Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Neuroinflammation ; 19(1): 288, 2022 Dec 03.
Article in English | MEDLINE | ID: mdl-36463243

ABSTRACT

BACKGROUND: Adverse pathophysiological and behavioral outcomes related to mild traumatic brain injury (mTBI), posttraumatic stress disorder (PTSD), and chronic pain are common following blast exposure and contribute to decreased quality of life, but underlying mechanisms and prophylactic/treatment options remain limited. The dynorphin/kappa opioid receptor (KOR) system helps regulate behavioral and inflammatory responses to stress and injury; however, it has yet to be investigated as a potential mechanism in either humans or animals exposed to blast. We hypothesized that blast-induced KOR activation mediates adverse outcomes related to inflammation and affective behavioral response. METHODS: C57Bl/6 adult male mice were singly or repeatedly exposed to either sham (anesthesia only) or blast delivered by a pneumatic shock tube. The selective KOR antagonist norBNI or vehicle (saline) was administered 72 h prior to repetitive blast or sham exposure. Serum and brain were collected 10 min or 4 h post-exposure for dynorphin A-like immunoreactivity and cytokine measurements, respectively. At 1-month post-exposure, mice were tested in a series of behavioral assays related to adverse outcomes reported by humans with blast trauma. RESULTS: Repetitive but not single blast exposure resulted in increased brain dynorphin A-like immunoreactivity. norBNI pretreatment blocked or significantly reduced blast-induced increase in serum and brain cytokines, including IL-6, at 4 h post exposure and aversive/anxiety-like behavioral dysfunction at 1-month post-exposure. CONCLUSIONS: Our findings demonstrate a previously unreported role for the dynorphin/KOR system as a mediator of biochemical and behavioral dysfunction following repetitive blast exposure and highlight this system as a potential prophylactic/therapeutic treatment target.


Subject(s)
Blast Injuries , Dynorphins , Receptors, Opioid, kappa , Animals , Male , Mice , Blast Injuries/complications , Blast Injuries/genetics , Blast Injuries/immunology , Brain/immunology , Brain/physiology , Dynorphins/genetics , Dynorphins/immunology , Quality of Life , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/immunology
2.
Salvador; s.n; 2015. 132 p. ilus, tab.
Thesis in Portuguese | LILACS | ID: biblio-1000963

ABSTRACT

Alguns estudos sugerem que as vias opioidérgicas centrais parecem desempenhar um papel regulatório no controle da ingestão de água e sal em mamíferos. As ações dos opioides centrais sobre a regulação do controle hidroeletrolítico são mediadas por vários dos subtipos de receptores opioides. O papel dos receptores delta e kappa-opioides centrais neste processo não está adequadamente elucidado sendo necessário mais estudos que o esclareçam. Objetivo: Este estudo investigou o envolvimento dos receptores delta e kappa-opioides centrais no apetite por sódio em ratos depletados deste íon e em rato ativados centralmente com angiotensina. Material e Métodos: Foram utilizados ratos Wistar (270 ± 20 g), submetidos à cirurgia estereotáxica para implante de cânula guia no ventrículo lateral esquerdo (VL), no órgão subfornical (OSF), no núcleo preóptico mediano (MnPO) e no núcleo basolateral da amígdala (BLA). No protocolo de depleção de sódio os animais foram submetidos à injeção subcutânea de furosemida combinada com dieta hipossódica quatro dias após a cirurgia. Neste modelo de estudo os animais receberam injeção intracerebroventricular (i.c.v.) do antagonista delta-opioide naltrindole no quinto dia pós-cirúrgico, nas doses de 5, 10 e 20 nmol/2 μL e do antagonista kappa-opioide, norbinaltorfimina, injetado no OSF, MnPO e BLA, nas doses de 0,5, 1,0 e 2,0 nmol/0,2 μL...


Central opioid pathways seem to have an important role on the control of water and salt intake in mammals, and brain opioid peptides may influence hydroelectrolyte balance through a myriad of actions mediated by distinct opioid receptors. The specific role of central delta and kappa-opioid receptors (DOR and KOR) in this process is far from being fully understood. In the present work, we investigated the role of those receptors in the control of water and salt intake, in sodium-depleted rats and rats with activation central angiotensinergic. Method: Wistar male rats (250 ± 20 g) were used in the experiment after stereotaxic cannulation of the VL left, SFO, MnPO and BLA. To study the effect of the blockade of central DOR and KOR on water and salt intake in rats were sodium depleted by the concomitant use of s.c. injections of furosemide and were kept in hypossodic diet, five days after surgery. In the sixth day, they received i.c.v. injections of a selective delta-opioid receptor antagonist (naltrindole) at the doses of 5, 10 and 20 nmol/2 μL and injections in the SFO, MnPO and BLA of a selective kappa-opioid receptor antagonist (norbinaltorphimine) at the doses of 0.5, 1.0 and 2.0 nmol/0.2 μL...


Subject(s)
Animals , Appetite , Appetite/physiology , Appetite/immunology , Receptors, Opioid, delta/analysis , Receptors, Opioid, delta/classification , Receptors, Opioid, delta/isolation & purification , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/analysis , Receptors, Opioid, kappa/immunology
3.
PLoS One ; 7(9): e46348, 2012.
Article in English | MEDLINE | ID: mdl-23029489

ABSTRACT

G-protein coupled receptors (GPCRs) play a major role in a number of physiological and pathological processes. Thus, GPCRs have become the most frequent targets for development of new therapeutic drugs. In this context, the availability of highly specific antibodies may be decisive to obtain reliable findings on localization, function and medical relevance of GPCRs. However, the rapid and easy generation of highly selective anti-GPCR antibodies is still a challenge. Herein, we report that highly specific antibodies suitable for detection of GPCRs in native and unfolded forms can be elicited by immunizing animals against purified full length denatured recombinant GPCRs. Contrasting with the currently admitted postulate, our study shows that an active and well-folded GPCR is not required for the production of specific anti-GPCR antibodies. This new immunizing strategy validated with three different human GPCR (µ-opioid, κ-opioid, neuropeptide FF2 receptors) might be generalized to other members of the GPCR family.


Subject(s)
Antibodies/immunology , Immunoglobulin G/biosynthesis , Receptors, Neuropeptide/immunology , Receptors, Opioid, kappa/immunology , Receptors, Opioid, mu/immunology , Amino Acid Sequence , Animals , Antibodies/isolation & purification , Humans , Immunization , Immunoglobulin G/immunology , Immunoglobulin G/isolation & purification , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Pichia/genetics , Protein Denaturation , Protein Folding , Receptors, Neuropeptide/administration & dosage , Receptors, Neuropeptide/genetics , Receptors, Opioid, kappa/administration & dosage , Receptors, Opioid, kappa/genetics , Receptors, Opioid, mu/administration & dosage , Receptors, Opioid, mu/genetics , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology
4.
J Neuroimmune Pharmacol ; 6(4): 608-16, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21938479

ABSTRACT

Here, we evaluated the influence of endogenous opioid activation on immune responses by examining consequences of all three opioid receptor gene (mu, delta and kappa) inactivation. In triple-opioid receptor knockout mice, splenocytes and thymocytes numbers, lymphocyte subsets as well as proliferation and cytokines induced by in vitro stimulation of T lymphocytes were measured. Compared with wild-type mice, similar lymphocyte distribution in thymus and spleen as well as comparable T lymphocyte proliferation were observed, while lower levels of IL-2 and IFNγ as well as higher levels of IL-4 and IL-10 were found in triple-opioid receptor knockout mice. Together, our results indicate a shift from TH1 to TH2 cytokines in triple-opioid receptor knockout animals, suggesting that global endogenous opioid tone drives T lymphocytes toward a TH1 profile under non-pathological conditions.


Subject(s)
Opioid Peptides/immunology , Receptors, Opioid, delta/immunology , Receptors, Opioid, kappa/immunology , Receptors, Opioid, mu/immunology , Th1 Cells/immunology , Animals , Cytokines/biosynthesis , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Opioid, delta/deficiency , Receptors, Opioid, delta/genetics , Receptors, Opioid, kappa/deficiency , Receptors, Opioid, kappa/genetics , Receptors, Opioid, mu/deficiency , Receptors, Opioid, mu/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spleen/cytology , Spleen/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Thymus Gland/cytology , Thymus Gland/immunology
5.
J Neuroimmune Pharmacol ; 6(4): 528-39, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21850403

ABSTRACT

Anti-retroviral therapy (ART) has had a tremendous impact on the clinical outcomes of HIV-1 infected individuals. While ART has produced many tangible benefits, chronic, long-term consequences of HIV infection have grown in importance. HIV-1-associated neurocognitive disorder (HAND) represents a collection of neurological syndromes that have a wide range of functional cognitive impairments. HAND remains a serious threat to AIDS patients, and there currently remains no specific therapy for the neurological manifestations of HIV-1. Based upon work in other models of neuroinflammation, kappa opioid receptors (KOR) and synthetic cannabinoids have emerged as having neuroprotective properties and the ability to dampen pro-inflammatory responses of glial cells; properties that may have a positive influence in HIV-1 neuropathogenesis. The ability of KOR ligands to inhibit HIV-1 production in human microglial cells and CD4 T lymphocytes, demonstrate neuroprotection, and dampen chemokine production in astrocytes provides encouraging data to suggest that KOR ligands may emerge as potential therapeutic agents in HIV neuropathogenesis. Based upon findings that synthetic cannabinoids inhibit HIV-1 expression in human microglia and suppress production of inflammatory mediators such as nitric oxide (NO) in human astrocytes, as well as a substantial literature demonstrating neuroprotective properties of cannabinoids in other systems, synthetic cannabinoids have also emerged as potential therapeutic agents in HIV neuropathogenesis. This review focuses on these two classes of compounds and describes the immunomodulatory and neuroprotective properties attributed to each in the context of HIV neuropathogenesis.


Subject(s)
AIDS Dementia Complex/immunology , Analgesics, Opioid/pharmacology , Cannabinoids/pharmacology , HIV-1/drug effects , Neuroimmunomodulation/immunology , Neuroprotective Agents/pharmacology , AIDS Dementia Complex/prevention & control , Analgesics, Opioid/immunology , Animals , Cannabinoids/immunology , Humans , Neuroprotective Agents/immunology , Receptors, Opioid, kappa/immunology
6.
J Exp Med ; 208(3): 617-29, 2011 Mar 14.
Article in English | MEDLINE | ID: mdl-21357741

ABSTRACT

Little is know about the nature of peripheral B cell tolerance or how it may vary in distinct lineages. Although autoantibody transgenic studies indicate that anergy and apoptosis are involved, some studies claim that receptor editing occurs. To model peripheral B cell tolerance in a normal, polyclonal immune system, we generated transgenic mice expressing an Igκ-light chain-reactive superantigen targeted to the plasma membrane of hepatocytes (pAlb mice). In contrast to mice expressing κ superantigen ubiquitously, in which κ cells edit efficiently to λ, in pAlb mice, κ B cells underwent clonal deletion. Their κ cells failed to populate lymph nodes, and the remaining splenic κ cells were anergic, arrested at a semi-mature stage without undergoing receptor editing. In the liver, κ cells recognized superantigen, down-regulated surface Ig, and expressed active caspase 3, suggesting ongoing apoptosis at the site of B cell receptor ligand expression. Some, apparently mature, κ B1 and follicular B cells persisted in the peritoneum. BAFF (B cell-activating factor belonging to the tumor necrosis factor family) overexpression rescued splenic κ B cell maturation and allowed κ cells to populate lymph nodes. Our model facilitates analysis of tissue-specific autoimmunity, tolerance, and apoptosis in a polyclonal B cell population. The results suggest that deletion, not editing, is the major irreversible pathway of tolerance induction among peripheral B cells.


Subject(s)
B-Lymphocytes/immunology , Bacterial Outer Membrane Proteins/immunology , Clonal Deletion/immunology , Liver/immunology , Porins/immunology , Receptors, Opioid, kappa/immunology , Receptors, Virus/immunology , Superantigens/immunology , Animals , Apoptosis/immunology , Autoimmunity/immunology , Hepatocytes/immunology , Immunoglobulin kappa-Chains/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic/immunology
7.
Methods Mol Biol ; 717: 197-219, 2011.
Article in English | MEDLINE | ID: mdl-21370032

ABSTRACT

Psychiatric disorders including anxiety, depression, and addiction are both precipitated and exacerbated by severe or chronic stress exposure. While acutely, stress responses are adaptive, repeated exposure to stress can dysregulate the brain in such a way as to predispose the organism to both physiological and mental illness. Understanding the neuronal chemicals, cell types, and circuits involved in both normal and pathological stress responses are essential in developing new therapeutics for psychiatric diseases. Varying degrees of stressor exposure cause the release of a constellation of chemicals, including neuropeptides such as dynorphin. Neuropeptidergic release can be very difficult to directly measure with adequate spatial and temporal resolution. Moreover, the downstream consequences following release and receptor binding are numerous and also difficult to measure with cellular resolution. Following repeated stressor exposure, dynorphin is released, binds to the kappa opioid receptor (KOR), and causes activation of KOR. Agonist-activated KOR becomes a substrate for G protein receptor kinase (GRK), which phosphorylates the Ser369 residue at the C-terminal tail of the receptor in the first step in the ß-Arrestin-dependent desensitization cascade. Through the use of phospho--selective antibodies developed and validated in the laboratory, we have the tools, to assess with fine cellular resolution, the strength of behavioral stimulus required for release, time course of the release, and regional location of release. We have gone on to show that following KOR activation, both ERK 1/2 and p38 MAP kinase phosphorylation are increased through use of commercially available phospho-selective antibodies. Finally, we have identified that one effector of KOR/p38MAP kinase is K(IR) 3.1 and have developed a phospho-selective antibody against the Y12 motif of this channel. Much like KOR and p38 MAP kinase, phosphorylation of this potassium channel increases following repeated stress. The following chapter discusses immunohistochemical and quantification methods used for phospho-selective antibodies used in various brain regions following behavioral manipulations.


Subject(s)
Antibodies, Phospho-Specific/analysis , Immunohistochemistry/methods , Potassium Channels, Inwardly Rectifying/metabolism , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Antibodies, Phospho-Specific/immunology , Antibodies, Phospho-Specific/isolation & purification , Brain/metabolism , Brain/ultrastructure , Cell Line , Chromatography, Affinity/methods , Humans , Mice , Microscopy/methods , Mitogen-Activated Protein Kinase 1/analysis , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/analysis , Mitogen-Activated Protein Kinase 3/immunology , Mitogen-Activated Protein Kinase 3/metabolism , Potassium Channels, Inwardly Rectifying/analysis , Potassium Channels, Inwardly Rectifying/immunology , Rats , Receptors, Opioid, kappa/analysis , Receptors, Opioid, kappa/immunology , p38 Mitogen-Activated Protein Kinases/analysis , p38 Mitogen-Activated Protein Kinases/immunology
8.
Biosens Bioelectron ; 26(1): 195-201, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20621464

ABSTRACT

Label-free detection of the interaction between hexahistidine-tagged human κ-opioid receptor membrane protein and anti-His antibody is demonstrated in liquid by an optical microelectromechanical system utilizing electromagnetically actuated microresonators. Shift in resonance frequency due to accretion of mass on the sensitive surface of microresonators is monitored via an integrated optical readout. A frequency resolution of 2Hz is obtained. Together with a sensitivity of 7 ppm/(ng/ml) this leads to a minimum detectable antibody concentration of 5.7 ng/ml for a 50-kHz device. The measurement principle is shown to impart immunity to environmental noise, facilitate operation in liquid media and bring about the prospect for further miniaturization of actuator and readout leading to a portable biochemical sensor.


Subject(s)
Antibodies/analysis , Biosensing Techniques/instrumentation , Immunoassay/instrumentation , Micro-Electrical-Mechanical Systems/instrumentation , Optical Devices , Receptors, Opioid, kappa/analysis , Refractometry/instrumentation , Antibodies/immunology , Equipment Design , Equipment Failure Analysis , Receptors, Opioid, kappa/immunology
9.
Pain ; 136(3): 320-330, 2008 Jun.
Article in English | MEDLINE | ID: mdl-17764840

ABSTRACT

Acute microinjection of mu-, delta-, or kappa-opioid receptor (MOPr, DOPr, KOPr) agonists into the rostral ventromedial medulla (RVM) produces antinociception. Thermal antinociception produced by MOPr and DOPr agonists is potentiated during inflammation [Hurley RW, Hammond DL. The analgesic effects of supraspinal mu and delta opioid receptor agonists are potentiated during persistent inflammation. J Neurosci 2000;20:1249-59]. Whether this potentiation extends to other stimulus modalities or to KOPr agonists is unknown. To examine these issues, rats received a unilateral intraplantar injection of complete Freund's adjuvant (CFA). Antinociception produced by RVM infusion of the KOPr agonist, U69593, and the MOPr agonist, DAMGO, was tested 4h-2 weeks thereafter. Thermal paw withdrawal latencies (PWLs) were assessed using the Hargreaves method. Mechanical thresholds were determined with the Von Frey and Randall-Selitto method. PWLs of the inflamed paw were reduced 4h-2 weeks after CFA injection. Infusion of either U69593 or DAMGO increased PWLs in CFA treated rats. A bilateral enhancement of the response to both agonists was observed 2 weeks relative to 4h post-CFA injection. Mechanical thresholds of the inflamed paw were decreased for >2 weeks post-CFA injection. Infusion of either agonist elevated thresholds of the inflamed and non-inflamed paws of CFA-treated rats. The magnitude of these effects was greater 2 weeks post-CFA injection for DAMGO and increased progressively for U69593. These data demonstrate that RVM infusion of MOPr or KOPr agonists attenuates CFA-evoked thermal and tactile allodynia and that these effects increase during prolonged inflammation. The augmented response of the non-inflamed paw to agonists suggests that inflammation induces centrally-mediated neuroplastic changes which enhance MOPr- and KOPr-mediated antinociception.


Subject(s)
Encephalitis/immunology , Hyperalgesia/immunology , Medulla Oblongata/immunology , Pain/immunology , Receptors, Opioid, kappa/immunology , Receptors, Opioid, mu/immunology , Animals , Male , Rats , Rats, Sprague-Dawley , Touch/immunology
10.
Brain Behav Immun ; 19(2): 153-64, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15664788

ABSTRACT

Activation of the hypothalamic-pituitary-adrenal axis (HPA) and sympathetic nervous system by stress has been shown to modulate both innate and adaptive immunity during an experimental influenza A/PR8 viral infection. HPA activation alters levels of glucocorticoids (GC) and opioids which are associated with suppression of lymphoid cellularity and NK activity. These experiments were designed to investigate the role that stress-induced GC and opioids have in modulating NK activity during an influenza viral infection. C57BL/6 mice were treated daily with mifepristone (RU486), a GC receptor antagonist or naltrexone (NTX), a non-specific opioid receptor antagonist. Mice were infected intranasally with A/PR8 virus and underwent daily restraint stress (RST). Phenotypic analysis of splenic cell populations and NK cytotoxicity were assessed at 3 days post-infection. RST of infected mice significantly suppressed splenic CD3(-)DX5+ cellularity and NK cytolytic activity. RU486 administration fully restored splenic NK cellularity but not cytolytic activity. NTX administration restored NK cytolytic activity but not splenic NK cell number. A similar restoration in NK cytolytic activity was observed after administration of beta-funaltrexamine (FNA), a mu-specific opioid receptor antagonist, but not the delta- or kappa-specific opioid receptor antagonists naltrindole or nor-binaltorphimine, respectively. Co-administration of RU486 and NTX restored both NK cellularity and cytolytic activity. The restoration of RST-induced suppression of NK activity by RU486 and NTX or FNA suggests that glucocorticoids modulate NK cellularity and opioids that bind to the mu-opioid receptor modulate NK cytotoxicity during periods of stress and viral infection.


Subject(s)
Influenza A virus , Killer Cells, Natural/immunology , Neuroimmunomodulation/immunology , Orthomyxoviridae Infections/immunology , Stress, Physiological/immunology , Animals , Glucocorticoids/immunology , Hormone Antagonists/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/virology , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Mifepristone/pharmacology , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Neuroimmunomodulation/drug effects , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, delta/immunology , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, kappa/immunology , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/immunology , Restraint, Physical , Spleen/cytology , Spleen/immunology
11.
Neuroimmunomodulation ; 11(6): 392-403, 2004.
Article in English | MEDLINE | ID: mdl-15467355

ABSTRACT

OBJECTIVE: Methionine-enkephalin (MET) modulates various functions of macrophages related to both immune and inflammatory reactions in a naloxone reversible manner, suggesting that opioid receptors are involved in the regulation of macrophage activity. Since an endogenous opioid ligand might interact with more than one type of opioid receptor, the receptor interaction determines its effect on a particular function. METHODS: In the present study we have investigated the involvement of different opioid receptor types/subtypes in MET-induced modulation of H(2)O(2) and NO production in macrophages. Thioglycollate-elicited or resident rat peritoneal macrophages were treated in vitro with MET and/or specific antagonists of delta(1,2), delta(1), delta(2), mu and kappa opioid receptors. RESULTS: MET increased H(2)O(2)production in phorbol myristate acetate-stimulated rat peritoneal macrophages mainly through delta(1) opioid receptor. MET also enhanced NO production in rat peritoneal macrophages stimulated with lipopolysaccharide through delta(1) and mu opioid receptors. The blockade of mu and kappa receptor facilitated a potentiating effect of MET on H(2)O(2) release, and blockade of kappa receptor further raised the MET-induced increase of NO production in macrophages. CONCLUSION: It is concluded that both negative and positive functional interaction between delta, mu and kappa opioid receptors regulate the influence of MET on H(2)O(2) and NO production in rat peritoneal macrophages.


Subject(s)
Enkephalin, Methionine/pharmacology , Hydrogen Peroxide/metabolism , Macrophages, Peritoneal/drug effects , Nitric Oxide/metabolism , Receptors, Opioid/drug effects , Animals , Dose-Response Relationship, Drug , Enkephalin, Methionine/immunology , Inflammation/chemically induced , Inflammation/immunology , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Male , Narcotic Antagonists/pharmacology , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/immunology , Rats , Rats, Wistar , Receptors, Opioid/immunology , Receptors, Opioid, delta/drug effects , Receptors, Opioid, delta/immunology , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, kappa/immunology , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/immunology , Tetradecanoylphorbol Acetate/pharmacology , Up-Regulation/drug effects , Up-Regulation/immunology
12.
Cell Tissue Res ; 307(1): 23-33, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11810311

ABSTRACT

Opioid drugs have profound antidiarrheal and constipating actions in the intestinal tract and are effective in mitigating abdominal pain. Mediators of intestinal inflammation and allergy produce increased mucosal secretion, altered bowel motility and pain due to their ability to evoke enteric secretomotor reflexes through primary afferent neurons. In this study, the distribution of delta- and kappa-opioid receptor (DOR and KOR, respectively) immunoreactivities in chemically identified neurons of the porcine ileum was compared with that of the capsaicin-sensitive type 1 vanilloid receptor (VR1). DOR and VR1 immunoreactivities were observed to be highly localized in choline acetyltransferase (ChAT)- and calcitonin gene-related peptide (CGRP)-positive neurons and nerve fibers of the submucosal and myenteric plexuses and both receptors exhibited frequent colocalization. In the inner submucosal plexus, they also were colocalized in substance P (SP)-positive neurons. Neurons in the outer submucosal plexus expressed DOR immunoreactivity alone or in combination with VR1. KOR-immunoreactive neurons were found only in the myenteric plexus; these cells coexpressed immunoreactivity to ChAT, CGRP, vasoactive intestinal peptide (VIP) or nitric oxide synthase (NOS). In addition, some KOR-positive neurons coexpressed immunoreactivities to DOR and VR1. Based on their neurochemical coding, opioid and vanilloid receptor-immunoreactive neurons in the submucosal and myenteric plexuses may include primary afferents and constitute novel therapeutic targets for the palliation of painful intestinal inflammatory, hypersensitivity and dysmotility states.


Subject(s)
Ileum/innervation , Receptors, Drug/metabolism , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Animals , Female , Ileum/immunology , Ileum/metabolism , Immunohistochemistry , Male , Myenteric Plexus/immunology , Myenteric Plexus/metabolism , Neurons/metabolism , Receptors, Drug/immunology , Receptors, Opioid, delta/immunology , Receptors, Opioid, kappa/immunology , Submucous Plexus/immunology , Submucous Plexus/metabolism , Swine
14.
Synapse ; 42(3): 185-92, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11746715

ABSTRACT

The behavioral effects of psychostimulants, which are produced at least in part through inhibition of the dopamine transporter (DAT), are modulated by kappa-opioid receptors (KOR) in the nucleus accumbens (Acb). Using electron microscopic immunocytochemistry, we reveal that in the Acb KOR labeling is mainly, and DAT immunoreactivity is exclusively, presynaptic. From 400 KOR-labeled presynaptic structures, including axon terminals, intervaricosities, and small axons, 51% expressed DAT and 29% contacted another population of terminals exclusively labeled for DAT. Within axonal profiles that contained both antigens, DAT and KOR were prominently localized to plasma membrane segments that showed overlapping distributions of the respective immunogold-silver and immunoperoxidase markers. KOR labeling was also localized to membranes of small synaptic vesicles in terminals with or without DAT immunoreactivity. In addition, from 24 KOR-immunoreactive dendritic spines 42% received convergent input from DAT-containing varicosities and unlabeled terminals forming asymmetric, excitatory-type synapses. Our results provide the first ultrastructural evidence that in the Acb, KOR is localized to strategic sites for involvement in the direct presynaptic release and/or reuptake of dopamine. These data also suggest a role for KOR in the presynaptic modulation of other neurotransmitters and in the postsynaptic excitatory responses of single spiny neurons in the Acb. Dual actions on dopamine terminals and their targets in the Acb may account for KOR-mediated attenuation of drug reinforcement and sensitization.


Subject(s)
Axons/chemistry , Membrane Glycoproteins , Membrane Transport Proteins/analysis , Nerve Tissue Proteins , Nucleus Accumbens/chemistry , Receptors, Opioid, kappa/analysis , Animals , Axons/ultrastructure , Cell Membrane/chemistry , Cell Membrane/ultrastructure , Dendrites/chemistry , Dendrites/ultrastructure , Dopamine/physiology , Dopamine Plasma Membrane Transport Proteins , Immunohistochemistry , Membrane Transport Proteins/biosynthesis , Membrane Transport Proteins/immunology , Microscopy, Immunoelectron , Nucleus Accumbens/cytology , Rats , Receptors, Opioid, kappa/biosynthesis , Receptors, Opioid, kappa/immunology
15.
Neuroscience ; 90(1): 229-34, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10188949

ABSTRACT

Activation of kappa-opioid receptors in the rostral ventral medulla has been reported to attenuate analgesia induced by activation of mu-opioid receptors in the periaqueductal gray matter. Previous studies have suggested that the cells associated with this effect might contain serotonin. In the present study, we investigated the relationship of the cloned kappa-opioid receptor to spinally projecting neurons immunoreactive for serotonin or GABA. This was done by employing two-color immunofluorescence in combination with retrograde tract-tracing using Fluoro-Gold. In the rostral ventral medulla, neurons triple-labeled for the cloned kappa-opioid receptor, serotonin and Fluoro-Gold were observed; neurons double-labeled for the cloned kappa-opioid receptor and serotonin, or single-labeled for the cloned kappa-opioid receptor or for serotonin were also observed. In addition, cloned kappa-opioid receptor immunoreactivity was expressed in some cell profiles immunoreactive for GABA. The expression of the cloned kappa-opioid receptor in the spinal cord dorsal horn was not associated with processes immunoreactive for serotonin. Our findings suggest that kappa-opioid receptors in the rostral ventral medulla are positioned to directly control the activity of at least some serotonergic neurons projecting to the dorsal spinal cord. Thus, it appears possible that the anti-analgesic action resulting from microinjection of kappa-opioid agonists into the rostral ventral medulla is mediated, at least in part, by these neurons.


Subject(s)
Medulla Oblongata/metabolism , Pons/metabolism , Receptors, Opioid, kappa/metabolism , Serotonin/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Immunohistochemistry , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/immunology
16.
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
17.
Ross Fiziol Zh Im I M Sechenova ; 84(4): 385-90, 1998 Apr.
Article in Russian | MEDLINE | ID: mdl-9742618

ABSTRACT

Activation of the opioid receptors by delta-agonist DSLET and by kappa-agonist rimorphin led to a significant inhibition of plaque-forming and rosette-forming cells in the CBA mice. On the other hand, mu-agonist DAGO stimulated the immune response on the 4th and 5th days after immunization with SRBC (5 x 10(8)). Lesion of the hypothalamo-hypophyseal connection prevented immuno-stimulating as well as immuno-depressive effects. The latter seems to be due to an interaction with the serotoninergic system, whereas immuno-stimulating effect involves the dopaminergic system.


Subject(s)
Neuroimmunomodulation/drug effects , Receptors, Opioid/agonists , Animals , Cyproheptadine/pharmacology , Dopamine D2 Receptor Antagonists , Dynorphins/pharmacology , Endorphins/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Enkephalin, Leucine/analogs & derivatives , Enkephalin, Leucine/pharmacology , Enkephalins/pharmacology , Haloperidol/pharmacology , Male , Mice , Mice, Inbred CBA , Neuroimmunomodulation/immunology , Pituitary Gland/immunology , Pituitary Gland/physiology , Receptor, Serotonin, 5-HT2A , Receptors, Dopamine D2/immunology , Receptors, Opioid/immunology , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/immunology , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/immunology , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/immunology , Receptors, Serotonin/drug effects , Receptors, Serotonin/immunology , Spleen/cytology , Spleen/drug effects , Spleen/immunology
18.
Glia ; 22(3): 249-59, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9482211

ABSTRACT

The diversity of opioid receptor expression was examined in astrocytes in low-density and non-dividing (confluent) cultures from the cerebral cortex, hippocampus, cerebellum, and striatum of 1-day-old mice. Mu, delta, and kappa opioid receptor expression was assessed in individual cells immunocytochemically, by using flow cytometry, and functionally by examining agonist-induced changes in intracellular calcium ([Ca2+]i). Significant spatial and temporal differences were evident in the pattern of expression of mu, delta, and kappa receptors among astrocytes. In low-density cultures, greater proportions of astrocytes expressed mu-opioid receptor immunoreactivity in the cerebral cortex and hippocampus (26-34%) than in the cerebellum or striatum (7-12%). At confluence, a greater percentage of astrocytes in cerebellar (26%) and striatal (30%) cultures expressed mu-immunoreactivity. Fewer astrocytes possessed delta-immunoreactivity in low-density striatal cultures (8%) compared to other regions (16-22%). The proportion of delta receptor-expressing astrocytes declined in the cerebellum but increased in the hippocampus. Kappa-opioid receptors were uniformly expressed by 27-34% of astrocytes from all regions, except in cortical cultures, where the proportion of kappa expressing cells was 38% at low-density and decreased to 22% at confluence. Selective mu (PLO 17; H-Tyr-Pro-Phe (N-Me) -D-Pro-NH2, delta ([D-Pen2, D-Pen5] enkephalin), or kappa (U50,488H; trans-(+/-)-3,4-Dichloro-N-methyl-N-[2-(1-pyrrolidinyl) cyclohexyl] benzeneacetamide methanesulfonate) opioid receptor agonists increased [Ca2+]i in subpopulations of astrocytes indicating the presence of functional receptors. Lastly, opioid receptor immunofluorescence varied during the cell division cycle. A greater proportion of astrocytes in the G2/M phase of the cell cycle were mu or delta receptor immunofluorescent than at G0/G1. When astrocytes were reversibly arrested in G1, significantly fewer cells expressed delta receptor immunofluorescence; however, upon reentry into the cell cycle immunofluorescent cells reappeared. In conclusion, opioid phenotype varies considerably among individual cultured astrocytes, and this diversity was determined by regional and developmental (age and cell cycle dependent) differences in the brain. These in vitro findings suggest astroglia contribute to regional and developmental idiosyncrasies in opioid function within the brain.


Subject(s)
Astrocytes/chemistry , Cell Cycle/physiology , Receptors, Opioid, delta/biosynthesis , Receptors, Opioid, kappa/biosynthesis , Receptors, Opioid, mu/biosynthesis , Receptors, Opioid/biosynthesis , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Age Factors , Analgesics/pharmacology , Analgesics, Non-Narcotic/pharmacology , Animals , Antibody Specificity , Astrocytes/cytology , Astrocytes/metabolism , Brain Chemistry/physiology , Calcium/metabolism , Cell Division/drug effects , Cells, Cultured , Cerebellum/chemistry , Cerebellum/cytology , Cerebral Cortex/chemistry , Cerebral Cortex/cytology , Corpus Striatum/chemistry , Corpus Striatum/cytology , Endorphins/pharmacology , Enkephalin, D-Penicillamine (2,5)- , Enkephalins/pharmacology , Flow Cytometry , Hippocampus/chemistry , Hippocampus/cytology , Mice , Mice, Inbred ICR , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Receptors, Opioid/analysis , Receptors, Opioid/immunology , Receptors, Opioid, delta/analysis , Receptors, Opioid, delta/immunology , Receptors, Opioid, kappa/analysis , Receptors, Opioid, kappa/immunology , Receptors, Opioid, mu/analysis , Receptors, Opioid, mu/immunology
19.
J Immunol ; 158(4): 1670-80, 1997 Feb 15.
Article in English | MEDLINE | ID: mdl-9029103

ABSTRACT

Site-directed polyclonal Abs specific for a synthetic peptide with sequence homology to the predicted N-terminal sequence of the human kappa opioid receptor [anti-kappa R-(33-52)] are capable of binding to normal human cells and cell lines expressing mRNA specific for the human kappa receptor. Flow cytometric analysis of 1) a neuronal cell line (NT2), 2) blood-derived CD14+ monocytes, 3) monocyte-like cell lines (U937 and THP 1), 4) blood-derived CD3+ T cells and a T cell line, and 5) human B cell lines bound anti-kappa R-(33-52) in a specific manner. Anti-kappa R-(33-52) was also found to specifically neutralize the immunosuppressive activities associated with the kappa R-selective agonist U50,488H. This antiserum was found to block U50,488H-mediated inhibition of 1) Staphylococcus aureus Cowen strain I-induced B and T lymphocyte proliferation, 2) PHA-induced T lymphocyte proliferation, and 3) S. aureus Cowen strain I-induced IgG production. However, this antiserum failed to neutralize mu R-selective agonist (Tyr-D-Ala-Gly-NMe-Phe-Gly-ol)-mediated suppression of IgG synthesis. Finally, the kappa R-selective antagonist nor-binaltorphimine hydrochloride inhibits the binding of anti-kappa R-(33-52) to the U937 cell line. These results suggest that anti-kappa R-(33-52) specifically interacts with the human kappa R molecule. Studies conducted with anti-kappa R-(33-52) indicated that this antiserum effectively blocked U50,488H-mediated immunosuppression, but by itself did not enhance or suppress lymphocyte activation. These data suggest that anti-kappa R-(33-52) 1) does not interact with the effector binding site of the receptor, but sterically interferes with U50,488H binding to the receptor; and/or 2) the antiserum interacts with a secondary binding site that is important for ligand binding, but may not be involved in signal transduction.


Subject(s)
Antibody Specificity , Immune Sera/chemistry , Immune Sera/pharmacology , Peptide Fragments/immunology , Receptors, Opioid, kappa/immunology , Adult , Amino Acid Sequence , Antibodies, Blocking/pharmacology , Base Sequence , Binding Sites, Antibody , Cell Line , Female , Humans , Immune Sera/biosynthesis , Immunoglobulin G/biosynthesis , Immunosuppressive Agents/antagonists & inhibitors , Immunosuppressive Agents/immunology , Jurkat Cells , Lymphocyte Activation , Lymphoma, Large B-Cell, Diffuse , Male , Molecular Sequence Data , Monocytes , Neuroblastoma , Peptide Fragments/chemistry , RNA, Messenger/biosynthesis , Receptors, Opioid, kappa/biosynthesis , Receptors, Opioid, kappa/chemistry , Receptors, Opioid, kappa/genetics
20.
J Immunol ; 158(3): 1377-82, 1997 Feb 01.
Article in English | MEDLINE | ID: mdl-9013982

ABSTRACT

A conformationally biased decapeptide agonist of human C5a anaphylatoxin (YSFKPMPLaR) was used as a molecular adjuvant in stimulating Ab responses against peptide epitopes derived from human MUC1 glycoprotein and the human mu and kappa opioid receptors. C57BL6 mice were immunized with the MUC1 epitope (YKQGGFLGL); the C5a agonist (YSFKPMPLaR); YSFKPMPLaR and YKQGGFLGL together, but unconjugated; a C5a-active, MUC1 epitope construct (YKQGGFLGLYSFKPMPLaR); and a C5a-inactive, reversed moiety construct (YSFKPMPLaRYKQGGFLGL). High Ab titers specific for the MUC1 epitope were observed only in mice immunized with the C5a-active epitope construct. Similar results were obtained in BALB/c mice immunized with the C5a-active, MUC1 epitope construct. Abs from the sera of the C57BL6 mice were predominately of the IgG2a, IgG2b, and IgM isotypes and were reactive against human recombinant MUC1 and MUC1 expressed by the Panc-1 M1F.15 pancreatic cell line. When compared with the corresponding KLH-epitope conjugates in C57BL6 mice, the epitope-C5a agonist constructs produced titers of specific IgG Abs of isotypes distinct from those generated by the keyhole limpet hemocyanin-epitope conjugates. Rabbits immunized with a mu opioid receptor epitope-C5a agonist construct (GDLSDPCGNRTNLGGRDSLYSFKPMPLaR) or a kappa opioid receptor epitope-C5a agonist construct (FPGWAEPDSNGSEDAQLYSFKPMPLaR) generated high titer, epitope-specific Ab responses. Ab titers generated in response to the opioid epitope-C5a agonist constructs were comparable to those generated by the opioid KLH-epitope conjugates. The results of this study are discussed in terms of possible mechanisms by which the conformationally biased C5a agonist serves as a molecular adjuvant.


Subject(s)
Adjuvants, Immunologic , Anaphylatoxins/chemistry , Complement C5a/agonists , Mucin-1/immunology , Adjuvants, Immunologic/chemistry , Amino Acid Sequence , Animals , Epitope Mapping , Humans , Immunologic Techniques , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Peptides/chemistry , Protein Conformation , Rabbits , Receptors, Opioid, kappa/immunology , Receptors, Opioid, mu/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...