Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Immunity ; 15(4): 533-43, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11672536

ABSTRACT

Mice without secreted TNF but with functional, normally regulated and expressed membrane-bound TNF (memTNF(Delta/Delta) mice) were created by knocking-in the uncleavable Delta 1-9,K11E TNF allele. In contrast to TNF-deficient mice (TNF(-/-)), memTNF supported many features of lymphoid organ structure, except generation of primary B cell follicles. Splenic chemokine expression was near normal. MemTNF-induced apoptosis was mediated through both TNF-R1 and TNF-R2. That memTNF is suboptimal for development of inflammation was revealed in experimental autoimmune encephalomyelitis. Disease severity was reduced in memTNF(Delta/Delta) mice relative to wild-type mice, and the nature of spinal cord infiltrates resembled that in TNF(-/-) mice. We conclude that memTNF supports many processes underlying lymphoid tissue structure, but secreted TNF is needed for optimal inflammatory lesion development.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/etiology , Spleen/immunology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis , Cells, Cultured , Chemokines/biosynthesis , Chemokines/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Targeting , Germinal Center/cytology , Lipopolysaccharides/pharmacology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , Mice, Knockout , RNA, Messenger/biosynthesis , Receptors, Tumor Necrosis Factor/physiology , Sequence Deletion , Shock/etiology , Spleen/anatomy & histology , Spleen/drug effects , Tumor Necrosis Factor-alpha/genetics
2.
J Exp Med ; 194(3): 343-54, 2001 Aug 06.
Article in English | MEDLINE | ID: mdl-11489953

ABSTRACT

The stimulation of interferon (IFN)-gamma by interleukin (IL)-12 has been shown to provide protection from intracellular pathogens such as Listeria monocytogenes. Tumor necrosis factor (TNF) is also a major player in the resolution of Listeria infections and is suggested to have more global effects than can be explained by the induction of IFN-gamma alone. Since IL-18 synergizes with IL-12 to induce IFN-gamma production by natural killer and T helper (Th)1 cells, we determined its role in responses to Listeria. IL-18 appeared to be even more potent than either IL-12 or IFN-gamma for protection against this pathogen and IL-18 enhanced bacterial clearance in the complete absence of IFN-gamma. Indeed IL-18 was comparable to TNF in its ability to resolve the infection and showed a lowered protective capacity in the absence of TNF. Moreover, IL-18 induced macrophages to secrete both TNF and nitric oxide after a Listeria infection. IL-18 was also essential for optimal IFN-gamma production by antigen-specific T cells. Therefore, IL-18 operates via its effects on both the innate immune response, including macrophages, as well as on Th1 cells, to protect against Listeria.


Subject(s)
Interferon-gamma/biosynthesis , Interleukin-18/physiology , Listeria monocytogenes/immunology , Animals , Antibodies, Monoclonal/pharmacology , Female , Immunologic Memory , Interleukin-12/physiology , Interleukin-18/pharmacology , Interleukin-18 Receptor alpha Subunit , Listeria monocytogenes/pathogenicity , Listeriosis/etiology , Listeriosis/immunology , Listeriosis/pathology , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, SCID , Mice, Transgenic , Neutralization Tests , Receptors, Interleukin/immunology , Receptors, Interleukin-18 , Recombinant Proteins/pharmacology , Th1 Cells/immunology , Tumor Necrosis Factor-alpha/physiology
3.
Science ; 290(5497): 1768-71, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11099416

ABSTRACT

OX2 (CD200) is a broadly expressed membrane glycoprotein, shown here to be important for regulation of the macrophage lineage. In mice lacking CD200, macrophage lineage cells, including brain microglia, exhibited an activated phenotype and were more numerous. Upon facial nerve transection, damaged CD200-deficient neurons elicited an accelerated microglial response. Lack of CD200 resulted in a more rapid onset of experimental autoimmune encephalomyelitis (EAE). Outside the brain, disruption of CD200-CD200 receptor interaction precipitated susceptibility to collagen-induced arthritis (CIA) in mice normally resistant to this disease. Thus, in diverse tissues OX2 delivers an inhibitory signal for the macrophage lineage.


Subject(s)
Antigens, Surface/metabolism , Down-Regulation , Macrophages/physiology , Animals , Antigens, CD , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Cell Lineage , Central Nervous System/immunology , Central Nervous System/pathology , Denervation , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Facial Nerve , Gene Targeting , Joints/immunology , Joints/pathology , Lymph Nodes/cytology , Macrophage Activation , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Microglia/physiology , Neurons/physiology , Rats , Receptors, Immunologic/metabolism , Spleen/cytology
5.
J Neuroimmunol ; 84(1): 14-23, 1998 Apr 01.
Article in English | MEDLINE | ID: mdl-9600704

ABSTRACT

IFN-beta has recently been shown to exert remarkable beneficial effects on disease development in patients with early stage relapsing-remitting MS. The specific immune mechanism(s) by which IFN-beta ameliorates this human demyelinating disease is at present undefined. One potential mechanism may reside in the antiproliferative activity of IFN-beta which may inhibit the expansion of autoaggressive T cells thereby limiting disease progression. In the present study we investigated whether the administration of recombinant rat IFN-beta (rrIFN-beta) to Lewis rats with actively induced experimental autoimmune encephalomyelitis (EAE) inhibits the expansion of encephalitogenic T cells in lymphoid organs and as such may contribute to suppression of disease activity in this widely used animal model for MS. Our data show that daily administrations of > or = 3 x 10(5) u rrIFN-beta to EAE rats, starting two days before MBP sensitization and continued for 10 days led to a dramatic and dose-dependent reduction in encephalitogenic T cells in both spleen and inguinal lymph nodes at day 8 post-immunization (p.i.). However, the rrIFN-beta-mediated reduction in effector T cells did not ameliorate paralytic disease as expected but significantly enhanced the severity of EAE. Analyses of lymphoid organs in the remission phase of EAE revealed strongly elevated numbers of encephalitogenic T cells in rrIFN-beta-treated versus control rats suggesting a rapid reversal of the antiproliferative action of rrIFN-beta followed by an overshoot in the subsequent expansion of these effector T cells. In conformity with higher numbers of encephalitogenic T cells and worsening of disease, animals also showed significantly greater perivascular inflammation in the CNS. The relevance of our findings in relation to the beneficial effects of IFN-beta in MS is discussed.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Interferon-beta/administration & dosage , Lymph Nodes/drug effects , Spleen/drug effects , Animals , Disease Models, Animal , Female , Lymph Nodes/physiology , Multiple Sclerosis/metabolism , Rats , Rats, Inbred Lew , Recurrence , Spleen/physiology
6.
Immunol Cell Biol ; 76(1): 65-73, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9553778

ABSTRACT

Secreted and cell surface molecules that control the nature of immune responses or directly mediate tissue damage are important targets for therapeutic intervention of autoimmune inflammatory disorders. The precise aetiology of multiple sclerosis (MS), an inflammatory demyelinating disease of the central nervous system is unknown and therapeutic interventions for this disease are limited. However, as it is believed that MS is autoimmune in nature, it seems likely that targeting of the immune system, either to deviate it down a more benign pathway or to directly block tissue damaging effector mechanisms, may enable this disease to be controlled. In this overview we examine three alternative approaches to therapy of MS: cytokine-directed immune deviation, immunoregulation by type I interferons and blockade of TNF. Underlying rationales for the application of these approaches to the treatment of MS are based largely on studies in the disease model experimental autoimmune encephalomyelitis. These studies are reviewed.


Subject(s)
Cytokines/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/therapy , Multiple Sclerosis/therapy , Animals , Humans , Tumor Necrosis Factor-alpha/immunology
7.
J Neuroimmunol ; 78(1-2): 184-97, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9307244

ABSTRACT

The non-hormone secreting folliculo-stellate (FS) cells in the anterior pituitary (AP) appear heterogeneous. Some of these cells have been described as having a neuroectodermal origin and being glial, while some others have been suggested to be monocytic or dendritic cells (DC). We have analyzed here the hematopoietic origin of interstitial cell populations in the AP. In the rat AP, the relative densities of S100+ FS cells and major histocompatibility complex (MHC) class II-expressing DC-like cells show a parallel increase in the postnatal period between the age of 3 weeks to 2 months. We first looked for the presence of donor derived cells in the AP of lethally irradiated bone marrow (BM)-transplanted rats. Donor derived myeloid cells carrying the n haplotype of the MHC class I antigen (RT1.An) reacting with the OX27 moAb, could not be detected in the AP three months after transplantation. It appeared, however, that OX27+ DC-like cells a-priori were virtually absent from the rat AP. Therefore this transplantation model was not suitable for our studies. We then turned to a model of transgenic mice expressing a suicide gene in subpopulations of dendritic cells. Mice were lethally irradiated and received a BM transplant from the transgenic animals, with or without a treatment with ganciclovir (GCV) that specifically kills the dividing cells expressing the suicide gene. This model has already been used to identify and delete mainly dendritic cell populations, viz N418+ and ER-BMDM1+ dendritic cells in the marginal zones of the spleen and in the thymic medulla. We observed in the AP a 30% reduction of the ER-BMDM1+ FS-like cells and a 50-100% reduction of interstitial cells expressing the F4/80, Mac-1 and MOMA-1 markers in the mice receiving the transgenic BM and treated with GCV, compared to control mice that were not treated with GCV or that received non-transgenic BM. When a treatment with granulocyte-macrophage colony-stimulating factor (GM-CSF) was initiated during the GCV treatment, we observed an even stronger reduction of the above-mentioned interstitial cell populations. These data indicate that in the mouse AP a population of stellate cells exists with a hematopoietic origin, that expresses markers of myeloid cells, and that has a rapid turnover.


Subject(s)
Hematopoiesis/physiology , Pituitary Gland, Anterior/cytology , Animals , Biomarkers , Bone Marrow Transplantation , Cell Line , Chimera , Dendritic Cells/cytology , Dendritic Cells/immunology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Histocompatibility Antigens Class I/analysis , Histocompatibility Antigens Class II/analysis , Macrophages/immunology , Male , Mice , Mice, Transgenic , Pituitary Gland, Anterior/drug effects , Pituitary Gland, Anterior/immunology , Rats , Rats, Inbred Strains , Whole-Body Irradiation
8.
J Neuropathol Exp Neurol ; 56(1): 10-20, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8990125

ABSTRACT

The cellular localization and distribution of inducible and constitutive nitric oxide synthase (iNOS/cNOS) was determined in tissue sections from multiple sclerosis (MS) and control brain and spinal cord. Immunocytochemical techniques were applied using specific iNOS- and cNOS-directed antibodies. In addition, NADPH-diaphorase histochemistry was performed. To establish the identity of iNOS-, cNOS- and NADPH-diaphorase-positive cells single and double staining was performed on tissue sections with the macrophage marker KP1 (CD68) and with the astrocyte marker glial fibrillary acidic protein (GFAP). Areas of myelin breakdown and demyelination were determined using a staining for neutral lipids, Oil Red O (ORO). Furthermore, macrophages isolated from active demyelinating MS lesions were stained for iNOS, cNOS, KP1 and ORO. In active MS lesions strong iNOS immunoreactivity was found exclusively in perivascular and parenchymal macrophages distributed within regions of active demyelination. In these active MS lesions immunoreactivity for cNOS was also found in macrophages. Macrophages isolated from active MS lesions also showed immunoreactivity for iNOS and cNOS. Moreover, these isolated macrophages produced nitric oxide (NO; >30 microM) in vitro. NADPH-diaphorase activity was detected in KP1-positive perivascular and parenchymal macrophages and in GFAP-positive reactive astrocytes in active MS lesions and in reactive astrocytes located in the hypercellular rims of chronic active MS lesions. cNOS-positive reactive astrocytes were detected in both active and chronic active MS lesions. Inside chronic active lesions some residual macrophages were weakly iNOS-positive. In control brain and spinal cord no iNOS immunoreactivity could be detected. These results suggests an important role for human macrophages capable of producing the free radical nitric oxide (NO), which may contribute to the cytotoxicity of oligodendrocytes and destruction of myelin in MS brain and spinal cord.


Subject(s)
Brain/enzymology , Demyelinating Diseases , Gene Expression Regulation, Enzymologic , Isoenzymes/biosynthesis , Multiple Sclerosis/enzymology , Nerve Tissue Proteins/biosynthesis , Nitric Oxide Synthase/biosynthesis , Spinal Cord/enzymology , Aged , Aged, 80 and over , Astrocytes/enzymology , Biomarkers , Brain/pathology , Enzyme Induction , Female , Humans , Isoenzymes/genetics , Macrophages/enzymology , Male , Middle Aged , Multiple Sclerosis/pathology , NADPH Dehydrogenase/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/enzymology , Nitric Oxide Synthase/classification , Nitric Oxide Synthase/genetics , Spinal Cord/pathology
9.
J Immunol ; 157(12): 5721-31, 1996 Dec 15.
Article in English | MEDLINE | ID: mdl-8955226

ABSTRACT

The mechanism of action underlying the beneficial effect of IFN-beta in multiple sclerosis (MS) is not understood. To date, little information is available on the effects of IFN-beta in experimental autoimmune encephalomyelitis (EAE), the animal correlate of the human disease MS. Therefore, we investigated the effects of recombinant rat IFN-beta (rrIFN-beta) on EAE in Lewis rats with emphasis on a treatment regimen during the paralytic phase of the disease. The results indicated that rrIFN-beta dose-dependently inhibited disease activity with complete prevention at a s.c. dose of 300,000 U/day, provided that treatment was continued for 3 wk. Discontinuation of treatment on day 17 postimmunization resulted in a protracted and relapsing disease course with strongly enhanced clinical severity. Detailed immunohistology of central nervous system (CNS) tissue of protected animals revealed an almost complete absence of CNS lesions and a >90% reduction in the number of infiltrating leukocytes. Accordingly, isolation of mononuclear cells from spinal cord tissue of successfully treated EAE rats revealed a reduction of approximately 95% in the number of cells that produce IFN-gamma in response to the encephalitogenic peptide MBP63-88. Furthermore, rrIFN-beta significantly enhanced serum corticosterone levels, which showed an inverse relationship with disease activity. We show that rrIFN-beta can have both beneficial and detrimental effects on disease activity dependent on the timing and the duration of treatment. Beneficial effects on EAE are associated with inhibition of the extravasation of blood-derived mononuclear cells in the CNS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Interferon-beta/administration & dosage , Animals , Autoantibodies/immunology , Body Weight , Corticosterone/biosynthesis , Dose-Response Relationship, Drug , Drug Administration Schedule , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Interferon-gamma/biosynthesis , Lymphoid Tissue/pathology , Myelin Basic Protein/immunology , Rats , Rats, Inbred Lew , Spinal Cord/immunology , Spinal Cord/pathology , Time Factors
10.
J Neuroimmunol ; 70(2): 145-52, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8898723

ABSTRACT

Demyelination of axons in the central nervous system (CNS) during multiple sclerosis (MS) and its animal model experimental allergic encephalomyelitis (EAE) is a result of phagocytosis and digestion by macrophages (M phi) and the local release of inflammatory mediators like tumor necrosis factor-alpha (TNF-alpha) and nitric oxide (NO). We have investigated the process of myelin phagocytosis by M phi in vitro using flow cytometric analysis. The binding and uptake of CNS-derived myelin was dose dependent, was abolished in the presence of EDTA and was enhanced after opsonization with complement. The phagocytosis of opsonized myelin could be inhibited by antibodies directed against complement receptor type 3 (CR3). Furthermore, CR3 also contributes to phagocytosis of non-opsonized myelin, e.g. under serum-free conditions. The phagocytosis of CNS-derived myelin induced the production of substantial amounts of TNF-alpha and NO by the M phi. Our results indicate an important role for CR3 in myelin phagocytosis. The induction of TNF-alpha and NO which accompanies this phagocytosis may further contribute to the overall process of demyelination during MS or EAE.


Subject(s)
Macrophage-1 Antigen/physiology , Macrophages/immunology , Myelin Sheath/immunology , Animals , Brain/immunology , Complement C3/metabolism , Flow Cytometry , Immunity, Cellular , Microscopy, Electron , Nitric Oxide/biosynthesis , Phagocytosis , Rats , Tumor Necrosis Factor-alpha/biosynthesis
11.
Clin Exp Immunol ; 103(3): 467-74, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8608648

ABSTRACT

Macrophages constitute a large proportion of the inflammatory cells that infiltrate the central nervous system (CNS) of animals with EAE. Through the production of inflammatory mediators these infiltrating macrophages can contribute to the regulation of the immune reaction within the CNS, that eventually results in neurological deficits associated with EAE. NO, a free radical produced by macrophages and other cell types, has been put forward as such an immune mediator. In the present study we show that macrophages isolated from the CNS of Lewis rats with clinical signs of EAE produce elevated amounts of NO. We treated rats, in which EAE was induced, with N(omega) -nitro-L-arginine-methylester or N(g)-monomethyl-L-arginine, inhibitors of NO synthase, either systemically via intraperitoneal injection, or intracerebrally via a cannula placed in the lateral ventricle. Both treatments resulted in a marked aggravation of clinical signs of EAE. These data point to an important role of NO, produced by infiltrating macrophages, as an immune-suppressor in the disease process during EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Enzyme Inhibitors/toxicity , Nitric Oxide Synthase/antagonists & inhibitors , Animals , Arginine/analogs & derivatives , Arginine/toxicity , Brain/cytology , Cells, Cultured , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/metabolism , Enzyme Inhibitors/pharmacology , Female , Guinea Pigs , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , NG-Nitroarginine Methyl Ester , Nitric Oxide/biosynthesis , Nitric Oxide/immunology , Rats , Rats, Inbred Lew
12.
Histochem J ; 28(2): 83-97, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8737290

ABSTRACT

In this review the role of various subpopulations of macrophages in the pathogenesis of experimental autoimmune encephalomyetitis is discussed. Immunohistochemistry with macrophage markers shows that in this disease different populations of macrophages (i.e. perivascular cells, microglia and infiltrating blood-borne macrophages) are present in the central nervous system. These subpopulations partially overlap in some functional activity while other activities seem to be restricted to a distinct subpopulation, indicating that these subpopulations have different roles in the pathogenesis of encephalomyelitis. The studies discussed in this review reveal that immunocytochemical and morphological studies, combined with new techniques such as in situ nick translation and experimental approaches like the use of bone marrow chimeras and macrophage depletion techniques, give valuable information about the types and functions of cells involved in central nervous system inflammation. The review is divided in three parts. In the first part the experimental autoimmune encephalomyelitis model is introduced. The second part gives an overview of the origin, morphology and functions of the various subpopulations. In the third part the role of these subpopulations is discussed in relation to the various stages (i.e. preclinical, clinical and recovery) of the experimental disease.


Subject(s)
Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Macrophages/immunology , Animals , Humans , Inflammation/immunology , Microglia/immunology , Multiple Sclerosis/immunology
13.
Clin Exp Immunol ; 100(2): 344-51, 1995 May.
Article in English | MEDLINE | ID: mdl-7743675

ABSTRACT

About 50% of the mononuclear cells in the perivascular lesions in the central nervous system (CNS) of rats suffering from experimental allergic encephalomyelitis (EAE) are blood-borne macrophages. In this study we investigated the role of these macrophages in different variants of EAE, using a liposome-mediated macrophage depletion technique. Intravenously injected liposomes containing dichloromethylene diphosphonate (Cl2MDP) are ingested by macrophages and cause temporary and selective elimination of these cells. Macrophage depletion during EAE induced by a T cell line specific for myelin basic protein (MBP; T cell-EAE) suppresses development of neurological signs of EAE. T cell-EAE with pronounced demyelination as induced by an additionally injected MoAb directed against myelin oligodendrocyte glycoprotein (MOG) was also significantly ameliorated after macrophage depletion. During chronic relapsing EAE (CR-EAE) the occurrence of relapses was prevented or suppressed, provided that the liposomes were injected before the initiation of a putative relapse. A chronic progressive course of CR-EAE was not modified by Cl2MDP containing liposome treatment. Histologic examination of the CNS of liposome-treated animals confirmed decreased infiltration of macrophages into the parenchyma in the rats with T cell and AD-EAE, whereas T cells were still present.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Macrophages/immunology , Animals , Clodronic Acid/pharmacology , Cyclosporine/pharmacology , Demyelinating Diseases/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Male , Rats , Rats, Inbred Lew , T-Lymphocytes/immunology
14.
J Neuroimmunol ; 56(2): 207-17, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7860716

ABSTRACT

During experimental allergic encephalomyelitis (EAE), both blood-borne macrophages as well as activated, resident microglial cells are considered to be involved in inflammatory reactions in the central nervous system (CNS), resulting in the neurological deficits common to EAE. Both cell types can produce multiple mediators of tissue damage, among which are the reactive oxygen species (ROS). In this study we show that macrophages and microglial cells, isolated from the CNS of Lewis rats with clinical signs of EAE, exhibited significantly elevated spontaneous and phorbol myristate acetate (PMA)-inducible levels of ROS compared to similar cells isolated from healthy controls, sham (complete Freund's adjuvant, CFA)-immunized rats as well as rats sacrificed before the manifestation of clinical signs of EAE. However, during clinical EAE, peripheral blood mononuclear cells (PBMC) did not show increased spontaneous nor PMA-inducible ROS production compared to controls. In vivo treatment of EAE with catalase, which scavenges the ROS H2O2, markedly suppressed the severity of the disease as compared to sham (albumin)-treated controls. In contrast, superoxide dismutase had no effect on clinical signs. Our studies point at a putative functional role for ROS, and in particular H2O2, in the pathogenesis of EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/etiology , Reactive Oxygen Species/toxicity , Animals , Catalase/pharmacology , Female , Guinea Pigs , Hydrogen Peroxide/metabolism , Leukocytes/metabolism , Luminescent Measurements , Male , Microglia/metabolism , Rats , Rats, Inbred Lew , Tetradecanoylphorbol Acetate/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis
15.
Eur J Pharmacol ; 195(2): 295-300, 1991 Mar 26.
Article in English | MEDLINE | ID: mdl-1651867

ABSTRACT

The beta-adrenoceptor-sensitive adenylate cyclase in primary cultures of rat striatal neurons was inhibited by opioids, unlike that in rat striatal slices. Isoprenaline (1 microM)-stimulated cyclic AMP production was dose dependently inhibited by the mu-opioid receptor agonist. [D-Ala2,MePhe4,Gly-ol5]enkephalin (DAGO, EC50 = 0.02 microM, 36% inhibition), and only slightly reduced by relatively high concentrations of the delta-opioid receptor agonist, [D-penicillamine2, D-penicillamine5]enkephalin (DPDPE, 1 microM). The highly selective and potent delta-opioid receptor agonist. [D-Ser2(O-tert-butyl),Leu5]enkephalyl-Thr6 (DSTBULET), and the kappa-opioid receptor agonist, U50-488, were ineffective in concentrations up to 3 microM. Naloxone reversed equally well the inhibitory effects of DPDPE and of DAGO, indicating the involvement of functional mu-opioid receptors. The isoprenaline (1 microM)-stimulated adenylate cyclase activity in cultured glial cells, which exceeded that in neurons about 10-fold, was not affected by opioids. Therefore, opioids were ineffective in rat brain slices probably due to the fact that cyclic AMP production induced by beta-adrenoceptor activation occurs primarily in the glial cells, where it is not subject to inhibition by opioids. These data indicate for the first time the existence of an interaction between functional mu-opioid receptors and beta-adrenoceptors on striatal neurons of the rat.


Subject(s)
Adenylyl Cyclase Inhibitors , Corpus Striatum/physiology , Neurons/physiology , Receptors, Adrenergic, beta/physiology , Receptors, Opioid/physiology , Analgesics/pharmacology , Animals , Astrocytes/metabolism , Cells, Cultured , Corpus Striatum/cytology , Corpus Striatum/enzymology , Cyclic AMP/biosynthesis , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Enkephalin, D-Penicillamine (2,5)- , Enkephalins/pharmacology , Isoproterenol/pharmacology , Male , Naloxone/pharmacology , Neuroglia/metabolism , Neurons/enzymology , Norepinephrine/metabolism , Oligopeptides/pharmacology , Rats , Rats, Inbred Strains , Receptors, Opioid, mu
SELECTION OF CITATIONS
SEARCH DETAIL
...