Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 252
Filter
1.
Neuroscience ; 152(3): 753-60, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18304750

ABSTRACT

Cannabinoid receptor activation has been shown to modulate both neurotransmission (CB(1)) and neuroinflammatory (CB(2)) responses. There are conflicting reports in the literature describing the influence of cannabinoid receptor activation on ischemic/reperfusion injury. The goal of this study was to evaluate how changing the balance between CB(1) and CB(2) activation following cerebral ischemia influences outcome. CB(1) and CB(2) expression were tested at different times after transient middle cerebral artery occlusion (MCAO) in mice by real-time RT-PCR. Animals subjected to 1 h MCAO were randomly assigned to receive different treatments: a CB(1) antagonist, a CB(2) antagonist, a CB(2) agonist, a CB(1) antagonist plus CB(2) agonist, a CB(2) antagonist plus CB(2) agonist or an equal volume of vehicle as control. Cerebral blood flow was continuously monitored during ischemia; cerebral infarction and neurological deficit were tested 24 h after MCAO. Cerebral CB(1) and CB(2) mRNA expression undertook dynamic changes during cerebral ischemia. The selective CB(1) antagonist significantly decreased cerebral infarction by 47%; the selective CB(2) antagonist increased infarction by 26% after 1 h MCAO followed by 23 h reperfusion in mice. The most striking changes were obtained by combining a CB(1) antagonist with a CB(2) agonist. This combination elevated the cerebral blood flow during ischemia and reduced infarction by 75%. In conclusion, during cerebral ischemia/reperfusion injury, inhibition of CB(1) receptor activation is protective while inhibition of CB(2) receptor activation is detrimental. The greatest degree of neuroprotection was obtained by combining an inhibitor of CB(1) activation with an exogenous CB(2) agonist.


Subject(s)
Brain Ischemia/drug therapy , Cannabinoid Receptor Modulators/pharmacology , Neuroprotective Agents/pharmacology , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Reperfusion Injury/drug therapy , Animals , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Cannabinoid Receptor Modulators/metabolism , Cannabinoid Receptor Modulators/therapeutic use , Cannabinoids/pharmacology , Cannabinoids/therapeutic use , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Cerebral Infarction/drug therapy , Cerebral Infarction/metabolism , Cerebral Infarction/prevention & control , Drug Combinations , Drug Synergism , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/physiopathology , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/therapeutic use , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/drug effects , Receptor, Cannabinoid, CB2/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology , Time Factors , Treatment Outcome
4.
Sex Transm Infect ; 78(2): 83-6, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12081189

ABSTRACT

The first ever national strategy for sexual health and HIV in England was published in July 2001 and proposes a comprehensive and holistic model for dealing with an increasing public health problem. The strategy covers the issues of prevention, service provision, commissioning of services, and the necessary requirements to support change. This paper concentrates on service issues and developments in relation to genitourinary medicine/HIV services, and outlines a model for providing these which attempts to do so around patients' needs, delivered through comprehensive and interlinked local networks of services.


Subject(s)
Comprehensive Health Care/organization & administration , Continuity of Patient Care/organization & administration , HIV Infections/therapy , National Health Programs/organization & administration , Venereology/organization & administration , England , Female , Holistic Health , Humans , Male , Sexually Transmitted Diseases/therapy
6.
Int Immunopharmacol ; 1(11): 2001-9, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11606031

ABSTRACT

Previously, our laboratory has shown that morphine given by implantation of a 75-mg slow-release pellet for 48 h suppresses murine splenic antibody responses to sheep red blood cells (SRBCs) in a plaque-forming cell (PFC) assay. However, the use of slow-release pellets for such studies is limited, as these pellets are only available in fixed doses and similar pellets for kappa and delta agonists have not been developed. In the present study, we investigated the feasibility of administering opioids via Alzet osmotic minipumps to assess their immunomodulatory effects. Groups of mice received minipumps dispensing morphine sulfate, which has primary activity at the mu opioid receptor; U50,488H, which is a kappa-selective agonist; deltorphin II, which is a delta2-selective agonist; or DPDPE, which has greater selectivity for delta1 than delta, receptors. Morphine, U50,488H and deltorphin II were all immunosuppressive, with biphasic dose-response curves exhibiting maximal (approximately 50%) suppression of the PFC response at doses of 0.5 to 2 mg/kg/day 48 h after pump implantation. Further, immunosuppression by morphine sulfate, U50,488H or deltorphin II was blocked by simultaneous implantation of a minipump administering the opioid receptor-selective antagonists CTAP (1 mg/kg/day), nor-binaltorphimine (5 mg/kg/day), or naltriben (3 mg/kg/day), respectively. DPDPE was inactive at doses lower than 10 mg/kg/day. We conclude that osmotic minipumps are a practical and useful way of administering opioids to study their effects on the immune system, and give further evidence that immunosuppression induced in vivo by opioid agonists is mediated not only via mu, but also via kappa and delta2 opioid receptors.


Subject(s)
Antibody Formation/drug effects , Immunosuppressive Agents/pharmacology , Naltrexone/analogs & derivatives , Receptors, Opioid, delta/agonists , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/agonists , Spleen/drug effects , Spleen/immunology , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/antagonists & inhibitors , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Analgesics, Opioid/antagonists & inhibitors , Analgesics, Opioid/pharmacology , Animals , Depression, Chemical , Dose-Response Relationship, Drug , Female , Infusion Pumps , Mice , Mice, Inbred C3H , Morphine/antagonists & inhibitors , Morphine/pharmacology , Naltrexone/pharmacology , Neuroimmunomodulation , Oligopeptides/antagonists & inhibitors , Oligopeptides/pharmacology , Osmosis , Peptide Fragments , Peptides/pharmacology , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, mu/antagonists & inhibitors , Somatostatin
7.
J Immunol ; 167(7): 3677-81, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11564781

ABSTRACT

In this study, we investigated the effect of morphine on the mucosal immune system using fragment cultures of ileal segments, Peyer's patches (PPs), and mesenteric lymph nodes. Mice were implanted s.c. with a morphine slow release pellet. Control groups received a naltrexone slow release pellet, a placebo pellet, or both a morphine and a naltrexone pellet. After 48 h, mice were orally immunized with cholera toxin (CT) and were boosted orally 1 wk later. Animals were sacrificed 1 wk after the booster immunization, and PPs, mesenteric lymph nodes, and ileal segments were cultured in 24-well plates for 12 days. Morphine resulted in a highly significant inhibition of CT-specific IgA and IgG production in fragment culture supernatants of all three tissues compared with placebo. Naltrexone blocked the reduction in Ab levels induced by morphine, indicating that the effect is opioid receptor mediated. Morphine did not significantly alter total IgA levels in any of the tissue culture supernatants. Morphine also inhibited CT-specific IgA and IgG levels in serum. By flow cytometry, morphine did not alter the lymphoid cell composition in PPs compared with placebo. The effect of morphine on TGF-beta, IL-5, and IL-6 mRNA expression in PPs and ileal segments was determined following oral immunization with CT. Morphine significantly decreased TGF-beta mRNA compared with that in the placebo group, and naltrexone blocked this effect. These results indicate that morphine inhibits Ag-specific IgA responses in gut-associated lymphoid tissue at least partially through the inhibition of TGF-beta, a putative IgA switch factor, in the gastrointestinal tract.


Subject(s)
Cholera Toxin/pharmacology , Immunity, Mucosal , Immunoglobulin A/biosynthesis , Intestinal Mucosa/immunology , Morphine/pharmacology , Transforming Growth Factor beta/biosynthesis , Administration, Oral , Animals , Cholera Toxin/administration & dosage , Drug Antagonism , Ileum/drug effects , Ileum/immunology , Immunoglobulin G/biosynthesis , Lymph Nodes/drug effects , Lymph Nodes/immunology , Mesentery , Mice , Mice, Inbred C3H , Organ Culture Techniques , Peyer's Patches/drug effects , Peyer's Patches/immunology , RNA, Messenger/biosynthesis , Transforming Growth Factor beta/genetics
8.
Brain Res ; 904(2): 252-8, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11406123

ABSTRACT

Orphanin/nociceptin (OFQ/N), a 17-amino-acid peptide, is an endogenous peptide, the receptor for which is similar to mu-, delta- and kappa-opioid receptors ( approximately 65% homology). Reports indicate that OFQ/N can block the antinociception induced by mu-, delta- and kappa-opioid agonists in the rat and in the mouse, indicating that there is a functional interaction between opioid receptors and OFQ/N receptors in the nervous system. It is well known that activation of the mu- and kappa-opioid receptors results in hyperthermia and hypothermia, respectively, in Sprague-Dawley rats. The present studies were designed to examine effects of OFQ/N on body temperature (Tb) and explore whether the mechanism of T(b) change induced by OFQ/N involved the opioid system. The results show that (1) i.c.v. injection of a high dose of OFQ/N (9-18 micro g) produces hypothermia in adult rats; (2) OFQ/N (1.8 micro g, i.c.v., t=+30 s after morphine) can decrease morphine-induced hyperthermia; (3) neither the opioid receptor antagonist, naloxone (10 mg/kg, s.c., t=-15 s before OFQ/N) nor the kappa-opioid receptor antagonist nor-BNI (1 micro g/5 microl, i.c.v., t=-30 s before OFQ/N) reduces the hypothermia induced by i.c.v. injection of OFQ/N at dose of 18 micro g (P>0.05); (4) 60 micro g/5 microl AS oligo (i.c.v. treatment on days 1, 3 and 5) against OFQ/N receptors significantly reduces the hypothermia induced by i.c.v. injection of 9 micro g OFQ/N (P<0.01). These results suggest that the hypothermia induced by i.c.v. injection of a high dose of OFQ/N (9 or 18 micro g) is mediated, at least partially, by its own receptor, independent or downstream of opioid receptors in the rat brain and that OFQ/N probably acts as a physiological antagonist to reduce morphine-induced hyperthermia.


Subject(s)
Hypothermia/chemically induced , Opioid Peptides/physiology , Animals , Body Temperature/drug effects , Body Temperature/physiology , Injections, Intraventricular , Male , Narcotic Antagonists , Opioid Peptides/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Opioid/agonists , Receptors, Opioid/physiology , Vasodilator Agents/pharmacology , Nociceptin
10.
Brain Res ; 894(2): 266-73, 2001 Mar 16.
Article in English | MEDLINE | ID: mdl-11251200

ABSTRACT

The effect of central and peripheral administration of a nitric oxide synthase inhibitor, N-nitro-L-arginine methyl ester (L-NAME), on morphine hyperthermia was studied in male Sprague-Dawley rats. The first series of experiments examined the effect of subcutaneous (s.c.) administration of L-NAME on the hyperthermia induced by morphine given s.c. in doses of 4 and 15 mg/kg. L-NAME, at a s.c. dose of 50 mg/kg, per se, had no influence on body temperature (T(b)). Coadministration of L-NAME (50 mg/kg, s.c.) with the higher dose of morphine (15 mg/kg, s.c.) caused a significant suppression of morphine hyperthermia during the first 30 min and then produced hypothermia. In contrast, s.c. injection of L-NAME (50 mg/kg, s.c.) failed to alter the hyperthermic response induced by the lower dose of morphine (4 mg/kg). In the second series of experiments, we investigated the effect of intracerebroventricular (i.c.v.) administration of L-NAME on the hyperthermia induced by morphine given s.c. L-NAME, itself, given i.c.v. at a dose of 1 mg did not evoke any change in T(b). Intracerebroventricular administration of L-NAME (1 mg) blocked the hyperthermia induced by 15 mg/kg morphine during the first 30 min and induced a slight hypothermia but did not alter the hyperthermia induced by 4 mg/kg morphine. The results indicate that either central or peripheral NO synthesis is required for the production of hyperthermia induced by 15 mg/kg of morphine. However, NO synthesis does not seem to be involved in the hyperthermic process induced by 4 mg/kg of morphine.


Subject(s)
Enzyme Inhibitors/pharmacology , Fever/drug therapy , Morphine , NG-Nitroarginine Methyl Ester/pharmacology , Narcotics , Nitric Oxide Synthase/antagonists & inhibitors , Animals , Fever/chemically induced , Injections, Intraventricular , Injections, Subcutaneous , Male , Nitric Oxide Synthase/metabolism , Rats , Rats, Sprague-Dawley
12.
J Leukoc Biol ; 68(5): 723-8, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11073113

ABSTRACT

In this study we investigated the capacity of morphine to modulate expression of cytokines in peritoneal macrophages. Mice were implanted subcutaneously with a 75-mg morphine slow-release pellet, and 48 h later resident peritoneal macrophages were harvested. Control groups received placebo pellets, naltrexone pellets, or morphine plus naltrexone pellets. Adherent cells were stimulated with lipopolysaccharide (LPS: 10 microg/mL) plus interferon-gamma (IFN-gamma: 100 units/mL) to induce cytokine production. After 24 h RNA was extracted for analysis of cytokine mRNA levels by reverse transcriptase-polymerase chain reaction, or supernatants were collected after 48 h for determination of cytokine production by enzyme-linked immunosorbent assay (ELISA). Morphine enhanced mRNA expression of interleukin (IL)-12 p40 and tumor necrosis factor alpha (TNF-alpha) compared with controls, whereas IL-10 levels were unchanged by drug treatment. ELISA data showed that both IL-12 p40 and p70 were increased by morphine. The enhancement of IL-12 at both the mRNA and protein levels was antagonized by naltrexone, indicating that the modulation of this cytokine by morphine is via a classic opioid receptor. These results are particularly interesting in light of our previous observation that 48 h after morphine pellet implantation, the peritoneal cavity is colonized with gram-negative and other enteric bacteria. The enhancement of IL-12 by morphine might be related to morphine-induced sepsis.


Subject(s)
Analgesics, Opioid/pharmacology , Interleukin-12/biosynthesis , Macrophages, Peritoneal/drug effects , Morphine/pharmacology , Adjuvants, Immunologic/biosynthesis , Adjuvants, Immunologic/genetics , Analgesics, Opioid/antagonists & inhibitors , Animals , Corynebacterium , Delayed-Action Preparations , Female , Inflammation Mediators , Interferon-gamma/pharmacology , Interleukin-10/biosynthesis , Interleukin-10/genetics , Interleukin-12/genetics , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C3H , Morphine/antagonists & inhibitors , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Stimulation, Chemical , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
13.
J Immunol ; 165(11): 6519-24, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11086093

ABSTRACT

Strong evidence for the direct modulation of the immune system by opioids is well documented. Mu-opioids have been shown to alter the release of cytokines important for both host defense and the inflammatory response. Proinflammatory chemokines monocyte chemoattractant protein-1 (MCP-1), RANTES, and IFN-gamma-inducible protein-10 (IP-10) play crucial roles in cell-mediated immune responses, proinflammatory reactions, and viral infections. In this report, we show that [D-Ala(2),N:-Me-Phe(4),Gly-ol(5)]enkephalin (DAMGO), a mu-opioid-selective agonist, augments the expression in human PBMCs of MCP-1, RANTES, and IP-10 at both the mRNA and protein levels. Because of the proposed relationship between opioid abuse and HIV-1 infection, we also examined the impact of DAMGO on chemokine expression in HIV-infected cells. Our results show that DAMGO administration induces a significant increase in RANTES and IP-10 expression, while MCP-1 protein levels remain unaffected in PBMCs infected with the HIV-1 strain. In contrast, we show a dichotomous effect of DAMGO treatment on IP-10 protein levels expressed by T- and M-tropic HIV-infected PBMCs. The differential modulation of chemokine expression in T- and M-tropic HIV-1-infected PBMCs by opioids supports a detrimental role for opioids during HIV-1 infection. Modulation of chemokine expression may enhance trafficking of potential noninfected target cells to the site of active infection, thus directly contributing to HIV-1 replication and disease progression to AIDS.


Subject(s)
Chemokine CCL2/biosynthesis , Chemokine CCL5/biosynthesis , Chemokines, CXC/biosynthesis , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Receptors, Opioid, mu/physiology , Cells, Cultured , Chemokine CCL2/blood , Chemokine CCL2/genetics , Chemokine CCL5/blood , Chemokine CCL5/genetics , Chemokine CXCL10 , Chemokines, CXC/blood , Chemokines, CXC/genetics , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Humans , Interferon-gamma/physiology , Leukocytes, Mononuclear/drug effects , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Peptides/pharmacology , Phytohemagglutinins/pharmacology , RNA, Messenger/biosynthesis , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/blood , Up-Regulation/drug effects , Up-Regulation/genetics , Up-Regulation/immunology
14.
Microvasc Res ; 60(2): 121-30, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10964586

ABSTRACT

Gram-negative sepsis and subsequent endotoxic shock after surgery remain problematic in the United States and throughout the world. While morphine is widely prescribed for postoperative trauma pain management, there are reports that morphine may compromise the immune system and contribute to postoperative sepsis. The current study tested the hypothesis that morphine attenuates leukocyte rolling and sticking in both arterioles and venules via nitric oxide production. Nude mice implanted with slow-release morphine pellets were used in this study. The dorsal skinfold chamber model for intravital fluorescence microscopy on awake mice was used. Leukocyte/endothelial interactions were evaluated after bolus injection of oxidized low density lipoprotein. Morphine was found to significantly attenuate leukocyte rolling and sticking in both the arterial and venular side of the microcirculation. This attenuation was reversed by simultaneous implantation of naloxone pellets. The mechanisms of this attenuation were further investigated by administration of the nitric oxide synthase inhibitors NG-nitro-l-arginine (NOLA) and aminoguanidine (AG) in drinking water. NOLA was found to significantly reverse this morphine-induced attenuation of leukocyte rolling and sticking in both arterioles and venules. However, AG did not have the same effect. The results indicate that morphine interferes with leukocyte/endothelial cell interactions via stimulation of nitric oxide production.


Subject(s)
Analgesics, Opioid/pharmacology , Cell Communication/drug effects , Cell Communication/physiology , Endothelium, Vascular/physiology , Leukocytes/physiology , Morphine/pharmacology , Animals , Cell Adhesion/drug effects , Cell Adhesion/physiology , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Guanidines/pharmacology , Leukocytes/cytology , Mice , Mice, Hairless , Nitroarginine/pharmacology
15.
Eur J Pharmacol ; 401(2): 161-5, 2000 Aug 04.
Article in English | MEDLINE | ID: mdl-10924921

ABSTRACT

The endogenous opioid system has been found to be involved in fever caused by pyrogens. In the present study, we have investigated the role of the mu-opioid receptor in the brain in fever induced by lipopolysaccharide. Rats were microinjected with 1 microg of the mu-opioid receptor-selective antagonist, cyclic D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH(2) (CTAP), into the preoptic anterior hypothalamus. Thirty minutes later, lipopolysaccharide (50 microg/kg) was injected intraperitoneally (i.p.). CTAP reduced by 1 degrees C the fever induced by lipopolysaccharide. However, it did not affect lipopolysaccharide fever when it was given 3 h after lipopolysaccharide injection. These data indicate that mu-opioid receptors within the preoptic anterior hypothalamus mediate the initiation of lipopolysaccharide fever and suggest that the opioid system is involved in the pathogenesis of fever in rats.


Subject(s)
Fever/prevention & control , Lipopolysaccharides/adverse effects , Narcotic Antagonists/pharmacology , Peptides/pharmacology , Receptors, Opioid, mu/antagonists & inhibitors , Amino Acid Sequence , Animals , Body Temperature/drug effects , Dose-Response Relationship, Drug , Fever/chemically induced , Hypothalamus, Anterior/drug effects , Hypothalamus, Anterior/metabolism , Male , Peptide Fragments , Preoptic Area/drug effects , Preoptic Area/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/metabolism , Somatostatin , Time Factors
16.
J Infect Dis ; 181(4): 1350-8, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10762566

ABSTRACT

This study examined the effect of morphine on oral infection with virulent Salmonella typhimurium. Animals were treated with a 75-mg slow-release morphine pellet followed by inoculation with salmonellae. Morphine markedly sensitized mice to oral infection, as assessed by survival, mean survival time, and colony culture. By 24 h after Salmonella inoculation, morphine-treated mice had a 105-fold difference in number of organisms in the Peyer's patches, compared with controls. The opioid antagonist naltrexone significantly blocked Salmonella colonization in Peyer's patches and reduced Salmonella burden in other organs, indicating that morphine acts at least in part via an opioid receptor-mediated pathway. The data show that morphine markedly potentiates Salmonella infection at the gastrointestinal portal of entry and enhances subsequent dissemination of Salmonella organisms. The results have implications for potentiating gastrointestinal opportunistic infections in intravenous drug abusers and in opioid-medicated postsurgical patients.


Subject(s)
Morphine/pharmacology , Mouth Diseases/microbiology , Salmonella Infections, Animal/etiology , Animals , Cytokines/biosynthesis , Cytokines/genetics , Disease Susceptibility , Female , Germ-Free Life , Mice , Mice, Inbred C3H , Mouth/microbiology , Mouth Diseases/etiology , Naltrexone/pharmacology , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Peyer's Patches/microbiology , RNA, Messenger/biosynthesis , Salmonella typhimurium
17.
J Eur Acad Dermatol Venereol ; 14(5): 370-7, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11305378

ABSTRACT

Sexually transmitted infections and HIV in Europe present considerable social and medical problems and are not always adequately controlled. The recent trends for sexually transmitted infections and HIV in Western and Eastern Europe are reviewed.


Subject(s)
HIV Infections/epidemiology , Sexually Transmitted Diseases, Bacterial/epidemiology , Sexually Transmitted Diseases, Viral/epidemiology , Adolescent , Adult , Europe/epidemiology , Female , HIV Infections/transmission , Humans , Incidence , Male , Prevalence , United Kingdom/epidemiology
20.
J Neuroimmunol ; 91(1-2): 55-62, 1998 Nov 02.
Article in English | MEDLINE | ID: mdl-9846819

ABSTRACT

We have found that the immature T cell lines R1.1 and DPK and the macrophage lines P388D1 and WEHI-3 also express kappa-opioid receptor (KOR) mRNA. Characterization of the KOR transcripts in both brain tissue and these T cells has revealed both the normal full-length as well as a truncated form of the mRNA. Our results show that the truncated transcript lacks the second exon. Primary macrophages express this truncated form of the transcript in the absence of detectable levels of the full-length form. These results suggest a degree of heterogeneity in the expression of the opioid receptors which has not previously been reported.


Subject(s)
Macrophages, Peritoneal/chemistry , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/immunology , T-Lymphocytes/chemistry , Animals , Brain Chemistry/immunology , Gene Expression/immunology , Macrophages, Peritoneal/immunology , Mice , Mice, Transgenic , Molecular Sequence Data , Oligonucleotide Probes , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , T-Lymphocytes/immunology , Thymoma , Transcription, Genetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...