Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 416
Filter
1.
Med J Malaysia ; 70(3): 208-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26248787

ABSTRACT

The purpose of this case study is to report the use of oral Bovril (a food supplement which contains arginine) as an alternative test for growth hormone stimulation test. We performed oral Bovril test in 3 patients -- one with suspected growth hormone deficiency in whom insulin tolerance test could not be performed (subject A), one sex-matched control (subject B), and one with confirmed growth hormone deficiency (subject C). 14g/m(2) of oral Bovril was mixed with 150ml of warm water and was given to all three subjects. Blood for growth hormone was taken at baseline, and every 30 minutes till 150 minutes after ingestion of oral Bovril. The ingestion of oral Bovril showed a positive response in subjects A and B, with highest growth hormone levels of 28.4mIU/L and 42.0mIU/L respectively at 150 minutes. Subject C had suppressed growth hormone throughout the test. Oral Bovril is readily available and is a safe alternative for standard growth hormone stimulation test.

2.
Med J Malaysia ; 67(1): 133-5, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22582570

ABSTRACT

Wasp stings can present in various ways, ranging from mild self-limiting illness to severe multi organ failure with a potentially fatal outcome. We report a case of multiple wasp stings leading to acute renal failure needing prolonged dialysis support and posterior reversible encephalopathy syndrome.


Subject(s)
Acute Kidney Injury/etiology , Insect Bites and Stings/complications , Posterior Leukoencephalopathy Syndrome/etiology , Wasps , Adult , Animals , Female , Humans
3.
J Pharmacol Exp Ther ; 334(3): 739-45, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20554907

ABSTRACT

Direct injection of double-stranded adeno-associated virus type 2 (dsAAV2) with a mu-opioid receptor (MOR) mutant [S4.45(196)A], and a reporter protein (enhanced green fluorescent protein) into the spinal cord (S2/S3) dorsal horn region of ICR mice resulted in antinociceptive responses to systemic injection of opioid antagonist naloxone without altering the acute agonist morphine responses and no measurable tolerance or dependence development during subchronic naloxone treatment. To develop further such mutant MORs into a therapeutic agent in pain management, a less invasive method for virus delivery is needed. Thus, in current studies, the dsAAV2 was locally injected into the subarachnoid space of the spinal cord by intrathecal administration. Instead of using the MORS196A mutant, we constructed the dsAAV2 vector with the MORS196ACSTA mutant, a receptor mutant in which naloxone has been shown to exhibit full agonistic properties in vitro. After 2 weeks of virus injection, naloxone (10 mg/kg s.c.) elicited antinociceptive effect (determined by tail-flick test) without tolerance (10 mg/kg s.c., b.i.d. for 6 days) and significant withdrawal symptoms. On the other hand, subchronic treatment with morphine (10 mg/kg s.c., b.i.d.) for 6 days induced significant tolerance (4.8-fold) and withdrawal symptoms. Furthermore, we found that morphine, but not naloxone, induced the rewarding effects (determined by conditioned place preference test). These data suggest that local expression of MORS196ACSTA in spinal cord and systemic administration of naloxone has the potential to be developed into a new strategy in the management of pain without addiction liability.


Subject(s)
Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Pain Management , Pain Measurement/drug effects , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/genetics , Animals , Conditioning, Operant/drug effects , Drug Tolerance , Gene Transfer Techniques , Genetic Therapy , Injections, Spinal , Male , Mice , Mice, Inbred ICR , Mutation , Opioid-Related Disorders/psychology , Reward , Subarachnoid Space
4.
Proc Natl Acad Sci U S A ; 104(50): 20096-101, 2007 Dec 11.
Article in English | MEDLINE | ID: mdl-18056815

ABSTRACT

We previously reported that mutations in the mu-opioid receptor (MOR), S196L or S196A, rendered MOR responsive to the opioid antagonist naloxone without altering the agonist phenotype. Subsequently, a mouse strain carrying the S196A mutation exhibited in vivo naloxone antinociceptive activity without the development of tolerance. In this study we investigated the possibility of combining the in vivo site-directed delivery of MORS196A and systemic naloxone administration as a paradigm for pain management. Double-stranded adenoassociated virus type 2 (dsAAV2) was used to deliver MORS196A-EGFP by injecting the virus into the spinal cord (S2/S3) dorsal horn region of ICR mice. MORS196A-EGFP fluorescence colocalized with some calcitonin gene-related peptide and neuron-specific protein immunoreactivity in the superficial layers of the dorsal horn 1 week after injection and lasted for at least 6 months. In mice injected with the mutant receptor, morphine induced similar antinociceptive responses and tolerance development or precipitated withdrawal symptoms and reward effects, similar to those in the control mice (saline injected into the spinal cord). Conversely, in the dsAAV2-injected mice, naloxone produced antinociceptive effects at the spinal level but not at the supraspinal level, whereas naloxone had no measurable effect on the control mice. Furthermore, the chronic administration of naloxone to mice injected with dsAAV2-MORS196A-EGFP did not induce tolerance, dependence, or reward responses. Thus, our current approach to activate a mutant receptor, but not the endogenous receptor, with an opioid antagonist represents an alternative to the use of traditional opioid agonists for pain management.


Subject(s)
Dependovirus/genetics , Genes, Reporter/genetics , Pain Measurement , Receptors, Opioid, mu/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Animals , Dependovirus/classification , Drug Tolerance , Gene Expression Regulation , Gene Transfer Techniques , Male , Mice , Morphine/therapeutic use , Mutation/genetics , Naloxone/pharmacology , Pain/drug therapy , Pain/genetics , Pain/metabolism , Receptors, Opioid, mu/genetics , Serine/genetics , Serine/metabolism , Time Factors
5.
Science ; 162(3859): 1290-2, 1968 Dec 13.
Article in English | MEDLINE | ID: mdl-5748987

ABSTRACT

Tolerance and physical dependence development to morphine in mice can be prevented by concomitant administration of cycloheximide. The fact that the rate of synthesis of brain 5-hydroxytryptamine (5HT) increases with tolerance to morphine suggests that the protein involved may be associated with 5HT synthesis. Inhibition of this synthesis with p-chlorophenylalanine markedly decreases tolerance and physical dependence development to morphine.


Subject(s)
Brain/metabolism , Drug Tolerance , Morphine Dependence , Morphine/pharmacology , Serotonin/biosynthesis , Animals , Antimetabolites/pharmacology , Cycloheximide/pharmacology , Depression, Chemical , Humans , Mice , Morphine Dependence/prevention & control , Movement Disorders/chemically induced , Narcotic Antagonists , Pargyline/pharmacology , Phenylalanine/pharmacology , Substance Withdrawal Syndrome
6.
Science ; 177(4049): 616-7, 1972 Aug 18.
Article in English | MEDLINE | ID: mdl-4558902

ABSTRACT

Naloxone hydrochloride, an opioid antagonist, was applied to several discrete brain regions of morphine-dependent rats to precipitate abstinence. Severe withdrawal signs were elicited after administration in the thalamus but not in neocortical, hippocampal, hypothalamic, or tegmental areas of the brain.


Subject(s)
Brain/drug effects , Morphinans/pharmacology , Morphine Dependence , Narcotic Antagonists/pharmacology , Animals , Cerebral Cortex/drug effects , Diencephalon/drug effects , Hippocampus/drug effects , Humans , Hypothalamus/drug effects , Male , Mesencephalon/drug effects , Morphine/pharmacology , Naloxone/administration & dosage , Naloxone/pharmacology , Rats , Stereotaxic Techniques , Substance Withdrawal Syndrome/chemically induced , Thalamus/drug effects
7.
Science ; 207(4426): 75-6, 1980 Jan 04.
Article in English | MEDLINE | ID: mdl-6243189

ABSTRACT

Morphine and beta-endorphin inhibit the shaking response of pentobarbital-anesthetized rats to ice water. Stereotaxically guided administration of antibodies to cerebroside sulfate into the periaqueductal gray region, the most sensitive brain region in which to demonstrate inhibition of this response, antagonizes the effect of morphine and beta-endorphin. These results suggest that cerebroside sulfate may be an integral component of an opiate receptor in rat brain.


Subject(s)
Brain/immunology , Endorphins/antagonists & inhibitors , Morphine/antagonists & inhibitors , Receptors, Opioid/immunology , Sulfoglycosphingolipids/immunology , Animals , Antigen-Antibody Reactions , Behavior, Animal/drug effects , Biological Assay , Cerebral Aqueduct , Male , Pentobarbital/pharmacology , Rats
8.
Neuroscience ; 151(4): 1034-41, 2008 Feb 19.
Article in English | MEDLINE | ID: mdl-18201839

ABSTRACT

The gene of mouse kappa opioid receptor (KOR) utilizes two promoters, P1 and P2. P1 is active in various brain areas and constitutively in P19 mouse embryonal carcinoma cells. P2 is active in limited brain stem areas of adult animals and only in late differentiated cells of P19 induced for neuronal differentiation in the presence of nerve growth factor (NGF). NGF response of P2 was found to be mediated by a specific binding site for transcription factor activation protein 2 (AP2) located in P2. Electrophoretic gel shift assay showed specific binding of this AP2 site by AP2beta, but not AP2alpha. Knockdown of endogenous AP2beta with siRNA abolished the stimulating effect of NGF on the expression of transcripts driven by P2. Binding of endogenous AP2beta on the endogenous KOR P2 chromatin region was also confirmed by chromatin immunoprecipitation. The effect of NGF was inhibited by LY2942002 (phosphatidylinositol 3-kinase, PI3K inhibitor), suggesting that PI3K was involved in signaling pathway mediating the effect of NGF stimulation on KOR P2. The chromatin of P2 in P19 was found to be specifically modified following NGF stimulation, which included demethylation at Lys9 and dimethylation at Lys4 of histone H3 and was consistent with the increased recruitment of RNA polymerase II to this promoter. This study presents the first evidence for epigenetic changes occurred on a specific KOR promoter triggered by NGF in cells undergoing neuronal differentiation. This epigenetic change is mediated by recruited AP2beta to this promoter and involves the PI3K system.


Subject(s)
Cell Differentiation/physiology , Epigenesis, Genetic/physiology , Neurons/metabolism , Receptors, Opioid, kappa/genetics , Animals , Cell Differentiation/drug effects , Chromatin Immunoprecipitation/methods , Dose-Response Relationship, Drug , Drug Interactions , Electrophoresis, Polyacrylamide Gel , Electrophoretic Mobility Shift Assay/methods , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , Luciferases/metabolism , Mice , Models, Biological , Nerve Growth Factor/pharmacology , Neurons/drug effects , PC12 Cells/drug effects , Promoter Regions, Genetic/drug effects , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Rats , Receptors, Opioid, kappa/metabolism , Transfection , Tretinoin/pharmacology
9.
Int Rev Cell Mol Biol ; 327: 89-161, 2016.
Article in English | MEDLINE | ID: mdl-27692181

ABSTRACT

Treatment of chronic pain has created a "silent epidemic," a term that describes the serious public health problem of the abuse of opioid painkillers and other prescription drugs. Conventional pharmacotherapy is limited by the loss of effectiveness in the long-term and by potentially lethal side effects. Efforts need to be focused on the development of nonpharmacological approaches. As significant progress is made in the viral vector technology, gene therapy involving recombinant viruses as vehicles may become a viable alternative for treatment of severe pain. Virus-based gene therapy has several advantages: (1) the transfer of a therapeutic gene to produce/release bioactive therapeutic molecules in a specific location in the nervous system thus minimizing the risks of off-target side effects, and (2) sustained long-term production of the therapeutic agent. This review compiles recently developed strategies for gene therapy targeting specific mechanisms of specific chronic pain conditions. A few successful studies on animal models of chronic pain have been translated to human clinical trials.


Subject(s)
Chronic Pain/genetics , Chronic Pain/therapy , Genetic Therapy , Analgesics, Opioid/therapeutic use , Animals , Chronic Pain/complications , Chronic Pain/physiopathology , Humans , Inflammation/complications , Inflammation/pathology , Neuralgia/complications , Neuralgia/genetics , Neuralgia/physiopathology , Neuralgia/therapy , Nociception
10.
J Neurosci ; 21(5): 1590-9, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11222649

ABSTRACT

The effect of retinoids on the expression of kappa opioid receptor (KOR) gene was examined in normal and transgenic animals. KOR-lacZ transgene expression was specifically elevated in KOR-positive areas of the developing CNS by depleting vitamin A from animal diets. The endogenous KOR mRNA species, including all three isoforms, were also upregulated by depleting vitamin A in developing animals. Change in the expression of isoforms a and b is similar in prenatal stages but differs during postnatal development. Interestingly, upregulation of isoform c is most significant postnatally. The regulation of KOR gene by vitamin A was substantiated in a mouse embryonal carcinoma P19 culture system in which retinoic acid (RA), the most potent ingredient of vitamin A, was able to suppress the expression of all the three KOR isoforms and KOR protein. The RA-mediated suppression was blocked by an RA receptor antagonist and a histone deacetylase (HDAC) inhibitor. By using a reporter transfection assay in P19 cells, the potential genetic element responsible for RA-mediated suppression of KOR gene expression was located to intron 1 of the mouse KOR gene, which could also be blocked by HDAC inhibitor. Furthermore, suppression of KOR gene expression by RA in P19 cells appeared to be an indirect event and required protein synthesis. A role of RA in KOR gene regulation during developmental stages was discussed.


Subject(s)
Gene Expression Regulation/drug effects , Receptors, Opioid, kappa/metabolism , Tretinoin/metabolism , Animals , Diet , Embryonal Carcinoma Stem Cells , Enzyme Inhibitors/pharmacology , Female , Genes, Reporter , Histone Deacetylase Inhibitors , Introns/drug effects , Male , Mice , Mice, Transgenic , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Organ Specificity , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Receptors, Opioid, kappa/genetics , Receptors, Retinoic Acid/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Regulatory Sequences, Nucleic Acid/drug effects , Transfection , Transgenes , Tretinoin/pharmacology , Tumor Cells, Cultured , Vitamin A Deficiency/metabolism
11.
J Leukoc Biol ; 70(4): 527-36, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11590188

ABSTRACT

The major objective of this paper is to characterize the mechanism by which morphine modulates lymphocyte function and if these effects are mediated through the mu-opioid receptor. We evaluated the in vitro effects of morphine on lymphocytes that were freshly isolated from lymph nodes from wild type (WT) and mu-opioid receptor knock-out (MORKO) mice. Results show that morphine inhibits Con A-induced lymph node T-cell proliferation and IL-2 and IFN-gamma synthesis in a dose-dependent manner. This effect was abolished in lymph node cells isolated from MORKO mice. The inhibition of T-cell function with low-dose morphine was associated with an increase in caspase-3- and caspase-8-mediated apoptosis. The inhibition of T-cell function with high-dose morphine was associated with an increase in the inducible NO synthase mRNA expression. N(G)-nitro-L-arginine methyl ester (L-NAME) antagonized the apoptosis induced by high-dose morphine. Our results suggest that low-dose morphine, through the mu-opioid receptor, can induce lymph node lymphocyte apoptosis through the cleavage activity of caspase-3 and caspase-8. Morphine at high doses induces NO release. This effect of morphine is also mediated through the mu-opioid receptor present on the surface of macrophages.


Subject(s)
Caspases/physiology , Lymph Nodes/immunology , Morphine/pharmacology , Nitric Oxide/physiology , T-Lymphocytes/immunology , Animals , Apoptosis , Caspase 3 , Caspase 8 , Caspase 9 , Cells, Cultured , Cytokines/biosynthesis , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Knockout , Morphine/administration & dosage , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , RNA, Messenger/biosynthesis , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/physiology
12.
Gene ; 117(2): 249-54, 1992 Aug 15.
Article in English | MEDLINE | ID: mdl-1339369

ABSTRACT

A rat brain cDNA library was screened with probes constructed from portions of the cDNA (OBCAM) encoding the opioid-binding cell adhesion molecule (OBCAM). Three clones of interest were isolated and sequenced. The largest clone, DUZ1, had a putative open reading frame (ORF) essentially identical to OBCAM in its C-terminal 318 amino acids (aa), but differing in its N-terminal aa (20 vs. 27 in OBCAM), and in all of its 5'-noncoding regions. The other clones, SG8 and SG13, had a putative ORF essentially identical to that of OBCAM, but differed from each other in a portion of their 5'-noncoding region. This study suggests that there is a family of OBCAM-like genes.


Subject(s)
Multigene Family/genetics , Receptors, Opioid/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA/genetics , Gene Library , Molecular Sequence Data , Rats , Receptors, Opioid/chemistry , Receptors, Opioid, delta
13.
Gene ; 152(2): 227-32, 1995 Jan 23.
Article in English | MEDLINE | ID: mdl-7835705

ABSTRACT

A subtraction cDNA library was constructed from control hybrid NG108-15 (mouse neuroblastoma x rat glioma) cells and NG108-15 cells which had been treated for 48 h with the delta-opioid agonist D-Ala2-D-Leu5 enkephalin (DADLE) to down-regulate the delta-opioid receptor on these cells. Among the clones isolated from this library was NGD16-4, a 2768-bp clone encoding a putative 64-kDa protein containing 14 tandemly repeated zinc fingers (Zf) with high homology to the Krüppel family of Zf proteins. NGD16-4 also contains a region homologous to the A element of the Krüppel Associated Box (KRAB) domain, a domain recently linked to transcriptional repression. Southern and Northern analyses indicate that NGD16-4 is derived from the mouse genome. Northern analysis also demonstrates that expression of NGD16-4 mRNA is much higher in several mouse neuroblastoma cell lines than in mouse brain or other tissues. Although the function of NGD16-4 is unclear, the expression pattern of NGD16-4 indicates a possible association with the processes of differentiation or transformation in the mouse.


Subject(s)
DNA-Binding Proteins/genetics , Zinc Fingers , Amino Acid Sequence , Base Sequence , Blotting, Northern , Blotting, Southern , DNA, Complementary , Glioma , Molecular Sequence Data , Neuroblastoma , Sequence Homology, Amino Acid , Tumor Cells, Cultured
14.
FEBS Lett ; 287(1-2): 93-6, 1991 Aug 05.
Article in English | MEDLINE | ID: mdl-1652466

ABSTRACT

Mouse thymocytes incubated in vitro with increasing concentrations of interleukin-1 (IL-1) in the presence of phytohemagglutinin (PHA) exhibited a dose-dependent increase in cell proliferation, as measured by [3H]thymidine incorporation. Under these conditions, there was a parallel dose-dependent increase in specific [3H]morphine binding, with a maximum increase of approximately 5-fold over basal levels. The binding sites differ from classical opioid receptors in that they are not stereo-selective. Interleukin-2 was ineffective in promoting either cell proliferation or enhanced opioid binding, but the effects of IL-1 could be mimicked by phorbol myristate acetate (PMA), suggesting the involvement of tyrosine phosphorylation. These results indicate that morphine-binding sites on immune cells can be regulated by cytokine activation.


Subject(s)
Interleukin-1/pharmacology , Lymphocyte Activation , Morphine/metabolism , T-Lymphocytes/metabolism , Animals , Cell Division , Feedback , Mice , Mice, Inbred ICR , Phytohemagglutinins/pharmacology , Receptors, Opioid/physiology , T-Lymphocytes/cytology , Tetradecanoylphorbol Acetate/pharmacology
15.
Neuropharmacology ; 26(7B): 917-21, 1987 Jul.
Article in English | MEDLINE | ID: mdl-2821434

ABSTRACT

The purification of the mu-opioid receptor has proved to be more difficult than most other receptors for at least two reasons. First, they are easily inactivated by most detergents and the second problem is the heterogeneous existence of the opioid receptor. Despite these problems, this laboratory, and several others, have recently reported purification of the mu-opioid receptor. The 58,000 molecular weight of the purified mu-opioid receptor agrees closely with the value reported by other investigators. In addition, it was found that this purified mu-opioid receptor was associated with a phosphatase activity. While the relevance of this phosphatase activity to opioid receptor action remains to be determined, it is interesting to note that this activity was stimulated by Mn++ and Mg+ ions, both of which also increase opioid binding.


Subject(s)
Receptors, Opioid/isolation & purification , Animals , Cattle , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Enkephalins/metabolism , Glycosylation , Lipid Metabolism , Magnesium/pharmacology , Manganese/pharmacology , Molecular Weight , Phosphoric Monoester Hydrolases/metabolism , Rats , Receptors, Opioid/metabolism , Receptors, Opioid, mu
16.
Neuropharmacology ; 47 Suppl 1: 300-11, 2004.
Article in English | MEDLINE | ID: mdl-15464146

ABSTRACT

Drug addiction has great social and economical implications. In order to resolve this problem, the molecular and cellular basis for drug addiction must be elucidated. For the past three decades, our research has focused on elucidating the molecular mechanisms behind morphine tolerance and dependence. Although there are many working hypotheses, it is our premise that cellular modulation of the receptor signaling, either via transcriptional or post-translational control of the receptor, is the basis for morphine tolerance and dependence. Thus, in the current review, we will summarize our recent work on the transcriptional and post-translational control of the opioid receptor, with special emphasis on the mu-opioid receptor, which is demonstrated to mediate the in vivo functions of morphine.


Subject(s)
Narcotics/pharmacology , Opioid-Related Disorders/genetics , Opioid-Related Disorders/physiopathology , Receptors, Opioid/biosynthesis , Receptors, Opioid/metabolism , Receptors, Opioid/physiology , Transcription, Genetic/genetics , Animals , Drug Tolerance , Humans , Mice , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Opioid/drug effects , Receptors, Opioid/genetics , Signal Transduction/drug effects
17.
J Med Chem ; 20(3): 325-8, 1977 Mar.
Article in English | MEDLINE | ID: mdl-845862

ABSTRACT

The solid-phase synthesis of human beta-endorphin is described. A yield of 32% is achieved based on starting resin. The synthetic product behaves as a homogeneous peptide in partition chromatography, paper electrophoresis, thin-layer chromatography, disc electrophoresis, amino acid composition, and a tryptic map. The synthetic beta h-endorphin possesses antinociceptive properties as estimated by the tail-flick, hot-plate, and writhing tests in mice. When applied centrally, beta h-endorphin is 17-48 times more potent than morphine. It is 3.4 times more potent than morphine when injected intravenously. The analgesic responses are blocked by the specific opiate antagonist, naloxone.


Subject(s)
Analgesics/chemical synthesis , Peptides/chemical synthesis , Amino Acid Sequence , Animals , Male , Methods , Mice , Mice, Inbred ICR , Morphine/pharmacology , Naloxone/pharmacology , Peptides/antagonists & inhibitors , Peptides/pharmacology , Reaction Time/drug effects
18.
J Neuroimmunol ; 65(2): 167-70, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8964899

ABSTRACT

A delta opioid receptor complementary DNA (cDNA) was cloned by expression of cDNA library from activated thymocytes in Cos 7 cells. The deduced amino acid sequence of this receptor was similar to that described in the brain. As analyzed by southern blot hybridization, the delta opioid receptor transcripts are constitutively expressed in unactivated thymocytes. In addition, neither kappa nor mu opioid receptor transcripts were detected in thymocytes, suggesting tissue-specific opioid receptor gene expression in the immune system. The studies represent the first report of a full-length opioid receptor in the immune system.


Subject(s)
Cloning, Molecular , DNA, Complementary/genetics , Receptors, Opioid, delta/genetics , Thymus Gland/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blotting, Southern , Brain/metabolism , Mice , Mice, Inbred ICR , Molecular Probes/genetics , Molecular Sequence Data , Polymerase Chain Reaction , Thymus Gland/cytology , Transcription, Genetic
19.
J Neuroimmunol ; 116(2): 147-55, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11438169

ABSTRACT

Mu-opioid receptor knockout mice (MORKO), were used to address two questions: (1) if morphine induced decrease in thymic weight and cell distribution is mediated by the mu-opioid receptor and (2) the role of corticosteroids in morphine mediated alteration in thymic cell distribution. Our result show that morphine mediated increase in plasma corticosterone is mediated by the mu-opioid receptor since morphine at doses as high as 25 mg/kg-body weight does not increase plasma corticosterone levels in the MORKO. In addition, we have also shown that morphine treatment results in the differentiation of CD4+CD8+ (double positive cells) to single positive CD4+ cells while dexamethasone treatment results in the deletion of CD4+CD8+ (double positive) cells.


Subject(s)
Analgesics, Opioid/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Morphine/pharmacology , Pituitary Gland/drug effects , Receptors, Opioid, mu/genetics , Thymus Gland/cytology , Adrenocorticotropic Hormone/blood , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Corticosterone/blood , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Glucocorticoids/pharmacology , Hypothalamo-Hypophyseal System/immunology , Mice , Mice, Knockout , Organ Size , Pituitary Gland/immunology , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, mu/immunology , Thymus Gland/drug effects
20.
J Neuroimmunol ; 64(1): 29-36, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8598387

ABSTRACT

The existence of receptors for opioid compounds on cells of the immune system has long been hypothesized, but has been very difficult to demonstrate unequivocally. We have used reverse-transcription polymerase chain reaction to obtain cDNA clones from the human MOLT-4 and CEM-3 T-leukemic cell lines which are nearly identical to portions of the delta and kappa opioid receptor cDNAs recently isolated from human brain and placenta, respectively. Northern analyses with riboprobes derived from the delta and kappa opioid receptor clones indicate these sequences are expressed at low levels in human peripheral blood lymphocytes and in several human lymphoid cell lines. Sequences corresponding to the mu opioid receptor cDNA were not detected in this study. The results suggest that delta and kappa opioid receptors may be responsible for mediating some direct effects of opioids in immune cells.


Subject(s)
Gene Expression Regulation , Leukemia/metabolism , Lymphocytes/metabolism , Receptors, Opioid/genetics , Base Sequence , Blotting, Northern , Cells, Cultured , DNA, Complementary/analysis , Humans , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL