Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
1.
Neurotox Res ; 37(4): 800-814, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32026358

ABSTRACT

Disturbances in the function of the mesostriatal dopamine system may contribute to the development and maintenance of chronic pain, including its sensory and emotional/cognitive aspects. In the present study, we assessed the influence of chronic constriction injury (CCI) of the sciatic nerve on the expression of genes coding for dopamine and opioid receptors as well as opioid propeptides in the mouse mesostriatal system, particularly in the nucleus accumbens. We demonstrated bilateral increases in mRNA levels of the dopamine D1 and D2 receptors (the latter accompanied by elevated protein level), opioid propeptides proenkephalin and prodynorphin, as well as delta and kappa (but not mu) opioid receptors in the nucleus accumbens at 7 to 14 days after CCI. These results show that CCI-induced neuropathic pain is accompanied by a major transcriptional dysregulation of molecules involved in dopaminergic and opioidergic signaling in the striatum/nucleus accumbens. Possible functional consequences of these changes include opposite effects of upregulated enkephalin/delta opioid receptor signaling vs. dynorphin/kappa opioid receptor signaling, with the former most likely having an analgesic effect and the latter exacerbating pain and contributing to pain-related negative emotional states.


Subject(s)
Neuralgia/metabolism , Pain Measurement/methods , Prosencephalon/metabolism , Receptors, Dopamine/biosynthesis , Receptors, Opioid, delta/biosynthesis , Receptors, Opioid, kappa/biosynthesis , Animals , Corpus Striatum/metabolism , Enkephalins/biosynthesis , Enkephalins/genetics , Gene Expression , Male , Mice , Neuralgia/genetics , Protein Precursors/biosynthesis , Protein Precursors/genetics , Receptors, Dopamine/genetics , Receptors, Opioid, delta/genetics , Receptors, Opioid, kappa/genetics , Receptors, Opioid, mu/biosynthesis , Receptors, Opioid, mu/genetics
2.
Biomed Pharmacother ; 109: 938-944, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30551548

ABSTRACT

Social isolation stress (SIS) as a type of chronic stress could induce depressive- and anxiety-like behaviors. Our study evaluates the role of opioid system on negative behavioral impacts of SIS in male NMRI mice. We investigated effects of morphine, a nonselective opioid receptor (OR) agonist, naltrexone (NLX), an OR antagonist, naltrindole (NLT), a delta opioid receptor (DOR) antagonist, SNC80, a DOR agonist, U-69593, a kappa opioid receptor (KOR) agonist, nor-Binaltorphimine, a selective KOR antagonist and cyprodime hydrochloride a selective mu opioid receptor (MOR) antagonist on depressive- and anxiety-like behaviors. Using RT-PCR we evaluated ORs gene expression in mice brain. Our findings showed that SIS induced anxiety- and depressive-like behavior in the forced swimming test, open field test, splash test and hole-board test. Moreover, administration of SNC-80 significantly mitigated anxiety- and depressive-like behaviors. NLT decreased grooming-activity in the splash test. Excitingly, administration of agents affecting KOR failed to alter the negative effects of SIS. RT-PCR demonstrated that MOR and KOR gene expression decreased in socially isolated mice; however, SIS did not affect DORs expression. Our findings suggest that SIS at least in part, probably via altering endogenous opioids particularly MORs and KORs but not DORs mediated negative impacts on behavior; also, it could be concluded that DORs might be considered as a novel target for studying depression and anxiety.


Subject(s)
Receptors, Opioid, delta/biosynthesis , Receptors, Opioid, kappa/biosynthesis , Receptors, Opioid, mu/biosynthesis , Social Isolation/psychology , Stress, Psychological/metabolism , Stress, Psychological/psychology , Analgesics, Opioid/pharmacology , Animals , Anxiety/metabolism , Anxiety/psychology , Depression/metabolism , Depression/psychology , Male , Mice , Narcotic Antagonists/pharmacology , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors
3.
J Neuroimmunol ; 321: 12-23, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29957382

ABSTRACT

Regulation of µ-, δ- and κ-opioid receptor protein level in spleen lymphocytes when stimulated by mitogen is not known. To answer the question whether these cells do express opioid receptor (OR) proteins, primary, fresh rat spleen lymphocytes were prepared and stimulated for 48 h with mitogenic dose of Con A. The unstimulated lymphocytes did not express µ- and δ-OR proteins in detectable amounts, however, stimulation with Con A resulted in appearance of clearly detectable immunoblot signals of both µ-OR and δ-OR. κ-OR were detected already in primary cells and increased 2.4-fold in Con A-stimulated cells. These results were supported by data obtained by flow cytometry analysis indicating a dramatic increase in number of µ-, δ- and κ-OR expressing cells after mitogen stimulation. The newly synthesized µ-, δ- and κ-OR in Con A-stimulated spleen lymphocytes were present in the cells interior and not functionally mature, at least in terms of their ability to enhance activity of trimeric G proteins determined by three different protocols of agonist-stimulated, high-affinity [35S]GTPγS binding assay. The up-regulation of µ-, δ- and κ-OR was associated with specific decrease of their cognate trimeric G proteins, Gi1α/Gi2α; the other Gα and Gß subunits were unchanged. The level of ß-arrestin-1/2 was also decreased in Con A-stimulated splenocytes. We conclude that up-regulation of OR expression level in spleen lymphocytes by Con A proceeds in conjunction with down-regulation of their intracellular signaling partners, Gi1α/Gi2α proteins and ß-arrestin-1/2. These regulatory proteins are expressed in high amounts already in unstimulated cells and decreased by mitogen stimulation.


Subject(s)
Lymphocytes/metabolism , Receptors, Opioid, delta/biosynthesis , Receptors, Opioid, kappa/biosynthesis , Receptors, Opioid, mu/biosynthesis , Spleen/metabolism , Animals , Concanavalin A/pharmacology , Lymphocytes/drug effects , Male , Mitogens/pharmacology , Rats , Rats, Wistar , Receptors, Opioid, delta/drug effects , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, mu/drug effects , Spleen/cytology , Spleen/drug effects , Up-Regulation
4.
Neuron ; 98(1): 90-108.e5, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29576387

ABSTRACT

Cellular interactions between delta and mu opioid receptors (DORs and MORs), including heteromerization, are thought to regulate opioid analgesia. However, the identity of the nociceptive neurons in which such interactions could occur in vivo remains elusive. Here we show that DOR-MOR co-expression is limited to small populations of excitatory interneurons and projection neurons in the spinal cord dorsal horn and unexpectedly predominates in ventral horn motor circuits. Similarly, DOR-MOR co-expression is rare in parabrachial, amygdalar, and cortical brain regions processing nociceptive information. We further demonstrate that in the discrete DOR-MOR co-expressing nociceptive neurons, the two receptors internalize and function independently. Finally, conditional knockout experiments revealed that DORs selectively regulate mechanical pain by controlling the excitability of somatostatin-positive dorsal horn interneurons. Collectively, our results illuminate the functional organization of DORs and MORs in CNS pain circuits and reappraise the importance of DOR-MOR cellular interactions for developing novel opioid analgesics.


Subject(s)
Anterior Horn Cells/metabolism , Nerve Net/metabolism , Pain/metabolism , Posterior Horn Cells/metabolism , Receptors, Opioid, delta/biosynthesis , Receptors, Opioid, mu/biosynthesis , Animals , Anterior Horn Cells/chemistry , Anterior Horn Cells/pathology , Central Nervous System/chemistry , Central Nervous System/metabolism , Central Nervous System/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Nerve Net/chemistry , Nerve Net/pathology , Pain/pathology , Pain Measurement/methods , Posterior Horn Cells/chemistry , Posterior Horn Cells/pathology , Receptors, Opioid, delta/genetics , Receptors, Opioid, mu/genetics
5.
Br J Pharmacol ; 175(13): 2622-2634, 2018 07.
Article in English | MEDLINE | ID: mdl-29579315

ABSTRACT

BACKGROUND AND PURPOSE: To better understand opioid signalling in visceral nociceptors, we examined the expression and selective activation of µ and δ opioid receptors by dorsal root ganglia (DRG) neurons innervating the mouse colon. EXPERIMENTAL APPROACH: DRG neurons projecting to the colon were identified by retrograde tracing. δ receptor-GFP reporter mice, in situ hybridization, single-cell RT-PCR and µ receptor-specific antibodies were used to characterize expression of µ and δ receptors. Voltage-gated Ca2+ currents and neuronal excitability were recorded in small diameter nociceptive neurons (capacitance <30 pF) by patch clamp and ex vivo single-unit afferent recordings were obtained from the colon. KEY RESULTS: In situ hybridization of oprm1 expression in Fast Blue-labelled DRG neurons was observed in 61% of neurons. µ and δ receptors were expressed by 36-46% of colon DRG neurons, and co-expressed by ~25% of neurons. µ and δ receptor agonists inhibited Ca2+ currents in DRG, effects blocked by opioid antagonists. One or both agonists inhibited action potential firing by colonic afferent endings. Incubation of neurons with supernatants from inflamed colon segments inhibited Ca2+ currents and neuronal excitability. Antagonists of µ, but not δ receptors, inhibited the effects of these supernatant on Ca2+ currents, whereas both antagonists inhibited their actions on neuronal excitability. CONCLUSIONS AND IMPLICATIONS: A significant number of small diameter colonic nociceptors co-express µ and δ receptors and are inhibited by agonists and endogenous opioids in inflamed tissues. Thus, opioids that act at µ or δ receptors, or their heterodimers may be effective in treating visceral pain.


Subject(s)
Colon/metabolism , Nociceptors/metabolism , Receptors, Opioid, delta/biosynthesis , Receptors, Opioid, mu/biosynthesis , Animals , Gene Expression Profiling , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Inbred C57BL , Receptors, Opioid, delta/genetics , Receptors, Opioid, mu/genetics
6.
Handb Exp Pharmacol ; 247: 227-260, 2018.
Article in English | MEDLINE | ID: mdl-28035528

ABSTRACT

Delta opioid receptor (DOR) displays a unique, highly conserved, structure and an original pattern of distribution in the central nervous system, pointing to a distinct and specific functional role among opioid peptide receptors. Over the last 15 years, in vivo pharmacology and genetic models have allowed significant advances in the understanding of this role. In this review, we will focus on the involvement of DOR in modulating different types of hippocampal- and striatal-dependent learning processes as well as motor function, motivation, and reward. Remarkably, DOR seems to play a key role in balancing hippocampal and striatal functions, with major implications for the control of cognitive performance and motor function under healthy and pathological conditions.


Subject(s)
Learning/physiology , Motivation/physiology , Receptors, Opioid, delta/physiology , Animals , Humans , Learning/drug effects , Motivation/drug effects , Receptors, Opioid, delta/biosynthesis , Receptors, Opioid, delta/drug effects , Reward
7.
Handb Exp Pharmacol ; 247: 87-114, 2018.
Article in English | MEDLINE | ID: mdl-28993838

ABSTRACT

The functional diversity of primary afferent neurons of the dorsal root ganglia (DRG) generates a variety of qualitatively and quantitatively distinct somatosensory experiences, from shooting pain to pleasant touch. In recent years, the identification of dozens of genetic markers specifically expressed by subpopulations of DRG neurons has dramatically improved our understanding of this diversity and provided the tools to manipulate their activity and uncover their molecular identity and function. Opioid receptors have long been known to be expressed by discrete populations of DRG neurons, in which they regulate cell excitability and neurotransmitter release. We review recent insights into the identity of the DRG neurons that express the delta opioid receptor (DOR) and the ion channel mechanisms that DOR engages in these cells to regulate sensory input. We highlight recent findings derived from DORGFP reporter mice and from in situ hybridization and RNA sequencing studies in wild-type mice that revealed DOR presence in cutaneous mechanosensory afferents eliciting touch and implicated in tactile allodynia. Mechanistically, we describe how DOR modulates opening of voltage-gated calcium channels (VGCCs) to control glutamatergic neurotransmission between somatosensory neurons and postsynaptic neurons in the spinal cord dorsal horn. We additionally discuss other potential signaling mechanisms, including those involving potassium channels, which DOR may engage to fine tune somatosensation. We conclude by discussing how this knowledge may explain the analgesic properties of DOR agonists against mechanical pain and uncovers an unanticipated specialized function for DOR in cutaneous mechanosensation.


Subject(s)
Neurons, Afferent/metabolism , Receptors, Opioid, delta/biosynthesis , Sensory Receptor Cells/metabolism , Animals , Ganglia, Spinal/metabolism , Humans , Pain/drug therapy , Pain/physiopathology , Receptors, Opioid, delta/agonists
8.
J Neurosci Res ; 94(12): 1531-1545, 2016 12.
Article in English | MEDLINE | ID: mdl-27661001

ABSTRACT

Brain injuries, such as cerebral hypoxia-ischemia (H-I), induce a regenerative response from the neural stem/progenitors (NSPs) of the subventricular zone (SVZ); however, the mechanisms that regulate this expansion have not yet been fully elucidated. The Notch- Delta-Serrate-Lag2 (DSL) signaling pathway is considered essential for the maintenance of neural stem cells, but it is not known if it is necessary for the expansion of the NSPs subsequent to perinatal H-I injury. Therefore, the aim of this study was to investigate whether this pathway contributes to NSP expansion in the SVZ after H-I and, if so, to establish whether this pathway is directly induced by H-I or regulated by paracrine factors. Here we report that Notch1 receptor induction and one of its ligands, Delta-like 1, precedes NSP expansion after perinatal H-I in P6 rat pups and that this increase occurs specifically in the most medial cell layers of the SVZ where the stem cells reside. Pharmacologically inhibiting Notch signaling in vivo diminished NSP expansion. With an in vitro model of H-I, Notch1 was not induced directly by hypoxia, but was stimulated by soluble factors, specifically leukemia inhibitory factor, produced by astrocytes within the SVZ. These data confirm the importance both of the Notch-DSL signaling pathway in the expansion of NSPs after H-I and in the role of the support cells in their niche. They further support the body of evidence that indicates that leukemia inhibitory factor is a key injury-induced cytokine that is stimulating the regenerative response of the NSPs. © 2016 Wiley Periodicals, Inc.


Subject(s)
Astrocytes/metabolism , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Leukemia Inhibitory Factor/biosynthesis , Nerve Regeneration , Neural Stem Cells , Animals , Cytokines/metabolism , Diamines/pharmacology , Female , Lateral Ventricles/pathology , Pregnancy , Rats , Rats, Wistar , Receptor, Notch1/biosynthesis , Receptor, Notch1/genetics , Receptors, Opioid, delta/biosynthesis , Signal Transduction , Thiazoles/pharmacology
9.
Oncol Rep ; 36(5): 2579-2586, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27665747

ABSTRACT

δ­opioid receptor (DOR) belongs to the family of G protein­coupled receptors (GPCRs). Numerous studies have shown that DOR is widely distributed in human peripheral tissues and is closely related to the development and progression of certain malignant tumours. However, there is controversy in the literature regarding whether DOR has an impact on the development and progression of human breast cancer. The present study comprehensively elaborates on the biological functions of DOR by determining the distribution of DOR expression in breast cancer tissues and cells and by further verifying the effects of DOR on breast cancer progression. DOR was found to be highly expressed in human breast cancer tissues and cells. In addition, the high expression level of DOR positively correlated with tumour grade and clinical stage and negatively correlated with breast cancer metastasis and prognosis. Upregulating and activating DOR promoted the proliferation of human breast cancer cells in a concentration­dependent manner within a specific concentration range, whereas downregulating or inhibiting DOR activation significantly suppressed cell proliferation. The majority of tumour cells were arrested in G1 phase, and some cells exhibited apoptosis. DOR upregulation and activation induced protein kinase C (PKC) activation, resulting in increased phosphorylation levels of extracellular signal­regulated kinases (ERKs). After inhibition of the PKC/ERK signalling pathway, the effects of DOR on breast cancer were significantly attenuated in vivo and in vitro. In summary, DOR is highly expressed in breast cancer and is closely related to its progression. These results suggest that DOR may serve as a potential biomarker for the early diagnosis of breast cancer and may be a viable molecular target for therapeutic intervention.


Subject(s)
Breast Neoplasms/genetics , Cell Proliferation/genetics , Protein Kinase C/genetics , Receptors, Opioid, delta/biosynthesis , Transcriptional Activation/genetics , Adult , Aged , Animals , Apoptosis/genetics , Breast Neoplasms/pathology , Cell Cycle Checkpoints/genetics , Disease Progression , Female , Humans , MAP Kinase Signaling System/genetics , MCF-7 Cells , Mice , Middle Aged , Phosphorylation , Protein Kinase C/biosynthesis , Receptors, Opioid, delta/genetics , Xenograft Model Antitumor Assays
10.
Cancer Lett ; 344(2): 212-22, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24291668

ABSTRACT

The aim of this study was to investigate the immunological mechanisms by which synthetic methionine enkephalin (MENK) exerts therapeutic effects on tumor growth. Our findings in vivo or in vitro show that MENK treatment either in vivo or in vitro could up-regulate the percentages of CD8+T cells, induce markers of activated T cells, increased cytotoxic activity against mouse S180 tumor cells and increase secretion of IFNγ. In addition, the adoptively transferred CD8+T cells, after either in vitro or in vivo treatment with MENK, result in significantly increased survival of S180 tumor-bearing mice and significant shrinkage in tumor growth. Opioid receptors are detected on normal CD8+T cells and exposure to MENK leads to increased expression of opioid receptors. Interaction between MENK and the opioid receptors on CD8+T cells appears to be essential for the activation of CTL, since the addition of naltrexone (NTX), an opioid receptor antagonist, significantly inhibits all of the effects of MENK. The evidence obtained indicates that the MENK-induced T cell signaling is associated with a significant up-regulation of Ca2+ influx into the cytoplasm and the translocation of NFAT2 into nucleus, and these signaling effects are also inhibited by naltrexone.


Subject(s)
Enkephalin, Methionine/pharmacology , Immunotherapy, Adoptive/methods , Sarcoma 180/immunology , Sarcoma 180/therapy , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Calcium/metabolism , Cell Nucleus/immunology , Cell Nucleus/metabolism , Enkephalin, Methionine/immunology , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred C57BL , Random Allocation , Receptors, Opioid, delta/biosynthesis , Receptors, Opioid, delta/immunology , Receptors, Opioid, mu/biosynthesis , Receptors, Opioid, mu/immunology , Sarcoma 180/drug therapy
11.
Methods Mol Biol ; 995: 43-54, 2013.
Article in English | MEDLINE | ID: mdl-23494371

ABSTRACT

Calcium signaling plays a major role in the function of cells. Measurement of intracellular calcium mobilization is a robust assay that can be performed in a high-throughput manner to study the effect of compounds on potential drug targets. Pharmaceutical companies frequently use calcium signaling assays to screen compound libraries on G-protein-coupled receptors (GPCRs). In this chapter we describe the application of FLIPR technology to the evaluation of GPCR-induced calcium mobilization. We also include the implications of GPCR hetero-oligomerization and the identification of heteromeric receptors as novel drug targets on high-throughput calcium screening.


Subject(s)
Benzylidene Compounds/pharmacology , Drug Evaluation, Preclinical/methods , Enkephalin, D-Penicillamine (2,5)-/pharmacology , High-Throughput Screening Assays/methods , Naltrexone/analogs & derivatives , Receptors, Opioid, delta/agonists , Calcium Signaling , Cell Culture Techniques , HEK293 Cells , Humans , Naltrexone/pharmacology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/biosynthesis , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, delta/biosynthesis , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Spectrometry, Fluorescence , Transfection
12.
J Neurochem ; 124(6): 808-20, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23286559

ABSTRACT

Although biochemical and physiological evidence suggests a strong interaction between striatal CB1 cannabinoid (CB1 R) and D2 dopamine (D2 R) receptors, the mechanisms are poorly understood. We targeted medium spiny neurons of the indirect pathway using shRNA to knockdown either CB1 R or D2 R. Chronic reduction in either receptor resulted in deficits in gene and protein expression for the alternative receptor and concomitantly increased expression of the cannabinoid receptor interacting protein 1a (CRIP1a), suggesting a novel role for CRIP1a in dopaminergic systems. Both CB1 R and D2 R knockdown reduced striatal dopaminergic-stimulated [(35) S]GTPγS binding, and D2 R knockdown reduced pallidal WIN55212-2-stimulated [(35) S]GTPγS binding. Decreased D2 R and CB1 R activity was associated with decreased striatal phosphoERK. A decrease in mRNA for opioid peptide precursors pDYN and pENK accompanied knockdown of CB1 Rs or D2 Rs, and over-expression of CRIP1a. Down-regulation in opioid peptide mRNAs was followed in time by increased DOR1 but not MOR1 expression, leading to increased [D-Pen2, D-Pen5]-enkephalin-stimulated [(35) S]GTPγS binding in the striatum. We conclude that mechanisms intrinsic to striatal medium spiny neurons or extrinsic via the indirect pathway adjust for changes in CB1 R or D2 R levels by modifying the expression and signaling capabilities of the alternative receptor as well as CRIP1a and the DELTA opioid system.


Subject(s)
Carrier Proteins/biosynthesis , Corpus Striatum/metabolism , Receptor, Cannabinoid, CB1/physiology , Receptors, Dopamine D2/physiology , Receptors, Opioid, delta/biosynthesis , Animals , Carrier Proteins/genetics , Carrier Proteins/physiology , Dopamine D2 Receptor Antagonists , Gene Expression Regulation , Gene Knockdown Techniques/methods , Male , Mice , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptors, Opioid, delta/genetics
13.
Cancer Immunol Immunother ; 61(10): 1699-711, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22392190

ABSTRACT

Methionine enkephalin (MENK), the endogenous neuropeptide, is known to exert direct effects on the neuroendocrine and the immune systems and participates in regulation of various functions of cells related to both the innate and adaptive immune systems. Dendritic cells (DCs) play important role in initiating and regulating T cell responses. The aim of this work is to investigate the effects of MENK on differentiation, maturation, and function of DCs derived from murine bone marrow progenitors (BM-derived DCs). Our result showed that MENK could induce BM-derived DCs to polarize predominantly to mDC subtype, rather than pDC both in vivo and in vitro, and this was in favor of Th1 response. BM-derived DCs, after treatment with MENK, up-regulated the expressions of MHC class II and key costimulatory molecules. Result by RT-PCR showed MENK could increase expressions of delta and kappa receptors on BM-derived DCs. Also MENK promoted BM-derived DCs to secret higher levels of proinflammatory cytokines of IL-12p70, TNF-α. Furthermore, differentiated BM-derived DCs treated with MENK displayed higher activity to induce allogeneic T cell proliferation and MENK also inhibited tumor growth in vivo and induced apoptosis of tumor cells in vitro. Thus, it is concluded that MENK could be an effective inducer of BM-derived DCs and might be a new therapeutic agent for cancer, as well as other immune handicapped disease. Also we may consider MENK as a potential adjuvant in vaccine preparation.


Subject(s)
Bone Marrow Cells/drug effects , Cell Differentiation/drug effects , Dendritic Cells/drug effects , Enkephalin, Methionine/pharmacology , Neurotransmitter Agents/pharmacology , Stem Cells/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cytokines/metabolism , Female , Genes, MHC Class II/drug effects , Humans , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Opioid, delta/biosynthesis , Receptors, Opioid, kappa/biosynthesis , Up-Regulation/drug effects
14.
Cereb Cortex ; 22(5): 1215-23, 2012 May.
Article in English | MEDLINE | ID: mdl-21810780

ABSTRACT

Certain cognitive deficits in schizophrenia have been linked to dysfunction of prefrontal cortical (PFC) γ-aminobutyric acid (GABA) neurons and appear neurodevelopmental in nature. Since opioids suppress GABA neuron activity, we conducted the first study to determine 1) whether the µ opioid receptor (MOR), δ opioid receptor (DOR), and opioid ligand proenkephalin are altered in the PFC of a large cohort of schizophrenia subjects and 2) the postnatal developmental trajectory in monkey PFC of opioid markers that are altered in schizophrenia. We used quantitative polymerase chain reaction to measure mRNA levels from 42 schizophrenia and 42 matched healthy comparison subjects; 18 monkeys chronically exposed to haloperidol, olanzapine, or placebo; and 49 monkeys aged 1 week-11.5 years. We found higher levels for MOR mRNA (+27%) in schizophrenia but no differences in DOR or proenkephalin mRNAs. Elevated MOR mRNA levels in schizophrenia did not appear to be explained by substance abuse, psychotropic medications, or illness chronicity. Finally, MOR mRNA levels declined through early postnatal development, stabilized shortly before adolescence and increased across adulthood in monkey PFC. In schizophrenia, higher MOR mRNA levels may contribute to suppressed PFC GABA neuron activity and might be attributable to alterations in the postnatal developmental trajectory of MOR signaling.


Subject(s)
Enkephalins/biosynthesis , Prefrontal Cortex/metabolism , Protein Precursors/biosynthesis , Receptors, Opioid, delta/biosynthesis , Receptors, Opioid, mu/biosynthesis , Schizophrenia/metabolism , Animals , Antipsychotic Agents/pharmacology , Benzodiazepines/pharmacology , Female , Haloperidol/pharmacology , Humans , Macaca fascicularis , Male , Middle Aged , Olanzapine , Polymerase Chain Reaction , Prefrontal Cortex/drug effects , Prefrontal Cortex/growth & development , RNA, Messenger/analysis , Schizophrenia/physiopathology
15.
Neuroscience ; 179: 9-22, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21277946

ABSTRACT

The hippocampal formation (HF) is an important site at which stress circuits and endogenous opioid systems intersect, likely playing a critical role in the interaction between stress and drug addiction. Prior study findings suggest that the stress-related neuropeptide corticotropin releasing factor (CRF) and the delta opioid receptor (DOR) may localize to similar neuronal populations within HF lamina. Here, hippocampal sections of male and cycling female adult Sprague-Dawley rats were processed for immunolabeling using antisera directed against the DOR and CRF peptide, as well as interneuron subtype markers somatostatin or parvalbumin, and analyzed by fluorescence and electron microscopy. Both DOR- and CRF-labeling was observed in interneurons in the CA1, CA3, and dentate hilus. Males and normal cycling females displayed a similar number of CRF immunoreactive neurons co-labeled with DOR and a similar average number of CRF-labeled neurons in the dentate hilus and stratum oriens of CA1 and CA3. In addition, 70% of DOR/CRF dual-labeled neurons in the hilar region co-labeled with somatostatin, suggesting a role for these interneurons in regulating perforant path input to dentate granule cells. Ultrastructural analysis of CRF-labeled axon terminals within the hilar region revealed that proestrus females have a similar number of CRF-labeled axon terminals that contain DORs compared to males but an increased number of CRF-labeled axon terminals without DORs. Taken together, these findings suggest that while DORs are anatomically positioned to modulate CRF immunoreactive interneuron activity and CRF peptide release, their ability to exert such regulatory activity may be compromised in females when estrogen levels are high.


Subject(s)
Corticotropin-Releasing Hormone/analysis , Hippocampus/metabolism , Interneurons/chemistry , Receptors, Opioid, delta/analysis , Animals , Corticotropin-Releasing Hormone/biosynthesis , Female , Fluorescent Antibody Technique , Immunohistochemistry , Interneurons/metabolism , Interneurons/ultrastructure , Male , Microscopy, Electron, Transmission , Proestrus/physiology , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/biosynthesis , Sex Characteristics
16.
FEBS J ; 277(13): 2815-29, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20528919

ABSTRACT

Sarco(endo)plasmic reticulum calcium ATPase (SERCA)2b maintains the cellular Ca(2+) homeostasis by transferring Ca(2+) from the cytosol to the lumen of the endoplasmic reticulum (ER). Recently, SERCA2b has also been shown to be involved in the biosynthesis of secreted and membrane proteins via direct protein-protein interactions, involving components of the ER folding and quality-control machinery, as well as newly synthesized G protein-coupled receptors. Here we demonstrate that the human delta opioid receptor (hdeltaOR) exists in a ternary complex with SERCA2b and the ER molecular chaperone calnexin. The interaction between SERCA2b and hdeltaOR in vivo did not require calnexin as it was independent of the C-terminal calnexin-interacting domain of SERCA2b. However, the receptor was able to mediate co-immunoprecipitation of calnexin with the C-terminally truncated SERCA2b. The association of SERCA2b with hdeltaOR was regulated in vitro by Ca(2+) and ATP in a manner that was opposite to the calnexin-hdeltaOR interaction. Importantly, co-expression of the catalytically inactive SERCA2b(D351A) or calnexin binding-compromised SERCA2bDeltaC mutants with the receptor decreased the expression of mature receptors in a manner that did not directly relate to changes in the ER Ca(2+) concentration. We conclude that dynamic interactions among SERCA2b, calnexin and the hdeltaOR precursor orchestrate receptor biogenesis and are regulated by Ca(2+) and ATP. We further hypothesize that the primary role of SERCA2b in this process is to act as a Ca(2+) sensor in the vicinity of active translocons, integrating protein folding with local fluctuations of ER Ca(2+) levels.


Subject(s)
Calnexin/metabolism , Receptors, Opioid, delta/biosynthesis , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Adenosine Triphosphate/metabolism , Calcium/metabolism , Humans
17.
J Pharmacol Exp Ther ; 334(3): 887-96, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20498253

ABSTRACT

Both delta-opioid receptor (DOPr) and cannabinoid-2 receptor (CB2R) agonists attenuate neuropathic pain, but the precise mechanism implicated in these effects is not completely elucidated. We investigated whether nitric oxide synthesized by neuronal (NOS1) or inducible (NOS2) nitric-oxide synthases could modulate DOPr and/or CB2R antiallodynic and antihyperalgesic effects through the peripheral nitric oxide-cGMP-protein kinase G (PKG) pathway activation and affect their expression during neuropathic pain. In wild-type (WT) mice at 21 days after chronic constriction of sciatic nerve, we evaluated the effects of [d-Pen(2),d-Pen(5)]-enkephalin (DPDPE); (2-methyl-1-propyl-1H-indol-3-yl)-1-naphthalenylmethanone (JWH-015); and a NOS1 [N-[(4S)-4-amino-5-[(2-aminoethyl)amino]pentyl]-N'-nitroguanidine tris(trifluoroacetate) salt; NANT], NOS2 [l-N(6)-(1-iminoethyl)-lysine; l-NIL], l-guanylate cyclase [1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one; ODQ], or PKG [(Rp)-8-(para-chlorophenylthio)guanosine-3',5'-cyclic monophosphorothioate; Rp-8-pCPT-cGMPs] inhibitor administered alone or combined. Expression of DOPr and CB2R mRNA in the spinal cord and dorsal root ganglia of naive and nerve-injured WT, NOS1-knockout (KO), and NOS2-KO mice, also was assessed. The subplantar administration of NANT, l-NIL, ODQ, or Rp-8-pCPT-cGMPs dose-dependently inhibited neuropathic pain and enhanced the local effects of DPDPE or JWH-015. Moreover, although the basal levels of DOPr and CB2R mRNA were similar between WT and NOS-KO animals, nerve injury only decreased (DOPr) or increased (CB2R) their expression in the dorsal root ganglia of WT and NOS2-KO mice, and not in NOS1-KO mice. Results suggest that inactivation of the nitric oxide-cGMP-PKG peripheral pathway triggered by NOS1 and NOS2 enhanced the peripheral actions of DOPr and CB2R agonists and that nitric oxide synthesized by NOS1 is implicated in the peripheral regulation of DOPr and CB2R gene transcription during neuropathic pain.


Subject(s)
Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Nitric Oxide/physiology , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/metabolism , Receptor, Cannabinoid, CB2/biosynthesis , Receptors, Opioid, delta/biosynthesis , Animals , Cyclic GMP-Dependent Protein Kinases/metabolism , Ganglia, Spinal/metabolism , Hot Temperature , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type II/genetics , Pain Measurement/drug effects , Physical Stimulation , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Sciatic Neuropathy/pathology , Signal Transduction/drug effects , Spinal Cord/metabolism
18.
J Neurosci ; 30(13): 4735-45, 2010 Mar 31.
Article in English | MEDLINE | ID: mdl-20357124

ABSTRACT

By sustained activation of mu-opioid receptors (MORs), chronic opioids cause analgesic tolerance, physical dependence, and opioid addiction, common clinical problems for which an effective treatment is still lacking. Chronic opioids recruit delta-opioid receptors (DORs) to plasma membrane through exocytotic trafficking, but the role of this new DOR and its interaction with existing MOR in brain functions and in these clinical problems remain largely unknown. In this study, we investigated the mechanisms underlying synaptic and behavioral actions of chronic morphine-induced DORs and their interaction with MORs in nucleus raphe magnus (NRM) neurons important for opioid analgesia. We found that the emerged DOR inhibited GABAergic IPSCs through both the phospholipase A(2) (PLA(2)) and cAMP/protein kinase A (PKA) signaling pathways. MOR inhibition of IPSCs, normally mediated predominantly by the PLA(2) pathway, was additionally mediated by the cAMP/PKA pathway, with MOR potency significantly increased after chronic morphine treatment. Isobologram analysis revealed a synergistic DOR-MOR interaction in their IPSC inhibition, which was dependent on upregulated activities of both the PLA(2) and cAMP/PKA pathways. Furthermore, DOR and MOR agonists microinjected into the NRM in vivo also produced a PLA(2)-dependent synergism in their antinociceptive effects. These findings suggest that the cAMP/PKA pathway, upregulated by chronic opioids, becomes more important in the mechanisms of both MOR and DOR inhibition of GABA synaptic transmission after chronic opioid exposure, and DORs and MORs are synergic both synaptically and behaviorally in producing analgesic effects in a PLA(2)-dependent fashion, supporting the potential therapeutic use of DOR agonists in pain management under chronic opioid conditions.


Subject(s)
Receptors, Opioid, delta/physiology , Receptors, Opioid, mu/physiology , Synapses/physiology , Analgesics, Opioid/pharmacology , Animals , Brain Stem/drug effects , Brain Stem/metabolism , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Drug Synergism , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Morphine/pharmacology , Patch-Clamp Techniques , Phospholipases A2/metabolism , Phospholipases A2/physiology , Presynaptic Terminals/physiology , Rats , Rats, Wistar , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/biosynthesis , Receptors, Opioid, mu/agonists , Signal Transduction , Synapses/drug effects , Up-Regulation , gamma-Aminobutyric Acid/metabolism
19.
Neuroscience ; 168(2): 531-42, 2010 Jun 30.
Article in English | MEDLINE | ID: mdl-20167252

ABSTRACT

The delta opioid receptor (DOR) agonist [D-Ala2, D-Leu5] enkephalin (DADLE) has been implicated as a novel neuroprotective agent in the CNS. The current study was designed to evaluate the effects of intracerebroventricular (ICV) application of DADLE on neurological outcomes following asphyxial cardiac arrest (CA) in rats. Male Sprague-Dawley rats were randomly assigned to four groups: Sham group, CA group, DADLE group (DADLE+CA), and Naltrindole group (Naltrindole and DADLE+CA). All drugs were administered into the left cerebroventricle 30 min before CA. CA was induced by 8-min asphyxiation and the animals were resuscitated with a standardized method. DOR protein expression in the hippocampus was significantly increased in the CA group at 1 h after restoration of spontaneous circulation (ROSC) compared with the Sham group. As time progressed, expression of DOR proteins decreased gradually in the CA group. Treatment with DADLE alone or co-administration with Naltrindole reversed the down-regulation of DOR proteins in the hippocampus induced by CA at 24 h after ROSC. Compared with the CA group, the DADLE group had persistently better neurological functional recovery, as assessed by neurological deficit score (NDS) and Morris water maze trials. The number of surviving hippocampal CA1 neurons in the DADLE group was significantly higher than those in the CA group. However, administration of Naltrindole abolished most of the neuroprotective effects of DADLE. We conclude that ICV administration of DADLE 30 min before asphyxial CA has significant protective effects in attenuating hippocampal CA1 neuronal damage and neurological impairments, and that DADLE executes its effects mainly through DOR.


Subject(s)
Asphyxia/complications , Enkephalin, Leucine-2-Alanine/therapeutic use , Heart Arrest/drug therapy , Neuroprotective Agents/therapeutic use , Receptors, Opioid, delta/agonists , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , Cell Survival/drug effects , Enkephalin, Leucine-2-Alanine/administration & dosage , Heart Arrest/etiology , Heart Arrest/pathology , Heart Arrest/physiopathology , Injections, Intraventricular , Male , Maze Learning/drug effects , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/administration & dosage , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, delta/biosynthesis
20.
J Pharmacol Exp Ther ; 332(1): 255-65, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19828880

ABSTRACT

Tricyclic antidepressants (TCAs) have been reported to interact with the opioid system, but their pharmacological activity at opioid receptors has not yet been elucidated. In the present study, we investigated the actions of amoxapine, amitriptyline, nortriptyline, desipramine, and imipramine at distinct cloned and native opioid receptors. In Chinese hamster ovary (CHO) cells expressing delta-opioid receptors (CHO/DOR), TCAs displaced [3H]naltrindole binding and stimulated guanosine 5'-O-(3-[(35)S]thio)triphosphate ([(35)S]GTPgammaS) binding at micromolar concentrations with amoxapine displaying the highest potency and efficacy. Amoxapine and amitriptyline inhibited cyclic AMP formation and induced the phosphorylation of signaling molecules along the extracellular signal-regulated kinase 1/2 (ERK1/2) and phosphatidylinositol-3 kinase pathways. Amoxapine also activated delta-opioid receptors in rat dorsal striatum and nucleus accumbens and human frontal cortex. In CHO cells expressing kappa-opioid receptors (CHO/KOR), TCAs, but not amoxapine, exhibited higher receptor affinity and more potent stimulation of [(35)S]GTPgammaS binding than in CHO/DOR and effectively inhibited cyclic AMP accumulation. Amitriptyline regulated ERK1/2 phosphorylation and activity in CHO/KOR and C6 glioma cells endogenously expressing kappa-opioid receptors, and this effect was attenuated by the kappa-opioid antagonist nor-binaltorphimine. In rat nucleus accumbens, amitriptyline slightly inhibited adenylyl cyclase activity and counteracted the inhibitory effect of the full kappa agonist trans-(-)-3,4dichloro-N-methyl-N-[2-(1-pyrrolidinyl)cyclohexyl]benzeneacetamide (U50,488). At the cloned mu-opioid receptor, TCAs showed low affinity and no significant agonist activity. These results show that TCAs differentially regulate opioid receptors with a preferential agonist activity on either delta or kappa subtypes and suggest that this property may contribute to their therapeutic and/or side effects.


Subject(s)
Antidepressive Agents, Tricyclic/pharmacology , Receptors, Opioid, delta/agonists , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/agonists , Adult , Aged , Animals , CHO Cells , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Humans , Male , Middle Aged , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Protein Binding , Radioligand Assay , Rats , Receptors, Opioid, delta/biosynthesis , Receptors, Opioid, kappa/biosynthesis , Receptors, Opioid, mu/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...