Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 70: 236-46, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23415633

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARγ) is emerging as a new pharmacotherapeutic target for chronic pain. When oral (3-30 mg/kg/day in chow for 7 wk) or twice-daily intraperitoneal (1-10 mg/kg/day for 2 wk) administration began before spared nerve injury (SNI), pioglitazone, a PPARγ agonist, dose-dependently prevented multiple behavioral signs of somatosensory hypersensitivity. The highest dose of intraperitoneal pioglitazone did not produce ataxia or reductions in transient mechanical and heat nociception, indicating that inhibitory effects on hypersensitivity were not secondary to adverse drug-induced behaviors or antinociception. Inhibitory effects on hypersensitivity persisted at least one week beyond cessation of pioglitazone administration, suggestive of long-lasting effects on gene expression. Blockade of PPARγ with GW9662, an irreversible and selective PPARγ antagonist, dose-dependently reduced the inhibitory effect of pioglitazone on hypersensitivity, indicating a PPARγ-dependent action. Remarkably, a single preemptive injection of pioglitazone 15 min before SNI attenuated hypersensitivity for at least 2 weeks; this was enhanced with a second injection delivered 12 h after SNI. Pioglitazone injections beginning after SNI also reduced hypersensitivity, albeit to a lesser degree than preemptive treatment. Intraperitoneal pioglitazone significantly reduced the nerve injury-induced up-regulation of cd11b, GFAP, and p-p38 in the dorsal horn, indicating a mechanism of action involving spinal microglia and/or astrocyte activation. Oral pioglitazone significantly reduced touch stimulus-evoked phospho-extracellular signal-related kinase (p-ERK) in lamina I-II, indicating a mechanism of action involving inhibition of central sensitization. We conclude that pioglitazone reduces spinal glial and stimulus-evoked p-ERK activation and that PPARγ activation blocks the development of and reduces established neuropathic pain.


Subject(s)
Neuralgia/physiopathology , PPAR gamma/physiology , Thiazolidinediones/therapeutic use , Anilides/pharmacology , Animals , Ataxia/chemically induced , CD11b Antigen/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Extracellular Signal-Regulated MAP Kinases/metabolism , Glial Fibrillary Acidic Protein/metabolism , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Hyperalgesia/prevention & control , Male , Neuralgia/drug therapy , Neuralgia/prevention & control , PPAR gamma/agonists , PPAR gamma/antagonists & inhibitors , Pioglitazone , Posterior Horn Cells/drug effects , Posterior Horn Cells/metabolism , Rats , Thiazolidinediones/administration & dosage , Thiazolidinediones/antagonists & inhibitors , Thiazolidinediones/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Neuroscience ; 165(3): 914-22, 2010 Feb 03.
Article in English | MEDLINE | ID: mdl-19879928

ABSTRACT

We used a new computer-assisted method to precisely localize and efficiently quantify increases in neuropeptide Y immunoreactivity (NPY-ir) along the mediolateral axis of the L4 dorsal horn (DH) following transection of either the tibial and common peroneal nerves (thus sparing the sural branch, spared nerve injury (SNI)), the tibial nerve, or the common peroneal and sural nerves. Two weeks after SNI, NPY-ir increased within the tibial and peroneal innervation territories; however, NPY-ir in the central-lateral region (innervated by the spared sural nerve) was indistinguishable from that of sham. Conversely, transection of the sural and common peroneal nerves induced an increase in NPY-ir in the central-lateral region, while leaving the medial region (innervated by the tibial nerve) unaffected. All nerve injuries increased NPY-ir in dorsal root ganglia (DRG) and nucleus gracilis (NG). By 24 weeks, both NPY-ir upregulation in the DH and hyper-responsivity to cold and noxious mechanical stimuli had resolved. Conversely, NPY-ir in DRG and NG, and hypersensitivity to non-noxious static mechanical stimuli, did not resolve within 24 weeks. Over this time course, the average cross-sectional area of NPY-immunoreactive DRG neurons increased by 151 mum(2). We conclude that the upregulation of NPY after SNI is restricted to medial zones of the DH, and therefore cannot act directly upon synapses within the more lateral (sural) zones to control sural nerve hypersensitivity. Instead, we suggest that NPY in the medial DH tonically inhibits hypersensitivity by interrupting mechanisms of central sensitization and integration of sensory signals at the spinal and supraspinal levels.


Subject(s)
Neurons/metabolism , Neuropeptide Y/metabolism , Pain/metabolism , Posterior Horn Cells/metabolism , Sciatic Nerve/injuries , Spinal Cord/metabolism , Animals , Cold Temperature , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Lumbar Vertebrae , Male , Medulla Oblongata/metabolism , Medulla Oblongata/pathology , Neurons/pathology , Pain/etiology , Peroneal Nerve/injuries , Physical Stimulation , Posterior Horn Cells/pathology , Rats , Rats, Sprague-Dawley , Spinal Cord/pathology , Sural Nerve/injuries , Tibial Nerve/injuries , Time Factors
3.
Neuroscience ; 151(4): 948-54, 2008 Feb 19.
Article in English | MEDLINE | ID: mdl-18222040

ABSTRACT

The medial thalamus contains abundant mu-opioid receptors and is activated by acute morphine administration. However, the role of the medial thalamus in the rewarding effects of morphine is unclear. The present study examined whether mu-opioid receptors of the medial thalamus influenced the acquisition and expression of morphine-induced conditioned place preference (CPP) in rats. An unbiased apparatus and biased subject assignment were used. Administration of morphine in increasing doses (2 mg/kg, 4 mg/kg, 6 mg/kg, 10 mg/kg, s.c.) was paired with an initially non-preferred chamber and saline administration was paired with an initially preferred chamber. Conditioning trials were conducted twice daily for 4 days. Microinjection of the irreversible mu-opioid receptor antagonist, beta-funaltrexamine (5 microg/rat), into the medial thalamus 23 h prior to each morphine conditioning completely blocked the acquisition of CPP. However, microinjection of beta-funaltrexamine into the medial thalamus after morphine conditioning trials, but 23 h prior to a test session, had no effect on the expression of CPP. It is concluded that mu-opioid receptors in the rat medial thalamus are involved in the acquisition, but not expression, of morphine-induced CPP.


Subject(s)
Conditioning, Operant/drug effects , Morphine/pharmacology , Narcotics/pharmacology , Receptors, Opioid/physiology , Thalamus/drug effects , Animals , Behavior, Animal/drug effects , Conditioning, Operant/physiology , Drug Administration Routes , Drug Interactions , Male , Microinjections/methods , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Rats , Rats, Sprague-Dawley , Thalamus/physiology
4.
Neuroscience ; 135(3): 887-96, 2005.
Article in English | MEDLINE | ID: mdl-16154285

ABSTRACT

Endomorphins represent a group of endogenous opioid peptides with high affinity for the mu-opioid receptor. In the brainstem, Endomorphin-2 is found in trigeminal dorsal horn and the nuclei of the solitary tract, suggesting its presence in both nociceptive and visceral primary afferents. If Endomorphin-2 were an endogenous ligand for the mu-opioid receptor, we would expect to find the receptor at cellular sites in close association with the peptide. We used dual-labeling immunocytochemistry combined with electron microscopy to examine interactions between Endomorphin-2-immunoreactive and mu-opioid receptor-immunoreactive profiles within the nuclei of the solitary tract in the rat. Endomorphin-2-immunoreactivity was found primarily in unmyelinated axons and axon terminals in nuclei of the solitary tract and the majority of these terminals contained dense core vesicles. Endomorphin-2-immunoreactive axon terminals often formed asymmetric synapses with dendritic spines lacking mu-opioid receptor-immunoreactivity, but mu-opioid receptor-immunoreactivity was found in many of the larger dendritic targets of Endomorphin-2-immunoreactive terminals. Thus, mu-opioid receptor-immunoreactivity was found in the postsynaptic targets of Endomorphin-2-immunoreactive axon terminals, consistent with the hypothesis that Endomorphin-2 is an endogenous ligand for this receptor within the nuclei of the solitary tract. A small number of Endomorphin-2-immunoreactive somata, dendrites, and axon terminals also contained mu-opioid receptor-immunoreactivity. Cells that contain both the opioid peptide and its receptor may be a substrate for potential autoregulation of nuclei of the solitary tract neurons by opioid ligands. Finally, using tract tracing and confocal microscopy, we found Endomorphin-2-immunoreactivity in a subset of vagal afferents. Together these findings support the hypothesis that Endomorphin-2 is a ligand for the mu-opioid receptor within nuclei of the solitary tract and that the peptide is at least partially derived from primary visceral afferents.


Subject(s)
Dendrites/metabolism , Oligopeptides/physiology , Presynaptic Terminals/metabolism , Receptors, Opioid, mu/metabolism , Solitary Nucleus/metabolism , Animals , Dendrites/physiology , Dendrites/ultrastructure , Immunohistochemistry , Male , Microscopy, Confocal , Microscopy, Electron , Neurons, Afferent/metabolism , Presynaptic Terminals/ultrastructure , Rats , Rats, Sprague-Dawley , Solitary Nucleus/chemistry , Solitary Nucleus/ultrastructure
5.
Neuroscience ; 134(4): 1205-16, 2005.
Article in English | MEDLINE | ID: mdl-16039798

ABSTRACT

Transgenic mice ectopically expressing nerve growth factor in oligodendrocytes have high levels of nerve growth factor immunoreactivity in the white matter of the spinal cord from birth until 2 months of age. The nerve growth factor over-expression leads to the appearance of ectopic substance P containing sensory fibers in the white matter of the spinal cord that persist throughout the life of the animal. These transgenic mice have been found to display hypersensitivity to a thermal stimulus following a sensitizing pinch stimulus known to release endogenous substance P. Surprisingly, this hypersensitivity is completely reversed following the administration of morphine, to the extent that transgenic mice become less sensitive to pain than the wild type mice given morphine. Endomorphin-2, an endogenous opioid peptide, has been found co-localized with substance P in primary sensory fibers in the spinal cord. In this study, we show that the ectopic fibers also express endomorphin-2, and describe the postnatal development of such expression, as detected by immunocytochemistry. We confirmed that endomorphin-2 expression starts later in the postnatal period than substance P. Surprisingly, transgenic animals had delayed appearance of endomorphin-2 in the superficial dorsal horn, compared with wild type, and expressed particularly high levels of endomorphin-2 immunoreactivity in the ectopic fibers from postnatal days 10-30, coinciding with the peak of nerve growth factor expression in oligodendrocytes. Endomorphin-2 immunoreactivity was still readily detected in ectopic fibers of 120-day-old animals. Furthermore, we detected immunoreactivity for the mu-opioid receptor in the ectopic fibers, where it was co-localized with endomorphin-2 immunoreactivity. In the superficial dorsal horn, there were no apparent differences in the distribution and intensity of mu-opioid receptor immunoreactivity between wild type and transgenic animals. Taken together, these data could provide an explanation for the enhanced effect of opioid analgesics in transgenic mice, when compared with control mice, as well as provide the basis for studies of the postnatal development of the hyperalgesia and allodynia demonstrated by these animals.


Subject(s)
Nerve Growth Factor/biosynthesis , Neurons, Afferent/metabolism , Oligodendroglia/metabolism , Oligopeptides/biosynthesis , Pain/physiopathology , Spinal Cord/metabolism , Animals , Animals, Newborn , Female , Immunohistochemistry , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Receptors, Opioid, mu/metabolism , Substance P/biosynthesis
6.
Brain Res ; 969(1-2): 126-34, 2003 Apr 18.
Article in English | MEDLINE | ID: mdl-12676373

ABSTRACT

Endomorphins are endogenous opioid peptides with high affinity and selectivity for the mu-opioid receptor. In the present study, we examined the morphology of the endomorphin 2-like immunoreactive (EM2-LI) neurons in the hypothalamus at the light and electron microscopic levels. At the light microscopic level, EM2-LI neurons were found mostly distributed in the regions between the dorsomedial and ventromedial hypothalamic nuclei and the region near the third ventricle. At the electron microscopic level, EM2-LI perikarya could be divided into two groups. Type I perikarya contained relatively undeveloped endoplasmic reticulum and Golgi apparatus while type II perikarya contained well-developed rough-surfaced endoplasmic reticulum and Golgi apparatus. Both type I and type II neurons contained numerous EM2-LI dense-cored vesicles. Type II perikarya and dendrites received synapses and showed immunoreactivity in the endoplasmic reticulum and Golgi apparatus. EM2-LI axon terminals formed synapses with both immunonegative and immunopositive dendrites. In some cases, the axon terminals contained both immunonegative and immunopositive dense-cored vesicles. EM2-LI neurons often had synaptic relationships with neurons containing immunonegative dense-cored vesicles. Myelinated axon shafts containing EM2-LI were also found. This first demonstration of the ultrastructure and synaptic relationships of EM2-LI neurons in the hypothalamus provides morphological evidence that suggests (1) endomorphin 2-containing neurons modulate physiological function through synaptic relationships; (2) endomorphin 2 may coexist with other neurotransmitters in the same neurons; and (3) endomorphin 2-containing neurons could modulate other endomorphin 2-containing neurons as well as those containing other neurotransmitters.


Subject(s)
Neurons/metabolism , Oligopeptides/metabolism , Synapses/ultrastructure , Animals , Hypothalamus, Posterior/cytology , Hypothalamus, Posterior/metabolism , Hypothalamus, Posterior/ultrastructure , Immunohistochemistry , Inclusion Bodies/metabolism , Male , Microscopy, Immunoelectron , Neural Pathways/metabolism , Neural Pathways/ultrastructure , Neurons/cytology , Neurons/ultrastructure , Rats , Rats, Wistar , Synapses/metabolism
7.
Eur J Neurosci ; 16(6): 1075-82, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12383236

ABSTRACT

The present study characterizes the relationship between the endogenous mu opioid peptides endomorphin-1 (EM-1) and endomorphin-2 (EM-2) and several splice variants of the cloned mu opioid receptor (MOR-1) encoded by the mu opioid receptor gene (Oprm). Confocal laser microscopy revealed that fibers containing EM-2-like immunoreactivity (-LI) were distributed in close apposition to fibers showing MOR-1-LI (exon 4-LI) and to MOR-1C-LI (exons 7/8/9-LI) in the superficial laminae of the lumbar spinal cord. We also observed colocalization of EM-2-LI and MOR-1-LI in a few fibers of lamina II, and colocalization of EM-2-LI and MOR-1C-LI in laminae I-II, and V-VI. To assess the functional relevance of the MOR-1 variants in endomorphin analgesia, we examined the effects of antisense treatments that targeted individual exons within the Oprm1 gene on EM-1 and EM-2 analgesia in the tail flick test. This antisense mapping study implied mu opioid receptor mechanisms for the endomorphins are distinct from those of morphine or morphine-6beta-glucuronide (M6G).


Subject(s)
Alternative Splicing/genetics , Oligopeptides/metabolism , Pain/metabolism , Posterior Horn Cells/metabolism , Receptors, Opioid, mu/metabolism , Amino Acid Sequence/genetics , Animals , Base Sequence/genetics , Exons/genetics , Immunohistochemistry , Male , Mice , Mice, Inbred Strains , Oligopeptides/genetics , Oligopeptides/pharmacology , Pain/drug therapy , Pain/physiopathology , Posterior Horn Cells/cytology , Posterior Horn Cells/drug effects , Protein Structure, Tertiary/genetics , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/genetics
8.
Mol Cell Neurosci ; 20(4): 695-711, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12213449

ABSTRACT

UNLABELLED: 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. CONCLUSION: alpha9/alpha10nAChR is a target for modulation by endomorphin-1 and dynorphin B, efferent cotransmitters in the inner ear.


Subject(s)
Dynorphins/physiology , Ear, Inner/physiology , Endorphins/physiology , Neurotransmitter Agents/physiology , Oligopeptides/physiology , Receptors, Nicotinic/metabolism , Acetylcholine/pharmacology , Animals , Anura , Cochlea/drug effects , Cochlea/physiology , Dynorphins/pharmacology , Electric Conductivity , Endorphins/pharmacology , Enkephalin, D-Penicillamine (2,5)-/pharmacology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/physiology , In Vitro Techniques , Narcotic Antagonists , Oligopeptides/pharmacology , Oocytes/drug effects , Oocytes/metabolism , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/chemistry , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/physiology , Saccule and Utricle/cytology , Saccule and Utricle/drug effects , Saccule and Utricle/physiology , Synapses/drug effects , Synapses/physiology , Xenopus laevis
9.
Neuroscience ; 113(3): 593-605, 2002.
Article in English | MEDLINE | ID: mdl-12150779

ABSTRACT

Endomorphin-2 is a newly discovered endogenous opioid peptide with high affinity and selectivity for the micro-opioid receptor, and potent analgesic activity, particularly in the spinal cord. Using immunoelectron microscopy, we examined the ultrastructure of the endomorphin-2-like immunoreactive processes and their synaptic relationships in the spinal cord. Endomorphin-2-like immunopositive dense-cored vesicles were observed in many axon terminals, and, in a few cases, were observed together with immunonegative dense-cored vesicles. Immunopositive axons with or without myelination were also observed. The endomorphin-2-like immunoreactive axon terminals formed synapses with both immunopositive and immunonegative processes. Most synapses were asymmetrical, but symmetrical synapses were also found. Examples of axo-dendritic, axo-somatic and axo-axonic contacts were observed. This first demonstration of the ultrastructure and synaptic relationships of endomorphin-2-like immunoreactive axon terminals in the spinal cord dorsal horn provides morphological evidence that this peptide functions as a transmitter regulating pain processes.


Subject(s)
Microscopy, Immunoelectron , Oligopeptides/analysis , Posterior Horn Cells/chemistry , Posterior Horn Cells/ultrastructure , Animals , Cervical Vertebrae , Male , Oligopeptides/immunology , Presynaptic Terminals/chemistry , Presynaptic Terminals/ultrastructure , Rats , Rats, Wistar , Receptors, Opioid, mu/agonists , Synapses/chemistry , Synapses/ultrastructure
10.
Brain Res ; 914(1-2): 74-80, 2001 Sep 28.
Article in English | MEDLINE | ID: mdl-11578599

ABSTRACT

The present study examined the effect of endomorphin-1 (EM1), an endogenous opioid with a high affinity for the mu opiate receptor, on conditioned defeat. Conditioned defeat is a phenomenon in which hamsters that have been defeated subsequently fail to exhibit normal territorial aggression and instead display submissive/defensive behaviors even when paired with a non-aggressive intruder. In experiment 1, animals were placed in the home cage of a larger resident for 15 min and were defeated. After 24 h, animals received a 3-microl injection of EM1 (0.0, 0.3, 3.0, or 10 nmol) into the left lateral cerebral ventricle 5 min before a smaller non-aggressive intruder was placed in the home cage of the experimental animal. In experiment 2, animals were infused with EM1 immediately after the initial defeat and were paired with a non-aggressive intruder 24 h later as in experiment 1. EM1 reduced the duration of submissive/defensive behavior in experiment 1 (P<0.05) but not in experiment 2 (P>0.05). These data support the hypothesis that the highly selective mu receptor agonist endomorphin-1 modulates the expression of conditioned defeat, but provides no support for the hypothesis that endomorphin-1 modulates the consolidation of conditioned defeat.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Conditioning, Psychological/drug effects , Fear/drug effects , Oligopeptides/pharmacology , Receptors, Opioid, mu/drug effects , Stress, Psychological/metabolism , Animals , Anxiety/metabolism , Anxiety/physiopathology , Behavior, Animal/physiology , Brain/metabolism , Conditioning, Psychological/physiology , Cricetinae , Depression/metabolism , Depression/physiopathology , Dose-Response Relationship, Drug , Fear/physiology , Injections, Intraventricular , Male , Mesocricetus , Oligopeptides/metabolism , Receptors, Opioid, mu/metabolism , Stress, Psychological/physiopathology
11.
Eur J Pharmacol ; 427(3): 203-10, 2001 Sep 21.
Article in English | MEDLINE | ID: mdl-11567650

ABSTRACT

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


Subject(s)
Analgesics/pharmacology , Naloxone/analogs & derivatives , Naltrexone/analogs & derivatives , Oligopeptides/pharmacology , Animals , Benzylidene Compounds/pharmacology , Dose-Response Relationship, Drug , Dynorphins/immunology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enkephalin, Leucine/immunology , Enkephalin, Methionine/immunology , Immune Sera/pharmacology , Injections, Spinal , Injections, Subcutaneous , Male , Mice , Naloxone/pharmacology , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Pain/prevention & control , Pain Measurement , Pain Threshold/drug effects , Peptide Fragments/immunology , Time Factors
12.
Life Sci ; 69(11): 1305-12, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11521754

ABSTRACT

The blood-brain barrier (BBB), composed of the microvessels of cerebral capillary endothelial cells, regulates the passage of peptides into the brain in several ways, mainly by saturable transport or passive diffusion. Here we describe an additional mechanism by which this regulatory function can occur. Cerebral microvessels were isolated from different regions of the brain and incubated with the mu-opiate selective endomorphin-1 (Tyr-Pro-Trp-Phe-NH2) or the opiate-modulating Tyr-MIF-1 (Tyr-Pro-Leu-Gly-NH2), both tetrapeptides selectively tritiated at the Pro. Degradation was determined by HPLC. For both peptides, the metabolism by microvessels from the cerebral cortex was much greater than that by microvessels from the hypothalamus or pons. For endomorphin-1, the least degradation was in the pons; for Tyr-MIF-1 there was no difference in metabolism by microvessels from the pons or hypothalamus. The results show a novel mechanism at the BBB by which the BBB can selectively regulate the activity of different peptides in different regions of the brain.


Subject(s)
Blood-Brain Barrier/physiology , Cerebral Cortex/metabolism , MSH Release-Inhibiting Hormone/metabolism , Oligopeptides/metabolism , Animals , Capillaries/metabolism , Cerebral Cortex/blood supply , Chromatography, High Pressure Liquid , Endothelium, Vascular/metabolism , Hypothalamus/blood supply , Hypothalamus/metabolism , MSH Release-Inhibiting Hormone/analogs & derivatives , Male , Pons/blood supply , Pons/metabolism , Rats , Rats, Sprague-Dawley
13.
Neuroscience ; 105(3): 773-8, 2001.
Article in English | MEDLINE | ID: mdl-11516840

ABSTRACT

Nerve injury often leads to chronic, sometimes excruciating, pain. The mechanisms contributing to this syndrome include neurochemical plasticity in neurons involved in the earliest stages of pain transmission. Endomorphin-2 (Tyr-Pro-Phe-Phe-NH(2)) is an endogenous morphine-like substance that binds to the mu-opioid receptor with high affinity and selectivity. Endomorphin-2-like immunoreactivity (LI) is present in the superficial layers of the dorsal horn in the spinal cord and in primary afferents, suggesting a role for this peptide in pain transmission. To determine whether spinal endomorphin-2-LI is altered in an animal model of chronic pain, the left sciatic nerve of Swiss Webster and ICR mice was ligated in a modified Seltzer model of nerve injury. Changes in endomorphin-2-LI were assessed by immunocytochemistry at 2, 4 and 14 days after nerve injury. The side of the spinal cord ipsilateral to the nerve injury exhibited a dramatic decrease in endomorphin-2-LI relative to the contralateral side and to control animals. The change was restricted to the medial dorsal horn in the lumbar segments innervated by the sciatic nerve. Substance P-LI showed a small decrease, while calcitonin gene-related peptide-LI was unchanged. Both thermal hyperalgesia, as evidenced by significantly decreased paw withdrawal latencies, and decreased endomorphin-2-LI were observed within 2 days of injury and were most pronounced at 2 weeks after injury. The decrease in endomorphin-2-LI during the development of chronic pain is consistent with the loss of an inhibitory influence on pain transmission. These results provide the first evidence that reduction of an endogenous opioid in primary afferents is associated with injury-induced chronic pain.


Subject(s)
Down-Regulation/physiology , Neuralgia/metabolism , Oligopeptides/metabolism , Peripheral Nervous System Diseases/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Animals , Calcitonin Gene-Related Peptide/metabolism , Chronic Disease , Functional Laterality/physiology , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Immunohistochemistry , Ligation , Male , Mice , Mice, Inbred ICR , Nerve Crush , Neuralgia/physiopathology , Pain Measurement , Pain Threshold/physiology , Peripheral Nervous System Diseases/physiopathology , Posterior Horn Cells/cytology , Posterior Horn Cells/metabolism , Reaction Time/physiology , Sciatic Nerve/surgery , Substance P/metabolism
14.
Exp Brain Res ; 139(1): 70-5, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11482845

ABSTRACT

Opiate-modulating tetrapeptides such as tyrosine-melanocyte-stimulating hormone-release inhibiting factor-1 (Tyr-MIF-1; Tyr-Pro-Leu-Gly-NH2) and Tyr-W-MIF-1 (Tyr-Pro-Trp-Gly-NH2) are saturably transported from brain to blood. We examined whether two recently described endogenous opiate tetrapeptides with similar structures, the mu-specific endomorphins, also are transported across the blood-brain barrier (BBB). We found that the efflux rates of endomorphin-1 (Tyr-Pro-Trp-Phe-NH2) and endomorphin-2 (Tyr-Pro-Phe-Phe-NH2) were each self-inhibited by an excess of the respective endomorphin, thereby defining saturable transport. Cross-inhibition of the transport of each endomorphin by the other indicated shared transport. By contrast, no inhibition of the efflux of either endomorphin resulted from coadministration of Tyr-MIF-1, indicating that peptide transport system-1 (PTS-1) was not involved. Tyr-W-MIF-1, which is partially transported by PTS-1, significantly (P<0.01) decreased the transport of endomorphin-1 and tended (P=0.051) to decrease the transport of endomorphin-2, consistent with its role as both an opiate and antiopiate. Although involved in modulation of pain, coinjection of calcitonin gene-related peptide or constriction of the sciatic nerve did not appear to inhibit endomorphin efflux. Thus, the results demonstrate the existence of a new efflux system across the BBB which saturably transports endomorphins from brain to blood.


Subject(s)
Blood-Brain Barrier/physiology , Brain/metabolism , Carrier Proteins/metabolism , MSH Release-Inhibiting Hormone/analogs & derivatives , Oligopeptides/metabolism , Receptors, Opioid, mu/metabolism , Animals , Binding, Competitive/drug effects , Binding, Competitive/physiology , Blood-Brain Barrier/drug effects , Brain/drug effects , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide/pharmacology , Carrier Proteins/drug effects , Iodine Radioisotopes/pharmacokinetics , Ligation , MSH Release-Inhibiting Hormone/metabolism , MSH Release-Inhibiting Hormone/pharmacokinetics , Male , Membrane Transport Proteins/drug effects , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred ICR , Oligopeptides/pharmacokinetics , Pain/metabolism , Pain/physiopathology , Radioligand Assay , Receptors, Opioid, mu/drug effects , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology , Sciatic Nerve/surgery
15.
Life Sci ; 69(1): 67-73, 2001 May 25.
Article in English | MEDLINE | ID: mdl-11411806

ABSTRACT

A saturable blood-to-brain transport system for leptin across the blood-brain barrier (BBB) has been observed in vivo. Since the main component of the non-fenestrated microvessels of the BBB is the endothelial cell, we established an in vitro culture system of these cerebrovascular cells to study leptin transport and to determine whether the self-inhibition of leptin transport characteristic of a saturable system occurs at this level. The results show that 125I-leptin crossed from the luminal to abluminal side of a monolayer of cerebral microvessel cells significantly faster than the albumin and lactalbumin controls. This transport of 125I-leptin across an in vitro BBB was significantly faster than in the opposite direction and was dose-relatedly inhibited by the addition of unlabeled leptin. Thus, the results establish that the saturable transport system for leptin across the BBB occurs at the level of the endothelial cells of the BBB.


Subject(s)
Blood-Brain Barrier/physiology , Cerebrovascular Circulation/physiology , Endothelium, Vascular/physiology , Leptin/metabolism , Microcirculation/physiology , Animals , Biological Transport , Cell Membrane/metabolism , Cells, Cultured , Kinetics , Lactalbumin/metabolism , Leptin/blood , Male , Rats , Rats, Sprague-Dawley , Serum Albumin/metabolism
16.
Neurotox Res ; 3(3): 249-53, 2001 Jul.
Article in English | MEDLINE | ID: mdl-15111249

ABSTRACT

The use of genetically modified cells to deliver growth factors has been proposed as a possible treatment for neurodegeneration, including Parkinson's disease. Here we demonstrate that the implantation of fibroblasts genetically modified to secrete fibroblast growth factor-1 (FGF) increased striatal dopamine concentrations in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mouse model of Parkinson's disease.

17.
Brain Res Bull ; 56(6): 563-7, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11786242

ABSTRACT

Recently, endomorphin-1 (Tyr-Pro-Trp-Phe-NH2; EM1), an endogenous peptide that has high affinity and selectivity for the mu-opiate receptor, has been shown to modulate emotional behavior in mice and social behavior in Syrian hamsters. Endomorphin-1 (EM1) is present throughout the central nervous system in rats, mice, and guinea pigs; however, the distribution of EM1 in hamsters has not been described. The purpose of the present study was to investigate the distribution of EM1-like immunoreactivity (EM1L-IR) in the limbic system of Syrian hamsters using immunocytochemistry. Perikarya containing EM1L-IR were present in the anterior area, dorsomedial, ventromedial, periventricular, posterior, and arcuate nuclei of the hypothalamus. Fibers expressing EM1L-IR were present in the nucleus accumbens, caudate putamen, septum, bed nucleus of the stria terminalis, amygdaloid complex, and hypothalamus. The distribution of EM1 suggests a potential endogenous role for this peptide in major processes modulated by opiates, including affective states and social behavior.


Subject(s)
Limbic System/metabolism , Mesocricetus/metabolism , Neurons/metabolism , Oligopeptides/metabolism , Animals , Axons/metabolism , Axons/ultrastructure , Behavior, Animal/physiology , Cricetinae , Emotions/physiology , Immunohistochemistry , Limbic System/cytology , Mesocricetus/anatomy & histology , Neurons/cytology , Social Behavior
18.
Neuroimmunomodulation ; 8(3): 148-53, 2000.
Article in English | MEDLINE | ID: mdl-11124581

ABSTRACT

OBJECTIVE: Cytokine signaling is the key to fighting infection. Fever is elicited by the production of inflammatory cytokines, particularly interleukin-1beta (IL-1beta), and the subsequent action of cytokines in the hypothalamus. In old age, the ability to produce fever in response to infection or to peripheral injections of IL-1beta is diminished. Intracerebroventricular injections of IL-1beta can still produce a normal fever response in the aged. A logical hypothesis to explain this discrepancy is that passage of IL-1beta across the blood-brain barrier (BBB) is altered. METHOD: We used a quantitative in vivo technique, which previously showed a saturable system transporting IL-1beta across the BBB, to investigate the speed at which radiolabeled IL-1beta crosses from blood to brain in mice of widely different ages. RESULTS: We found that passage of IL-1beta across the BBB was significantly decreased in old (23-month) mice as compared with young (2-month) or middle-aged (12-month) animals. Passage of IL-1beta across the blood-testis barrier was not significantly different among the groups. The passage of radiolabeled albumin across the BBB was not increased in any group, ruling out any disruption of the BBB by IL-1beta. CONCLUSION: These results provide a mechanism that could help explain why fever production is reduced in old age and suggest an important role for the BBB in regulating immune changes.


Subject(s)
Aging/immunology , Blood-Brain Barrier/immunology , Fever/immunology , Fever/metabolism , Interleukin-1/metabolism , Animals , Biological Transport, Active/immunology , Biological Transport, Active/physiology , Blood-Brain Barrier/physiology , Male , Mice , Mice, Inbred ICR
19.
Brain Res ; 881(1): 1-8, 2000 Oct 20.
Article in English | MEDLINE | ID: mdl-11033087

ABSTRACT

To determine the role of spinal mu-opioid receptor subtypes in antinociception induced by intrathecal (i.t.) injection of endomorphin-1 and -2, we assessed the effects of beta-funaltrexamine (a selective mu-opioid receptor antagonist) naloxonazine (a selective antagonist at the mu(1)-opioid receptor) and a novel receptor antagonist (3-methoxynaltrexone) using the paw-withdrawal test. Antinociception of i.t. endomorphins and [D-Ala(2), MePhe(4), Gly(ol)(5)]enkephalin (DAMGO) was completely reversed by pretreatment with beta-funaltrexamine (40 mg/kg s.c.). Pretreatment with a variety of doses of i.t. or s.c. naloxonazine 24 h before testing antagonized the antinociception of endomorphin-1, -2 and DAMGO. Judging from the ID(50) values of naloxonazine, the antinociceptive effect of endomorphin-2 was more sensitive to naloxonazine than that of endomorphin-1 or DAMGO. The selective morphine-6beta-glucuronide antagonist, 3-methoxynaltrexone, which blocked endomorphin-2-induced antinociception at each dose (0.25 mg/kg s.c. or 2.5 ng i.t.) that was inactive against DAMGO, did not affect endomorphin-1-induced antinociception but shifted the dose-response curve of endomorphin-2 3-fold to the right. These findings may be interpreted as indicative of the existence of a novel mu-opioid receptor subtype in spinal sites, where antinociception of morphine-6beta-glucuronide and endomorphin-2 are antagonized by 3-methoxynaltrexone. The present results suggest that endomorphin-1 and endomorphin-2 may produce antinociception through different subtypes of mu-opioid receptor.


Subject(s)
Heroin/agonists , Naloxone/analogs & derivatives , Naltrexone/analogs & derivatives , Oligopeptides/antagonists & inhibitors , Pain Measurement/drug effects , Receptors, Opioid, mu/antagonists & inhibitors , Analgesics, Opioid/antagonists & inhibitors , Animals , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/antagonists & inhibitors , Injections, Spinal , Male , Mice , Naloxone/pharmacology , Naltrexone/pharmacology , Receptors, Opioid, mu/agonists
20.
Am J Respir Crit Care Med ; 162(3 Pt 1): 994-9, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10988119

ABSTRACT

The novel endogenous mu-opioid receptor (MOR) agonists endomorphin 1 (EM1) and 2 (EM2) were tested for their cardiorespiratory effects in conscious, freely behaving rats. After systemic (intravenous) administration of EM1, EM2, or the selective MOR agonist DAMGO, analgesia, minute ventilation (V E), heart rate (HR) and mean arterial blood pressure (BP) were measured. The threshold dose for analgesia was similar for all 3 peptides ( approximately 900 nmol/kg). All 3 compounds elicited biphasic V E responses, with marked, short-lived V E depressions (4-6 s) followed by more sustained V E increases (10-12 min). However, compared with responses elicited by EM2 or DAMGO, EM1 decreased V E only at higher doses, and produced greater V E stimulation. Morphine produced a V E decrease, but no subsequent V E increase. EM2 and DAMGO decreased HR and BP, while EM1 decreased HR, but did not decrease BP in conscious rats at doses up to 9,600 nmol/kg. In anesthetized rats, all 3 peptides decreased HR and BP. The decreases in V E, HR, and BP were blocked by the MOR antagonist, naloxone HCI (NIx). Only the HR and BP responses, however, were blocked by naloxone-methiodide (MeNIx), indicating central mediation of V E responses and peripheral mediation of cardiovascular responses. We conclude that MOR-selective compounds vary in their cardiorespiratory response characteristics which could be linked to differential cellular actions. The results support the concept that the analgesic, respiratory, and cardiovascular effects of MOR agonists can be dissociated and that EM1-like compounds could provide the basis for novel, safer analgesics.


Subject(s)
Analgesics, Opioid/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Heart Rate/drug effects , Morphine/pharmacology , Oligopeptides/pharmacology , Respiration/drug effects , Animals , Blood Pressure/drug effects , Dose-Response Relationship, Drug , Humans , Infant, Newborn , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...