but significant, increase of extracellular dynorphin B and aspartate levels in the neostriatum and substantia nigra. No effect was observed on the other neurotransmitters investigated. A 6-hydroxydopamine lesion of the nigrostriatal dopamine pathway did not affect the increases in dynorphin B and aspartate levels produced by local administration of cholecystokinin-8S. Basal extracellular GABA levels were increased significantly in both the neostriatum and substantia nigra ipsilateral to the lesion. Nigral glutamate and aspartate levels were also increased in the lesioned substantia nigra, but in the lesioned neostriatum aspartate levels were decreased. The cholecystokinin-B antagonist L-365,260 (20 mg/kg, s.c.), but not the cholecystokinin-A antagonist L-364,718 (devazepide; 20 mg/kg, s.c.), significantly inhibited the effect of cholecystokinin-8S on striatal dynorphin B and aspartate levels. In the substantia nigra, however, the effect of cholecystokinin-8S on dynorphin B and aspartate levels was inhibited to a similar extent by both L-365,260 and L-364,718. Pretreatment with L-364,718, but not with L-365.260, prevented the increase in nigral dopamine levels produced by nigral cholecystokinin-8S administration. Taken together, these results suggest that cholecystokinin-8S modulates dynorphin B and aspartate release in the neostriatum and substantia nigra of the rat via different receptor mechanisms. In the neostriatum, the effect of cholecystokinin-8S on dynorphin B and aspartate release is mediated via the cholecystokinin-B receptor subtype, while in the substantia nigra, cholecystokinin-8S modulates dynorphi Topics: 3,4-Dihydroxyphenylacetic Acid; Animals; Aspartic Acid; Cholecystokinin; Dopamine; Dynorphins; Endorphins; gamma-Aminobutyric Acid; Glutamic Acid; Homovanillic Acid; Kinetics; Male; Neostriatum; Neurons; Neurotransmitter Agents; Organ Specificity; Oxidopamine; Rats; Rats, Sprague-Dawley; Receptor, Cholecystokinin B; Receptors, Cholecystokinin; Substantia Nigra; Time Factors | 1996 |
Inhibition of dynorphin-converting enzymes prolongs the antinociceptive effect of intrathecally administered dynorphin in the mouse formalin test.
The effects of peptidase inhibitors on the antinociceptive induced by intrathecally (i.t.) administered by dynorphin A and dynorphin B in the mouse formalin test were examined. When administered i.t. 5 min before the injection of 0.5% formalin solution into the dorsal surface of a hindpaw, dynorphin A (0.5-2 nmol) and dynorphin B (2-8 nmol) produced a dose-dependent and significant reduction of the paw-licking response. Dynorphin A (2 nmol) and dynorphin B (8 nmol)-induced antinociception disappeared completely within 90 min and 60 min, respectively. p-Hydroxymercuribenzoate, a cysteine proteinase inhibitor, and phosphoramidon, and endopeptidase 24.11 inhibitor simultaneously administered with dynorphin A or dynorphin B. Significantly prolonged antinociception induced by both dynorphins. However, captopril, and angiotensin-converting enzyme inhibitor, bestatin (a general aminopeptidase inhibitor) and a serine proteinase inhibitor phenylmethanesulfonyl fluoride, were active. Dynorphin converting enzyme(s) transform dynorphin-related peptides to [Leu5]enkephalin and [Leu5]enkephalin-Arg6. Neither [Leu5]enkephalin nor [Leu5]enkephalin-Arg6, even at high dose (10 nmol), produced any antinociceptive effect. However, [Leu5[enkephalin-Arg6, but not [Leu5]enkephalin, produced a significant antinociceptive effect when co-administered with phosphoramidon. Therefore, the prolongation of the antinociception induced by both dynorphins in the presence of phosphoramidon, may be due to inhibition of [Leu5]enkephalin-Arg6 degradation. The present results indicate that dynorphin-converting enzyme(s) may be important enzyme(s) responsible for terminating dynorphin-A- and dynorphin-B-induced antinociception at the spinal cord level in mice. Topics: Analgesia; Animals; Cysteine Endopeptidases; Dynorphins; Endorphins; Injections, Spinal; Male; Mice; Pain Measurement | 1996 |
Effect of morphine on dynorphin B and GABA release in the basal ganglia of rats.
In vivo microdialysis was used to study the effects of systemic, as well as intracerebral administration of morphine and naloxone on dynorphin B release in neostriatum and substantia nigra of rats. The release of dopamine (DA), gamma-aminobutyric acid (GABA), glutamate (Glu) and aspartate (Asp) was also investigated. Systemic injection of morphine (1 mg/kg s.c.) induced long-lasting increases in extracellular dynorphin B and GABA levels in the substantia nigra, whereas DA, Glu and Asp levels, measured in the same region, were not significantly affected. No effect on striatal neurotransmitter levels was observed following systemic morphine administration. Local perfusion of the substantia nigra with morphine (100 microM) through the microdialysis probe also increased nigral dynorphin B and GABA levels. Perfusion of the neostriatum with morphine (100 microM) significantly increased GABA and dynorphin B levels in the ipsilateral substantia nigra, but no effect was observed locally. Naloxone blocked the effect of systemic morphine administration on nigral dynorphin B and GABA release, already at a dose of 0.2 mg/kg s.c. Naloxone alone, given either systemically (0.2-4 mg/kg s.c.) or intracerebrally (1-100 microM), did not affect dynorphin B or amino acid levels, either in neostriatum or in substantia nigra. However, naloxone produced a concentration-dependent increase in DA levels. The present results indicate that systemic morphine administration stimulates the release of dynorphin B in the substantia nigra, probably by activating the mu-subtype of opioid receptor, since the effect of morphine on nigral dynorphin B and GABA was antagonized by a low dose of naloxone. The increase in extracellular DA levels produced by high concentrations of naloxone, both in neostriatum and substantia nigra, indicates a disinhibitory effect of this drug on DA release, probably via a non-mu subtype of opioid receptors located on nigro-striatal DA neurones. Topics: Animals; Basal Ganglia; Drug Combinations; Dynorphins; Endorphins; gamma-Aminobutyric Acid; Injections; Injections, Subcutaneous; Male; Microdialysis; Morphine; Naloxone; Narcotic Antagonists; Perfusion; Rats; Rats, Sprague-Dawley | 1996 |
Purification and characterization of a secreted arginine-specific dibasic cleaving enzyme from EL-4 cells.
A secreted dibasic cleaving peptidase capable of converting dynorphins into Leu-enkephalin-Arg6 was purified from the medium of EL-4 mouse thymoma cells. The enzyme is a novel metalloendopeptidase with a neutral pH optimum (6.9) and a molecular weight of approximately 130 000. The dibasic cleaving enzyme was completely inhibited in the presence of 20-50 mM amine buffers, 0.1 mM EDTA, 0.5 mM 1,10-phenanthroline, 0.5 mM N-ethylmaleimide, and 1mM DTNB. Unlike the Kex2 family of proteases, Ca2+ did not activate the endopeptidase, but high concentrations (1 mM) of metal ions such as Cu2+, Ni2+, Zn2+, and Co2+ completely inhibited the enzyme. Inhibition was not seen with 0.2 mM TLCK, 1 mM DTT, and 1 mM PMSF. The enzyme will cleave Arg-Arg and Arg-Lys bonds, but not Lys-Arg or Lys-Lys bonds in identical environments, and no aminopeptidase or carboxypeptidase activity was seen. The size of the substrate does not seem to be a determining factor, since dynorphin A(1-12) is cleaved at a rate similar to prodynorphin B(228-256) containing 29 amino acids. The identity of the residues on either side of the cleavage site influences the rate of processing, as noted by different rates of cleavage for the same size peptides dynorphin A(1-13) vs dynorphin A(1-9) vs beta-neoendorphin. The presence of proline in the P3' (alpha-neoendorphin), P4' (dynorphin A(1-11)), or P5' (bovine adrenal medulla dodecapeptide) position does not prevent cleavage, but neurotensin and its (1-11) fragment containing both P2 and P2' proline residues are not cleaved. Topics: Amino Acid Sequence; Animals; Arginine; Cations, Divalent; Cysteine Endopeptidases; Dynorphins; Endorphins; Enzyme Inhibitors; Hydrogen-Ion Concentration; Metalloendopeptidases; Metals; Mice; Molecular Sequence Data; Protease Inhibitors; Protein Precursors; Substrate Specificity; Thymoma; Tromethamine; Tumor Cells, Cultured | 1995 |
Purification and characterization of a dynorphin-processing endopeptidase.
Dynorphin B (Dyn B-13, also known as rimorphin) is generated from Dyn B-29 (leumorphin) by the cleavage at a single Arg residue. An enzymatic activity capable of processing at this monobasic site has been previously reported in neurosecretory vesicles of the bovine pituitary and pituitary-derived cell lines. This enzyme termed "the dynorphin-converting enzyme" (DCE) has been purified to apparent homogeneity from the neurointermediate lobe of the bovine pituitary using hydrophobic chromatography on phenyl-Sepharose, preparative isoelectrofocusing in a granulated gel between pH 4 to 6.5, and non-denaturing electrophoresis on 5% polyacrylamide gel. DCE exhibits a pI of about 5.1 and a molecular mass of about 54 kDa under reducing conditions. DCE is a metallopeptidase and exhibits a neutral pH optimum. Specific Inhibitors of soluble metallopeptidases such as enkephalinase (EC 3.4.24.11) or enkephalin generating neutral endopeptidase (EC 3.4.24.15) do not inhibit DCE activity indicating that DCE is distinct from these two enzymes. Cleavage site determination with matrix-assisted laser desorption ionization time of flight (MALDITOF) mass spectrometry shows that DCE cleaves the Dyn B-29 N terminus to the Arg14 generating Dyn B-13 and Dyn B-(14-29). Among other peptides derived from Dyn B-29, DCE cleaves only those peptides that fit the predicted "consensus motif" for monobasic processing. These data are consistent with a broader role for the dynorphin converting enzyme in the biosynthesis of many peptide hormones and neuropeptides by processing at monobasic sites. Topics: Amino Acid Sequence; Animals; Binding Sites; Cattle; Dynorphins; Endorphins; Enkephalins; Isoelectric Point; Kinetics; Molecular Sequence Data; Molecular Weight; Peptide Fragments; Pituitary Gland; Protein Precursors; Protein Processing, Post-Translational; Serine Endopeptidases; Substrate Specificity | 1995 |
The effects of morphine treatment and morphine withdrawal on the dynorphin and enkephalin systems in Sprague-Dawley rats.
The effect of morphine tolerance and withdrawal on prodynorphin peptides was studied in relevant brain areas and in the pituitary gland of male Sprague-Dawley rats, and compared with effects on the proenkephalin-derived peptide Met-enkephalin. After 8 days of morphine injections (twice daily), dynorphin A and B levels increased in the nucleus accumbens and dynorphin A levels increased also in the striatum. Morphine treatment increased striatal Met-enkephalin. Leu-enkephalinArg6 levels were reduced in the ventral tegmental area (VTA). Morphine-treated rats had very low Leu-enkephalinArg6 levels in the hippocampus as compared to saline control rats. Comparison of the relative amounts of dynorphin peptides and the shorter prodynorphin-derived peptides, Leu-enkephalinArg6 and Leu-enkephalin, revealed a relative increase in dynorphin peptides versus shorter fragments in the nucleus accumbens, VTA and hippocampus. Morphine-tolerant rats had lower levels of dynorphin A in both lobes of the pituitary gland, whereas hypothalamic dynorphin levels were unaffected by morphine. Leu-enkephalinArg6 levels were reduced in the hypothalamus, but not changed in the pituitary gland. Naloxone-precipitated withdrawal accentuated the increase in dynorphin A and B levels in the accumbens and dynorphin A levels in the striatum, while inducing an increase in enkephalin levels in the accumbens and Met-enkephalin in the VTA. In the hippocampus, Leu-enkephalinArg6 levels remained low in the withdrawal state. The low dynorphin levels in the anterior part of the pituitary gland were reversed by naloxone, whereas the low dynorphin A levels in the neurointermediate lobe were 0ven lower in the withdrawal state.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: Analysis of Variance; Animals; Drug Tolerance; Dynorphins; Endorphins; Enkephalin, Leucine; Male; Morphine; Naloxone; Peptides; Rats; Rats, Sprague-Dawley; Substance Withdrawal Syndrome | 1995 |
Processing of prodynorphin-derived peptides in striatal extracts. Identification by electrospray ionization mass spectrometry linked to size-exclusion chromatography.
Proteolytic processing of prodynorphin-derived peptides in rat brain was studied with the help of high performance size exclusion chromatography (SEC) connected to electrospray ionization mass spectrometry. Extracts from rat striatum were incubated with individual synthetic dynorphin peptides. Dynorphin A was the most resistant to proteolytic cleavage, converting slowly to Leu-enkephalin (0.3 pmol/min), whereas dynorphin B was processed to this pentapeptide at a 10(4)-fold higher rate. Minor cleavage was also observed between Arg6-Arg7. Alphaneoendorphin was also rapidly metabolized to Leu-enkephalin (6 nmol/min) and, to a lesser extent, to Leu-enkephalinArg6. This new strategy for studying peptidases can easily be adapted to identification of components present in body fluids. Topics: Animals; Anti-Bacterial Agents; Chromatography, Gel; Corpus Striatum; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalins; Male; Mass Spectrometry; Peptide Hydrolases; Peptides; Protein Precursors; Rats; Sensitivity and Specificity; Tissue Extracts | 1995 |
Phorbol ester regulation of opioid peptide gene expression in myocardial cells. Role of nuclear protein kinase.
Opioid peptide gene expression was characterized in adult rat ventricular cardiac myocytes that had been cultured in the absence or the presence of phorbol 12-myristate 13-acetate. The phorbol ester induced a concentration- and time-dependent increase of prodynorphin mRNA, the maximal effect being reached after 4 h of treatment. The increase in mRNA expression was suppressed by incubation of cardiomyocytes with staurosporine, a putative protein kinase C inhibitor, and was not observed when the cells were cultured in the presence of the inactive phorbol ester 4 alpha-phorbol 12,13-didecanoate. Incubation of cardiac myocytes with phorbol 12-myristate 13-acetate also elicited a specific and staurosporine-sensitive increase in immunoreactive dynorphin B, a biologically active end product of the precursor, both in the myocardial cells and in the culture medium. In vitro run-off transcription assays indicated that transcription of the prodynorphin gene was increased both in nuclei isolated from phorbol ester-treated myocytes and in nuclei isolated from control cells and then exposed to phorbol 12-myristate 13-acetate. No transcriptional effect was observed when cardiac myocytes or isolated nuclei where exposed to 4 alpha-phorbol 12,13-didecanoate. The phorbol ester-induced increase in prodynorphin gene transcription was prevented by pretreatment of myocytes or isolated nuclei with staurosporine, suggesting that myocardial opioid gene expression may be regulated by nuclear protein kinase C. In this regard, cardiac myocytes expressed protein kinase C-alpha, -delta, -epsilon, and -zeta, as shown by immunoblotting. Only protein kinase C-delta and protein kinase C-epsilon were expressed in nuclei that have been isolated from control myocytes, suggesting that these two isotypes of the enzyme may be part of the signal transduction pathway involved in the effect elicited by the phorbol ester on opioid gene transcription in isolated nuclei. The incubation of myocardial nuclei isolated from control cells in the presence of a protein kinase C activator induced the phosphorylation of the myristoylated alanine-rich protein kinase C substrate peptide, a specific fluorescent substrate of the enzyme. The possibility that prodynorphin gene expression may control the heart function through autocrine or paracrine mechanisms is discussed. Topics: Alkaloids; Animals; Cell Nucleus; Cells, Cultured; Dose-Response Relationship, Drug; Dynorphins; Endorphins; Enkephalins; Enzyme Inhibitors; Gene Expression; Heart Ventricles; Isoenzymes; Kinetics; Male; Myocardium; Phorbol Esters; Protein Kinase C; Protein Precursors; Rats; Rats, Wistar; Staurosporine; Tetradecanoylphorbol Acetate; Transcription, Genetic | 1995 |
[Structural organization of rimorphin and its synthetic analogs].
The spatial structure and conformations of rimorphin were investigated using theoretical conformational analysis. The spatial organization of the peptide can be described by a set of 11 low-energy conformations of the backbone. By solving the reverse conformational problem, a number of modified amino acid sequences ([Ala2], [Ala3], [MePhe9], [MeLys10], [MeVal11], and [MeVal12]-analogs of rimorphin) were determined that have spatial structures corresponding to the set of low-energy conformations and should, therefore, possess physiological activity. Topics: Amino Acid Sequence; Dynorphins; Endorphins; Molecular Sequence Data; Protein Conformation; Structure-Activity Relationship | 1995 |
Occurrence of secretogranin II in the prolactin-immunoreactive neurons of the rat lateral hypothalamus: an in situ hybridization and immunocytochemical study.
The occurrence of secretogranin II in a neuron population of the rat lateral hypothalamus specifically detected by an anti-serum to ovine prolactin was examined. As this population was previously reported to synthesize dynorphin, the distribution of neurons recognized by ovine prolactin-, dynorphin B- and secretogranin II anti-sera was investigated on adjacent sections of hypothalami. The prolactin immunoreactive neurons were the only cells in the lateral hypothalamus to be stained by secretogranin II anti-serum. Moreover, coupling immunocytochemical detection and in situ hybridization with an oligonucleotide probe complementary to secretogranin II mRNA showed that these neurons expressed the secretogranin II gene. These new findings should help to study the physiological role of the prolactin immunoreactive neurons of the lateral hypothalamus. Topics: Animals; Chromogranins; Dynorphins; Endorphins; Hypothalamus; Immunohistochemistry; In Situ Hybridization; Male; Neurons; Peptides; Prolactin; Proteins; Rats; Rats, Sprague-Dawley; RNA, Messenger | 1995 |
Basic peptides can be imported into yeast mitochondria by two distinct targeting pathways. Involvement of the peptide-sensitive channel of the outer membrane.
The interaction of several basic peptides with yeast mitochondria has been analyzed. The peptides were selected for their ability to block a cationic channel of the outer membrane, the peptide-sensitive channel. These peptides possess common characteristics, such as a net positive charge superior to 2 and the capacity to form amphiphilic structures. They can be divided into two classes as follows: peptides of class I derived from mitochondrial signal peptides, such as the presequence of cytochrome c oxidase subunit IV, e.g. pCyt OX IV (1-12) Y; and peptides of class II unrelated to the mitochondria, such as dynorphin B (1-13). Class I peptides inhibited the translocation of a chimeric protein, cytochrome b2-DHFR, into the mitochondrial matrix, whereas peptides of class II failed to inhibit this import. Experiments with iodinated pCyt OX IV (1-12) Y and dynorphin B (1-13) showed, however, that both types of peptides were imported into yeast mitochondria in vitro and subsequently degraded. At 30 degrees C, two import mechanisms could be distinguished; the mitochondrial presequences (class I) were translocated into the matrix in a temperature- and potential-sensitive manner, probably along the general import pathway, while class II dynorphin B (1-13) was imported into the intermembrane space by a process that was neither temperature- nor potential-sensitive. At 0 degree C, both peptides were imported in a class II manner. The class II characteristics suggested the existence of a direct pathway into the intermembrane space, which may be associated with the peptide-sensitive channel. This hypothesis is substantiated by the competition for the import into the mitochondria between peptides of the two classes. The import of pCyt OX (1-12) Y was inhibited at 30 degrees C only by peptides of class I, IV whereas, at 0 degree C, this import was also inhibited by peptides of class II. Import of peptides of the latter class was inhibited by peptides of the two classes both at 0 degree C and 30 degrees C. Topics: Amino Acid Sequence; Binding Sites; Biological Transport; Dynorphins; Electron Transport Complex IV; Endorphins; Hydrogen-Ion Concentration; Intracellular Membranes; Ion Channels; L-Lactate Dehydrogenase; L-Lactate Dehydrogenase (Cytochrome); Membrane Potentials; Mitochondria; Molecular Sequence Data; Peptide Fragments; Peptides; Porins; Recombinant Fusion Proteins; Saccharomyces cerevisiae; Temperature; Tetrahydrofolate Dehydrogenase; Trypsin | 1994 |
Dynorphin B is present in sensory and parasympathetic nerves innervating pial arteries.
Dynorphin B (dyn B) in trigeminal ganglion cells and in perivascular nerve fibers in pial arteries was demonstrated in rat, guinea-pig, and monkey by immunohistochemistry. The pathway from the trigeminal ganglion, which runs via the nasociliary nerve and ethmoidal foramen to the pial arteries, was shown in rat by retrograde tracer technique and nerve section. In the guinea-pig the peptide was demonstrated to coexist with substance P and calcitonin gene-related peptide in neurons of the trigeminal ganglion and pial nerve fibers, i.e., it was present in cerebrovascular sensory nerves with primarily nociceptive function. Another finding in guinea-pig was a coexistence of dyn B with vasoactive intestinal polypeptide in the pial nerve fibers and neurons of the sphenopalatine ganglion, indicating a presence also in parasympathetic nerves to the cerebral vessels. No vasomotor effect of dyn B could be detected in isolated segments of rat pial arteries, which rules out a direct postsynaptic effect on vascular tone. The peptide did not display a prejunctional modulatory action on the adrenergic nerves present in the vessels. The function of dyn B in the cerebrovascular nerves is discussed. Topics: Animals; Calcitonin Gene-Related Peptide; Dynorphins; Endorphins; Ganglia, Parasympathetic; Guinea Pigs; Male; Neurons, Afferent; Norepinephrine; Parasympathetic Nervous System; Pia Mater; Rats; Rats, Sprague-Dawley; Saimiri; Species Specificity; Substance P; Sympathectomy; Trigeminal Ganglion; Trigeminal Nerve Injuries; Vascular Resistance; Vasoactive Intestinal Peptide | 1994 |
An in vitro polysome display system for identifying ligands from very large peptide libraries.
We have used an in vitro protein synthesis system to construct a very large library of peptides displayed on polysomes. A pool of DNA sequences encoding 10(12) random decapeptides was incubated in an Escherichia coli S30 coupled transcription/translation system. Polysomes were isolated and screened by affinity selection of the nascent peptides on an immobilized monoclonal antibody specific for the peptide dynorphin B. The mRNA from the enriched pool of polysomes was recovered, copied into cDNA, and amplified by the polymerase chain reaction (PCR) to produce template for the next round of in vitro synthesis and selection. A portion of the amplified template from each round was cloned into a filamentous phagemid vector to determine the specificity of peptide binding by phage ELISA and to sequence the DNA. After four rounds of affinity selection, the majority of clones encoded peptides that bound specifically to the antibody and contained a consensus sequence that is similar to the known epitope for the antibody. Synthetic peptides corresponding to several of these sequences have binding affinities ranging from 7 to 140 nM. The in vitro system described here has the potential to screen peptide libraries that are three to six orders of magnitude larger than current biological peptide display systems. Topics: Amino Acid Sequence; Antibodies, Monoclonal; Base Sequence; DNA Primers; Dynorphins; Endorphins; Epitopes; Escherichia coli; In Vitro Techniques; Ligands; Molecular Sequence Data; Peptides; Polyribosomes; Protein Binding; Recombinant Fusion Proteins; Structure-Activity Relationship | 1994 |
Dentate granule cells as a central cardioregulatory site in the rat.
Dentate granule cells can be selectively destroyed by intrahippocampal injections of colchicine. This study evaluates the consequences of granule cell destruction on blood pressure regulation in the normotensive Wistar-Kyoto (WKY) and spontaneously hypertensive rat (SHR). Bilateral destruction of dentate granule cells at 6 weeks of age produced a significant increase in blood pressure in the WKY that lasted for approximately 3 weeks, and a biphasic effect (increase then decrease) in the SHR that resulted in a significant hypotensive period that persisted for 6 weeks. Granule cell destruction at 11 weeks produced a maximal hypertension in the SHR that preceded age-matched controls by 4 weeks, but produced only a small transient increase in WKY blood pressure. Dentate granule cells are the exclusive source of prodynorphin-derived peptides in the hippocampal formation and their synthesis is regulated by glucocorticoids. Evidence suggests glucocorticoids may be involved in the regulation of blood pressure and hypertension. We determined that chronic high levels of corticosterone significantly reduced hippocampal dynorphin B levels in normotensive Sprague-Dawley rats. In addition, we confirmed that naive SHRs also contain significantly lower levels of hippocampal dynorphin B. These results suggest (i) that dentate granule cells represent a discrete neural site that may exert a tonic inhibitory influence on blood pressure, (ii) that dentate granule cells are not required for the full expression of hypertension in the SHR, and (iii) that chronic high levels of corticosterone can reduce dynorphin B levels in the dentate granule cells of normotensive rats. Topics: Aging; Animals; Blood Pressure; Colchicine; Corticosterone; Dynorphins; Endorphins; Hippocampus; Hypertension; Male; Rats; Rats, Inbred SHR; Rats, Inbred WKY; Rats, Sprague-Dawley | 1994 |
Prodynorphin-derived peptide expression in primate cortex and striatum.
The distributions of four prodynorphin-derived peptides, dynorphin A (1-17), dynorphin A (1-8), dynorphin B, and alpha-neo-endorphin were determined in 10 cortical regions and the striatum of the old world monkey (Macaca nemestrina). alpha-neo-endorphin was the most abundant peptide in both cortex and striatum. The concentrations of all four peptides were significantly greater in the striatum compared to the cortex. In general, concentrations of each peptide tended to be higher in allocortex than in neocortex. Possible inter- and intradomain processing differences, as estimated by ratios of these peptides, did not vary within cortex, but the intradomain peptide ratio, dyn A (1-17)/dyn A (1-8), was significantly greater in cortex than in striatum. These results indicate that prodynorphin is, in some ways, uniquely processed in the primate. Particularly unusual is the relatively low abundance of prodynorphin-derived products in the cortex, in the face of moderately high levels of kappa opiate receptor expression. Topics: Animals; Cerebral Cortex; Corpus Striatum; Dynorphins; Endorphins; Enkephalins; Macaca nemestrina; Peptide Fragments; Protein Precursors; Tissue Distribution | 1994 |
Modulation of striatal aspartate and dynorphin B release by cholecystokinin (CCK-8) studied in vivo with microdialysis.
Sulphated cholecystokinin-8 (CCK-8) given into the neostriatum of the rat by in vivo microdialysis produced a concentration-dependent (1-100 microM) increase in extracellular aspartate (Asp) and dynorphin B (Dyn B), but not in glutamate, GABA or dopamine levels. The increase in Asp levels produced by 10 microM CCK-8 was approximately 10 fold and was inhibited (approximately 50%) by the CCKB antagonist L-365,260 (20 mg kg-1, i.p.), while the increase in Dyn B (approximately 2 fold) was totally abolished. Both increases were inhibited (approximately 50%) by local infusion of 10 microM of tetrodotoxin (TTX). Thus, CCK exerts modulatory effects in the basal ganglia, possibly by interacting with local neostriatal neurones releasing Asp, and with Dyn B-containing neurones projecting to the pars reticulata of the substantia nigra. Topics: Animals; Aspartic Acid; Benzodiazepinones; Corpus Striatum; Dynorphins; Endorphins; Male; Microdialysis; Phenylurea Compounds; Rats; Rats, Sprague-Dawley; Receptors, Cholecystokinin; Sincalide; Tetrodotoxin | 1994 |
The striatonigral dynorphin pathway of the rat studied with in vivo microdialysis--I. Effects of K(+)-depolarization, lesions and peptidase inhibition.
Extracellular levels of dynorphin B were analysed with in vivo microdialysis in the neostriatum and substantia nigra of halothane-anaesthetized rats. Dopamine and its metabolites, 3,4-dihydroxyphenyl-acetic acid and homovanillic acid, as well as GABA were simultaneously monitored. Chromatographic analysis revealed that the dynorphin B-like immunoreactivity measured in perfusates collected under basal and K(+)-depolarizing conditions co-eluted with synthetic dynorphin B. Dynorphin B, GABA and dopamine levels were Ca(2+)-dependently increased by K(+)-depolarization, while 3,4-dihydroxyphenylacetic acid and homovanillic acid levels were decreased. Dopamine and its metabolites, but not dynorphin B or GABA levels, were significantly decreased after a unilateral 6-hydroxydopamine injection into the left medial forebrain bundle. In contrast, following a unilateral injection of ibotenic acid into the striatum, dynorphin B and GABA levels were decreased by > 50% in striatum and substantia nigra on the lesioned side, whereas no significant changes were observed in basal dopamine levels. The inclusion of the peptidase inhibitor captopril (50-500 microM) into the nigral perfusion medium produced a concentration-dependent increase in nigral extracellular levels of dynorphin B. In the striatum, a delayed increase in dynorphin B and GABA levels could be observed following the nigral captopril administration, but this effect was not concentration-dependent. Thus, we demonstrate that extracellular levels of dynorphin B, dopamine and GABA can simultaneously be monitored with in vivo microdialysis. Extracellular dynorphin B appears to originate from neurons, since the levels were (i) increased in a Ca(2+)-dependent manner by K(+)-depolarization, and (ii) decreased by a selective lesion of the striatum, known to contain cell bodies of dynorphin neurons in the striatonigral pathway. Furthermore, (iii) the increase in nigral dynorphin B levels by peptidase inhibition suggests the presence of clearance mechanisms for the released dynorphin peptides. Topics: Animals; Captopril; Chromatography; Corpus Striatum; Dopamine; Dynorphins; Electrophysiology; Endorphins; Extracellular Space; gamma-Aminobutyric Acid; Male; Microdialysis; Neural Pathways; Potassium; Rats; Rats, Sprague-Dawley; Substantia Nigra | 1994 |
The striatonigral dynorphin pathway of the rat studied with in vivo microdialysis--II. Effects of dopamine D1 and D2 receptor agonists.
In vivo microdialysis was used to study the effect of intracerebral administration of dopamine agonists on dynorphin B release in the striatum and substantia nigra of rats. The release of dopamine and GABA was also investigated. Administration of the dopamine D1 agonist SKF 38393 (10-100 microM) into the striatum increased extracellular dynorphin B and GABA levels in the ipsilateral substantia nigra, in a concentration-dependent manner. After a short-lasting increase, nigral dopamine levels were significantly decreased after the highest concentration of striatal SKF 38393. An increase in striatal dynorphin B, GABA and dopamine levels was also observed. When SKF 38393 (10 microM) was administered into the substantia nigra, nigral dynorphin B and GABA, but not dopamine levels increased. No significant effects were observed on striatal levels. Administration of the dopamine D2 agonist, quinpirole (100 microM), into the striatum decreased dopamine levels in both striatum and substantia nigra, while no effect was observed on striatal or nigral dynorphin B and GABA levels. Quinpirole (10-100 microM) given into the substantia nigra, decreased striatal dopamine levels in a concentration manner. In the nigra, a short-lasting increase in dopamine levels was observed following the highest concentration of nigral quinpirole (100 microM). The effect was followed by a decrease in dopamine levels. No significant effects were observed on striatal or nigral dynorphin B and GABA levels. The results show that stimulation of D1 receptors in striatum and substantia nigra leads to activation of the striatonigral dynorphin pathway. A parallel effect could also be seen on nigral GABA release.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: Animals; Corpus Striatum; Dopamine; Dynorphins; Endorphins; gamma-Aminobutyric Acid; Male; Microdialysis; Neural Pathways; Rats; Rats, Sprague-Dawley; Receptors, Dopamine D1; Receptors, Dopamine D2; Substantia Nigra | 1994 |
Voltage-dependent effects of opioid peptides on hippocampal CA3 pyramidal neurons in vitro.
Opioid peptides, and especially the dynorphins, have been localized to several circuits in the CA3 hippocampal region, yet electrophysiological studies often find mixed effects of opiates on the excitability of CA3 neurons. Reasoning that these mixed effects might involve voltage-dependent actions, we tested the effect of several opiates on CA3 pyramidal neurons using single-electrode voltage-clamp recording in a slice preparation of rat hippocampus. In most CA3 neurons, the voltage-dependent K+ current known as the M-current (IM) was uniquely sensitive to the opioid peptides, with the direction of response dependent upon the opiate type and concentration. Thus, an opiate selective for kappa receptors, U-50,488H, significantly augmented IM. The kappa-selective agonists dynorphin A and dynorphin B, which exist in mossy fiber afferents to CA3 pyramidal neurons, also markedly augmented IM at low concentrations (20-100 nM). By contrast, dynorphin A at higher concentrations (1-1.5 microM) often reduced IM. Similarly, several opiates [e.g., D-Ala2,D-Leu5-enkephalin: (DADL), [D-Pen2,5]-enkephalin (DPDPE)] known to act on the delta receptor subtypes reduced the M-current, with partial reversal of this effect by naloxone. Neither the selective mu-receptor agonist [D-Ala2, NMe-Phe4, Gly-ol]-enkephalin (DAMGO) nor the nonopioid fragment of dynorphin, des-Tyr-dynorphin, consistently altered IM. These opiate effects on IM were accompanied by changes in conductance and holding current consistent with their respective effects on IM. Dynorphin A did not measurably affect the Q-current, a conductance known to contribute to inward rectification in hippocampal pyramidal neurons. The opiate effects on IM were not altered by pretreatment with Cs+ (which blocks IQ) or Ca2+ channel blockers. The opposing effects of the dynorphins (both A and B) and DADL on IM were antagonized by naloxone (1-3 microM), and the dynorphin-induced augmentations of IM were usually reversed by the kappa receptor antagonist norbinaltorphimine. These results suggest that the opiates can have opposing effects on the same voltage-dependent K+ channel type (the M channel) in the rat CA3 pyramidal neuron, with the direction of the response depending on which receptor subtype is activated. These data not only help explain the mixed effects of opiates seen in other studies, but also suggest a potential postsynaptic function for the endogenous opiates contained in the CA3 mossy fibers. Topics: 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer; Animals; Barium Compounds; Cadmium; Carbachol; Chlorides; Computer Simulation; Dynorphins; Electrophysiology; Endorphins; Enkephalin, D-Penicillamine (2,5)-; Enkephalin, Leucine-2-Alanine; Enkephalins; Hippocampus; In Vitro Techniques; Male; Membrane Potentials; Models, Neurological; Naloxone; Narcotics; Neurons; Pyramidal Tracts; Pyrrolidines; Rats; Rats, Sprague-Dawley | 1994 |
Facilitation of self-stimulation of ventral tegmentum by microinjection of opioid receptor subtype agonists.
Intracranial self-stimulation (ICSS) evoked from the ventral tegmental area-substantia nigra (VTA-SN) and lateral hypothalamus-medial forebrain bundle (LH-MFB) was assessed following microinjections of mu (Tyr-D-Ala2-N-Me-Phe4-Gly5ol: DAGO), delta-(D-Ala2, D-Met5)-enkephalin: DADME) or kappa (Dynorphin-B or Rimorphin) opioid receptor subtype agonists or saline into either VTA-SN or LH-MFB. The current intensity was fixed at an optimum level to obtain 60-70% of the maximum asymptotic response rate. DAGO (5 micrograms/0.5 microliters), DADME (2 micrograms/0.5 microliters) or Dynorphin B (0.5 microgram/0.5 microliters) injected into VTA-SN facilitated the self-stimulation rates of VTA-SN by 27%, 32%, and 59%, respectively. These microinjections did not alter the self-stimulation of LH-MFB when effects of these injections were still persisting in VTA-SN. Similar doses of these opioid receptor agonists injected into LH-MFB had no significant effect on the self-stimulation rates of either LH-MFB or VTA-SN. The facilitatory effects of DADME were completely abolished by naloxone (30 mg/kg IP). Taken together, these results suggest that all three opioid receptor subtypes of ventral tegmentum and not of lateral hypothalamus are involved in the electrically evoked self-stimulation of VTA-SN. Topics: Analgesics; Animals; Brain Mapping; Dose-Response Relationship, Drug; Dynorphins; Electric Stimulation; Endorphins; Enkephalin, Ala(2)-MePhe(4)-Gly(5)-; Enkephalin, Methionine; Enkephalins; Hypothalamic Area, Lateral; Male; Medial Forebrain Bundle; Motivation; Naloxone; Neural Pathways; Rats; Rats, Wistar; Receptors, Opioid; Self Stimulation; Substantia Nigra; Tegmentum Mesencephali; Ventral Tegmental Area | 1994 |
Evidence for the expression of dynorphin gene in the prolactin-immunoreactive neurons of the rat lateral hypothalamus.
Dynorphin B (DYN B) immunoreactivity was recently reported in a population of prolactin (PRL)-immunoreactive neurons of the rat lateral hypothalamus (LH). By coupling immunocytochemistry and in situ hybridization using two synthetic oligonucleotide probes complementary to DYN mRNA, a hybridization signal was observed over the neurons of the LH exhibiting both DYN- and PRL-like immunoreactivities. Our results clearly demonstrate that these neurons contain the mRNA encoding preproDYN and are able to synthesize authentic DYN B in colocalization with a peptide related to PRL. Topics: Animals; Autoradiography; Base Sequence; Dynorphins; Endorphins; Gene Expression; Histocytochemistry; Hypothalamic Area, Lateral; In Situ Hybridization; Male; Molecular Sequence Data; Neurons; Oligonucleotide Probes; Prolactin; Rats; RNA, Messenger | 1994 |
[The serotonin-dependent suppression of the immune response by kappa-opiate receptors].
The modulating effect of the kappa-opiate agonist, rimorphin, on the immune response was studied. This opioid in all the doses tested (1, 10 and 100 mkg/kg) was found to cause inhibition of the immune reaction as compared to the control CBA mice which received saline only. The dissection of the pituitary stalk prevented the inhibitory effect of rimorphin on the immunity. The opioid-induced immunosuppression was shown to be due to interaction with the serotoninergic system since it had no effect after preliminary blockade of S-2 serotonin postsynaptic receptors with cyproheptadine. Topics: Animals; Cyproheptadine; Dose-Response Relationship, Drug; Dynorphins; Endorphins; Haloperidol; Immune Tolerance; Immunization; Mice; Mice, Inbred CBA; Pituitary Gland; Receptors, Opioid, kappa; Receptors, Serotonin; Serotonin | 1993 |
Transduction of a protein kinase C-generated signal into the long-lasting facilitation of glutamate release.
The present study investigated the delayed and persistent effects of 4 beta-phorbol 12,13-dibutyrate (PDBu) on the K(+)-evoked release of endogenous glutamate and dynorphin B-like immunoreactivity from a subcellular fraction (P3) that is enriched in hippocampal mossy fiber synaptosomes. It is demonstrated that the alpha, beta, gamma, epsilon, and zeta isoforms of protein kinase C (PKC) are present in the P3 fraction obtained using the guinea pig hippocampus as starting tissue. The K(+)-evoked release of glutamate was found to be selectively enhanced when mossy fiber-enriched synaptosomes were preincubated with PDBu for 15 minutes and extensively washed with a PDBu-free medium. The persistent enhancement of glutamate release observed under this condition was not reversed by the protein kinase inhibitor staurosporine and was desensitized to the potentiating effects of an acute reexposure to PDBu. The overall content and activity of PKC was not substantially altered during the initial 15 minutes of treatment with PDBu (10 microM). More prolonged pretreatments with PDBu altered the substrate specificity of PKC and decreased the content of all PKC isoforms, but did not reverse the facilitation of glutamate release that followed preincubation in the presence of PDBu. It is concluded that the persistent activation of PKC enhances K(+)-evoked glutamate release from hippocampal mossy fiber-enriched synaptosomes and that, once established, this presynaptic facilitation is sustained by a process that is no longer directly dependent on continued PKC phosphotransferase activity. Topics: Animals; Dynorphins; Endorphins; Glutamates; Glutamic Acid; Guinea Pigs; Hippocampus; Isoenzymes; Male; Nerve Fibers; Phorbol 12,13-Dibutyrate; Phosphorylation; Potassium; Protein Kinase C; Reaction Time; Signal Transduction; Subcellular Fractions; Synaptosomes | 1993 |
Apomorphine-susceptible and apomorphine-unsusceptible Wistar rats differ in novelty-induced changes in hippocampal dynorphin B expression and two-way active avoidance: a new key in the search for the role of the hippocampal-accumbens axis.
The present study examines two characteristic traits of the hippocampus in apomorphine-susceptible (APO-SUS) and apomorphine-unsusceptible (APO-UNSUS) Wistar rat lines. Since hippocampal mossy fibers contain among others dynorphin B as transmitter, a radioimmunoassay was used to analyze the hippocampal dynorphin B expression in response to novelty in these lines. Dynorphin B expression at the end of the baseline condition was greater in APO-SUS rats than in APO-UNSUS rats, while exposure to novelty decreased and increased the dynorphin B expression in APO-SUS and APO-UNSUS rats, respectively. These interline differences in dynorphin B expression could be due to (a) an interline difference in the size of the mossy fiber terminal fields, (b) an interline difference in the regulation of the firing rate of mossy fibers by corticosteroids, and/or (c) an interline difference in the release of corticosteroids in response to novelty. Since the size of the mossy fiber infra/intrapyramidal terminal field is inversely related to two-way active avoidance performance, APO-SUS and APO-UNSUS rats (n = 9 per line) were given this task: APO-UNSUS rats performed much better than APO-SUS rats. It is concluded that the neurochemical and behavioural function of the hippocampus significantly differs between lines. Given the already known interline differences in the function of the nucleus accumbens, the present results provide a new avenue in search for the functional relationship between the hippocampus and the nucleus accumbens. Topics: Animals; Apomorphine; Arousal; Avoidance Learning; Dominance, Cerebral; Dynorphins; Endorphins; Gene Expression; Hippocampus; Male; Motor Activity; Neural Pathways; Nucleus Accumbens; Rats; Rats, Wistar; Social Environment; Stereotyped Behavior | 1993 |
Expression of prodynorphin-derived peptides and mRNA in guinea-pig cortex.
The distributions and extent of processing of four prodynorphin-derived peptides (dynorphin A (1-17), dynorphin A (1-8), dynorphin B, and alpha-neoendorphin) were determined in ten regions of the cortex as well as in the striatum of the guinea-pig. There were significant differences between concentrations of these peptides in most cortical regions, with alpha-neoendorphin being several times more abundant than the other peptides, and dynorphin A (1-17) being present in the least amount. There were significant between-region differences in concentration for each peptide, although most regions had concentrations similar to those seen in the striatum. Concentrations of each peptide tended to be higher in piriform, entorhinal, motor, and auditory cortex than in other cortical regions. The extent of processing of prodynorphin varied across cortical regions as well, primarily due to the extent of processing to alpha-neoendorphin. Prodynorphin mRNA levels were not significantly different between cortical regions or from the amount observed in the striatum. Although specific regional variation exists, it appears that in general prodynorphin is expressed and processed in a similar manner in the cortex as in the striatum. Topics: Animals; Blotting, Northern; Cerebral Cortex; Corpus Striatum; Dynorphins; Endorphins; Enkephalins; Guinea Pigs; Male; Organ Specificity; Peptide Fragments; Protein Precursors; Radioimmunoassay; RNA, Messenger | 1993 |
Kappa-opioids decrease excitatory transmission in the dentate gyrus of the guinea pig hippocampus.
In the guinea pig hippocampus, kappa 1-opioid binding sites were primarily localized in the molecular layer of the dentate gyrus as shown by autoradiography using either the kappa 1-selective radioligand 3H-U69,593 or the nonselective radioligand 3H-diprenorphine in the presence of unlabeled mu- and delta-blocking ligands. In this region, the electrophysiological effects of kappa 1-receptor activation were identified using extracellular and intracellular recordings of dentate granule cell responses. The amplitude of the extracellularly recorded population spike was reduced by U69,593 with an EC50 of 26 nM; this effect was reversible and blocked by the opioid antagonist naloxone. The kappa 1-selective antagonist norbinaltorphimine also blocked the effect of U69,593 with an apparent equilibrium dissociation constant (Ki) of 0.26 nM determined by Schild analysis in the physiologic assay. This value agreed well with the Ki for norbinaltorphimine at kappa 1-binding sites measured by radioligand binding displacement (0.24 nM). These results indicate that the electrophysiologic response observed was likely mediated by kappa 1-receptors. As seen with U69,593, dynorphin B, an endogenous opioid peptide that is present in the dentate gyrus, also inhibited the population spike response. mu- and delta-selective opioid agonists had no effect on the amplitude of the maximally evoked response. Intracellular recordings of dentate granule cells showed no direct effects of U69,593 on the granule cells themselves. However, analysis of synaptic potentials revealed that U69,593 significantly reduced the amplitude of glutaminergic EPSPs evoked by afferent stimulation without affecting IPSP amplitudes. The specific effect of U69,593 application on granule cell EPSPs indicates that presynaptic kappa 1-receptor activation inhibits glutamate release from perforant path terminals in the molecular layer of the dentate gyrus. These results suggest that endogenous dynorphins present in the granule cells may act as feedback inhibitors of the major excitatory input to the dentate gyrus. Topics: Action Potentials; Animals; Benzeneacetamides; Diprenorphine; Dynorphins; Electrophysiology; Endorphins; Guinea Pigs; Hippocampus; Male; Naloxone; Naltrexone; Narcotic Antagonists; Pyrrolidines; Receptors, Opioid; Receptors, Opioid, kappa; Synapses | 1992 |
Kainic acid depresses the ex vivo release of dynorphin B and glutamate from rat hippocampal mossy fiber synaptosomes.
This experiment examined the effects of intracerebroventricularly (i.c.v.) administered kainic acid (KA) on the subsequent ex vivo release of L-glutamate (Glu) and dynorphin B-like immunoreactivity (Dyn B-LI) from isolated rat hippocampal mossy fiber (MF) synaptosomes at 4.5 h, 20 h or 48 h after administration of 0.5 microgram/microliter KA. The Dyn B-LI content in the synaptosomal fraction initially decreased at 4.5 h and then rebounded and remained elevated above control levels at 20 h and 48 h. The K(+)-evoked release of Dyn B-LI from the synaptosomes was markedly depressed at 4.5 h after KA and remained significantly below control levels at 20 h and 48 h. In contrast, KA caused no change in the K(+)-evoked release of Glu at 4.5 h as compared to control levels, but did result in a significant decrease in Glu release at 20 h and 48 h. These data indicate a persistent effect of i.c.v. KA on neurotransmission at MF-CA3 synapses in rat hippocampus, resulting in a suppression of the release of Glu as well as the opioid peptide, Dyn B. Topics: Animals; Calcium; Dynorphins; Endorphins; Exocytosis; Glutamates; Glutamic Acid; Hippocampus; In Vitro Techniques; Injections, Intraventricular; Kainic Acid; Male; Protein Precursors; Rats; Rats, Inbred Strains; RNA, Messenger; Synaptosomes | 1992 |
Adenosine modulation of dynorphin B release by hippocampal synaptosomes.
A rat hippocampal preparation enriched in mossy fiber synaptosomes was employed in an attempt to expose any relationship between endogenous adenosine and the release of dynorphin B-like immunoreactivity (DynB-LI). Presumptive blockade of purinergic receptors increased the spontaneous release of DynB-LI, and reducing synaptic adenosine by exogenous adenosine deaminase increased the K(+)-evoked release. Evoked release of DynB-LI was reduced by inhibitors of adenosine uptake and 5'-nucleotidase. Taken together, these data suggest that adenosine endogenous to hippocampal mossy fiber synaptosomes serves to inhibit the release of one of the peptide neuromodulators of this preparation, and provide support for the concept of autoregulation of release. Topics: 5'-Nucleotidase; Adenosine; Adenosine Deaminase; Animals; Dynorphins; Endorphins; Hippocampus; Male; Nerve Endings; Neurotransmitter Agents; Rats; Rats, Inbred Strains; Synaptosomes | 1992 |
Peptides for calling? An immunohistochemical study of the avian n. intercollicularis.
We used two phylogenetically distant avian species (dark-eyed junco, hyemalis, Passeriformes; domestic pigeon, Columba livia, Columbiformes) to determine the immunocytochemical distribution of opioid (leucine-enkephalin, dynorphin B) and non-opioid (adrenocorticotropic hormone) peptides in the n. intercollicularis (ICo), a midbrain region which plays a central role in the control of vocalizations. We found that, in both species, the peptides under study are present as fibers and terminal-like structures, and are similarly distributed. The n. dorsomedialis intercollicularis division contains much less immunoreactivity than the rest of the ICo. Based on this and previous studies, we propose that opioid peptides, possibly transported from the preoptic and hypothalamic regions of the diencephalon to the ICo, regulate vocal behavior by altering respiratory function rather than syringeal mechanisms. Topics: Adrenocorticotropic Hormone; Animals; Birds; Columbidae; Dynorphins; Endorphins; Enkephalin, Leucine; Female; Immune Sera; Immunohistochemistry; Male; Mesencephalon; Species Specificity; Vocalization, Animal | 1992 |
Kappa opioid agonists inhibit transmitter release from guinea pig hippocampal mossy fiber synaptosomes.
Opioid agonists specific for the mu, delta, and kappa opioid receptor subtypes were tested for their ability to modulate potassium-evoked release of L-glutamate and dynorphin B-like immunoreactivity from guinea pig hippocampal mossy fiber synaptosomes. The kappa opioid agonists U-62,066E and (-) ethylketocyclazocine, but not the mu agonist [D-Ala2,N-MePhe4,Gly5-ol]-enkephalin (DAGO) nor the delta agonist [D-Pen2,5]enkephalin (DPDE), inhibited the potassium-evoked release of L-glutamate and dynorphin B-like immunoreactivity. U-62,066E, but not DAGO or DPDE, also inhibited the potassium-evoked rise in mossy fiber synaptosomal cytosolic Ca2+ levels, indicating a possible mechanism for kappa agonist inhibition of transmitter release. DAGO and DPDE were found to be without any effect on cytosolic Ca2+ levels or transmitter release in this preparation. The U-62,066E inhibition of the potassium-evoked rise in synaptosomal cytosolic Ca2+ levels was partially attenuated by the opioid antagonist quadazocine and insensitive to the delta-opioid specific antagonist ICI 174,864 and the mu opioid-preferring antagonists naloxone and naltrexone. Quadazocine also reversed U-62,066E inhibition of the potassium-evoked release of L-glutamate, but not dynorphin B-like immunoreactivity. These results suggest that kappa opioid agonists inhibit transmitter release from mossy fiber terminals through both kappa opioid and non-kappa opioid receptor mediated mechanisms. Topics: Animals; Azocines; Calcium; Dynorphins; Endorphins; Enkephalin, Ala(2)-MePhe(4)-Gly(5)-; Enkephalin, D-Penicillamine (2,5)-; Enkephalins; Ethylketocyclazocine; Glutamates; Glutamic Acid; Guinea Pigs; Hippocampus; Male; Potassium; Pyrrolidines; Receptors, Opioid; Receptors, Opioid, delta; Receptors, Opioid, kappa; Receptors, Opioid, mu; Synaptosomes | 1992 |
Dynorphin-processing endopeptidase in the rat anterior pituitary lactotrophic cell line, GH4C1.
Several peptide hormones and neurotransmitters are produced by cleavage at the monobasic processing sites. An endoprotease capable of cleaving a dynorphin peptide at the monobasic processing site is secreted from the rat anterior pituitary lactotrophic cell line, GH4C1. When characterized by fast protein liquid chromatography using an ion exchange column, the majority of the endoprotease activity elutes as a single symmetrical peak around 0.3 M NaCl. The protease inhibitor profile suggests that the activity is due to putative thiol protease. These enzymatic properties are similar to a monobasic processing enzyme previously found in bovine pituitary and in the rat brain. The secretory pathway which contains the enzyme activity in GH4C1 cells was characterized by stimulation of secretion by thyrotropin releasing hormone, forskolin, phorbol ester, or potassium chloride. The secretion of the enzyme activity was substantially increased by these compounds suggesting that the GH4C1 cells secrete the activity via the regulated pathway. A hormonal treatment of the GH4C1 cells which has been previously shown to produce a substantial increase in the number of secretory granules and ir-prolactin has been found in this study to elevate this enzyme activity 2-fold. This increase is similar to that seen in the carboxypeptidase E activity, another putative peptide hormone processing enzyme activity. These data suggest that the peptide processing activity is regulated to a small but significant extent and is coordinately regulated with carboxypeptidase E activity. Topics: Amino Acid Sequence; Animals; Cattle; Cell Line; Chromatography, High Pressure Liquid; Colforsin; Dynorphins; Endopeptidases; Endorphins; Hydrogen-Ion Concentration; Kinetics; Molecular Sequence Data; Pituitary Gland, Anterior; Potassium Chloride; Prolactin; Protease Inhibitors; Rats; Tetradecanoylphorbol Acetate; Thyrotropin-Releasing Hormone | 1992 |
Dynorphin A-(1-17) and dynorphin B are released from in vitro superfused rat hypothalami. Effects of depolarizing agents and ovariectomy.
We measured the release of immunoreactive (ir) dynorphin (dyn) A-(1-17) and dyn B from the rat hypothalamus by an in vitro superfusion technique. The system was validated on the basis of the recovery and stability of radiolabeled peptides added to the superfused hypothalami. These were detected as authentic peptides by reverse-phase high-performance liquid chromatography (rp-HPLC) only in the presence of a cocktail of peptidase inhibitors added to the superfusion medium. We observed spontaneous release of ir-dyn B, evaluated by a validated radioimmunoassay in the superfusates, that was increased by potassium and veratridine depolarization. It was calcium-dependent and tetrodotoxin-sensitive. We could not evaluate ir-dyn A-(1-17) directly in the superfusates, because the peptidase inhibitors added to the medium significantly altered the tracer-antibody reaction. To obviate this problem, pooled superfusate samples were purified on C18 cartridges and assayed by rp-HPLC. Rp-HPLC analysis of superfusates revealed two molecular forms with the same retention time as authentic dyn A-(1-17) and dyn B which were four times higher in K(+)-stimulated fractions. We could not detect dyn A-(1-32), comprising dyn A-(1-17) and dyn B, even though this peptide is recognized by the antibodies used in this study and is detected in acetic acid extracts of the rat hypothalamus. The spontaneous and K(+)-evoked release of ir-dyn A-(1-17) and ir-dyn B were significantly higher in 2-week ovariectomized rats, in parallel with the increase of their content in the anterior hypothalamus preoptic area. Topics: Animals; Chromatography, High Pressure Liquid; Dynorphins; Endorphins; Female; Hypothalamus; In Vitro Techniques; Male; Neuromuscular Depolarizing Agents; Ovariectomy; Ovary; Perfusion; Radioimmunoassay; Rats; Rats, Inbred Strains | 1992 |
Dynorphin B-like immunoreactivity in gastroduodenal biopsy specimens from gallstone patients.
Dynorphin B-like immunoreactivity (ir-dyn B) was measured by a validated radio-immunoassay in gastroduodenal biopsy specimens from control and gallstone patients. Levels were significantly lower in acetic acid extracts of specimens of the transverse portion of the duodenum from gallstone patients. Gel permeation chromatography showed that almost all ir-dyn B in duodenal samples corresponded to a molecular form co-eluting with authentic dyn B. Duodenal extracts from gallstone patients had less of this form. Reverse-phase high performance liquid chromatography of the pooled gel chromatography fractions showed up a molecular form with the same retention time as synthetic dyn B which was significantly less in fractions from duodenal extracts of gallstone patients. These results indicate the occurrence of dyn B in the human gastrointestinal tract; however, at this stage of our understanding, no causal relationship can be demonstrated with functional alterations of the biliary tree. Topics: Adult; Aged; Cholelithiasis; Duodenum; Dynorphins; Endorphins; Female; Gastric Mucosa; Humans; Male; Middle Aged; Radioimmunoassay; Tissue Distribution | 1992 |
A genetic-correlational study of hippocampal structural variation and variation in exploratory activities of mice.
Our previous work provided evidence that hippocampal opioid peptides form an important neurochemical substrate underlying the gene-dependent exploratory behavior of mice. A prominent hippocampal opioid is dynorphin B, which resides in the mossy fibers exclusively. In order to seek support for causal relationships between dynorphinergic hippocampal mechanisms and exploration, a quantitative-genetic method was chosen. For this purpose, mice from the inbred strains C57BL/6, DBA/2, BLN, and CPB-K were used. Their hippocampal mossy fiber projections were visualized by means of immunohistochemistry, using a highly specific anti-dynorphin B antiserum. The additive-genetic correlations that were estimated suggest pleiotropic gene effects on locomotion, rearing-up, wall-leaning, and several intra- and infrapyramidal mossy fiber (iipMF) variables. Long iipMF, in particular, were found to be associated with high exploratory activity. Topics: Animals; Arousal; Brain Mapping; Dynorphins; Endorphins; Exploratory Behavior; Genetic Variation; Hippocampus; Male; Mice; Mice, Inbred Strains; Motor Activity; Nerve Fibers; Social Environment; Species Specificity | 1991 |
A genetic-correlational study of hippocampal neurochemical variation and variation in exploratory activities of mice.
Previously, we have demonstrated that hippocampal mossy fibers, containing the opioid peptide dynorphin B, are functionally connected with the gene-dependent exploratory behavior of mice. In order to seek further evidence of causal relationships between dynorphin B action and exploration, a quantitative-genetic method was chosen. For this purpose, mice from the inbred strains C57BL/6, DBA/2, BLN, and CPB-K were used. By means of radioimmunoassay, the hippocampal level of dynorphin B was monitored in mice that had been exposed to environmental novelty, as compared to naive animals. Clear evidence was obtained that novelty induces the release of hippocampal dynorphin B. Furthermore, low tissue content was found to be causally connected with high exploratory scores. Topics: Animals; Arousal; Dominance, Cerebral; Dynorphins; Endorphins; Exploratory Behavior; Genetic Variation; Hippocampus; Male; Mice; Mice, Inbred Strains; Motor Activity; Radioimmunoassay; Social Environment; Species Specificity | 1991 |
Isoforms of a dynorphin B converting enzyme isolated by column electrophoresis in agarose suspension.
A dynorphin B converting enzyme previously purified from bovine spinal cord was subjected to column electrophoresis in agarose suspension. By this technique combined with HPLC gel filtration it was possible to resolve and recover several isoforms of the proteinase. All these isoenzymes were associated with a similar molecular size but apparently they differed with regard to their net charges. No significant difference between their inhibitory profiles or their Km values for the release of Leu-enkephalin-Arg6 from dynorphin B was observed. Topics: Animals; Cattle; Chromatography, Gel; Chromatography, High Pressure Liquid; Cysteine Endopeptidases; Dynorphins; Electrophoresis, Agar Gel; Endorphins; Enkephalin, Leucine; Hydroxymercuribenzoates; Isoelectric Point; Isoenzymes; Kinetics; Molecular Weight; Spinal Cord | 1991 |
Approach to studying proteinase specificity by continuous-flow fast atom bombardment mass spectrometry and high-performance liquid chromatography combined with photodiode-array ultraviolet detection.
Fast atom bombardment mass spectrometry (FAB-MS) and high-performance liquid chromatography using a photodiode-array ultraviolet detector were applied to study a dynorphin-converting endopeptidase from the human pituitary gland. The specificity of the enzyme was tested towards various opioid peptides derived from the prodynorphin precursor, i.e. dynorphin A, dynorphin B and alpha-neoendorphin. Peptide fragments were analysed directly by continuous-flow FAB-MS and those containing aromatic amino acids were detected independently by the photodiode-array ultraviolet detector. The results obtained suggest a similar processing of these structure-related substrates and it appears that the enzyme recognizes the dibasic stretch in their sequence. It is also clear from this study that the combination of the above techniques provides a powerful tool for studies of enzymatic conversion among the prodynorphin-derived peptides and it should be applicable to studies of similar mechanisms in other peptide systems. Topics: Amino Acid Sequence; Chromatography, High Pressure Liquid; Dynorphins; Endorphins; Humans; Molecular Sequence Data; Peptide Fragments; Pituitary Gland; Protein Precursors; Serine Endopeptidases; Spectrometry, Mass, Fast Atom Bombardment; Substrate Specificity | 1991 |
Studies of specificity and inhibition of human cerebrospinal fluid dynorphin converting enzyme.
Dynorphin-converting activity was recently discovered in human cerebrospinal fluid. This enzyme (hCSF-DCE) cleaves dynorphin A, dynorphin B and alpha-neoendorphin to release Leu-enkephalin-Arg6. To characterize the enzyme further we used several protease inhibitors, including N-peptidyl-O-acyl hydroxylamines which are known to act as potent irreversible inhibitors of serine and cysteine proteinases. No irreversible inactivation occurred but strong, reversible effects on the dynorphin-converting activity by some of the inhibitors tested could be observed. Although, hCSF-DCE binds its substrates (dynorphin A and B) in the microM-mM concentration range, it exhibits high specificity in recognizing and cleaving the linkage between the two basic amino acids in the substrate sequence. Topics: Amino Acid Sequence; Cysteine Endopeptidases; Dose-Response Relationship, Drug; Dynorphins; Endorphins; Enkephalin, Leucine; Humans; Hydroxylamines; Molecular Sequence Data; Peptides; Protease Inhibitors; Substrate Specificity | 1991 |
Levels of dynorphin peptides, substance P and CGRP in the spinal cord after subchronic administration of morphine in the rat.
Rats were rendered dependent on morphine by repeated injections of morphine, in increasing doses for 14 days and sacrificed. Levels of peptides in the dorsal spinal cord and dorsal root ganglia were analyzed in rats decapitated 2 hr, 24 hr (acute abstinent) or 7 days (late abstinent) respectively, after the last injection of drug. Dynorphin A was significantly decreased in rats abstinent for 24 hr, while dynorphin B remained unaffected. Substance P and CGRP, both putative transmitters in nociceptive primary afferent neurones, and partly existing together in the same neurone, were affected differently. Significantly less substance P but unchanged levels of CGRP were detected in rats abstinent for 24 hr, while on the other hand, CGRP but not levels of substance P, were increased 2 hr after the final injection. In dorsal root ganglia, levels of substance P were lower at 2 hr, while levels of CGRP were unaffected. In late (7 days) abstinence, no effect of opiate on any peptide was detected. Topics: Animals; Calcitonin Gene-Related Peptide; Dynorphins; Endorphins; Male; Morphine; Morphine Dependence; Radioimmunoassay; Rats; Rats, Inbred Strains; Spinal Cord; Substance P; Time Factors | 1991 |
High density of zinc-containing and dynorphin B- and substance P-immunoreactive terminals in the marginal division of the rat striatum.
A distinct subdivision of the striatum has recently been described which is located at the caudomedial margin of the striatum, surrounding the rostrolateral edge of the globus pallidus. This "marginal division" has an internal organization and an efferent distribution which is distinct from the rest of the striatum. The striatum contains moderately high levels of zinc and the neuropeptides enkephalin, dynorphin and substance P. In the present study we have examined the distribution of histologically detectable zinc and of dynorphin B- and substance P-immunoreactivity in the marginal division of the striatum. Each of these substances was more dense within the confines of the marginal division than in the rest of the striatum. These data provide further evidence that the marginal division is a structurally distinct subdivision of the striatum. Topics: Animals; Corpus Striatum; Dynorphins; Endorphins; Female; Immunohistochemistry; Nerve Endings; Rats; Rats, Inbred Strains; Substance P; Zinc | 1990 |
Optimization of non-radioactive in situ hybridization: image analysis of varying pretreatment, hybridization and probe labelling conditions.
Using detection of proopiomelanocortin (POMC) mRNA in rat pituitary as a model, varying conditions of tissue pretreatment, hybridization and probe labelling have been tested. Results were evaluated both by visual assessment and by image analysis of coded specimens. Good correlations between visual gradation, optical densities and cell area percentages were obtained. However, determinations of optical densities (or pixel values) provided most detailed information. The data obtained emphasize the interdependence of fixation and permeabilization conditions and clearly show that the stronger the primary fixation, the more efficient the permeabilization by proteinase K must be. The hybridization temperature is also of importance and temperatures between 40-45 degrees C produced the best signal to noise ratio. The POMC-directed 24-mer probe had a theoretical melting point (Tm) of 49.4 degrees C (in the absence of formamide) and four individual experimental determinations of Tm produced a mean value of 48.9 degrees C. Detection of the biotinylated probe was best accomplished with monoclonal antibiotin antibodies and the alkaline phosphatase-anti-alkaline phosphatase (APAAP) system. Short washes at high-stringency (0.1 x SSC, 45 degrees C) produced an optimal signal to noise ratio. Inclusion of 50% formamide in the hybridization buffer produced an enhanced signal to noise ratio, in spite of a higher background staining. The probe employed for most studies was a synthetic 24-mer oligodeoxynucleotide, complementary to the MSH[4-11]-coding region of POMC mRNA. It was labelled with biotinylated dUTP and unlabelled dCTP using terminal transferase. Chromatographical analyses revealed the labelled probe to be heterogeneous in tail length.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: Animals; Biotin; Cell Membrane Permeability; Deoxycytosine Nucleotides; Deoxyuracil Nucleotides; Dynorphins; Endorphins; Evaluation Studies as Topic; Fixatives; Formamides; Image Processing, Computer-Assisted; Male; Methods; Nucleic Acid Hybridization; Peptide Hydrolases; Pro-Opiomelanocortin; Rats; Rats, Inbred Strains; RNA, Messenger; Temperature | 1990 |
Measurement and chromatographic characterization of prodynorphin-derived peptides in the guinea-pig ileum.
Guinea-pig ileum was dissected and the mucosa, submucosa and external musculature extracted with aqueous acetic acid for measurement of four prodynorphin-derived peptides, namely dynorphin A 1-8, dynorphin A 1-17, dynorphin B, and alpha-neoendorphin. The peptide-like immunoreactive material extracted from the external musculature was characterized by multi-dimensional chromatographic analysis and compared to synthetic porcine standards. The chromatographic methods utilized were: reversed-phase high performance liquid chromatography (RP-HPLC), using two different eluants; cation exchange high performance liquid chromatography (CE-HPLC) and gel filtration chromatography. The dynorphin A 1-8-like immunoreactive material was homogeneous and coeluted with the standard in all chromatographic modes. The dynorphin A 1-17-like and dynorphin B-like immunoreactive material was heterogeneous but showed a peak that coeluted with synthetic standard in all chromatographic modes. The alpha-neoendorphin-like immunoreactive material also appeared to be heterogeneous with the major component on CE-HPLC coeluting with the synthetic peptide standard while the major component on RP-HPLC eluted differently. It was concluded that the guinea-pig ileum contains immunoreactivity for peptides derived from all coding regions of the prodynorphin gene and that these peptides may be present in multiple immunoreactive forms. Topics: Animals; Chromatography, Gel; Chromatography, High Pressure Liquid; Dynorphins; Endorphins; Enkephalins; Guinea Pigs; Ileum; Peptide Fragments; Protein Precursors; Radioimmunoassay | 1989 |
Distribution of dynorphin B and methionine-enkephalin in the mouse hippocampus: influence of genotype.
Immunohistochemical techniques were used to localize dynorphin B and methionine-enkephalin in the mouse hippocampus. Methionine-enkephalin-like immunoreactivity was found within the somata of interneurons distributed mainly in and around the CA1 stratum pyramidale and stratum granulosum as well as in the mossy fibers. Dynorphin B appeared to be confined to the mossy fiber pathway. In addition, we observed a difference between the inbred mouse strains DBA/2 and C57BL/6 with regard to the areas of the dynorphinergic mossy fiber projections: the intra- and infrapyramidal terminal fields were larger in the latter group. Topics: Animals; Dynorphins; Endorphins; Enkephalin, Methionine; Genotype; Hippocampus; Immunohistochemistry; Male; Mice; Mice, Inbred C57BL; Mice, Inbred DBA | 1989 |
A novel bovine spinal cord endoprotease with high specificity for dynorphin B.
An endopeptidase that converts the opioid peptide dynorphin B (Tyr-Gly-Gly-Phe-Leu-Arg-aRg-Gln-Phe-Lys-Val-Val-Thr) to its bioactive fragment Leu-enkephalin-Arg6 was isolated from bovine spinal cord. The enzyme was purified about 230-fold from a concentrated spinal cord extract. Upon sodium dodecyl sulfate-polyacrylamide gel electrophoresis, it stained as a protein of Mr 55,000. The purified enzyme is optimally active at around pH7 and has essential thiol groups. It appears to be highly specific for dynorphin B (Km = 11 microM) but not for alpha-neoendorphin or dynorphin A, two other opioids included in the prodynorphin precursor. From its specificity, molecular size, and inhibitory spectrum, this enzyme is different from other known dynorphin-converting or -degrading enzymes and appears to be a unique and novel endoprotease. Topics: Amino Acid Sequence; Animals; Cations, Divalent; Cattle; Chelating Agents; Chromatography, High Pressure Liquid; Dynorphins; Electrophoresis, Polyacrylamide Gel; Endopeptidases; Endorphins; Enkephalin, Leucine; Hydrogen-Ion Concentration; Molecular Sequence Data; Molecular Weight; Protease Inhibitors; Spinal Cord; Substrate Specificity; Sulfhydryl Reagents | 1989 |
Hypothermia elicited by some prodynorphin-derived peptides: opioid and non-opioid actions.
Prodynorphin-derived peptides were tested for their effects on body temperature after intracerebroventricular administration to unrestrained male rats. Dynorphin A (Dyn A) (5 and 10 nmol) and Dynorphin A-(1-32) (Dyn A-(1-32) (2.5 and 5 nmol) lowered body temperature with a maximum approximately 30 min after administration. Dyn B (up to 50 nmol) did not induce hypothermia. Lower doses of all peptides did not alter body temperature. The hypothermic effect was significantly, but not completely prevented by MR1452 (30 nmol), a preferential antagonist of the kappa receptor, administered intracerebroventricularly. Naloxone, a mu receptor antagonist, naltrexone, its long acting analog up to doses of 100 nmol, as well as MR1453, the (+)-enantiomer of kappa antagonist MR1452 with no opioid binding properties, did not prevent the hypothermic effect. Moreover, episodic barrel rolling and bizarre postures elicited by Dyn A and Dyn A-(1-32) were reduced in rats pretreated i.c.v. with MR1452 (30 nmol), but not with naloxone (up to 100 nmol). Interestingly, des-Tyr-Dynorphin A (Dyn A-(2-17)), a fragment with virtually no opioid binding potential, was 4 times less potent that Dyn A in inducing hypothermia. These findings are consistent with the hypothesis that prodynorphin-derived peptides effects are not exclusively opioids in nature. Topics: Animals; Benzomorphans; Body Temperature; Dynorphins; Endorphins; Hypothermia; Kinetics; Male; Motor Activity; Naloxone; Naltrexone; Peptide Fragments; Rats; Rats, Inbred Strains; Receptors, Opioid | 1989 |
Prodynorphin peptide distribution in the forebrain of the Syrian hamster and rat: a comparative study with antisera against dynorphin A, dynorphin B, and the C-terminus of the prodynorphin precursor molecule.
The neuroanatomical distribution of the prodynorphin precursor molecule in the forebrain of the male Syrian hamster (Mesocricetus auratus) has been studied with a novel antiserum directed against the C-terminus of the leumorphin [dynorphin B (1-29)] peptide product. C-peptide staining in sections from colchicine-treated hamsters is compared to staining in sections from untreated animals. In addition, the pattern of C-peptide immunostaining in hamster brain is compared to that in the rat brain. Finally, the C-peptide immunolabeling patterns in hamsters and rats are compared to those obtained with antisera to dynorphin A (1-17) and dynorphin B (1-13). Areas of heaviest prodynorphin immunoreactivity in the hamster include the hippocampal formation, lateral septum, bed nucleus of the stria terminalis, medial preoptic area, medial and central amygdaloid nuclei, ventral pallidum, substantia nigra, and numerous hypothalamic nuclei. Although this C-peptide staining pattern is similar to dynorphin staining reported previously in the rat, several species differences are apparent. Whereas moderate dentate gyrus granule cell staining and no CA4 cell staining have been reported in the rat hippocampal formation, intense immunostaining in the dentate gyrus and CA4 cell labeling are observed in the hamster. In addition, the medial preoptic area, bed nucleus of the stria terminalis, and medial nucleus of the amygdala stain lightly for prodynorphin-containing fibers and cells in the rat, compared to heavy cell and fiber staining in the hamster in all three of these regions. In the rat there is no differential staining between tissues processed with the C-peptide, dynorphin A, and dynorphin B antisera, but numerous areas of the hamster brain show striking differences. In most hamster brain areas containing prodynorphin peptides, the C-peptide antiserum immunolabels more cells and fibers than the dynorphin B antiserum, which in turn labels more cells and fibers than dynorphin A antiserum. However, exceptions to this hierarchy of staining intensity are found in the lateral hypothalamus, substantia nigra, arcuate nucleus, and habenula. The differences in staining patterns between rat and hamster are greatest when C-peptide antiserum is used; apparent species differences are present, though less pronounced, in dynorphin B- and dynorphin A-immunostained material. Topics: Animals; Brain Chemistry; Colchicine; Cricetinae; Dynorphins; Endorphins; Enkephalins; Immune Sera; Immunoenzyme Techniques; Male; Mesocricetus; Peptide Fragments; Perfusion; Protein Precursors; Rats; Rats, Inbred Strains; Species Specificity | 1989 |
A method permitting precise trimming of resin-embedded tissue for ultrathin sectioning in pre-embedding immunoelectronmicroscopy.
Due to the high complexity of the mammalian central nervous system, sampling of immunohistochemically processed brain tissues for electronmicroscopy requires an accurate and reliable technique. For this reason, the flat-embedding method, which allows light microscopical examination of tissue before sampling, is generally employed. Because of the osmification process, however, the tissue is blackish and opaque which hampers light microscopical selection of tissue areas of interest. We have found that tissue translucency is highly improved by an osmification process using an osmium tetroxide-ferrocyanide mixture. We describe a transilluminated chuck that enables visualization of immunostaining in specimens mounted on a trimming instrument, thus allowing for extremely precise sampling of the tissue. Topics: Animals; Brain; Dynorphins; Endorphins; Immunohistochemistry; Mice; Microscopy, Electron; Microtomy | 1989 |
Dopamine-independent motor behavior following microinjection of rimorphin in the substantia nigra.
The motor-activating effects of rimorphin, an opioid peptide derived from prodynorphin, were examined in the substantia nigra pars reticulata of rats. Unilateral microinjections of rimorphin produced dose-dependent contralateral rotational behavior that was antagonized by naloxone, suggesting that these effects were mediated by opiate receptors. Lesions of midbrain dopamine cells with 6-hydroxydopamine (6-OHDA) produced a 95% or greater depletion of tyrosine hydroxylase in the striatum ipsilateral to the lesion, but failed to reduce the number of circles made by the rats. In addition to an overall preservation of rimorphin-induced circling in animals with 6-OHDA lesions, 50% of these rats exhibited circling that was at least 2 standard deviations above the mean of animals without lesions. The motor activating effects of rimorphin, thus, appear to occur independently of the nigrostriatal dopamine system; these effects may instead be mediated by GABAergic efferents in the pars reticulata. Topics: Animals; Corpus Striatum; Dopamine; Dynorphins; Endorphins; Hydroxydopamines; Male; Microinjections; Motor Activity; Naloxone; Oxidopamine; Rats; Rats, Inbred Strains; Substantia Nigra; Tyrosine 3-Monooxygenase | 1988 |
Distribution of opioid peptides in the preoptic region: immunohistochemical evidence for a steroid-sensitive enkephalin sexual dimorphism.
The distribution of cells and fibers that contain opioid peptides within the preoptic region of the rat was examined immunohistochemically. Cells and/or fibers that contain peptides derived from each of the three major opioid peptide families were differentially stained by using antisera that recognize unique derivatives of each precursor molecule and do not cross-react with members of the other opioid peptide families. A beta-endorphin (beta E) antiserum was used to stain fibers that contain peptides derived from the proopiomelanocortin molecule, and dynorphin-containing cells were identified by using an antiserum directed toward dynorphin B (Dyn B) that does not show detectable cross-reactivity with enkephalin-related peptides. An antiserum raised against peptide E (PE), which does not appear to cross-react significantly with dynorphin peptides, was used to localize enkephalin cells and fibers. Each family of opioid peptides showed a unique distribution in the preoptic region. beta E-immunoreactive fibers were primarily localized to the preoptic part of the periventricular nucleus, with moderate densities of fibers contained in the anteroventral periventricular nucleus (AVPv) and medial preoptic nucleus (MPN). Dyn B-immunoreactive fibers showed a somewhat more uniform distribution throughout the region, and only a few Dyn B-stained cells bodies were found within the medial preoptic area. In contrast, the preoptic region contained hundreds of PE-immunoreactive cells, which were particularly numerous within the AVPv, MPN, and anterodorsal preoptic nucleus. The AVPv and MPN also contained discretely localized plexuses of PE-stained fibers. Although the overall distributions of opioid peptide-containing fibers within the preoptic region were quite similar in male and female rats, differential distributions of fibers were found in certain nuclei such as the AVPv and MPN, and they were correlated with previously identified cytoarchitectonic sexual dimorphisms. Such differential distributions were particularly distinct for enkephalin-containing fibers. Although the AVPv is larger in female rats, it contained more PE-immunoreactive cell bodies in male rats, and we have shown here that this sexual dimorphism appears to be at least partially dependent on perinatal levels of gonadal steroids. In contrast, no difference in the number of PE-stained cells was found within the anterodorsal preoptic nucleus of male and female animals, indicating that sexual differences Topics: Animals; Animals, Newborn; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalins; Female; Immunohistochemistry; Male; Ovariectomy; Peptides; Preoptic Area; Rats; Rats, Inbred Strains; Sex Characteristics; Steroids | 1988 |
Role of the ventromedial hypothalamus in the regulation of adenohypophyseal immunoreactive dynorphin in the rat.
In this study, we have examined the role of the dorsomedial (DMH), ventromedial (VMH) and arcuate (ARH) nuclei of the hypothalamus in the control of hypothalamic and pituitary immunoreactive (ir) dynorphin (Dyn) A and ir-Dyn B in the rat, by evaluating the effect of discrete, bilateral radiofrequency lesions in these structures. Lesions limited to the VMH reduced the content of ir-Dyn in the anterior pituitary but not in the neurointermediate lobe or in the hypothalamus. Gel chromatographic analysis of anterior pituitary extracts confirmed that ir-Dyn is mainly associated with high molecular weight forms containing Dyn A and Dyn B in their sequence. Anterior pituitary extracts of VMH-lesioned rats displayed a clearly lower proportion of these forms. Destruction of the DMH affected only the hypothalamic content of ir-Dyn; ablation of the ARH did not cause any significant change. Our results suggest that ablation of the VMH may disrupt critical neuronal connections to the median eminence originating in this nucleus or crossing it and participating in control of the adenohypophyseal pool of ir-Dyn. Topics: Animals; Dynorphins; Endorphins; Hypothalamus; Male; Neural Pathways; Pituitary Gland, Anterior; Radio Waves; Rats; Rats, Inbred Strains; Ventromedial Hypothalamic Nucleus | 1988 |
Distribution of opiate receptor subtypes and enkephalin and dynorphin immunoreactivity in the hippocampus of squirrel, guinea pig, rat, and hamster.
The distribution of enkephalin and dynorphin immunoreactivity in the hippocampus of four rodent species (gray squirrel, guinea pig, rat, and hamster) is compared with the pattern of opiate receptor subtypes (mu, delta, and kappa). The distribution of opioid peptides is fairly consistent in the anterior hippocampus of these four species. Intense immunoreactivity for dynorphin and enkephalin is found in the hilus of the dentate gyrus and in the mossy fiber system. Occasional immunoreactive processes are seen in the dentate molecular layer and scattered throughout the CA1 and CA3 fields. In the rat and hamster, an additional plexus of enkephalinergic fibers straddles both sides of the hippocampal fissure. Cells immunoreactive for both opioid peptides are located in and just superficial to the dentate granule cell layer. Opiate receptors are variably distributed in these rodent species. In the squirrel, guinea pig, and hamster, mu and kappa binding is dense in the stratum lucidum of CA3 and the molecular layer of the dentate gyrus. In the rat, dense mu and kappa binding is localized within and adjacent to the pyramidal and granule cell layers. Delta receptor patterns show additional species differences. In the rat, the delta distribution is similar to the mu and kappa patterns. In the other species, the delta binding pattern is generally the inverse of the mu/kappa pattern: most areas of the hippocampus are enriched in delta sites, whereas the stratum lucidum and the pyramidal cell layer are receptor-sparse. Thus, the stratum lucidum--site of dense terminations of mossy fibers containing opioid peptides--is characterized by selectively sparse delta receptors in four species and by selectively dense kappa receptors in three species. The three receptor subtypes, taken either individually or together and compared to the peptides, are more variably and more widely distributed throughout the hippocampus and fail to show a correspondence with opioid-peptide-containing terminals. The mismatches suggest that receptor locations and densities are organized without relation to the sites of relevant transmitter release. Topics: Animals; Autoradiography; Cricetinae; Dynorphins; Endorphins; Enkephalins; Female; Guinea Pigs; Hippocampus; Histocytochemistry; Immunochemistry; Male; Mesocricetus; Rats; Receptors, Opioid; Sciuridae | 1987 |
Brain endo-oligopeptidase A, a putative enkephalin converting enzyme.
Endo-oligopeptidase A, highly purified from the cytosol fraction of bovine brain by immunoaffinity chromatography, has been characterized as a thiol endopeptidase. This enzyme, known to hydrolyze the Phe5-Ser6 bond of bradykinin and the Arg8-Arg9 bond of neurotensin, has been shown to produce, by a single cleavage, Leu5-enkephalin or Met5-enkephalin from small enkephalin-containing peptides. Enkephalin formation could be inhibited in a concentration-dependent manner by the alternative substrate bradykinin. The optimal substrate size was found to be eight to 13 amino acids, with enkephalin the only product released from precursors in which this sequence is immediately followed by a pair of basic residues. However, the specificity constants (kcat/Km) obtained for endo-oligopeptidase A hydrolysis of bradykinin, neurotensin, and dynorphin B are of the same order, a result indicating that the substrate amino acid sequence is not the only factor determining the cleavage site of this enzyme. Topics: Animals; Bradykinin; Brain; Cattle; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Cytosol; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalin, Methionine; Enkephalins; Metalloendopeptidases; Neurotensin; Peptide Fragments; Rats; Substrate Specificity; Sulfhydryl Reagents | 1987 |
Bilateral intranigral microinjection of morphine and opioid peptides produces antinociception in rats.
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 |
Characterization of big dynorphins from rat brain and spinal cord.
To examine the processing of products of the dynorphin gene in the central nervous system, immunoreactive (ir) dynorphin (Dyn) A, Dyn B, Dyn A-(1-8), alpha- and beta-neo-endorphin (alpha- and beta-Neo) in rat brain and spinal cord were measured, using specific antisera after gel filtration and high-performance liquid chromatography (HPLC). Three peaks of Mr about 8, 4, and 2 kDa for ir-Dyn A and ir-Dyn B, and one peak of Mr less than 2 kDa for ir-Dyn A-(1-8), ir-alpha-, and ir-beta-Neo were found both in the brain and in the spinal cord. The 8 kDa peak was recognized by Dyn A and Dyn B antisera and, after hydrolysis by proline-specific endopeptidase, by beta-Neo antiserum. The 8 kDa peak was recognized by a monoclonal antibody against the amino terminal sequence Tyr-Gly-Gly-Phe of all opioid peptides and by an antiserum directed toward the carboxyl terminus of Dyn B, indicating that it contains, from the amino terminal tyrosine of neo-endorphin to the carboxyl-terminal threonine of Dyn B, all 3 opioid peptide regions in the prodynorphin. By means of proline-specific endopeptidase hydrolysis, we also found a big dynorphin precursor (Mr approximately equal to 26 kDa) in both brain and spinal cord. Topics: Animals; beta-Endorphin; Brain Chemistry; Chromatography, Gel; Chromatography, High Pressure Liquid; Dynorphins; Endorphins; Male; Molecular Weight; Peptide Fragments; Protein Precursors; Rats; Rats, Inbred Strains; Spinal Cord | 1987 |
Immunoreactive dynorphin B in sacral primary afferent fibers of the cat.
Immunocytochemical analysis of the distribution of dynorphin B terminals in the sacral spinal cord of the cat revealed a pattern of staining very similar to that produced with antisera directed against the primary afferent derived, putative neurotransmitter, vasoactive intestinal polypeptide. Labeled axons and terminals were concentrated in lamina I and V and there was dense fiber staining in the tract of Lissauer. Of particular interest was the presence of immunoreactive axons in attached dorsal rootlets. To specifically focus on the possibility that some of the sacral primary afferent fibers are dynorphin-immunoreactive, we first tried to increase perikaryal labeling in the sacral dorsal root ganglia by topical treatment with colchicine. This did not produce immunoreactive labeling of cell bodies in the ganglia. Unilateral multiple dorsal rhizotomy (L5 to coccygeal 1), however, significantly decreased the staining of dynorphin-immunoreactive axons and terminals in the tract of Lissauer and in the dorsal horn of sacral segments ipsilateral to the deafferentation. No changes were detected in the lumbar cord. Finally, radioimmunoassay of caudal lumbar and sacral dorsal root ganglia was performed. Measurable immunoreactivity was found in all ganglia assayed, but, consistent with the histochemical analysis, sacral ganglia contained the highest concentration of immunoreactive dynorphin B. These data indicate that a significant component of the sacral spinal cord dynorphin terminal immunoreactivity derives from primary afferent fibers.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: Animals; Axons; Cats; Colchicine; Dynorphins; Endorphins; Enkephalin, Leucine; Ganglia, Spinal; Histocytochemistry; Neurons, Afferent; Radioimmunoassay; Sacrum; Spinal Cord; Staining and Labeling | 1986 |
Opiate receptors, neuropeptides in CNS and CSF of two Macaca species with different responsiveness to opiates.
Of two related Macaca species, the rhesus (M. mulatta), acquires opiate tolerance and dependence more readily than the cynomolgus (M. fascicularis). In the cynomolgus, mu-opiate receptors were significantly fewer in the caudate nucleus and globus pallidus; delta-sites were fewer in the thalamus. kappa-Sites showed no species difference. The levels of [Met5]enkephalin, substance P and dynorphin B in various brain areas were comparable. On the other hand, receptor-assayed endorphin activity was higher in CSF of cynomolgus than rhesus monkeys. Topics: Animals; Central Nervous System; Dynorphins; Endorphins; Enkephalin, Methionine; Macaca; Macaca fascicularis; Macaca mulatta; Nerve Tissue Proteins; Radioimmunoassay; Receptors, Opioid; Species Specificity; Substance P | 1986 |
Dynorphin B-containing perivascular axons and sensory neurotransmitter mechanisms in brain blood vessels.
This is the first report demonstrating the existence of opiate-containing nerve fibers surrounding brain blood vessels. Dynorphin B, a tridecapeptide and potent opiate analgesic, was visualized by immunohistochemistry in guinea pig cerebral arteries comprising the circle of Willis and was measured by radioimmunoassay in canine middle cerebral arteries. This peptide, reportedly present in dorsal root ganglion cells, was observed by others to decrease the depolarization-induced release of substance P from primary sensory axons and, by so doing, to retard the development of neurogenic inflammation in target tissues. Consistent with an indirect action of dynorphin B, this peptide did not relax precontracted canine middle cerebral or basilar artery segments when added in vitro, nor did it modulate receptor-mediated relaxation on the addition of substance P. The presence of opiate-containing axons in or near trigeminovascular nerve fibers suggests novel mechanisms related to the modulation of pain possibly emanating from cerebral vessels. Topics: Animals; Arteries; Axons; Basilar Artery; Bombesin; Brain; Carotid Arteries; Cerebellum; Cerebral Arteries; Circle of Willis; Dogs; Dynorphins; Endorphins; Female; Guinea Pigs; Neurons, Afferent; Neurotransmitter Agents; Pia Mater; Somatostatin; Substance P; Trigeminal Nerve | 1986 |
Dynorphin- and enkephalin-like immunoreactivity is altered in limbic-basal ganglia regions of rat brain after repeated electroconvulsive shock.
In an attempt to determine whether the opioid peptides derived from prodynorphin participate in the effects of electroconvulsive shock (ECS), we used radioimmunoassay and immunocytochemistry to measure dynorphin-like immunoreactivity (DN-LI) in various rat brain regions after repeated ECS treatments. Ten daily ECSs caused a significant increase in dynorphin A (1-8)-LI in most limbic-basal ganglia structures, including hypothalamus (50%), striatum (30%), and septum (30%). No significant change was found in the frontal cortex or the neurointermediate lobe of the pituitary. In contrast, 10 ECS treatments depleted DN-LI in hippocampal mossy fibers by 64%. A detailed time-course study revealed that a single shock caused a small but significant increase in hippocampal DN-LI, whereas three consecutive shocks depleted DN-LI by 30%. The maximal decrease in DN-LI was reached after six daily ECSs. The level of DN-LI in the hippocampus partly recovered, but remained lower than the control value 4, 7, and 14 d after the cessation of six daily ECSs (50, 77, and 83% of control value, respectively). In contrast with the ECS-induced depletion of hippocampal dynorphin, 10 daily ECSs caused a significant increase (40%) in (Met5)-enkephalin-LI in the hippocampus, as well as in other limbic-basal ganglia structures. Immunocytochemistry revealed that enkephalin-LI was increased in the perforant pathway, which is presynaptic to the dynorphin-containing mossy fiber pathway in the hippocampus. These observations suggest that different mechanisms may regulate these two opioid peptide systems in the hippocampus.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: Animals; Basal Ganglia; Brain Chemistry; Dynorphins; Electroshock; Endorphins; Enkephalin, Leucine; Enkephalins; Hippocampus; Immunologic Techniques; Male; Radioimmunoassay; Rats; Rats, Inbred F344; Time Factors; Tissue Distribution | 1986 |
Peptides derived from prodynorphin are decreased in basal ganglia of Huntington's disease brains.
The contents of methionine-enkephalin-Arg-Gly-Leu, dynorphin A, dynorphin B and alpha-neoendorphin have been measured in both control and Huntington's disease brains obtained postmortem. All 4 peptides were significantly reduced in the caudate nucleus and putamen of Huntington's disease compared with the control group. No differences were observed in frontal cortex or hypothalamus. Immunocytochemistry showed a marked depletion of dynorphin-like immunoreactivity in Huntington's disease substantia nigra. Topics: Basal Ganglia; Caudate Nucleus; Dynorphins; Endorphins; Enkephalins; Globus Pallidus; Humans; Huntington Disease; Protein Precursors; Putamen; Substantia Nigra | 1986 |
Conversion of leumorphin (dynorphin B-29) to dynorphin B and dynorphin B-14 by thiol protease activity.
Dynorphin B (rimorphin) is formed from leumorphin (dynorphin B-29) by the action of a thiol protease from rat brain membranes, in a single step. This represents a "single-arginine cleavage" between threonine-13 and arginine-14 of the substrate. We have observed that in addition to dynorphin B, dynorphin B-14 is formed from dynorphin B-29. Among the various protease inhibitors tested, none except p-chloromercuribenzensulfonic acid inhibited the formation of the two products. Both temperature and pH had similar effects on the formation of dynorphin B-14 and dynorphin B. The inhibitory potencies of adrenocorticotropic hormone, peptide E, and dynorphin A were virtually identical for the formation of the two products. These results suggest that the same enzyme may be responsible for the formation of dynorphin B-14 and dynorphin B. Topics: Adrenocorticotropic Hormone; Cysteine Endopeptidases; Dynorphins; Endopeptidases; Endorphins; Enkephalins; Hydrogen-Ion Concentration; Peptides; Protease Inhibitors; Protein Precursors; Temperature | 1986 |
Distribution of dynorphin and enkephalin peptides in the rat brain.
The neuroanatomical distribution of dynorphin B-like immunoreactivity (DYN-B) was studied in the adult male and female albino rat. The distribution of DYN B in colchicine- and noncolchicine-treated animals was also compared to that of another opioid peptide derived from the prodynorphin precursor dynorphin A (1-8) (DYN 1-8), and an opioid peptide derived from the proenkephalin precursor met-enkephalin-arg-gly-leu (MERGL). DYN B cell bodies were present in nonpyramidal cells of neo- and allocortices, medium-sized cells of the caudate-putamen, nucleus accumbens, lateral part of the central nucleus of the amygdala, bed nucleus of the stria terminalis, preoptic area, and in sectors of nearly every hypothalamic nucleus and area, medial pretectal area, and nucleus of the optic tract, periaqueductal gray, raphe nuclei, cuneiform nucleus, sagulum, retrorubral nucleus, peripeduncular nucleus, lateral terminal nucleus, pedunculopontine nucleus, mesencephalic trigeminal nucleus, parabigeminal nucleus, dorsal nucleus of the lateral lemniscus, lateral superior olivary nucleus, superior paraolivary nucleus, medial superior olivary nucleus, ventral nucleus of the trapezoid body, lateral dorsal tegmental nucleus, accessory trigeminal nucleus, solitary nucleus, nucleus ambiguus, paratrigeminal nucleus, area postrema, lateral reticular nucleus, and ventrolateral region of the reticular formation. Fiber systems are present that conform to many of the known output systems of these nuclei, including major descending pathways (e.g., striatonigral, striatopallidal, reticulospinal, hypothalamospinal pathways), short projection systems (e.g., mossy fibers in hippocampus, hypothalamo-hypophyseal pathways), and local circuit pathways (e.g., in cortex, hypothalamus). The distribution of MERGL was, with a few notable exceptions, in the same nuclei as DYN B. From these neuroanatomical data, it appears that the dynorphin and enkephalin peptides are strategically located in brain regions that regulate extrapyramidal motor function, cardiovascular and water balance systems, eating, sensory processing, and pain perception. Topics: Animals; Brain; Diencephalon; Dynorphins; Endorphins; Enkephalin, Methionine; Female; Fluorescent Antibody Technique; Male; Medulla Oblongata; Mesencephalon; Peptide Fragments; Pons; Rats; Telencephalon | 1986 |
Identification of the tridecapeptide dynorphin B (rimorphin) within perikarya of rat duodenum.
Using an immunofluorescence microscopic staining technique, the opioid peptide dynorphin B (rimorphin) was revealed within neuronal cell bodies of the rat duodenum. Dynorphin B immunoreactive perikarya were revealed in the myenteric and submucousal plexus as well as in the longitudinal muscle layer. They were large in diameter and round in shape and they contained a large round nucleus. Because no dynorphin B immunofluorescent nerve fibre and terminal could be noted it might be that dynorphin B is further cleaved by proteases into the bioactive opioid pentapeptide Leu-enkephalin and dynorphin B(6-13). These findings might also indicate that dynorphin B is processed within duodenal perikarya and that it has important physiological roles in the rat duodenum. Topics: Animals; Duodenum; Dynorphins; Endorphins; Fluorescent Antibody Technique; Male; Microscopy, Fluorescence; Neurons; Rats; Rats, Inbred Strains | 1986 |
Localization of dynorphin gene product-immunoreactivity in neurons from spinal cord and dorsal root ganglia.
Using spinal cord and dorsal root ganglion cell cultures, we have studied the immuno-histochemical distribution of several peptide products of the dynorphin gene. With antibody directed toward the midregion of dynorphin A, peptide-immunoreactivity was found exclusively in the cell bodies of spinal cord neurons. Antibody directed toward the amino- or carboxy-terminus of dynorphin A revealed peptide-immunoreactivity in the neurites, as well as perikarya. Spinal cord neurons also expressed dynorphin B- and alpha-neo-endorphin-immunoreactivities in both cell bodies and neurites. Dorsal root ganglion neurons cultured from embryonic tissue expressed dynorphin A-(1-13)-, dynorphin A-(9-17)- and dynorphin B-immunoreactivities in their perikarya. Sensory neurons obtained from dissociated adult ganglia similarly expressed dynorphin-immunoreactivity immediately upon inoculation into culture. Embryonic and adult murine sensory ganglia from the sacral region more frequently expressed dynorphin than did cells obtained from other spinal levels. Expression of dynorphin-immunoreactivity by sensory neurons was not influenced by elevated levels of Nerve Growth Factor or spinal cord conditioned medium. These data indicate that intrinsic spinal cord neurons may modulate sensory and spinal function in rather subtle ways via the expression of several different opioid peptide products of the dynorphin gene, in addition to the opioid peptides produced by the proenkephalin A gene. Beyond this, the observation of dynorphin-related peptides in dorsal root ganglion neurons suggests that these opioid peptides may have a specialized role in primary afferent neurotransmission. Topics: Animals; Antibodies; Cells, Cultured; Dynorphins; Endorphins; Fluorescent Antibody Technique; Ganglia, Spinal; Gene Expression Regulation; Mice; Mice, Inbred C57BL; Spinal Cord | 1986 |
Response of rat pituitary anterior lobe prodynorphin products to changes in gonadal steroid environment.
The total content of rat pituitary anterior lobe (AL) immunoreactive (ir) dynorphin A (ir-Dyn A) and ir-dynorphin B (Dyn B) increased in male rats between 15 and 58 days of age, but there was little alteration in the concentration of ir-Dyn A or B expressed relative to protein content. Adult rats (90 days of age) had lower concentrations of these peptide immunoreactivities in the AL. Castration of 58-day-old male rats produced a testosterone-reversible loss of ir-Dyn A and B by 50-60% 3 days after surgery. Thereafter, the levels of these peptides gradually increased to 2.5 times the levels found in control animals at 1 month after castration. These effects of castration on AL dynorphin were not seen in 15-day-old rats and were much less marked in adults. Similar changes were seen in the levels of other prodynorphin products, alpha- and beta-neo-endorphin (ir-alpha-nEnd and ir-beta-nEnd), and ir-[Leu5]enkephalin (ir-LE). Administration of testosterone (100 micrograms/100 g BW) to castrated rats for 2 days largely prevented the drop in the levels of AL ir-Dyn A and B. Ovariectomy produced an increase in the levels of ir-Dyn A, Dyn B, alpha-nEnd, beta-nEnd, and LE 2 weeks after surgery, but, in contrast to castration, no significant decrease was seen 3 days after ovariectomy. These changes in AL content of dynorphin-related peptides after castration or ovariectomy directly reflect those previously reported for AL content of LH. The mechanisms regulating storage (and perhaps secretion) of AL peptides derived from prodynorphin may be similar to those regulating storage and secretion of LH and FSH in rat AL. AL ir-LE could potentially arise from proenkephalin A or prodynorphin (proenkephalin B). Ir-LE levels in AL were approximately 10 times higher than the levels of ir-[Met5]-enkephalinyl-Arg-Gly-Leu (ME-RGL) in male rat AL, and changes in ir-LE content after castration were very similar to those observed in other prodynorphin-derived peptides, but different from the effects of castration on ir-ME-RGL. It is possible that prodynorphin is a major source of AL ir-LE. Topics: Animals; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalins; Female; Gonadal Steroid Hormones; Male; Orchiectomy; Ovariectomy; Pituitary Gland, Anterior; Protein Precursors; Rats; Testosterone | 1986 |
[Effects of opiate and opioid peptides administered intrathecally on the pain threshold and micturition reflex in rats].
Topics: Animals; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalin, Leucine-2-Alanine; Female; Injections, Spinal; Morphine; Pain; Rats; Rats, Inbred Strains; Reflex; Sensory Thresholds; Urination | 1986 |
[Immunoreactive dynorphin B in the central nervous system of the mouse and its high performance liquid chromatograph (HPLC) analysis].
Topics: Animals; Central Nervous System; Chromatography, High Pressure Liquid; Dynorphins; Endorphins; Hypothalamus; Male; Mice; Peptide Fragments; Pituitary Gland | 1986 |
Inhibitory effects of rimorphin and dynorphin on insulin secretion from the isolated, perfused rat pancreas.
In order to settle the question about whether or not opioid peptides stimulate or inhibit insulin secretion, we studied effects of rimorphin and dynorphin, two members of the preproenkephalin B group, on glucose-induced insulin secretion in the isolated, perfused rat pancreas. These peptides (3.95 X 10(-8) M), like morphine (3.95 X 10(-8) M), significantly inhibited the glucose-induced insulin secretion. The inhibitory effect of rimorphin was attenuated by naloxone (1.2 X 10(-6) M) and phentolamine (10(-6) M), suggesting an involvement of adrenergic alpha receptors in the inhibition of glucose-induced insulin secretion mediated through specific opiate receptors. Rimorphin also inhibited glucose-induced insulin secretion even in the cysteamine-treated rat pancreas from which somatostatin had been depleted. Thus, somatostatin does not appear to play a major regulatory role in the insulin secretion in the pancreas. Topics: Animals; Dynorphins; Endorphins; Glucose; Insulin; Insulin Secretion; Male; Morphine; Pancreas; Phentolamine; Propranolol; Rats; Rats, Inbred Strains; Somatostatin | 1986 |
[Effects of ohmefentanyl on the content of endorphin in pituitary and brain areas of rats].
Topics: Analgesia; Animals; beta-Endorphin; Brain; Catalepsy; Dynorphins; Endorphins; Enkephalin, Methionine; Fentanyl; Male; Peptide Fragments; Pituitary Gland; Radioimmunoassay; Rats; Tissue Distribution | 1986 |
Methionine and leucine enkephalin in rat neurohypophysis: different responses to osmotic stimuli and T2 toxin.
Specific radioimmunoassays were used to measure the effects of hypertonic saline (salt loading), water deprivation, and trichothecene mycotoxin (T2 toxin) on the content of methionine enkephalin (ME), leucine enkephalin (LE), alpha-neoendorphin, dynorphin A, dynorphin B, vasopressin, and oxytocin in the rat posterior pituitary. Concentrations of vasopressin and oxytocin decreased in response to both osmotic stimuli and treatment with T2 toxin, but the decrease was greater with osmotic stimulations. Similarly, concentrations of LE and dynorphin-related peptides declined after salt loading and water deprivation; LE concentrations also decreased after treatment with T2 toxin. The concentration of ME decreased after water deprivation, did not change after salt loading, and increased after T2 toxin treatment. The differentiating effects of these stimuli on the content of immunoreactive LE and ME are consistent with the hypothesis that LE and ME may be localized in separate populations of nerve endings with different roles in the posterior pituitary. Topics: Animals; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalin, Methionine; Male; Osmosis; Oxytocin; Pituitary Gland, Posterior; Protein Precursors; Rats; Rats, Inbred Strains; Saline Solution, Hypertonic; Sesquiterpenes; T-2 Toxin; Vasopressins; Water Deprivation | 1985 |
Selective inactivation of opioid receptors in rat hippocampus demonstrates that dynorphin-A and -B may act on mu-receptors in the CA1 region.
Dynorphin-A1-17 and dynorphin-B increased the evoked response of hippocampal CA1 pyramidal cells, as did other opioids tested. Treatment of the hippocampal slice with beta-funaltrexamine, a mu-receptor selective antagonist, blocked the effects of normorphine, dynorphin-A and dynorphin-B, but did not change the response to D-Ala2, D-Leu5-enkephalin. The low potency of kappa selective agonists and the antagonism by beta-funaltrexamine of the dynorphins' effect indicate that kappa-opioid receptors may not be involved in these observed responses. Our data suggest that both mu- and delta-receptors are functionally represented and provide evidence that the dynorphins or their derivatives may also be agonists at the mu-receptor. Topics: Animals; Dynorphins; Endorphins; Hippocampus; In Vitro Techniques; Naltrexone; Rats; Receptors, Opioid; Receptors, Opioid, mu | 1985 |
Possible interactions between dynorphin and dopaminergic systems in rat basal ganglia and substantia nigra.
Chronic haloperidol treatment markedly increases dynorphin-related peptide contents in caudate-putamen, globus pallidus and substantia nigra. Leu-enkephalin levels follow dynorphin-related peptide concentrations in these areas while Met-enkephalin-related peptide contents are unchanged in the substantia nigra following a similar treatment. An acute haloperidol injection had no effect on any opioid peptide levels in the basal ganglia. This suggests that Leu-enkephalin is likely to be derived from prodynorphin in the rat striatonigral pathway. Moreover, the Leu-enkephalin/dynorphin projection appears to be under striatal dopaminergic control. Topics: Animals; Caudate Nucleus; Corpus Striatum; Dopamine; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalin, Methionine; Globus Pallidus; Haloperidol; Male; Putamen; Rats; Rats, Inbred Strains; Substantia Nigra | 1985 |
Proenkephalin and prodynorphin related neuropeptides in the cochlea.
Dynorphin B (rimorphin), a proenkephalin B (prodynorphin)-derived peptide, and met-enkephalin-Arg6, Gly7, Leu8 (met-enkephalin octapeptide), a proenkephalin A-derived peptide, were identified in the mammalian cochlea by specific radioimmunoassays. The antisera are directed against unique sequences in the peptides, and this immunoreactivity cannot be ascribed to cross-reaction with any other known opioid peptides. Met-enkephalin octapeptide and dynorphin B can for this reason serve as reliable markers for the proenkephalin A- and proenkephalin B-derived peptides, respectively. Lesion studies in the cochlea indicate that dynorphin B is confined to olivocochlear efferents. It has not been determined if the dynorphin-containing neurons are the same as those known to contain enkephalin-related peptides, or if they may be cholinergic. Different, presumably inhibitory, neurotransmitters or modulators in the olivocochlear fibers create the possibility of separately modulating the effects of inner or outer hair cells on auditory nerve activity, and so becoming able to study their individual actions in audition. The olivocochlear fiber-hair cell-eighth nerve interaction may provide a valuable model for a complex multi-transmitter synaptic junction. Topics: Animals; Cochlea; Dynorphins; Endorphins; Enkephalin, Methionine; Enkephalins; Female; Guinea Pigs; Hair Cells, Auditory; Nerve Tissue Proteins; Protein Precursors | 1985 |
Analysis of opioid and non-opioid end products of pro-dynorphin in the substantia nigra of the rat.
The substantia nigra is among the richest pro-dynorphin terminal field regions in the rat brain. We therefore contrasted processing in this area to the known processing in the posterior pituitary. Fractionation of acid extracts of the posterior pituitary by gel filtration followed by analysis by radioimmunoassay indicated that the molar ratio of dynorphin A(1-17) to dynorphin A(1-8) averaged 1:2. The levels of dynorphin A-related end products to alpha-neo-endorphin and bridge peptide (a 2K nonopioid end product of pro-dynorphin) were approximately equimolar; however, the levels of dynorphin B-sized material were 50% lower than dynorphin A levels. Similar analyses of acid extracts of the substantia nigra also indicated that the levels of dynorphin A, alpha-neo-endorphin, and bridge peptide were approximately equimolar. In this terminal field the levels of dynorphin B-sized material were approximately 60% lower than dynorphin A. A striking feature of the nigral system was that the molar ratio of dynorphin A(1-17) to dynorphin A(1-8) averaged 1:16. Thus, in the nigra, dynorphin A(1-17) is primarily a biosynthetic intermediate rather than as an end product. Topics: Animals; Dynorphins; Endorphins; Male; Peptide Fragments; Protein Precursors; Rats; Rats, Inbred Strains; Substantia Nigra | 1985 |
A general procedure for analysis of proenkephalin B derived opioid peptides.
Tryptic digestion followed by radioimmunoassay for (Leu)enkephalin-Arg6 has been used in this study as a general method to detect the presence of all possible products containing the enkephalin sequence from the opioid peptide prohormone, proenkephalin B. Tissue extracts of human hypothalamus and pituitary were examined. Gel filtration was used to separate the different precursor products according to molecular weight. The elution profile was also monitored with highly sensitive radioimmunoassays for dynorphin A and dynorphin B, respectively. Immunoreactive dynorphin A appeared in three peaks with the approximate molecular weight of 1000, 2000 and 5000. Immunoreactive dynorphin B partly occurred in other peaks, 1500, 5000 and 10 000 dalton. Profiles obtained by measuring immunoreactive (Leu)enkephalin-Arg6 in all fractions from gel filtration after trypsin digestion showed a more complex pattern compared to the profiles of immunoreactive dynorphin A and dynorphin B. The major peaks coincided with dynorphin A and dynorphin B but high levels of immunoreactive (Leu)enkephalin-Arg6 were also generated from higher molecular weight regions (MW greater than 5000). Topics: Chromatography, Gel; Chromatography, High Pressure Liquid; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalins; Humans; Hypothalamus; Molecular Weight; Pituitary Gland; Protein Precursors | 1985 |
Stimulation by leumorphin of prolactin secretion from the pituitary in rats.
The effect of leumorphin (LM), one of big leu-enkephalins derived from preproenkephalin B, on PRL secretion was studied in the rat in vivo and in vitro. Intracerebroventricular injection of synthetic porcine LM (0.06-6 nmol/rat) caused a dose-related increase in plasma PRL levels in urethane-anesthetized male rats and in conscious freely moving rats. Intravenous injection of LM (3 nmol/100 g BW) also raised plasma PRL levels in these animals. The plasma PRL response to intracerebroventricular LM (0.6 nmol/rat) was blunted by naloxone (125 micrograms/100 g BW, iv). The stimulating effect of LM on PRL release was the most potent among the peptides derived from preproenkephalin B. In in vitro studies, PRL release from superfused anterior pituitary cells was stimulated in a dose-related manner by LM (10(-9)-10(-6) M), and the effect was blunted by naloxone (10(-5) M). These results suggest that LM has a potent stimulating effect on PRL secretion from the pituitary in the rat by acting, at least in part, directly at the pituitary through an opiate receptor. Topics: Animals; beta-Endorphin; Dynorphins; Endorphins; Enkephalins; Injections, Intraventricular; Male; Naloxone; Pituitary Gland, Anterior; Prolactin; Protein Precursors; Rats; Swine; Thyrotropin-Releasing Hormone | 1985 |
Localization of dynorphin B-like and alpha-neoendorphin-like immunoreactivities in the guinea pig organ of Corti.
Antiserum to dynorphin B and antiserum to alpha-neoendorphin were used in an immunocytochemical examination of the guinea pig organ of Corti. Immunoreactive staining for these two proenkephalin B (prodynorphin)-derived peptides was seen in the lateral system of olivocochlear efferents in the organ of Corti: the inner spiral bundle, the tunnel spiral bundle and by the bases of inner hair cells. Immunoreactive staining with both antisera was also seen in efferent terminals on outer hair cells at or above the level of the nucleus, which may represent terminals of either the lateral or the medial system. No immunoreactive staining was seen in tunnel crossing fibers and at bases of outer hair cells corresponding to the medial system of efferents. The staining seen with antiserum to dynorphin B and to alpha-neoendorphin has similar distribution to that seen with antisera to methionine enkephalin; there may be co-localization of these neuropeptides in the lateral system of efferents. Choline acetyltransferase-like immunoreactivity (co-localized with enkephalin-like immunoreactivity in the lateral system in the brainstem) and glutamic acid decarboxylase (GAD)-like immunoreactivity have also been found in olivocochlear efferents. Further studies will be necessary to determine if the dynorphins are co-localized with other neurotransmitter candidates and what their interactions may be. Topics: Animals; Dynorphins; Endorphins; Guinea Pigs; Immunoenzyme Techniques; Nerve Endings; Neurons, Efferent; Organ of Corti; Protein Precursors | 1985 |
Isolation and characterization of an endogenous C-terminal fragment of the alpha-neo-endorphin/dynorphin precursor from bovine caudate nucleus.
Antibodies have been raised to a synthetic peptide corresponding to the C-terminal 15-amino acid residues of prodynorphin, the common precursor to the neo-endorphins and dynorphins. The amino acid sequence of the antigen was based on the sequence deduced from mRNA isolated and cloned from porcine hypothalamus (Kakidani, H., Y. Furutani, H. Takahashi, M. Noda, Y. Morimoto, T. Hirose, M. Asai, S. Inayama, S. Nakanishi, and S. Numa (1982) Nature 298: 245-248). Using a radioimmunoassay developed from these antibodies we have isolated an endogenous prodynorphin C-fragment from bovine caudate nucleus. The isolated peptide displayed characteristics on gel filtration similar to those of synthetic prodynorphin C-fragment predicted from the porcine mRNA sequence but had low cross-reactivity in the radioimmunoassay. Sequencing and amino acid analysis showed a substitution of serine for asparagine at position 6 in the porcine sequence. Dynorphin B (rimorphin), which is adjacent to prodynorphin C-fragment in the precursor, was isolated from the same extract. Amino acid analysis and elution position on a gel filtration column confirmed its structure as that previously characterized from bovine pituitary extracts. The release of prodynorphin C-fragment and the C-terminus of dynorphin B from the porcine precursor would require cleavage at a single arginine residue. However, a terminal arginine was not present on either of these prodynorphin peptides isolated from bovine caudate. The data would suggest that processing at a single arginine residue results in elimination of the arginine, a feature in common with processing at paired basic residues. Topics: Animals; Cattle; Caudate Nucleus; Dynorphins; Endorphins; Enkephalins; Protein Precursors; Rabbits; Radioimmunoassay | 1985 |
Neuropeptide processing by single-step cleavage: conversion of leumorphin (dynorphin B-29) to dynorphin B.
Dynorphin B (rimorphin) is formed from dynorphin B-29 (leumorphin) by the action of a thiol protease from rat brain membranes. This represents a "single-arginine cleavage" between threonine-13 and arginine-14 of the substrate. In isotope dilution experiments we find that the radioactivity from radiolabelled dynorphin B-29, which appears in dynorphin B during incubation with the enzyme preparation, is not diminished by addition of a high concentration of dynorphin B-Arg14. Moreover, in pulse-chase experiments, radioactivity that appeared in dynorphin B-Arg14 did not decrease, nor did the radioactivity in dynorphin B increase, after chasing with a high concentration of non-radioactive dynorphin B-29. These results indicate that although some dynorphin B-Arg14 is formed by the impure enzyme preparation, it is not an intermediate in the conversion of dynorphin B-29 to dynorphin B. Thus the formation of dynorphin B does not involve the action of a trypsin-like enzyme followed by removal of arginine-14 by a carboxypeptidase B-like enzyme. It appears that a single enzyme converts dynorphin B-29 to dynorphin B in a single step. Topics: Animals; Brain; Cell Membrane; Chromatography, High Pressure Liquid; Cysteine Endopeptidases; Dynorphins; Endopeptidases; Endorphins; Enkephalins; Iodine Radioisotopes; Kinetics; Protein Precursors; Rats | 1985 |
On the origin of Leu-enkephalin and Met-enkephalin in the rat neurohypophysis.
The posterior lobe of the pituitary contains large amounts of Leu- and Met-enkephalin (LE and ME, respectively). A marked depletion of ME (81.9%) and LE (94.5%) in the posterior pituitary occurred after transection of the pituitary stalk. This indicates that most, if not all, of the enkephalins are in processes of central neurons. In the present study, I attempted to determine the source(s) of the LE- and ME-containing fibers in the posterior pituitary by examining the effects of hypothalamic lesions or fiber transections on the LE and ME levels. Lesions of the hypothalamic paraventricular nuclei caused ME and LE levels in the posterior pituitary to decrease significantly (55.6% and 27.6%, respectively). Deafferentation of the medial basal hypothalamus (creating islands of tissue containing the ventromedial and arcuate nuclei) resulted in a marked reduction in LE (94.1%) and ME (54.7%). Treating neonatal rats with monosodium glutamate resulted in a selective destruction of arcuate nucleus neurons, but did not affect LE and ME concentrations in the posterior pituitary. Thus, about half of the ME in the posterior pituitary seems to be provided by neurons in the vicinity of the paraventricular and ventromedial nuclei, whereas only about one quarter of the LE in the posterior pituitary is in processes of the paraventricular nucleus neurons. The remainder of the LE is contributed to the posterior pituitary by neurons outside the medial basal hypothalamus, probably by the supraoptic nucleus neurons. These findings are consistent with the hypothesis that LE and ME may be localized in separate populations of nerve endings in the neurohypophysis and may have different roles. Topics: Animals; Arginine Vasopressin; beta-Endorphin; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalin, Methionine; Hypothalamus, Middle; Male; Paraventricular Hypothalamic Nucleus; Peptide Fragments; Pituitary Gland, Posterior; Protein Precursors; Rats; Rats, Inbred Strains; Rats, Inbred WKY; Sodium Glutamate | 1985 |
Endopeptidase in human cerebrospinal fluid which cleaves proenkephalin B opioid peptides at consecutive basic amino acids.
An endopeptidase releasing the common N-terminal hexapeptide, (Leu)-enkephalin-Arg6, from dynorphins A and B, and alpha-neoendorphin was purified from human cerebrospinal fluid. Purification involved ion-exchange chromatography (DEAE-Sepharose CL-6B), hydrophobic interaction chromatography (phenyl-Sepharose CL-4B) and molecular sieving (Sephadex G-100). The enzyme showed molecular heterogeneity. A major fraction had an apparent molecular weight of about 40,000. It had an optimum activity in the pH range of 6-8. The conversion of dynorphin A was not affected by EDTA or iodoacetate but strongly reduced in the presence of phenylmethyl-sulphonyl fluoride, suggesting the enzyme is a serine protease. Topics: Amino Acids; Chromatography, High Pressure Liquid; Chromatography, Ion Exchange; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalins; Humans; Hydrolysis; Protein Precursors; Radioimmunoassay; Serine Endopeptidases; Serine Proteinase Inhibitors | 1985 |
Steady state levels of pro-dynorphin-related end products in the striatum and substantia nigra of the adult rhesus monkey.
Analysis of an acid extract of the striatum of the rhesus monkey revealed that the molar ratio of dynorphin A(1-8)-sized material and dynorphin (A(1-17)-sized material is approximately 1:1. In addition, the molar ratios of the dynorphin A-related end products to both dynorphin B(1-13)-sized material and alpha-neo-endorphin-sized material were approximately 1:1. Fractionation of an acid extract of the substantia nigra by gel filtration and reverse phase HPLC revealed the following molar ratios for pro-dynorphin-related end products. The molar ratio of dynorphin A(1-8) to dynorphin A(1-17) is approximately 6:1. The molar ratios of dynorphin A-related end products to dynorphin B(1-13) and alpha-neo-endorphin were approximately 0.5 and 0.8, respectively. Comparisons between proteolytic processing patterns of pro-dynorphin in the striatum and the substantia nigra of the rhesus monkey are considered. In addition, comparisons between pro-dynorphin processing in the substantia nigra of the rhesus monkey and the substantia nigra of the rat are discussed. Topics: Animals; Chromatography, Gel; Chromatography, High Pressure Liquid; Corpus Striatum; Dynorphins; Endorphins; Enkephalins; Female; Macaca mulatta; Peptide Fragments; Protein Precursors; Radioimmunoassay; Substantia Nigra | 1985 |
Evidence for a selective processing of proenkephalin B into different opioid peptide forms in particular regions of rat brain and pituitary.
The distribution of five major products of proenkephalin B [dynorphin1-17, dynorphin B, dynorphin1-8, alpha-neo-endorphin and beta-neo-endorphin] was studied in regions of rat brain and pituitary. The distribution pattern of immunoreactive (ir) dynorphin B (= rimorphin) was found to be similar to that of ir-dynorphin1-17, with the highest concentrations being present in the posterior pituitary and the hypothalamus. HPLC and gel filtration showed the tridecapeptide dynorphin B to be the predominant immunoreactive species recognized by dynorphin B antibodies in all brain areas and in the posterior pituitary. In addition, two putative common precursor forms of dynorphin B and dynorphin1-17 with apparent molecular weights of 3,200 and 6,000 were detected in brain and the posterior pituitary. The 3,200 dalton species coeluted with dynorphin1-32 on HPLC. In contrast with all other tissues, anterior pituitary ir-dynorphin B and ir-dynorphin1-17 consisted exclusively of the 6,000 dalton species. Concentrations of dynorphin1-8 were several times higher than those of dynorphin1-17 in striatum, thalamus, and midbrain while posterior pituitary, hypothalamus, pons/medulla, and cortex contained roughly equal concentrations of these two opioid peptides. No dynorphin1-8 was detected in the anterior pituitary. Concentrations of beta-neo-endorphin were similar to those of alpha-neo-endorphin in the posterior pituitary. In contrast, in all brain tissues alpha-neo-endorphin was found to be the predominant peptide, with tissue levels in striatum and thalamus almost 20 times higher than those of beta-neo-endorphin. These findings indicate that differential proteolytic processing of proenkephalin B occurs within different regions of brain and pituitary. Moreover, evidence is provided that, in addition to the paired basic amino acids -Lys-Arg- as the "typical" cleavage site for peptide hormone precursors, other cleavage signals also seem to exist for the processing of proenkephalin B. Topics: Amino Acid Sequence; Animals; beta-Endorphin; Brain; Chromatography, High Pressure Liquid; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalins; Male; Molecular Weight; Organ Specificity; Peptide Fragments; Pituitary Gland; Pituitary Gland, Posterior; Protein Precursors; Radioimmunoassay; Rats; Rats, Inbred Strains | 1984 |
Opioid receptor selectivity of dynorphin gene products.
In the guinea-pig ileum myenteric plexus-longitudinal muscle preparation, products from the dynorphin gene fell into three groups according to their potency. Dynorphin A was the most potent; dynorphin-32, dynorphin B, dynorphin B-29 and alpha-neo-endorphin were about equipotent and 10 to 20 times less potent than dynorphin A; dynorphin A-(1-8) and beta-neo-endorphin were about 200 times less potent than dynorphin A. Dynorphin A (a kappa agonist) was about 10 times less sensitive to antagonism by naloxone (as measured by naloxone Ke) than was normorphine (a mu agonist). Ke values for dynorphin-32, dynorphin B and alpha-neo-endorphin were the same as for dynorphin A, indicating that these peptides are also highly selective kappa agonists. Dynorphin A-(1-8), dynorphin B-29 and beta-neo-endorphin had Ke values intermediate between dynorphin A and normorphine, suggesting that they interact at both kappa and mu receptors. Addition of peptidase inhibitors to the bathing medium increased the potencies of dynorphin B, dynorphin B-29, alpha-neo-endorphin, dynorphin A-(1-8) and beta-neo-endorphin, but not of dynorphin A, dynorphin-32 or normorphine. The inhibitors did not change the naloxone Ke for dynorphin A or normorphine, or for dynorphin B-29, dynorphin A-(1-8) and beta-neo-endorphin, suggesting that the intermediate values were not caused by degradation to products with different receptor selectivities from the parent compounds. Ke for dynorphin-32, dynorphin B and alpha-neo-endorphin changed from being the same as dynorphin A in the absence of inhibitors to intermediate between dynorphin A and normorphine in the presence of inhibitors.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: Animals; Dynorphins; Endorphins; Enkephalin, Leucine; Guinea Pigs; Ileum; In Vitro Techniques; Male; Muscle Contraction; Muscle, Smooth; Myenteric Plexus; Naloxone; Naltrexone; Peptide Fragments; Protein Precursors; Receptors, Opioid | 1984 |
Proenkephalin B (prodynorphin)-derived opioid peptides: evidence for a differential processing in lobes of the pituitary.
The distribution of peptides derived from the novel opioid peptide precursor proenkephalin B (prodynorphin) was studied in lobes of the pituitary with antibodies against alpha-neoendorphin (alpha-neo-E) beta-neoE, dynorphin (DYN)-(1-17), DYN-(1-8), and DYN B in combination with gel filtration and high pressure liquid chromatography. In the posterior pituitary, all five opioid peptides occurred in high and about equimolar concentrations, whereas putative precursor peptides were found in only minor quantities. In contrast, in the anterior pituitary immunoreactive (ir-) DYN-(1-17) and ir-DYN B consisted exclusively of a common precursor species with a mol wt of about 6000. Six thousand-dalton DYN may be comprised of the C-terminal portion of proenkephalin B, with the sequence of DYN-(1-17) at its N-terminus. Moreover, the major portions of ir-alpha-neo-E and ir-beta-neoE in the anterior pituitary were found to be of an apparent mol wt of 8000. These findings indicate a differential processing of the opioid peptide precursor proenkephalin B in the two lobes of the pituitary. The anterior pituitary seems to process proenkephalin B predominantly into high mol wt forms of neo-E and DYNs, whereas in the posterior pituitary proenkephalin B undergoes further proteolytic processing to the smaller opioid peptides alpha-neo-E, beta-neo-E, DYN-(1-17), DYN-(1-8), and DYN B. Thus, processing differences may enable the selective liberation of different (opioid) peptides with distinct biological properties from one precursor within different tissues. Topics: Animals; beta-Endorphin; Chromatography, High Pressure Liquid; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalins; Male; Molecular Weight; Peptide Fragments; Pituitary Gland; Pituitary Gland, Anterior; Pituitary Gland, Posterior; Protein Precursors; Radioimmunoassay; Rats; Rats, Inbred Strains; Tissue Distribution | 1984 |
Human leumorphin is a potent, kappa opioid receptor agonist.
The opioid activity and opioid receptor type specificity of synthetic human leumorphin were studied in vitro. Human leumorphin inhibited the contraction of the myenteric plexus-longitudinal muscle preparation of the guinea pig ileum and was similar in opioid potency to porcine leumorphin. This action of human leumorphin was antagonized less effectively by naloxone than by the opiate antagonist Mr 2266. Human leumorphin also inhibited the contraction of the rabbit vas deferens which has only the kappa-type opioid receptor. It is concluded that human leumorphin has potent opioid activity and acts as an agonist at the kappa-type opioid receptor, like porcine leumorphin and other peptides derived from preproenkephalin B. Topics: Animals; Benzomorphans; Depression, Chemical; Dynorphins; Endorphins; Enkephalins; Guinea Pigs; Humans; Ileum; Male; Naloxone; Protein Precursors; Rabbits; Receptors, Opioid; Receptors, Opioid, kappa; Swine; Vas Deferens | 1984 |
[Endogenous morphine-like substances: biogenesis, release and degradation].
Topics: Animals; DNA, Recombinant; Dynorphins; Endorphins; Enkephalins; Gene Expression Regulation; Humans; In Vitro Techniques; Pituitary Gland; Protease Inhibitors; Protein Biosynthesis | 1984 |
Primate model of Parkinson's disease: alterations in multiple opioid systems in the basal ganglia.
A motor disorder similar to idiopathic Parkinson's Disease develops in rhesus monkeys after several daily repeated doses of N-methyl-4-phenyl, 1,2,3,6-tetrahydropyridine (MPTP). The concentrations of peptides derived from proenkephalin A, proenkephalin B, substance P and somatostatin were measured by specific radioimmunoassays in the basal ganglia of MPTP-treated monkeys. In MPTP-treated monkeys, dynorphin B concentration was reduced in the caudate. In the putamen, the concentrations of peptides derived from both proenkephalin A and proenkephalin B were decreased. In the globus pallidus, the concentrations of all opioid peptides tend to be increased, reaching significance only for alpha-neo-endorphin. In the substantia nigra, only Met-enkephalin concentration was reduced, while other peptides derived from either proenkephalin A or proenkephalin B were not changed. Substance P and somatostatin were not changed in any brain area examined. Some of the symptoms associated with Parkinson's Disease may be related to altered activity of endogenous opiates in basal ganglia. Topics: Animals; Basal Ganglia; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalin, Methionine; Female; Macaca mulatta; Male; Nucleus Accumbens; Parkinson Disease; Protein Precursors; Substantia Nigra | 1984 |
Ontogenetic development of the pro-enkephalin B (= pro-dynorphin) opioid peptide system in the rat pituitary.
The postnatal development of several pro-enkephalin-B-derived opioid peptides - dynorphin 1-17, dynorphin 1-8, dynorphin B, alpha-neo-endorphin and beta-neo-endorphin - was examined in rat pituitary lobes. The concentrations of pro-enkephalin-B-derived peptides from the anterior pituitary were between 4- and 12-fold and those from the neurointermediate pituitary between 17- and 122-fold lower in newborn as compared to adult rats. Similarly, the concentrations of vasopressin in the neurointermediate pituitary increased 50-fold between birth and adulthood; those of oxytocin, however, increased more than 540-fold over this period. The molecular weight pattern of dynorphin 1-17, dynorphin 1-8, dynorphin B, alpha- and beta-neo-endorphin-immunoreactive peptides in the anterior and neurointermediate pituitary did not differ between 3-day-old pups and adult rats. In the neurointermediate pituitary, the major immunoreactive components had the same chromatographic properties as synthetic dynorphin 1-17, dynorphin 1-8, dynorphin B, alpha- and beta-neo-endorphin, respectively, on gel filtration and high-performance liquid chromatography (HPLC). This indicates that neonatal rats were already capable of processing the precursor pro-enkephalin B into these various opioid peptides. In newborn rats, however, the amount of dynorphin 1-8 in the neurointermediate pituitary was three times lower than that of its putative intermediate precursor peptide dynorphin 1-17. Similarly, the amount of beta-neo-endorphin was almost four times lower than that of its putative precursor alpha-neo-endorphin. In contrast, in the neurointermediate pituitary of adult rats, dynorphin 1-17 and dynorphin 1-8, in addition to a alpha- and beta-neo-endorphin, occurred in equimolar amounts.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: Animals; Animals, Newborn; Cell Differentiation; Chromatography, High Pressure Liquid; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalins; Female; Male; Molecular Weight; Oxytocin; Peptide Fragments; Peptides; Pituitary Gland, Anterior; Pituitary Gland, Posterior; Pregnancy; Protein Precursors; Rats; Rats, Inbred Strains; Vasopressins | 1984 |
Immunoreactive pro-enkephalin and prodynorphin products are differentially distributed within the nucleus of the solitary tract of the rat.
In this study we examined the distribution of two different endogenous opioid peptides in the nucleus of the solitary tract of the rat medulla. As a marker for immunoreactive enkephalin, we used an antiserum directed against one of the proenkephalin products, methionine enkephalin-arg-gly-leu (m-Enk). To identify immunoreactive dynorphin we used an antiserum directed against the prodynorphin product, dynorphin B (Dyn B). The PAP method was used on both colchicine and normal animals. Caudal to the obex, within the commissural nucleus, there is extensive overlap of both immunoreactive m-Enk and Dyn B terminals and cells. While the cells are morphologically similar, the immunoreactive dynorphin cells are somewhat larger. Rostral to the obex, there is a marked difference in the distribution of the two compounds. Immunoreactive m-Enk terminals are concentrated medial to the solitary tract; there is minimal staining laterally. In contrast, immunoreactive Dyn B terminals are concentrated lateral to the solitary tract. The rostral cellular distribution of the two opioid peptides follows a similar pattern. The morphology of the medially located m-Enk and laterally located Dyn B cells is also readily distinguished. The former are small, round cells with minimal dendritic labelling; the latter are larger, pyramidal neurons with prominent apical and basal dendrites. Since the medial and lateral nuclei of the solitary tract have been associated with cardiovascular and respiratory control, respectively, these data suggest that different endorphin families have different functional actions within the nucleus of the solitary tract. Topics: Animals; Cardiovascular System; Cats; Chemoreceptor Cells; Dynorphins; Endorphins; Enkephalin, Methionine; Medulla Oblongata; Neurons, Afferent; Pressoreceptors; Rats; Respiratory System; Species Specificity; Vagus Nerve | 1984 |
Localization and quantitation of dynorphin B in the rat hippocampus.
The present study measures the content of dynorphin B in the rat hippocampus, and localizes the dynorphins within the intrinsic hippocampal neuronal circuitry. The level of dynorphin B, which is representative of the prodynorphin-derived peptides, was markedly depleted by intrahippocampal injection of colchicine, which destroyed the great majority of the hippocampal granule cells and the associated mossy fiber pathway. The hippocampus contralateral to the injection demonstrated a slight, non-significant rise in dynorphin B levels after colchicine. Entorhinal cortical lesions ablating the perforant pathway input to the hippocampus did not significantly alter dynorphin B levels in the hippocampus. Unilateral fimbrial transection caused a small but significant increase in dynorphin B on the side of the lesion relative to the unlesioned side, but neither side was significantly different from control. Topics: Animals; Colchicine; Dynorphins; Endorphins; Enkephalin, Methionine; Hippocampus; Male; Radioimmunoassay; Rats; Rats, Inbred Strains | 1984 |
Analgesia induced by intrathecal injection of dynorphin B in the rat.
A dose-dependent analgesic effect of intrathecally injected dynorphin B was observed in rats using the tail flick as nociceptive test. Intrathecal injection of 20 nmol of dynorphin B increased the tail flick latency by 90 +/- 23%, an effect that lasted about 90 min. For the same degree of analgesia, dynorphin B was 50% more potent than morphine on a molar basis. The analgesic effect of this dose of dynorphin B was partially blocked by 10 mg/kg, but not by 1 mg/kg, of subcutaneous naloxone, showing a relative resistance to naloxone reversal as compared with morphine analgesia. The analgesia produced by dynorphin B was unchanged in morphine-tolerant rats but was significantly decreased in rats tolerant to ethylketazocine. These results suggest that dynorphin B produces its potent analgesic effect by activation of kappa rather than mu opioid receptors in the rat spinal cord. Topics: Analgesics; Animals; Cyclazocine; Dose-Response Relationship, Drug; Drug Tolerance; Dynorphins; Endorphins; Enkephalin, Leucine; Ethylketocyclazocine; Female; Injections, Spinal; Morphine; Naloxone; Rats | 1984 |
Distribution of immunoreactive dynorphin B in discrete areas of the rat brain and spinal cord.
The distribution of immunoreactive (ir)-dynorphin B in 101 microdissected rat brain and spinal cord regions was determined using a specific radioimmunoassay. The highest concentration of dynorphin B in brain was found in the substantia nigra (1106.2 fmol/mg protein). Very high concentrations of ir-dynorphin B (greater than 400 fmol/mg protein) were also found in the lateral preoptic area, parabrachial nuclei and globus pallidus. Relatively high concentrations of ir-dynorphin B (250-400 fmol/mg protein) were found in 19 nuclei, including the periaqueductal gray matter, anterior hypothalamic nucleus, median eminence, nucleus accumbens and hippocampus. Moderate levels of the peptide (between 100 and 250 fmol/mg protein) were found in 42 brain nuclei such as the perifornical nucleus, nucleus of the diagonal band, medial forebrain bundle, and dorsal premamillary nucleus. Low concentrations of ir-dynorphin B (less than 100 fmol/mg protein) were found in 28 brain areas, e.g. cerebral cortical structures (parietal, cingulate, frontal), claustrum, olfactory bulb, lateral and periventricular thalamic nuclei. The cerebellar cortex has the lowest dynorphin B concentration (53.7 fmol/mg protein). Spinal cord segments exhibit low or moderate (cervical segment) levels of the peptide. The neurointermediate lobe of the pituitary gland is extremely rich in ir-dynorphin B (11,047.1 fmol/mg protein). Topics: Animals; Brain Chemistry; Diencephalon; Dynorphins; Endorphins; Enkephalin, Leucine; Hypothalamus; Male; Medulla Oblongata; Mesencephalon; Pituitary Gland; Pons; Radioimmunoassay; Rats; Rats, Inbred Strains; Spinal Cord; Telencephalon | 1984 |
Rimorphin (dynorphin B) exists together with alpha-neo-endorphin and dynorphin (dynorphin A) in human hypothalamus.
Rimorphin (dynorphin B) has been demonstrated to exist together with alpha-neo-endorphin and dynorphin(1-17) (dynorphin A) in the human hypothalamus. The content of rimorphin was comparable to that of alpha-neo-endorphin and somewhat higher than that of dynorphin. This result is quite similar to the recent observations in bovine, porcine and rat neural tissues, suggesting that rimorphin is derived from preproenkephal in B together with alpha-neo-endorphin and dynorphin in man. Topics: Adult; Chromatography, High Pressure Liquid; Dynorphins; Endorphins; Enkephalin, Leucine; Female; Humans; Hypothalamus; Protein Precursors | 1983 |
Regional distribution of dynorphin and neo-endorphin peptides in rat brain, spinal cord, and pituitary.
Five products of the dynorphin gene--alpha-neo-endorphin, beta-neo-endorphin, dynorphin A, dynorphin A-(1-8), and dynorphin B--were measured in various regions of rat brain and in rat spinal cord and pituitary. Specific antisera were used, supplemented by gel permeation analysis and high performance liquid chromatography, confirming the presence of dynorphin-32, dynorphin A, and dynorphin B in rat brain. In whole brain, alpha-neo-endorphin, dynorphin A-(1-8), and dynorphin B are present in much greater amounts than beta-neo-endorphin or dynorphin A. Although a general parallelism was found in the distribution of the five peptides, there were also noteworthy exceptions, suggesting that differential processing may occur. Topics: Animals; beta-Endorphin; Brain Chemistry; Chromatography, Gel; Chromatography, High Pressure Liquid; Dynorphins; Endorphins; Enkephalin, Leucine; Immune Sera; Male; Peptide Fragments; Peptides; Pituitary Gland; Protein Precursors; Radioimmunoassay; Rats; Rats, Inbred Strains; Spinal Cord; Tissue Distribution | 1983 |
Comparison of the action of putative endogenous kappa-agonists, leumorphin and rimorphin in vitro.
Leumorphin and rimorphin (dynorphin B) were nearly equipotent in inhibiting the contraction of the myenteric plexus-longitudinal muscle preparation of the guinea pig ileum and the rabbit vas deferens. Leumorphin had long duration of action in vitro after wash-out, whereas the duration of action of rimorphin was relatively shorter. These action of leumorphin and rimophin were antagonized less effectively by naloxone than by Mr2266, an antagonist relatively specific for the kappa-receptor. Rimorphin as well as leumorphin inhibits the contraction of the rabbit vas deferens which contains solely kappa-receptors. It is concluded that leumorphin and rimorphin have equipotent opioid activity and act at the kappa-receptor, like other opioid peptides derived from preproenkephalin B. Topics: Animals; Biological Assay; Dynorphins; Electric Stimulation; Endorphins; Enkephalin, Leucine; Enkephalins; Guinea Pigs; Ileum; Male; Muscle Contraction; Muscle, Smooth; Naloxone; Protein Precursors; Rabbits; Receptors, Opioid; Receptors, Opioid, kappa; Vas Deferens | 1983 |
Synthesis of a newly isolated opioid tridecapeptide, rimorphin, from pituitary using a trifluoroacetic acid-thioanisole deprotection system.
Topics: Amino Acid Sequence; Analgesics; Animals; Dynorphins; Endorphins; Enkephalin, Leucine; Guinea Pigs; In Vitro Techniques; Mice; Muscle Contraction; Muscle, Smooth | 1983 |
Leumorphin is a novel endogenous opioid peptide derived from preproenkephalin B.
Using synthetic leumorphin, we obtained antisera for leumorphin and set up two radioimmunoassays (RIAs) with different specificities. Gel exclusion chromatography coupled with the two RIAs showed the existence of a considerable amount of leumorphin-like peptide in water extracts from porcine neuro-intermediate pituitaries. Reverse phase high performance liquid chromatography revealed that leumorphin-like peptide in the water extracts was indistinguishable from synthetic leumorphin. These results along with potent opioid activity of leumorphin indicate that leumorphin is a novel endogenous opioid peptide derived from preproenkephalin B. Topics: Animals; Chemical Phenomena; Chemistry; Chromatography, Gel; Chromatography, High Pressure Liquid; Dynorphins; Electrophoresis, Polyacrylamide Gel; Endorphins; Enkephalin, Leucine; Enkephalins; Pituitary Gland; Protein Precursors; Radioimmunoassay; Swine | 1983 |
Rimorphin, a unique, naturally occurring [Leu]enkephalin-containing peptide found in association with dynorphin and alpha-neo-endorphin.
The tridecapeptide NH2-Tyr-Gly-Gly-Phe-Leu-Arg-Arg-Gln-Phe-Lys-Val-Val-Thr-COOH has been purified from extracts of bovine posterior pituitary glands. This unique peptide, which has been given the name "rimorphin," is a major [Leu]enkephalin-containing peptide in all tissues examined that contain dynorphin and alpha-neo-endorphin. However, except for the initial hexapeptide sequence, it is structurally unrelated to the other two peptides. Topics: Amino Acid Sequence; Animals; Dynorphins; Endorphins; Enkephalin, Leucine; Hypothalamus; Pituitary Gland; Pituitary Gland, Posterior; Rats; Spinal Cord; Swine | 1982 |
A specific radioimmunoassay for the opioid peptide dynorphin B in neural tissues.
Dynorphin-32 was recently isolated from porcine pituitary and shown to consist of dynorphin A (the originally isolated dynorphin heptadecapeptide) at the amino terminus, followed by Lys-Arg (a putative signal for proteolytic cleavage) and then a tridecapeptide, dynorphin B, at the carboxyl terminus. Dynorphin B, like dynorphin A, contains leucine enkephalin. The present report describes and validates a radioimmunoassay for dynorphin B using an antiserum, "13S", that does not crossreact with other known opioid peptides. Immunoreactive dynorphin B was estimated in rat and porcine neural tissues and found to be 2-3 fold higher than reported values for dynorphin A. Distribution based on tissue concentration was similar to that of dynorphin A, with very high concentrations in neurointermediate pituitary. Small quantities of antiserum "13S" are available to investigators upon request. Topics: Animals; Brain Chemistry; Dynorphins; Endorphins; Enkephalin, Leucine; Immune Sera; Male; Pituitary Gland; Radioimmunoassay; Rats; Rats, Inbred Strains; Spinal Cord; Tissue Distribution | 1982 |