51
|
El-Hage N, Gurwell JA, Singh IN, Knapp PE, Nath A, Hauser KF. Synergistic increases in intracellular Ca2+, and the release of MCP-1, RANTES, and IL-6 by astrocytes treated with opiates and HIV-1 Tat. Glia 2005; 50:91-106. [PMID: 15630704 PMCID: PMC4301446 DOI: 10.1002/glia.20148] [Citation(s) in RCA: 182] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recent evidence suggests that injection drug users who abuse heroin are at increased risk of CNS complications from human immunodeficiency virus (HIV) infection. Opiate drugs may intrinsically alter the pathogenesis of HIV by directly modulating immune function and by directly modifying the CNS response to HIV. Despite this, the mechanisms by which opiates increase the neuropathogenesis of HIV are uncertain. In the present study, we describe the effect of morphine and the HIV-1 protein toxin Tat(1-72) on astroglial function in cultures derived from ICR mice. Astroglia maintain the blood-brain barrier and influence inflammatory signaling in the CNS. Astrocytes can express mu-opioid receptors, and are likely targets for abused opiates, which preferentially activate mu-opioid receptors. While Tat alone disrupts astrocyte function, when combined with morphine, Tat causes synergistic increases in [Ca(2+)](i). Moreover, astrocyte cultures treated with morphine and Tat showed exaggerated increases in chemokine release, including monocyte chemoattractant protein-1 (MCP-1) and regulated on activation, normal T cell expressed and secreted (RANTES), as well as interleukin-6 (IL-6). Morphine-Tat interactions were prevented by the mu-opioid receptor antagonist beta-funaltrexamine, or by immunoneutralizing Tat(1-72) or substituting a nontoxic, deletion mutant (Tat(Delta31-61)). Our findings suggest that opiates may increase the vulnerability of the CNS to viral entry (via recruitment of monocytes/macrophages) and ensuing HIV encephalitis by synergistically increasing MCP-1 and RANTES release by astrocytes. The results further suggest that astrocytes are key intermediaries in opiate-HIV interactions and disruptions in astroglial function and inflammatory signaling may contribute to an accelerated neuropathogenesis in HIV-infected individuals who abuse opiates.
Collapse
Affiliation(s)
- Nazira El-Hage
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, 40536 USA
| | - Julie A. Gurwell
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, 40536 USA
| | - Indrapal N. Singh
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, 40536 USA
| | - Pamela E. Knapp
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, 40536 USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky Medical Center, Lexington, KY, 40536 USA
| | - Avindra Nath
- Department of Neurology, Johns Hopkins University, Baltimore, MD, 21287 USA
| | - Kurt F. Hauser
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, 40536 USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky Medical Center, Lexington, KY, 40536 USA
- Address correspondence and reprint requests to Dr. Kurt F. Hauser, Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY 40536-0298 USA, ; Phone: (859) 323-6477; FAX: (859) 323-5946)
| |
Collapse
|
52
|
Atici S, Cinel L, Cinel I, Doruk N, Aktekin M, Akca A, Camdeviren H, Oral U. Opioid neurotoxicity: comparison of morphine and tramadol in an experimental rat model. Int J Neurosci 2005; 114:1001-11. [PMID: 15527204 DOI: 10.1080/00207450490461314] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Histopathologic changes in rat brain due to chronic use of morphine and/or tramadol in progressively increased doses were investigated in this study. Thirty male Wistar rats (180-220 g) were included and divided into three groups. Normal saline (1 ml/kg) was given intraperitoneally as placebo in the control group (n = 10). Morphine group (n = 10) received morphine intraperitoneally at a dose of 4 mg/kg/day for the first 10 days, 8 mg/kg/day between 11-20 days, and 12 mg/kg/day between 21-30 days. The tramadol group (n = 10) received the drug intraperitoneally at doses of 20, 40, and 80 mg/kg/day in the first, second, and the third 10 days of the study, respectively. All rats were decapitated on the 30th day and the brain was removed intact for histology. The presence and the number of red neurons, which are a histologic marker of apoptosis, were investigated in the parietal, frontal, temporal, occipital, entorhinal, pyriform, and hippocampal CA1, CA2, CA3 regions. Red neurons were found in morphine and tramadol groups but not in the control group. The total number of red neurons was not different in morphine and tramadol groups, but the numbers of red neurons were significantly higher in the temporal and occipital regions in tramadol group as compared with the morphine group (p < .05). In conclusion, chronic use of morphine and/or tramadol in increasing doses is found to cause red neuron degeneration in the rat brain, which probably contributes to cerebral dysfunction. These findings should be taken into consideration when chrome use of opioids is indicated.
Collapse
Affiliation(s)
- Sebnem Atici
- Department of Anesthesiology & Reanimation, Mersin University School of Medicine, Mersin, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|
53
|
Yuen JWF, So IYW, Kam AYF, Wong YH. Regulation of STAT3 by mu-opioid receptors in human neuroblastoma SH-SY5Y cells. Neuroreport 2004; 15:1431-5. [PMID: 15194868 DOI: 10.1097/01.wnr.0000130433.90962.6e] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Heptahelical opioid receptors are implicated in the transcriptional regulation of neuronal development. Here we demonstrated that activation of mu-opioid receptors in human neuroblastoma SH-SY5Y cells led to the activation of signal transducer and activator of transcription 3 (STAT3), a transcription factor central to the regulation of numerous biological processes. The mu-opioid-induced activation of STAT3 is sensitive to receptor was further shown to pertussis toxin treatment and required JAK and Src tyrosine kinases, but not phosphatidylinositol 3-kinase. This mu-opioid-induced response was mediated via the extracellular signal-regulated protein kinase in a Raf-1-independent manner. The present study provides a foundation to explore the importance of STAT3 signaling in the regulation of neuronal growth and differentiation by the mu-opioid receptor.
Collapse
Affiliation(s)
- Jessie W F Yuen
- Department of Biochemistry, the Molecular Neuroscience Center, and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | | | | | | |
Collapse
|
54
|
Khurdayan VK, Buch S, El-Hage N, Lutz SE, Goebel SM, Singh IN, Knapp PE, Turchan-Cholewo J, Nath A, Hauser KF. Preferential vulnerability of astroglia and glial precursors to combined opioid and HIV-1 Tat exposure in vitro. Eur J Neurosci 2004; 19:3171-82. [PMID: 15217373 PMCID: PMC4305445 DOI: 10.1111/j.0953-816x.2004.03461.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Human immunodeficiency virus (HIV)-1 infection can cause characteristic neural defects such as progressive motor dysfunction, striatal pathology and gliosis. Recent evidence suggests that HIV-induced pathogenesis is exacerbated by heroin abuse and that the synergistic neurotoxicity is a direct effect of heroin on the CNS, an alarming observation considering the high incidence of HIV infection with injection drug abuse. Although HIV infection results in neurodegeneration, neurons themselves are not directly infected. Instead, HIV affects microglia and astroglia, which subsequently contributes to the neurodegenerative changes. Opioid receptors are widely expressed by macroglia and macroglial precursors, and the activation of mu-opioid receptors can modulate programmed cell death, as well as the response of neural cells to cytotoxic insults. For this reason, we questioned whether opioid drugs might modify the vulnerability of macroglia and macroglial precursors to HIV-1 Tat protein. To address this problem, the effects of morphine and/or HIV Tat(1-72) on the viability of macroglia and macroglial precursors were assessed in mixed-glial cultures derived from mouse striatum. Our findings indicate that sustained exposure to morphine and Tat(1-72) viral protein induces the preferential death of glial precursors and some astrocytes. Moreover, the increased cell death is mediated by mu-opioid receptors and accompanied by the activation of caspase-3. Our results imply that opiates can enhance the cytotoxicity of HIV-1 Tat through direct actions on glial precursors and/or astroglia, suggesting novel cellular targets for HIV-opiate interactions.
Collapse
Affiliation(s)
- Valeriya K. Khurdayan
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Shreya Buch
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Nazira El-Hage
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Sarah E. Lutz
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Susan M. Goebel
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Indrapal N. Singh
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Pamela E. Knapp
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | | | - Avindra Nath
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Kurt F. Hauser
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
- Correspondence: Kurt F. Hauser, Ph.D., Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY 40536-0298 USA, , Phone: (859) 323-6477, FAX: (859) 323-5946
| |
Collapse
|
55
|
Knapp PE, Adams MH. Epidermal growth factor promotes oligodendrocyte process formation and regrowth after injury. Exp Cell Res 2004; 296:135-44. [PMID: 15149844 DOI: 10.1016/j.yexcr.2004.02.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2003] [Revised: 02/04/2004] [Indexed: 01/06/2023]
Abstract
Oligodendrocytes (OLs) form myelin within the central nervous system and are targets in numerous demyelinating diseases and injuries. OLs grown in culture maintain the developmental timetable which occurs in vivo and mature into cells with a relatively normal phenotype. In this study, cultured cells are used to test whether EGF can modulate process formation in OLs both before and after transection injury. EGF had no effect on the formation of new processes by OLs at any stage of development. To test the effect of EGF on process outgrowth after injury, mature OLs were selected and injured by laser transection of a single process, then imaged at 24-h intervals for 120 h. EGF promoted the recovery and regrowth of injured processes and also significantly increased outgrowth in uninjured processes. As well, it increased the number of new sprouts formed by OLs after injury. Results suggest that the effects of EGF on process outgrowth are a consequence of EGF interaction with a signaling pathway that is specifically activated within injured OLs. The potent effect of EGF on OL process formation after an injury suggests that modulation of the signaling pathways involved might provide a mechanism to promote remyelination.
Collapse
Affiliation(s)
- Pamela E Knapp
- Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington, KY 40536-0298, USA.
| | | |
Collapse
|
56
|
Mandyam CD, Norris RD, Eisch AJ. Chronic morphine induces premature mitosis of proliferating cells in the adult mouse subgranular zone. J Neurosci Res 2004; 76:783-94. [PMID: 15160390 DOI: 10.1002/jnr.20090] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The birth of cells with neurogenic potential in the adult brain is assessed commonly by detection of exogenous S phase markers, such as bromodeoxyuridine (BrdU). Analysis of other phases of the cell cycle, however, can provide insight into how external factors, such as opiates, influence the cycling of newly born cells. To this end, we examined the expression of two endogenous cell cycle markers in relation to BrdU: proliferating cell nuclear antigen (PCNA) and phosphorylated histone H3 (pHisH3). Two hours after one intraperitoneal BrdU injection, BrdU-, PCNA-, and pHisH3-immunoreactive (IR) cells exhibited similar distribution in the adult mouse subgranular zone (SGZ). Quantitative analysis within the SGZ revealed a relative abundance of cells labeled for PCNA > BrdU >> pHisH3. Similar to our reports in rat SGZ, chronic morphine treatment decreased BrdU- and PCNA-IR cells in mouse SGZ by 28 and 38%, respectively. We also show that pHisH3-IR cells are influenced by chronic morphine to a greater extent (58% decrease) than are BrdU- or PCNA-IR cells. Cell cycle phase analysis of SGZ BrdU-IR cells using triple labeling for BrdU, PCNA, and pHisH3 revealed premature mitosis in chronic morphine-treated mice. These results suggest that morphine-treated mice have a shorter Gap2/mitosis (G(2)/M) phase when compared to sham-treated mice. These findings demonstrate the power of using a combination of exogenous and endogenous cell cycle markers and nuclear morphology to track proliferating cells through different phases of the cell cycle and to reveal the regulation of cell cycle phase by chronic morphine.
Collapse
Affiliation(s)
- Chitra D Mandyam
- Department of Psychiatry, The University of Texas Southwestern Medical Center at Dallas, Texas 75390, USA
| | | | | |
Collapse
|
57
|
Hauser KF, Khurdayan VK, Goody RJ, Nath A, Saria A, Pauly JR. Selective vulnerability of cerebellar granule neuroblasts and their progeny to drugs with abuse liability. THE CEREBELLUM 2003; 2:184-95. [PMID: 14509568 PMCID: PMC4306667 DOI: 10.1080/14734220310016132] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Cerebellar development is shaped by the interplay of genetic and numerous environmental factors. Recent evidence suggests that cerebellar maturation is acutely sensitive to substances with abuse liability including alcohol, opioids, and nicotine. Assuming substance abuse disrupts cerebellar maturation, a central question is: what are the basic mechanisms underlying potential drug-induced developmental defects? Evidence reviewed herein suggests that the maturation of granule neurons and their progeny are intrinsically affected by several classes of substances with abuse liability. Although drug abuse is also likely to target directly other cerebellar neuron and glial types, such as Purkinje cells and Bergmann glia, findings in isolated granule neurons suggest that they are often the principle target for drug actions. Developmental events that are selectively disrupted by drug abuse in granule neurons and/or their neuroblast precursors include proliferation, migration, differentiation (including neurite elaboration and synapse formation), and programmed cell death. Moreover, different classes of drugs act through distinct molecular mechanisms thereby disrupting unique aspects of development. For example, drug-induced perturbations in: (i) neurotransmitter biogenesis; (ii) ligand and ion-gated receptor function and their coupling to intracellular effectors; (iii) neurotrophic factor biogenesis and signaling; and (iv) intercellular adhesion are all likely to have significant effects in shaping developmental outcome. In addition to identifying therapeutic strategies for drug abuse intervention, understanding the mechanisms by which drugs affect cellular maturation is likely to provide a better understanding of the neurochemical events that normally shape central nervous system development.
Collapse
Affiliation(s)
- Kurt F Hauser
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, Kentucky, 40536-0298, USA.
| | | | | | | | | | | |
Collapse
|
58
|
Persson AI, Thorlin T, Bull C, Eriksson PS. Opioid-induced proliferation through the MAPK pathway in cultures of adult hippocampal progenitors. Mol Cell Neurosci 2003; 23:360-72. [PMID: 12837621 DOI: 10.1016/s1044-7431(03)00061-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Administration of opioid agonists or antagonists has been reported to regulate proliferation or survival of neural progenitors in vivo. Here we report that beta-endorphin and selective mu-opioid receptor (MOR) and delta-opioid receptor (DOR) agonists stimulate proliferation of isolated rat adult hippocampal progenitors (AHPs). The AHPs were found to express DORs and MORs, but not kappa-opioid receptors. Incubation with beta-endorphin for 48 h increased the number of AHPs found in mitosis, the total DNA content, and the expression of proliferating cell nuclear antigen. This proliferative effect from beta-endorphin on AHPs was antagonized by naloxone. The beta-endorphin-induced proliferation was mediated through phosphorylation of extracellular signal-regulated kinases 1 and 2 and dependent on phosphatidylinositol 3-kinase and both intra- and extracellular calcium. These data suggest a role for the opioid system in the regulation of proliferation in progenitors from the adult hippocampus.
Collapse
Affiliation(s)
- Anders I Persson
- The Arvid Carlsson Institute for Neuroscience at Institute of Clinical Neuroscience, Sahlgrenska University Hospital, Göteborg University, Göteborg, Sweden
| | | | | | | |
Collapse
|
59
|
Pan ZZ. Kappa-opioid receptor-mediated enhancement of the hyperpolarization-activated current (I(h)) through mobilization of intracellular calcium in rat nucleus raphe magnus. J Physiol 2003; 548:765-75. [PMID: 12651920 PMCID: PMC2342905 DOI: 10.1113/jphysiol.2002.037622] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The hyperpolarization-activated current (Ih) is important in the control of resting membrane potential, in the regulation of network firing pattern and in the modulation of presynaptic transmitter release in central neurons. Recent studies on native and cloned Ih channels have demonstrated that the Ih channel is commonly modulated by cAMP through a positive shift in its voltage dependence without a change in its maximum current. The present study demonstrates that activation of kappa-opioid receptors enhances Ih by increasing its maximum current in brainstem neurons in the nucleus raphe magnus. Agents that interfere with the release of intracellular calcium from calcium stores altered the maximum Ih and significantly attenuated the kappa-receptor-mediated enhancement of Ih. These results suggest that kappa-opioid receptors enhance the maximum Ih by mobilizing intracellular calcium from calcium stores. This provides a physiological function for kappa-receptor-stimulated calcium release and may suggest another Ih-regulating mechanism by intracellular calcium in central neurons.
Collapse
Affiliation(s)
- Zhizhong Z Pan
- Department of Symptom Research, University of Texas-MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
60
|
Kam AYF, Chan ASL, Wong YH. Rac and Cdc42-dependent regulation of c-Jun N-terminal kinases by the delta-opioid receptor. J Neurochem 2003; 84:503-13. [PMID: 12558970 DOI: 10.1046/j.1471-4159.2003.01535.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Heptahelical opioid receptors utilize Gi proteins to regulate a multitude of effectors including the classical adenylyl cyclases and the more recently discovered mitogen-activated protein kinases (MAPKs). The c-Jun NH2-terminal kinases (JNKs) belong to one of three subgroups of MAPKs. In NG108-15 neuroblastoma x glioma hybrid cells that endogenously express delta-opioid receptors, delta-agonist dose-dependently stimulated JNK activity in a pertussis toxin-sensitive manner. By using COS-7 cells transiently transfected with the cDNAs of delta-opioid receptor and hemagglutinin (HA)-tagged JNK, we delineated the signaling components involved in this pathway. Sequestration of Gbetagamma subunits by transducin suppressed the opioid-induced JNK activity. The possible involvement of the small GTPases was also examined. Expression of dominant negative mutants of Rac and Cdc42 blocked the opioid-induced JNK activation, and a partial inhibition was observed in the presence of the dominant negative mutant of Ras. In contrast, the dominant negative mutant of Rho did not affect the opioid-induced JNK activation. In addition, the receptor-mediated JNK activation was dependent on Src family tyrosine kinases, but independent of phosphatidylinositol-3 kinase and EGF receptor tyrosine kinases. Collectively, these results demonstrate functional regulation of JNK by the delta-opioid receptor, and this pathway requires Gbetagamma, Src kinases and the small GTPases Rac and Cdc42.
Collapse
MESH Headings
- Animals
- COS Cells
- Cell Line
- Enkephalin, D-Penicillamine (2,5)-/pharmacology
- Enzyme Activation/drug effects
- GTP-Binding Proteins/metabolism
- Genes, Dominant
- Hybrid Cells/metabolism
- JNK Mitogen-Activated Protein Kinases
- Mice
- Mitogen-Activated Protein Kinases/genetics
- Mitogen-Activated Protein Kinases/metabolism
- Mutagenesis, Site-Directed
- Narcotics/pharmacology
- Protein Subunits/metabolism
- Rats
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Signal Transduction/genetics
- Signal Transduction/physiology
- Transfection
- cdc42 GTP-Binding Protein/genetics
- cdc42 GTP-Binding Protein/metabolism
- rac GTP-Binding Proteins/genetics
- rac GTP-Binding Proteins/metabolism
- ras Proteins/genetics
- ras Proteins/metabolism
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Angel Y F Kam
- Department of Biochemistry, the Molecular Neuroscience Center, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | | | | |
Collapse
|
61
|
Affiliation(s)
- Amelia J Eisch
- Department of Psychiatry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390-9070, USA.
| |
Collapse
|
62
|
Quillan JM, Carlson KW, Song C, Wang D, Sadée W. Differential effects of mu-opioid receptor ligands on Ca(2+) signaling. J Pharmacol Exp Ther 2002; 302:1002-12. [PMID: 12183657 DOI: 10.1124/jpet.302.3.1002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Activation of mu-opioid receptors (MORs) transfected into human embryonic kidney 293 cells, caused a multiphasic increase in cytosolic free Ca(2+) levels (Ca(2+)i). The first Ca(2+)i maximum (peak 1) between 5 and 7 s depended on the presence of extracellular Ca(2+) (Ca(2+)e). The second phase peaking at approximately 15 s (peak 2) was independent of Ca(2+)e and thus represents Ca(2+) release from intracellular stores. A decrease in temperature from 37 to 25 degrees C also caused reduction of peak 1 but not peak 2, suggesting that the two responses arise from mechanistically distinct pathways. A delayed Ca(2+)e-dependent third response phase is thought to represent capacitative Ca(2+)e influx evoked after release of Ca(2+) from internal stores. Agonists and antagonists of two major classes of opioid ligands, oxymorphinans (morphine and naloxone) and oripavines (etorphine and diprenorphine), had differential effects on Ca(2+) currents. Although morphine activated both phases with equal potency, etorphine was 20-fold less potent at stimulating peak 1 over peak 2. Similarly, the antagonists, naloxone and diprenorphine, blocked the Ca(2+) response to each agonist with greatly varying potencies. Specifically, concomitant injection of diprenorphine failed to affect peak 1 (thought to represent rapid Ca(2+)e influx) stimulated by morphine while fully blocking peak 2 (intracellular Ca(2+) release). However, diprenorphine potently inhibited peak 1 as well when added to the cells before morphine, indicating limited or slow access of diprenorphine to these morphine binding sites. The existence of multiple, functionally distinct binding site conformations could account for these findings. In conclusion, different opioid ligands can differentially affect Ca(2+) response patterns resulting from MOR activation.
Collapse
Affiliation(s)
- J Mark Quillan
- Department of Biopharmaceutical Sciences and Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA
| | | | | | | | | |
Collapse
|
63
|
Lohr C, Tucker E, Oland LA, Tolbert LP. Development of depolarization-induced calcium transients in insect glial cells is dependent on the presence of afferent axons. JOURNAL OF NEUROBIOLOGY 2002; 52:85-98. [PMID: 12124748 DOI: 10.1002/neu.10075] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Changes in the intracellular Ca(2+) concentration ([Ca(2+)](i)) induced by depolarization have been measured in glial cells acutely isolated from antennal lobes of the moth Manduca sexta at different postembryonic developmental stages. Depolarization of the glial cell membrane was elicited by increasing the external K(+) concentration from 4 to 25 mM. At midstage 5 and earlier stages, less than 20% of the cells responded to 25 mM K(+) (1 min) with a transient increase in [Ca(2+)](i) of approximately 40 nM. One day later, at late stage 5, 68% of the cells responded to 25 mM K(+), the amplitude of the [Ca(2+)](i) transients averaging 592 nM. At later stages, all cells responded to 25 mM K(+) with [Ca(2+)](i) transients with amplitudes not significantly different from those at late stage 5. In stage 6 glial cells isolated from deafferented antennal lobes, i.e., from antennal lobes chronically deprived of olfactory receptor axons, only 30% of the cells responded with [Ca(2+)](i) transients. The amplitudes of these [Ca(2+)](i) transients averaged 93 nM and were significantly smaller than those in normal stage 6 glial cells. [Ca(2+)](i) transients were greatly reduced in Ca(2+)-free, EGTA-buffered saline, and in the presence of the Ca(2+) channel blockers cadmium and verapamil. The results suggest that depolarization of the cell membrane induces Ca(2+) influx through voltage-activated Ca(2+) channels into antennal lobe glial cells. The development of the depolarization-induced Ca(2+) transients is rapid between midstage 5 and stage 6, and depends on the presence of afferent axons from the olfactory receptor cells in the antenna.
Collapse
Affiliation(s)
- Christian Lohr
- ARL Division of Neurobiology, University of Arizona, P.O. Box 210077, Tucson, Arizona 85721-0077, USA.
| | | | | | | |
Collapse
|
64
|
|
65
|
Seri B, Alvarez-Buylla A. Neural stem cells and the regulation of neurogenesis in the adult hippocampus. ACTA ACUST UNITED AC 2002; 2:11-16. [PMID: 25419197 DOI: 10.1016/s1566-2772(02)00004-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Neurogenesis continues in the hippocampal dentate gyrus of adult rodents and primates including humans. Neurons are born in the underlying subgranular layer (SGL) and move into the granule cell layer (GCL) to become mature granule neurons. Recent work indicates that the primary precursors for these new neurons correspond to radial astrocytes whose cell body is in the SGL and their processes traverse the GCL. These astrocytes divide to give rise to intermediate precursors, D cells that likely become mature granule neurons. Here we propose that the anatomy of radial astrocytes may allow for signals within the GCL to regulate neurogenesis in the SGL. Levels of neuronal activity within the granule cell layer may regulate the proliferation rates of radial astrocytes and determine the number of new neurons produced in the dentate gyrus.
Collapse
Affiliation(s)
- Bettina Seri
- Department of Neurosurgery Research, Box 0520, Brain Tumor Research Center, University of California, San Francisco, CA 94143, USA
| | - Arturo Alvarez-Buylla
- Department of Neurosurgery Research, Box 0520, Brain Tumor Research Center, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
66
|
Stiene-Martin A, Knapp PE, Martin K, Gurwell JA, Ryan S, Thornton SR, Smith FL, Hauser KF. Opioid system diversity in developing neurons, astroglia, and oligodendroglia in the subventricular zone and striatum: impact on gliogenesis in vivo. Glia 2001; 36:78-88. [PMID: 11571786 PMCID: PMC4303466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Accumulating evidence, obtained largely in vitro, indicates that opioids regulate the genesis of neurons and glia and their precursors in the nervous system. Despite this evidence, few studies have assessed opioid receptor expression in identified cells within germinal zones or examined opioid effects on gliogenesis in vivo. To address this question, the role of opioids was explored in the subventricular zone (SVZ) and/or striatum of 2-5-day-old and/or adult ICR mice. The results showed that subpopulations of neurons, astrocytes, and oligodendrocytes in the SVZ and striatum differentially express mu-, delta-, and/or kappa-receptor immunoreactivity in a cell type-specific and developmentally regulated manner. In addition, DNA synthesis was assessed by examining 5-bromo-2'-deoxyuridine (BrdU) incorporation into glial and nonglial precursors. Morphine (a preferential mu-agonist) significantly decreased the number of BrdU-labeled GFAP(+) cells compared with controls or mice co-treated with naltrexone plus morphine. Alternatively, in S100beta(+) cells, morphine did not significantly decrease BrdU incorporation; however, significant differences were noted between mice treated with morphine and those treated with morphine plus naltrexone. Most cells were GFAP(-)/S100beta(-). When BrdU incorporation was assessed within the total population (glia and nonglia), morphine had no net effect, but naltrexone alone markedly increased BrdU incorporation. This finding suggests that DNA synthesis in GFAP(-)/S100beta(-) cells is tonically suppressed by endogenous opioids. Assuming that S100beta and GFAP, respectively, distinguish among younger and older astroglia, this implies that astroglial replication becomes increasingly sensitive to morphine during maturation, and suggests that opioids differentially regulate the development of distinct subpopulations of glia and glial precursors.
Collapse
MESH Headings
- Aging/physiology
- Amino Acid Transport System X-AG/metabolism
- Animals
- Animals, Newborn/anatomy & histology
- Animals, Newborn/growth & development
- Animals, Newborn/metabolism
- Antigens, Differentiation/metabolism
- Antigens, Surface/metabolism
- Astrocytes/cytology
- Astrocytes/drug effects
- Astrocytes/metabolism
- Bromodeoxyuridine/pharmacokinetics
- Calcium-Binding Proteins/metabolism
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Cell Division/drug effects
- Cell Division/physiology
- Glial Fibrillary Acidic Protein/metabolism
- Immunohistochemistry
- Lateral Ventricles/cytology
- Lateral Ventricles/growth & development
- Lateral Ventricles/metabolism
- Mice
- Mice, Inbred ICR/anatomy & histology
- Mice, Inbred ICR/growth & development
- Mice, Inbred ICR/metabolism
- Morphine/pharmacology
- Naltrexone/pharmacology
- Neostriatum/cytology
- Neostriatum/growth & development
- Neostriatum/metabolism
- Nerve Growth Factors/metabolism
- Neurons/cytology
- Neurons/drug effects
- Neurons/metabolism
- Oligodendroglia/cytology
- Oligodendroglia/drug effects
- Oligodendroglia/metabolism
- Opioid Peptides/metabolism
- Receptors, Opioid/drug effects
- Receptors, Opioid/metabolism
- S100 Calcium Binding Protein beta Subunit
- S100 Proteins
Collapse
Affiliation(s)
- Anne Stiene-Martin
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine Lexington, Kentucky
| | - Pamela E. Knapp
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine Lexington, Kentucky
| | - Kenneth Martin
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine Lexington, Kentucky
| | - Julie A. Gurwell
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine Lexington, Kentucky
| | - Shawn Ryan
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine Lexington, Kentucky
| | - Suzanne R. Thornton
- Department of Pharmacology and Toxicology, Medical College of Virginia, Richmond, Virginia
| | - Forrest L. Smith
- Department of Pharmacology and Toxicology, Medical College of Virginia, Richmond, Virginia
| | - Kurt F. Hauser
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine Lexington, Kentucky
- Correspondence to: Kurt F. Hauser, Ph.D. Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY 40536-0298 USA, Phone: (606) 323-6477, FAX: (606) 323-5946,
| |
Collapse
|
67
|
Kim MS, Cheong YP, So HS, Lee KM, Kim TY, Oh J, Chung YT, Son Y, Kim BR, Park R. Protective effects of morphine in peroxynitrite-induced apoptosis of primary rat neonatal astrocytes: potential involvement of G protein and phosphatidylinositol 3-kinase (PI3 kinase). Biochem Pharmacol 2001; 61:779-86. [PMID: 11274962 DOI: 10.1016/s0006-2952(01)00541-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Opiates, such as morphine, have been used extensively in the clinical management of pain due to their potent analgesic effect. Astrocytes, representing a major non-neuronal cell population in the CNS, contain opioid receptors that are actively involved in several brain functions. This study was designed to evaluate the effects by which morphine, a preferential mu-opioid receptor agonist, contributes to cytotoxicity of nitric oxide (NO) species, including NO and peroxynitrite (ONOO-), in primary rat neonatal astrocytes. Primary astrocytes isolated from the cerebral cortex of 1- to 2-day-old Sprague-Dawley rats were treated with morphine, naloxone, and 3-morpholinosydnonimine (SIN-1), a donor of peroxynitrite. Morphine significantly protected primary rat astrocytes from apoptosis mediated by sodium nitroprusside, an NO donor, and SIN-1 in a dose-dependent manner, whereas it did not in other types of cells including C6 glioma, RAW 264.7, and HL-60 cells. Moreover, naloxone antagonized the protective effects of morphine on SIN-1-induced apoptosis. Morphine also inhibited the nuclear condensation and fragmentation of SIN-1-treated cells that was antagonized by naloxone pretreatment. The protective role of morphine in SIN-1-induced apoptosis was dependent on an intracellular antioxidant system such as GSH. Furthermore, the effects of morphine on SIN-1-induced cytotoxicity were prohibited by pretreatment with the G(i) protein inhibitor, pertussis toxin, and the phosphatidylinositol 3-kinase (PI3 kinase) inhibitors, wortmannin and LY294002. Taken together, these results suggest that morphine may protect primary rat astrocytes from apoptosis by NO species via the signaling cascades that involve both G protein and PI3 kinase.
Collapse
Affiliation(s)
- M S Kim
- Department of Microbiology, Wonkwang University School of Medicine, 344-2 Shinyong-dong, 570-749, Iksan Chonbuk, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Glasel JA. The effects of morphine on cell proliferation. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2001; 55:33-80. [PMID: 11127966 DOI: 10.1007/978-3-0348-8385-6_2] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
There is increasing evidence that endogenous opioid peptides ("enkephalins") and other neurotransmitters have widespread, receptor-mediated roles as growth regulators in non-neuronal cells and tissues. For example, it is now believed that enkephalins produced in placental trophoblast giant cells have multiple roles in supporting embryo growth, and in maternal adaptation to pregnancy. Since plant and synthetic narcotics (e.g., morphine) bind to the same receptors, the questions immediately arise: Do narcotics also have actions as growth regulators? If so, do these actions have physiological significance in addicts? Recent work on the first of these questions is covered in this review. While the greatest volume of research has been focused on the proliferative effects of narcotics for cells of the immune system, the roles of opioid peptides and narcotics on the growth of a variety of other cells has come under study recently.
Collapse
Affiliation(s)
- J A Glasel
- Global Scientific Consulting LLC, 15 Colton St., Farmington, CT 06032, USA
| |
Collapse
|
69
|
Wu VW, Mo Q, Yabe T, Schwartz JP, Robinson SE. Perinatal opioids reduce striatal nerve growth factor content in rat striatum. Eur J Pharmacol 2001; 414:211-4. [PMID: 11239921 DOI: 10.1016/s0014-2999(01)00807-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Both human and animal models indicate that perinatal methadone exposure produces a variety of short- and long-term neurobehavioral consequences, including disruption of normal development of striatal cholinergic neurons. Despite this, methadone maintenance is a standard method of managing pregnant heroin addicts, and the opioid receptor partial agonist buprenorphine is under evaluation for the same use. We now report that perinatal administration of either methadone or buprenorphine reduces the content of the neurotrophic factor nerve growth factor (NGF) in rat striatum, which may explain the behavioral deficits observed. Furthermore, although NGF content is reduced, there are no corresponding reductions in striatal NGF mRNA.
Collapse
Affiliation(s)
- V W Wu
- Neurotrophic Factors Section, NINDS, NIH, Bethesda, MD 20892-4126, USA
| | | | | | | | | |
Collapse
|
70
|
Stiene-Martin A, Knapp PE, Martin K, Gurwell JA, Ryan S, Thornton SR, Smith FL, Hauser KF. Opioid system diversity in developing neurons, astroglia, and oligodendroglia in the subventricular zone and striatum: Impact on gliogenesis in vivo. Glia 2001. [DOI: 10.1002/glia.1097] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
71
|
Ikeda M, Nelson CS, Shinagawa H, Shinoe T, Sugiyama T, Allen CN, Grandy DK, Yoshioka T. Cyclic AMP regulates the calcium transients released from IP(3)-sensitive stores by activation of rat kappa-opioid receptors expressed in CHO cells. Cell Calcium 2001; 29:39-48. [PMID: 11133354 DOI: 10.1054/ceca.2000.0161] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We analyzed intracellular Ca(2+)and cAMP levels in Chinese hamster ovary cells expressing a cloned rat kappa opioid receptor (CHO-kappa cells). Although expression of kappa(kappa)-opioid receptors was confirmed with a fluorescent dynorphin analog in almost all CHO-kappa cells, the kappa-specific agonists, U50488H or U69593, induced a Ca(2+) transient only in 35% of the cells. The Ca(2+) response occurred in all-or-none fashion and the half-maximal dosage of U50488H (812.1nM) was higher than that (3.2nM) to inhibit forskolin-stimulated cAMP. The kappa-receptors coupled to G(i/o)proteins since pertussis toxin significantly reduced the U50488H actions on intracellular Ca(2+) and cAMP. The Ca(2+) transient originates from IP(3)-sensitive internal stores since the Ca(2+) response was blocked by a PLC inhibitor (U73122) or by thapsigargin depletion of internal stores while removal of extracellular Ca(2+) had no effect. Interestingly, application of dibutyryl cAMP (+ 56.2%) or 8-bromo-cAMP (+ 174.7%) significantly increased the occurrence of U50488H-induced Ca(2+) mobilization while protein kinase A (PKA) inhibitors, Rp-cAMP (-32.3%) or myr-psi PKA (-73.9%) significantly reduced the response. Therefore, it was concluded that cAMP and PKA activity can regulate the Ca(2+) mobilization. These results suggest that the kappa receptor-linked cAMP cascade regulates the occurrence of kappa-opioid-mediated Ca(2+) mobilization.
Collapse
MESH Headings
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology
- 8-Bromo Cyclic Adenosine Monophosphate/pharmacology
- Analgesics/pharmacology
- Analgesics, Non-Narcotic/pharmacology
- Analgesics, Opioid/pharmacology
- Animals
- Benzeneacetamides
- Bucladesine/pharmacology
- CHO Cells
- Calcium/metabolism
- Calcium Signaling/drug effects
- Calcium Signaling/physiology
- Cloning, Molecular
- Colforsin/pharmacology
- Cricetinae
- Cyclic AMP/analogs & derivatives
- Cyclic AMP/metabolism
- Cyclic AMP/pharmacology
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Enkephalin, D-Penicillamine (2,5)-/pharmacology
- Enzyme Inhibitors/pharmacology
- Gene Expression/physiology
- Inositol 1,4,5-Trisphosphate/metabolism
- Peptides
- Pyrrolidines/pharmacology
- Rats
- Receptors, Opioid, kappa/analysis
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, kappa/metabolism
- Thionucleotides/pharmacology
- Transfection
Collapse
Affiliation(s)
- M Ikeda
- Advanced Research Institute for Science and Engineering, Waseda University, Okubo, Shinjuku-ku, Tokyo, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
72
|
Gurwell JA, Nath A, Sun Q, Zhang J, Martin KM, Chen Y, Hauser KF. Synergistic neurotoxicity of opioids and human immunodeficiency virus-1 Tat protein in striatal neurons in vitro. Neuroscience 2001; 102:555-63. [PMID: 11226693 PMCID: PMC4300203 DOI: 10.1016/s0306-4522(00)00461-9] [Citation(s) in RCA: 134] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Human immunodeficiency virus (HIV) infection selectively targets the striatum, a region rich in opioid receptor-expressing neural cells, resulting in gliosis and neuronal losses. Opioids can be neuroprotective or can promote neurodegeneration. To determine whether opioids modify the response of neurons to human immunodeficiency virus type 1 (HIV-1) Tat protein-induced neurotoxicity, neural cell cultures from mouse striatum were initially characterized for mu and/or kappa opioid receptor immunoreactivity. These cultures were continuously treated with morphine, the opioid antagonist naloxone, and/or HIV-1 Tat (1-72) protein, a non-neurotoxic HIV-1 Tat deletion mutant (TatDelta31-61) protein, or immunoneutralized HIV-1 Tat (1-72) protein. Neuronal and astrocyte viability was examined by ethidium monoazide exclusion, and by apoptotic changes in nuclear heterochromatin using Hoechst 33342. Morphine (10nM, 100nM or 1microM) significantly increased Tat-induced (100 or 200nM) neuronal losses by about two-fold at 24h following exposure. The synergistic effects of morphine and Tat were prevented by naloxone (3microM), indicating the involvement of opioid receptors. Furthermore, morphine was not toxic when combined with mutant Tat or immunoneutralized Tat. Neuronal losses were accompanied by chromatin condensation and pyknosis. Astrocyte viability was unaffected. These findings demonstrate that acute opioid exposure can exacerbate the neurodegenerative effect of HIV-1 Tat protein in striatal neurons, and infer a means by which opioids may hasten the progression of HIV-associated dementia.
Collapse
Affiliation(s)
- Julie A. Gurwell
- Department of Anatomy & Neurobiology University of Kentucky College of Medicine Lexington, Kentucky 40536-0298
| | - Avindra Nath
- Department of Neurology University of Kentucky College of Medicine Lexington, Kentucky 40536-0284
- Department of Microbiology & Immunology University of Kentucky College of Medicine Lexington, Kentucky 40536-0298
| | - Qinmiao Sun
- Department of Microbiology & Immunology University of Kentucky College of Medicine Lexington, Kentucky 40536-0298
| | - Jiayou Zhang
- Department of Microbiology & Immunology University of Kentucky College of Medicine Lexington, Kentucky 40536-0298
| | - Kenneth M. Martin
- Department of Anatomy & Neurobiology University of Kentucky College of Medicine Lexington, Kentucky 40536-0298
| | - Yan Chen
- Department of Anatomy & Neurobiology University of Kentucky College of Medicine Lexington, Kentucky 40536-0298
| | - Kurt F. Hauser
- Department of Anatomy & Neurobiology University of Kentucky College of Medicine Lexington, Kentucky 40536-0298
- Markey Cancer Center University of Kentucky Medical Center Lexington, Kentucky 40536-0084
- To whom correspondence should be addressed: Kurt F. Hauser, Ph.D. Department of Anatomy & Neurobiology University of Kentucky College of Medicine 800 Rose Street, Lexington, KY 40536-0298 Phone: (859) 323-6477; FAX: (859) 323-5946
| |
Collapse
|
73
|
Brüggemann I, Schulz S, Wiborny D, Höllt V. Colocalization of the mu-opioid receptor and calcium/calmodulin-dependent kinase II in distinct pain-processing brain regions. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2000; 85:239-50. [PMID: 11146127 DOI: 10.1016/s0169-328x(00)00265-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The mu-opioid receptor (MOR1) mediates the main analgesic effects of morphine and several other opioids. However, the clinical benefit of these drugs is limited by the development of tolerance and dependence. In vitro the mu-opioid receptor undergoes a rapid homologous desensitization during prolonged agonist exposure. We have recently identified the serine residues, Ser(261) and Ser(266), within the third intracellular loop as two consensus calcium/calmodulin-dependent protein kinase II (CaMKII) sites required for agonist-induced phosphorylation and desensitization of the mu-opioid receptor in HEK 293 cells. Since the specific pattern of mu-opioid receptor regulation in vivo is thought to depend on the cell- and tissue-specific complement of protein kinases, we examined the spatial relation between MOR1 and CaMKII in rat brain using specific antibodies. We found that MOR1 and CaMKII alpha which is a major CaMKII isoform expressed in the central nervous system co-exist in distinct pain-processing brain regions including the superficial layers of the spinal cord dorsal horn and dorsal root ganglia. At high power magnification it was evident that virtually all MOR1-expressing nociceptive spinal cord neurons also co-contain CaMKII. In naive or saline-treated animals the mu-opioid receptor was almost exclusively confined to the plasma membrane, while CaMKII was localized to vesicle-like structures throughout the cytoplasm. After subcutaneous administration of the mu-opioid receptor agonist, etorphine, a large proportion of the mu-opioid receptor proteins redistributed from the plasma membrane into the cytosol where it was frequently co-localized with CaMKII. Together, we identify CaMKII as a potential protein kinase, which by virtue of its colocalization with MOR1 may be in a position to phosphorylate the mu-opioid receptor and may thus contribute to the development of tolerance to opioid analgesics.
Collapse
Affiliation(s)
- I Brüggemann
- Department of Pharmacology and Toxicology, Otto-von-Guericke University, Leipziger Str. 44, Magdeburg 39120, Germany
| | | | | | | |
Collapse
|
74
|
Abstract
In the adult cerebellum, corticotropin releasing factor (CRF), that is localized in climbing fibers, mossy fibers, and a fine varicose plexus along the Purkinje cell layer, modulates the responsiveness of Purkinje cells to excitatory amino acids. During development, CRF has been detected in the primitive cerebellar anlage as early as embryonic day (E)10, and is continuously expressed throughout embryonic and postnatal cerebellar ontogeny. To investigate a possible trophic role for CRF during cerebellar development, cerebellar culture studies using E18 mouse embryos were carried out. In our culture paradigm, that used serum-free defined medium to suppress cell proliferation, CRF induced proliferation of cells in a dose-dependent manner in a range of concentrations between 0.1-10 microM. The proliferating cells were identified as astrocytes based on their expression of vimentin and GFAP. BrdU incorporation studies supported the proposed mitogenic effect of CRF on developing astrocytes. The mitogenic effects of CRF seemed to be primarily on immature astrocytes determined by their differential expression of vimentin and GFAP. Astrocytes at more advanced stages of development, as determined by the extent of process outgrowth and GFAP expression, incorporated less BrdU compared to immature astrocytes. CRF receptors were localized in astrocytes, and the proliferation of astrocytes induced by CRF was inhibited by astressin, a competitive CRF receptor antagonist. In conclusion, CRF induces proliferation of astrocytes derived from the developing cerebellum, that suggests a gliotrophic role for CRF during cerebellar development.
Collapse
Affiliation(s)
- B K Ha
- Department of Neuroscience, Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | |
Collapse
|
75
|
Eisch AJ, Barrot M, Schad CA, Self DW, Nestler EJ. Opiates inhibit neurogenesis in the adult rat hippocampus. Proc Natl Acad Sci U S A 2000; 97:7579-84. [PMID: 10840056 PMCID: PMC16588 DOI: 10.1073/pnas.120552597] [Citation(s) in RCA: 458] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Recent work implicates regulation of neurogenesis as a form of plasticity in the adult rat hippocampus. Given the known effects of opiates such as morphine and heroin on hippocampal function, we examined opiate regulation of neurogenesis in this brain region. Chronic administration of morphine decreased neurogenesis by 42% in the adult rat hippocampal granule cell layer. A similar effect was seen in rats after chronic self-administration of heroin. Opiate regulation of neurogenesis was not mediated by changes in circulating levels of glucocorticoids, because similar effects were seen in rats that received adrenalectomy and corticosterone replacement. These findings suggest that opiate regulation of neurogenesis in the adult rat hippocampus may be one mechanism by which drug exposure influences hippocampal function.
Collapse
Affiliation(s)
- A J Eisch
- Laboratory of Molecular Psychiatry and Yale Center for Genes and Behavior, Yale University School of Medicine, 34 Park Street, New Haven, CT 06508, USA
| | | | | | | | | |
Collapse
|
76
|
Tryoen-Toth P, Gavériaux-Ruff C, Labourdette G. Down-regulation of mu-opioid receptor expression in rat oligodendrocytes during their development in vitro. J Neurosci Res 2000; 60:10-20. [PMID: 10723064 DOI: 10.1002/(sici)1097-4547(20000401)60:1<10::aid-jnr2>3.0.co;2-o] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In the central nervous system, opioid receptors are found in neurons and also in glial cells. To gain more information on their presence and possibly on their function, we investigated the expression of mu-opioid receptors (MOR) during oligodendroglial cell development in two culture systems. In these models, during the first days, the cells are O-2A bipotential progenitor cells (also called OPCs; oligodendrocyte precursor cells), and then they differentiate into oligodendrocytes, which mature. In the first system, oligodendroglial cells, derived from newborn rat brain hemispheres, are grown in primary culture in the presence of a confluent layer of astrocytes, and they differentiate slowly. In the second, cells are specifically detached from the mixed cultures of the first system and are grown thereafter alone in secondary culture, a condition allowing a rapid cell differentiation. Under both conditions OPCs and immature oligodendrocytes were found to express a high level of MOR mRNA, whereas mature oligodendrocytes did not express it at all. The decrease of MOR expression during oligodendrocyte maturation was progressive, suggesting that it was not a primary effect of differentiation but an indirect secondary effect. Our study also shows that basic fibroblast growth factor (bFGF), which has been claimed by some authors to induce a dedifferentiation of the mature oligodendrocytes, and retinoic acid (RA), which had not been tested before, were not able to restore MOR expression in mature oligodendrocytes. These results indicate that bFGF and RA neither reverse the maturation process nor dedifferentiate the cells. However, RA was found to inhibit almost completely the expression of the myelin basic protein. The main result of this study is that MOR is expressed in progenitors and in immature oligodendrocytes, but not in mature oligodendrocytes. This suggests that MOR could be involved in some developmental process of the cells of the oligodendroglial lineage.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Blotting, Northern
- Blotting, Southern
- Cells, Cultured
- Down-Regulation
- Fibroblast Growth Factor 2/pharmacology
- Gene Expression Regulation, Developmental
- Oligodendroglia/metabolism
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
- Tretinoin/pharmacology
Collapse
Affiliation(s)
- P Tryoen-Toth
- Laboratory of Neurobiology of Development and Regeneration, UPR 1352 CNRS, Centre of Neurochemistry, Strasbourg, France
| | | | | |
Collapse
|
77
|
Hauser KF, Houdi AA, Turbek CS, Elde RP, Maxson W. Opioids intrinsically inhibit the genesis of mouse cerebellar granule neuron precursors in vitro: differential impact of mu and delta receptor activation on proliferation and neurite elongation. Eur J Neurosci 2000; 12:1281-93. [PMID: 10762357 PMCID: PMC4306580 DOI: 10.1046/j.1460-9568.2000.01015.x] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Although opioids are known to affect neurogenesis in vivo, it is uncertain the extent to which opioids directly or indirectly affect the proliferation, differentiation or death of neuronal precursors. To address these questions, the intrinsic role of the opioid system in neurogenesis was systematically explored in cerebellar external granular layer (EGL) neuronal precursors isolated from postnatal mice and maintained in vitro. Isolated neuronal precursors expressed proenkephalin-derived peptides, as well as specific mu and delta, but negligible kappa, opioid receptors. The developmental effects of opioids were highly selective. Morphine-induced mu receptor activation inhibited DNA synthesis, while a preferential delta2-receptor agonist ([D-Ala2]-deltorphin II) or Met-enkephalin, but not the delta1 agonist [D-Pen2, D-Pen5]-enkephalin, inhibited differentiation within the same neuronal population. If similar patterns occur in the developing cerebellum, spatiotemporal differences in endogenous mu and delta opioid ligand-receptor interactions may coordinate distinct aspects of granule neuron maturation. The data additionally suggest that perinatal exposure to opiate drugs of abuse directly interfere with cerebellar maturation by disrupting normal opioid signalling and inhibiting the proliferation of granule neuron precursors.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Antimetabolites/metabolism
- Antimetabolites/pharmacology
- Apoptosis/drug effects
- Apoptosis/physiology
- Bromodeoxyuridine/metabolism
- Bromodeoxyuridine/pharmacology
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Cell Division/drug effects
- Cell Division/physiology
- Cell Survival/drug effects
- Cell Survival/physiology
- Cells, Cultured
- Cerebellum/chemistry
- Cerebellum/cytology
- DNA/biosynthesis
- Enkephalin, D-Penicillamine (2,5)-/pharmacology
- Enkephalin, Methionine/pharmacology
- Enkephalins/analysis
- In Vitro Techniques
- Mice
- Microscopy, Electron
- Morphine/pharmacology
- Naloxone/pharmacology
- Narcotic Antagonists/pharmacology
- Neurites/physiology
- Neurites/ultrastructure
- Neurons/chemistry
- Neurons/metabolism
- Neurons/ultrastructure
- Oligopeptides/pharmacology
- Opioid-Related Disorders/metabolism
- Protein Precursors/analysis
- Receptors, Opioid, delta/analysis
- Receptors, Opioid, delta/immunology
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/analysis
- Receptors, Opioid, mu/immunology
- Receptors, Opioid, mu/metabolism
- Stem Cells/chemistry
- Stem Cells/metabolism
- Stem Cells/ultrastructure
Collapse
Affiliation(s)
- K F Hauser
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY 40536-0298, USA.
| | | | | | | | | |
Collapse
|
78
|
Bohn LM, Belcheva MM, Coscia CJ. Mitogenic signaling via endogenous kappa-opioid receptors in C6 glioma cells: evidence for the involvement of protein kinase C and the mitogen-activated protein kinase signaling cascade. J Neurochem 2000; 74:564-73. [PMID: 10646507 PMCID: PMC2504523 DOI: 10.1046/j.1471-4159.2000.740564.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
As reports on G protein-coupled receptor signal transduction mechanisms continue to emphasize potential differences in signaling due to relative receptor levels and cell type specificities, the need to study endogenously expressed receptors in appropriate model systems becomes increasingly important. Here we examine signal transduction mechanisms mediated by endogenous kappa-opioid receptors in C6 glioma cells, an astrocytic model system. We find that the kappa-opioid receptor-selective agonist U69,593 stimulates phospholipase C activity, extracellular signal-regulated kinase 1/2 phosphorylation, PYK2 phosphorylation, and DNA synthesis. U69,593-stimulated extracellular signal-regulated kinase 1/2 phosphorylation is shown to be upstream of DNA synthesis as inhibition of signaling components such as pertussis toxin-sensitive G proteins, L-type Ca2+ channels, phospholipase C, intracellular Ca2+ release, protein kinase C, and mitogen-activated protein or extracellular signal-regulated kinase kinase blocks both of these downstream events. In addition, by overexpressing dominant-negative or sequestering mutants, we provide evidence that extracellular signal-regulated kinase 1/2 phosphorylation is Ras-dependent and transduced by Gbetagamma subunits. In summary, we have delineated major features of the mechanism of the mitogenic action of an agonist of the endogenous kappa-opioid receptor in C6 glioma cells.
Collapse
Affiliation(s)
- L M Bohn
- E.A. Doisy Department of Biochemistry and Molecular Biology, St. Louis University School of Medicine, Missouri 63104, USA
| | | | | |
Collapse
|
79
|
Bohn LM, Belcheva MM, Coscia CJ. Mu-opioid agonist inhibition of kappa-opioid receptor-stimulated extracellular signal-regulated kinase phosphorylation is dynamin-dependent in C6 glioma cells. J Neurochem 2000; 74:574-81. [PMID: 10646508 PMCID: PMC2571950 DOI: 10.1046/j.1471-4159.2000.740574.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In previous studies we found that mu-opioids, acting via mu-opioid receptors, inhibit endothelin-stimulated C6 glioma cell growth. In the preceding article we show that the kappa-selective opioid agonist U69,593 acts as a mitogen with a potency similar to that of endothelin in the same astrocytic model system. Here we report that C6 cell treatment with mu-opioid agonists for 1 h results in the inhibition of kappa-opioid mitogenic signaling. The mu-selective agonist endomorphin-1 attenuates kappa-opioid-stimulated DNA synthesis, phosphoinositide turnover, and extracellular signal-regulated kinase phosphorylation. To investigate the role of receptor endocytosis in signaling, we have examined the effects of dynamin-1 and its GTPase-defective, dominant suppressor mutant (K44A) on opioid modulation of extracellular signal-regulated kinase phosphorylation in C6 cells. Overexpression of dynamin K44A in C6 cells does not affect kappa-opioid phosphorylation of extracellular signal-regulated kinase. However, it does block the inhibitory action on kappa-opioid signaling mediated by the kappa-opioid receptor. Our results are consistent with a growing body of evidence of the opposing actions of mu- and kappa-opioids and provide new insight into the role of opioid receptor trafficking in signaling.
Collapse
Affiliation(s)
- L M Bohn
- E.A. Doisy Department of Biochemistry and Molecular Biology, St. Louis University School of Medicine, Missouri 63104, USA
| | | | | |
Collapse
|
80
|
Drake CT, Milner TA. Mu opioid receptors are in somatodendritic and axonal compartments of GABAergic neurons in rat hippocampal formation. Brain Res 1999; 849:203-15. [PMID: 10592303 DOI: 10.1016/s0006-8993(99)01910-1] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Activation of mu opioid receptors (MORs) has a net excitatory effect in the hippocampal formation through inhibition of gamma-amino butyric acid (GABA)-containing interneurons. To determine the precise subcellular targets of MOR agonists, immunoreactivity against MOR1 and GABA was examined in single sections of the hippocampal formation prepared for dual-labeling electron microscopy. In both the CA1 region of hippocampus and the dentate gyrus, MOR-like immunoreactivity (-li) was present in neuronal somata, dendrites, axons, and axon terminals, as well as a very few glial processes. Axon terminals with MOR-li formed symmetric synapses with principal cell dendrites and somata. Many MOR-labeled profiles of all types also contained GABA-li, and the vast majority possessed the ultrastructural characteristics of interneurons. Additionally, in the dentate gyrus a very small proportion of granule cell dendrites contained MOR-li. MOR-li, identified using immunogold-silver particles, was often affiliated with the extrasynaptic regions of neuronal plasma membranes, consistent with responsiveness to diffusing endogenous neuropeptide ligands. Semiquantitative analysis of the distribution of MOR-li revealed significantly more "presynaptic" (axons and terminals) than "postsynaptic" (somata and dendrites) labeled profiles in most laminae. We conclude that in addition to previously described somatodendritic MOR-li, a substantial amount of MOR-li in hippocampal formation is presynaptic. Furthermore, MORs are almost exclusively in GABAergic interneurons.
Collapse
Affiliation(s)
- C T Drake
- Department of Neurology, Division of Neurobiology, Weill Medical College of Cornell University, 411 East 69th Street, New York, NY, USA.
| | | |
Collapse
|
81
|
Knapp PE, Ismaili S, Hauser KF, Ghandour MS. Abnormal Ca(2+) regulation in oligodendrocytes from the dysmyelinating jimpy mouse. Brain Res 1999; 847:332-7. [PMID: 10575104 DOI: 10.1016/s0006-8993(99)02012-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Jimpy (jp) is a point mutation in the gene on the X chromosome which codes for the major myelin proteolipid protein. Most oligodendrocytes (OLs) in the jp mouse undergo cell death at the time when they should be actively myelinating. Loss of mature OLs results in severe CNS dysmyelination. Dying jp OLs have the morphology of apoptotic cells but it is not clear how the mutation activates biochemical pathways which lead to programmed death of OLs in jp CNS. There is compelling evidence from a number of systems that high levels of intracellular Ca(2+) ([Ca2+]i) can activate downstream processes which result in both apoptotic and necrotic cell death. To determine whether [Ca2+](i) dysregulation might be involved in the death of jp OLs, we used ratiometric imaging to determine levels of [Ca2+](i) in OLs cultured from jp and normal CNS and in immortalized cell lines derived from jp and normal OLs. Immortalized jp OLs and OLs isolated directly from jp brain both showed a similar elevation in [Ca2+](i) ranging from 60% to 150% over control values. A higher baseline [Ca2+](i) in jp OLs might increase their vulnerability to other insults due to abnormal protein processing or changes in signaling pathways which act as a final trigger for cell death.
Collapse
Affiliation(s)
- P E Knapp
- Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, MS207 Chandler Medical Center, Lexington, KY 40536, USA.
| | | | | | | |
Collapse
|
82
|
Reznikov K, Hauser KF, Nazarevskaja G, Trunova Y, Derjabin V, Bakalkin G. Opioids modulate cell division in the germinal zone of the late embryonic neocortex. Eur J Neurosci 1999; 11:2711-9. [PMID: 10457167 DOI: 10.1046/j.1460-9568.1999.00680.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Opioid effects on cell division in the embryonic cerebral cortex were examined using two experimental approaches: (i) the presence of opioid receptors in the embryonic day 16 mouse neocortex was tested using immunohistochemical techniques; (ii) the values of the indices of [3H]thymidine pulse labelled cells and mitotic indices were estimated in the ventricular zone of the embryonic day 16 mouse neocortex 2.5, 4.5 and 8.5 h after administration to pregnant females of selected opioid receptor agonists or the opioid antagonist naloxone. The immunohistochemical study demonstrated that distinct subpopulations of the ventricular zone cells express mu, delta or kappa opioid receptors. Acute exposure of mouse embryos to mu, delta and kappa opioid receptor agonists or naloxone differentially affects the indices of [3H] thymidine pulse labelled cells and mitotic indices indicating changes in the cell cycle composition. Treatment with the mu opioid receptor agonist D-Ala2-MePhe4, Gly-ol5-enkephalin (DAGO), or the partially selective kappa opioid receptor agonist bremazocine, increased the [3H]thymidine labelling and mitotic indices. In contrast, the delta receptor agonist (D-Ser8)-leucine enkephalin-Thr (DSLET) produced a decrease in the labelled cell indices and mitotic indices. Naloxone provided a biphasic effect: a decrease in the values of labelled cell indices 2.5 h after naloxone administration, followed by an increase in the values of the indices at 4.5 and 8.5 h. These results suggest that the endogenous embryonic/maternal opioid systems are involved in the regulation of cell division in the ventricular zone of the late embryonic cortex.
Collapse
MESH Headings
- Animals
- Benzomorphans/pharmacology
- Cell Division/drug effects
- Cerebral Ventricles/embryology
- Embryo, Mammalian/cytology
- Embryo, Mammalian/metabolism
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-
- Enkephalin, Leucine/analogs & derivatives
- Enkephalin, Leucine/pharmacology
- Enkephalins/pharmacology
- Female
- Mice
- Mice, Inbred CBA
- Mitotic Index/drug effects
- Naloxone/pharmacology
- Narcotic Antagonists/pharmacology
- Narcotics/pharmacology
- Neocortex/embryology
- Pregnancy
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- K Reznikov
- Department of Clinical Neuroscience, Section of Alcohol and Drug Addiction Research, Karolinska Institute, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
83
|
Abstract
The distribution of mu opioid receptors was studied in human fetal spinal cords between 12-13 and 24-25 wk gestational ages. Autoradiographic localisation using [3H] DAMGO revealed the presence of mu receptors in the dorsal horn at all age groups with a higher density in the superficial laminae (I-II). A biphasic expression was noted. Receptor density increased in the dorsal horn, including the superficial laminae, between 12-13 and 16-17 wk. This could be associated with a spurt in neurogenesis. The density increased again at 24-25 wk in laminae I-II which resembled the adult pattern of distribution. A dramatic proliferation of cells was noted from the region of the ventricular zone between 16-17 and 24-25 wk. These were considered to be glial cells from their histological features. Mu receptor expression was noted over a large area of the spinal cord including the lateral funiculus at 24-25 wk. This may be due to receptor expression by glial cells. The study presents evidence of mu receptor expression by both neurons and glia during early development of human spinal cord.
Collapse
Affiliation(s)
- S B Ray
- Department of Anatomy, All India Institute of Medical Sciences, Ansari Nagar, New Delhi.
| | | |
Collapse
|
84
|
Nieto-Fernandez FE, Mattocks D, Cavani F, Salzet M, Stefano GB. Morphine coupling to invertebrate immunocyte nitric oxide release is dependent on intracellular calcium transients. Comp Biochem Physiol B Biochem Mol Biol 1999; 123:295-9. [PMID: 10481258 DOI: 10.1016/s0305-0491(99)00074-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Morphine significantly stimulated invertebrate immunocyte intracellular calcium level increases in a concentration-dependent manner in cells preloaded with Fura 2/AM. Morphine's action was blocked by prior exposure of the cells to the opiate receptor antagonist naloxone. Various opioid peptides did not exhibit this ability, indicating a morphine-mu 3 mediated process. In comparing the sequence of events concerning morphine's action in stimulating both [Ca2+]i and NO production in these cells, we found that the first event precedes the second by 42 +/- 7 s. The opiate stimulation of [Ca2+]i- was attenuated in cells leached of calcium. strongly suggesting that intracellular calcium levels regulate cNOS activity in invertebrate immunocytes.
Collapse
Affiliation(s)
- F E Nieto-Fernandez
- Neuroscience Research Institute, State University of New York at Old Westbury 11568-0210, USA.
| | | | | | | | | |
Collapse
|
85
|
Panagiotou S, Bakogeorgou E, Papakonstanti E, Hatzoglou A, Wallet F, Dussert C, Stournaras C, Martin PM, Castanas E. Opioid agonists modify breast cancer cell proliferation by blocking cells to the G2/M phase of the cycle: involvement of cytoskeletal elements. J Cell Biochem 1999; 73:204-11. [PMID: 10227383 DOI: 10.1002/(sici)1097-4644(19990501)73:2<204::aid-jcb6>3.0.co;2-v] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Opioids decrease cell proliferation in different systems including breast, prostate, lung, kidney, and intestine, through an interaction with opioid as well as other membrane-receptor systems (somatostatin, cholinergic), through an unidentified mechanism. Recently, we have reported an interaction of taxol with opioid membrane sites (BBRC 235, 201-204, 1997), and an involvement of opioids to the modification of actin cytoskeleton in renal OK cells (J Cell Biochem. [19981 70:60-69), indicating a possible action of the opioid effect. In the present work, we have examined the effect of two general opioid agonists (ethylketocyclazocine and etorphine) on the cell cycle, in human breast cancer T47D cells, as well as a possible modification of the cellular cytoskeleton under their action, in order to explain the antiproliferative effect of these agents. These two opioids produce a dose-dependent and reversible decrease of the proliferation of T47D cells, with a maximum attained at 10(-8) M. The addition of 10(-8) M of either opioid produced a significant increase of the number of cells arrested in the G2/M phase. Confocal laser microscopy revealed a modification of the actin and tubulin microfilaments, with a clear redistribution at the periphery of the cell, reversed by the addition of the general opioid antagonist diprenorphine. Furthermore, differences between the two opioids were obvious, attributed to the different receptor affinity of each agent. The observed redistribution of actin and tubulin cytoskeletal elements gives therefore a possible answer of the antiproliferative action of opioids. The modification of the cytoskeleton, directly involved to cell division, might provoke a "mechanical" obstacle, which could be the reason of the antiproliferative effect of these agonists. Furthermore, the observed tubulin-opioid interaction by opioids provides a possible explanation of the arrest at the G2/M phase of T47D cells under opioid treatment. Nevertheless, although the observed interaction of opioids with cytoskeletal elements gives a plausible answer of the antiproliferative effects of the agents, this might not be the only action of these agents in cell proliferation. Other, direct or indirect, genomic actions, which which remains to be elucidated, might be taken into consideration.
Collapse
Affiliation(s)
- S Panagiotou
- Laboratory of Experimental Cancerology, CJF 93-11, Marseille, France
| | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Fimiani C, Mattocks D, Cavani F, Salzet M, Deutsch DG, Pryor S, Bilfinger TV, Stefano GB. Morphine and anandamide stimulate intracellular calcium transients in human arterial endothelial cells: coupling to nitric oxide release. Cell Signal 1999; 11:189-93. [PMID: 10353693 DOI: 10.1016/s0898-6568(98)00060-6] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Both morphine and anandamide significantly stimulated cultured endothelial intracellular calcium level increases in a concentration-dependent manner in cells pre-loaded with fura 2/AM. Morphine is more potent than anandamide (approximately 275 vs. 135 nM [Ca]i), and the [Ca]i for both ligands was blocked by prior exposure of the cells to their respective receptor antagonist, i.e., naloxone and SR 171416A. Various opioid peptides did not exhibit this ability, indicating a morphine-mu3-mediated process. In comparing the sequence of events concerning morphine's and anandamide's action in stimulating both [Ca]i and nitric oxide production in endothelial cells, we found that the first event precedes the second by 40+/-8 sec. The opiate and cannabinoid stimulation of [Ca]i was attenuated in cells leeched of calcium, strongly suggesting that intracellular calcium levels regulate cNOS activity.
Collapse
Affiliation(s)
- C Fimiani
- Multidisciplinary Center for the Study of Aging, Neuroscience Research Institute, State University of New York at Old Westbury, 11568-0210, USA
| | | | | | | | | | | | | | | |
Collapse
|
87
|
Fimiani C, Liberty T, Aquirre AJ, Amin I, Ali N, Stefano GB. Opiate, cannabinoid, and eicosanoid signaling converges on common intracellular pathways nitric oxide coupling. Prostaglandins Other Lipid Mediat 1999; 57:23-34. [PMID: 10367294 DOI: 10.1016/s0090-6980(98)00068-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Scientific fields as they emerge initially appear to be unrelated to other projects even if they are in a similar area of interest. This is especially true in the case of opiate, cannabinoid, and eicosanoid signaling processes. In this limited speculative review, we attempt to examine aspects of their intracellular cascading signaling systems for their commonalities. We find intracellular calcium mobilization, nuclear factor kappa B involvement, adenylate cyclase activity, and, finally, constitutive nitric oxide release to be converging points for these signaling processes, occurring by separate and distinct receptor-mediated effector systems. Phosphokinase C, mitogen activated protein kinase, and cytosolic phospholipase A2 also represent points of common impact. In this regard, aspirin also appears to be involved in an aspect of this signaling convergence. We conclude that many of the physiological observations regarding the actions of these signaling molecules, for example, immunosuppression, neurotransmission, vasodilation, cellular adherence, and cytotoxicity, can now be understood by considering their converging biochemical cascades.
Collapse
Affiliation(s)
- C Fimiani
- Neuroscience Research Institute, State University of New York at Old Westbury, NY 11568, USA
| | | | | | | | | | | |
Collapse
|
88
|
Polakiewicz RD, Schieferl SM, Gingras AC, Sonenberg N, Comb MJ. mu-Opioid receptor activates signaling pathways implicated in cell survival and translational control. J Biol Chem 1998; 273:23534-41. [PMID: 9722592 DOI: 10.1074/jbc.273.36.23534] [Citation(s) in RCA: 151] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The mu-opioid receptor mediates the analgesic and addictive properties of morphine. Despite the clinical importance of this G-protein-coupled receptor and many years of pharmacological research, few intracellular signaling mechanisms triggered by morphine and other mu-opioid agonists have been described. We report that mu-opioid agonists stimulate three different effectors of a phosphoinositide 3-kinase (PI3K)-dependent signaling cascade. By using a cell line stably transfected with the mu-opioid receptor cDNA, we show that the specific agonist [D-Ala2,N-Me-Phe4,Gly5-ol]enkephalin (DAMGO) stimulates the activity of Akt, a serine/threonine protein kinase implicated in protecting neurons from apoptosis. Activation of Akt by DAMGO correlates with its phosphorylation at serine 473. The selective PI3K inhibitors wortmannin and LY294002 blocked phosphorylation of this site, previously shown to be necessary for Akt enzymatic activity. DAMGO also stimulates the phosphorylation of two other downstream effectors of PI3K, the p70 S6 kinase and the repressors of mRNA translation, 4E-BP1 and 4E-BP2. Upon mu-opioid receptor stimulation, p70 S6 kinase is activated and phosphorylated at threonine 389 and at threonine 421/serine 424. Phosphorylation of p70 S6 kinase and 4E-BP1 is also repressed by PI3K inhibitors as well as by rapamycin, the selective inhibitor of FRAP/mTOR. Consistent with these findings, DAMGO-stimulated phosphorylation of 4E-BP1 impairs its ability to bind the translation initiation factor eIF-4E. These results demonstrate that the mu-opioid receptor activates signaling pathways associated with neuronal survival and translational control, two processes implicated in neuronal development and synaptic plasticity.
Collapse
Affiliation(s)
- R D Polakiewicz
- Cell Signaling Laboratory, New England Biolabs, Beverly, Massachusetts 01915, USA
| | | | | | | | | |
Collapse
|
89
|
Opanashuk LA, Hauser KF. Opposing actions of the EGF family and opioids: heparin binding-epidermal growth factor (HB-EGF) protects mouse cerebellar neuroblasts against the antiproliferative effect of morphine. Brain Res 1998; 804:87-94. [PMID: 9729296 DOI: 10.1016/s0006-8993(98)00647-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Endogenous opioids and opiate drugs of abuse inhibit the proliferation of cerebellar external granular layer (EGL) neuroblasts by mechanisms that are incompletely understood. Opioids do not act alone, rather multiple extracellular factors regulate granule cell neurogenesis and these undoubtedly act in concert with opioids to shape developmental outcome. We examined whether, heparin binding-epidermal growth factor-like growth factor (HB-EGF), a recently described member of the epidermal growth factor (EGF) family, might compete with an inhibitory opioid signal. The results confirmed our ongoing studies that morphine inhibited neuroblast proliferation, while HB-EGF enhanced cell replication. HB-EGF not only counteracted the antiproliferative morphine signal, but invariably enhanced DNA synthesis irrespective of morphine treatment. Our findings suggest that regional and temporal differences in the availability of endogenous HB-EGF may serve to limit the response of EGL neuroblasts to opioids, and HB-EGF may be neuroprotective in opiate drug abuse. If similar responses occur in vivo, then the EGF family and the opioid system may represent distinct and contrasting components of an extracellular signaling system serving to coordinate EGL neurogenesis.
Collapse
Affiliation(s)
- L A Opanashuk
- Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY 40536-0084, USA
| | | |
Collapse
|
90
|
|
91
|
Thorlin T, Eriksson PS, Persson PA, Aberg ND, Hansson E, Rönnbäck L. Delta-opioid receptors on astroglial cells in primary culture: mobilization of intracellular free calcium via a pertussis sensitive G protein. Neuropharmacology 1998; 37:299-311. [PMID: 9681928 DOI: 10.1016/s0028-3908(98)00026-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Astrocytes in primary culture from rat cerebral cortex were probed concerning the expression of delta-opioid receptors and their coupling to changes in intracellular free calcium concentrations ([Ca2+]i). Fluo-3 or fura-2 based microspectrofluorometry was used for [Ca2+]i measurements on single astrocytes in a mixed astroglial-neuronal culture. Application of the selective delta-opioid receptor agonist, [D-Pen2, D-Pen5]-enkephalin (DPDPE), at concentrations ranging from 10 nM to 100 microM, induced concentration-dependent increases in [Ca2+]i (EC50 = 114 nM). The responses could be divided into two phases, with an initial spike in [Ca2+]i followed by either oscillations or a sustained elevation of [Ca2+]i. These effects were blocked by the selective delta-opioid receptor antagonist ICI 174864 (10 microM). The expression of delta-opioid receptors on astroglial cells was further verified immunohistochemically, using specific antibodies, and by Western blot analyses. Pre-treatment of the cells with pertussis toxin (100 ng/ml, 24 h) blocked the effects of delta-opioid receptor activation, consistent with a Gi- or Go-mediated response. The sustained elevation of [Ca2+]i was not observed in low extracellular Ca2+ and was partly blocked by nifedipine (1 microM), indicating the involvement of L-type Ca2+ channels. Stimulating neurons with DPDPE resulted in a decrease in [Ca2+]i, which may be consistent with the closure of the plasma membrane Ca2+ channels on these cells. The current results suggest a role for astrocytes in the response of the brain to delta-opioid peptides and that these opioid effects in part involve altered astrocytic intracellular Ca2+ homeostasis.
Collapse
Affiliation(s)
- T Thorlin
- Institute of Neurobiology, Department of Neurobiology, Göteborg University, Sweden.
| | | | | | | | | | | |
Collapse
|
92
|
Abstract
The astrocytoma cell line rat C6 glioma has been used as a model system to study the mechanism of various opioid actions. Nevertheless, the type of opioid receptor(s) involved has not been established. Here we demonstrate the presence of high-affinity U69,593, endomorphin-1, morphine, and beta-endorphin binding in desipramine (DMI)-treated C6 cell membranes by performing homologous and heterologous binding assays with [3H]U69,593, [3H]morphine, or 125I-beta-endorphin. Naive C6 cell membranes displayed U69,593 but neither endomorphin-1, morphine, nor beta-endorphin binding. Cross-linking of 125I-beta-endorphin to C6 membranes gave labeled bands characteristic of opioid receptors. Moreover, RT-PCR analysis of opioid receptor expression in control and DMI-treated C6 cells indicate that both kappa- and mu-opioid receptors are expressed. There does not appear to be a significant difference in the level of mu nor kappa receptor expression in naive versus C6 cells treated with DMI over a 20-h period. Collectively, the data indicate that kappa- and mu-opioid receptors are present in C6 glioma cells.
Collapse
Affiliation(s)
- L M Bohn
- E. A. Doisy Department of Biochemistry and Molecular Biology, St. Louis University School of Medicine, Missouri 63104, USA
| | | | | |
Collapse
|
93
|
Hauser KF, Harris-White ME, Jackson JA, Opanashuk LA, Carney JM. Opioids disrupt Ca2+ homeostasis and induce carbonyl oxyradical production in mouse astrocytes in vitro: transient increases and adaptation to sustained exposure. Exp Neurol 1998; 151:70-6. [PMID: 9582255 DOI: 10.1006/exnr.1998.6788] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pharmacologically distinct subpopulations of astroglia express mu, delta, and/or kappa opioid receptors. Activation of mu, delta, or kappa opioid receptors can destabilize intracellular calcium ([Ca2+]i) in astrocytes leading to cellular hypertrophy and reactive injury. To assess whether acute or sustained opioid exposure might adversely affect astroglial function by disrupting Ca2+ homeostasis or by producing reactive oxygen species, fura-2 and a novel fluorescent-tagged biotin-4-amidobenzoic hydrazide reagent, respectively, were used to detect [Ca2+]i and carbonyl oxidation products within individual murine astrocytes. Acute (3 h) exposure to mu; (H-Tyr-Pro-Phe (N-Me) -D-Pro-NH2; PLO17), delta ([D-Pen2, D-Pen5]-enkephalin), and kappa (trans-(+/-)-3, 4-dichloro-N-methyl-N-[2-(1-pyrr olidinyl) cyclohexyl] benzeneacetamide methanesulfonate; U50,488H) opioid agonists caused significant mean increases in [Ca2+]i and in the levels of oxidative products in astrocytes. In contrast, following 72 h of continuous opioid exposure, [Ca2+]i and carbonyl levels returned to normal, irrespective of opioid treatment. These preliminary findings indicate that opioids initially destabilize [Ca2+]i and increase reactive oxygen species in astrocytes; however, astrocytes later recover and adapt to sustained opioid exposure.
Collapse
MESH Headings
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology
- Analgesics/pharmacology
- Analgesics, Non-Narcotic/pharmacology
- Animals
- Astrocytes/chemistry
- Astrocytes/drug effects
- Astrocytes/metabolism
- Calcium/metabolism
- Drug Tolerance
- Endorphins/pharmacology
- Enkephalin, D-Penicillamine (2,5)-
- Enkephalins/pharmacology
- Homeostasis/drug effects
- Mice
- Mice, Inbred ICR
- Naloxone/pharmacology
- Narcotic Antagonists/pharmacology
- Narcotics/pharmacology
- Oxidative Stress/drug effects
- Reactive Oxygen Species/metabolism
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, mu/antagonists & inhibitors
Collapse
Affiliation(s)
- K F Hauser
- Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington, Kentucky 40536-0084, USA
| | | | | | | | | |
Collapse
|
94
|
Abstract
To characterize further the establishment of the opioid system during prenatal mouse development, we have examined the spatial and temporal expression patterns of mu, kappa, and delta opioid receptor mRNAs and find that the expression patterns of these mRNAs are distinct at all ages. Within the embryo, kappa is the first opioid receptor expressed, with transcripts detected in the gut epithelium as early as embryonic day 9.5 (E9.5). By E10.5, mu receptor expression is first detected in the facial-vestibulocochlear preganglion complex, whereas delta receptor mRNA is first detected at E12.5 in several peripheral tissues, including the olfactory epithelium, heart, limb bud, and tooth. In the brain, both mu and kappa mRNAs are first detected at E11.5 in the basal ganglia and midbrain, respectively. During mid-gestation and late gestation, the expression of both mu and kappa receptors extends to other brain regions that exhibit high expression in the adult, including the medial habenula, hypothalamus, pons, and medulla for mu and the basal ganglia, thalamus, hypothalamus, raphe, and ventral tegmental area for kappa. Thus by E17.5, many aspects of the adult expression patterns of mu and kappa receptors already have been established. Compared with mu and kappa, delta receptor mRNA expression in the brain begins relatively late, and the expression levels remain very low even at E19.5. In contrast to its late appearance in the brain, however, delta is the first opioid receptor expressed in the dorsal root ganglion, at E12.5, before its expression in the spinal cord begins at E15.5. Mu receptor is the first opioid receptor expressed in the spinal cord, at E11.5. These results extend previous ligand-binding data to significantly earlier ages and suggest that early developmental events in both neural and non-neural tissues may be modulated by opioid receptors. Several examples of possible autocrine and paracrine loops of opioid peptide and receptor expression have been identified, suggesting a role for these local circuits in developmental processes.
Collapse
|
95
|
Zhu Y, Hsu MS, Pintar JE. Developmental expression of the mu, kappa, and delta opioid receptor mRNAs in mouse. J Neurosci 1998; 18:2538-49. [PMID: 9502813 PMCID: PMC6793117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
To characterize further the establishment of the opioid system during prenatal mouse development, we have examined the spatial and temporal expression patterns of mu, kappa, and delta opioid receptor mRNAs and find that the expression patterns of these mRNAs are distinct at all ages. Within the embryo, kappa is the first opioid receptor expressed, with transcripts detected in the gut epithelium as early as embryonic day 9.5 (E9.5). By E10.5, mu receptor expression is first detected in the facial-vestibulocochlear preganglion complex, whereas delta receptor mRNA is first detected at E12.5 in several peripheral tissues, including the olfactory epithelium, heart, limb bud, and tooth. In the brain, both mu and kappa mRNAs are first detected at E11.5 in the basal ganglia and midbrain, respectively. During mid-gestation and late gestation, the expression of both mu and kappa receptors extends to other brain regions that exhibit high expression in the adult, including the medial habenula, hypothalamus, pons, and medulla for mu and the basal ganglia, thalamus, hypothalamus, raphe, and ventral tegmental area for kappa. Thus by E17.5, many aspects of the adult expression patterns of mu and kappa receptors already have been established. Compared with mu and kappa, delta receptor mRNA expression in the brain begins relatively late, and the expression levels remain very low even at E19.5. In contrast to its late appearance in the brain, however, delta is the first opioid receptor expressed in the dorsal root ganglion, at E12.5, before its expression in the spinal cord begins at E15.5. Mu receptor is the first opioid receptor expressed in the spinal cord, at E11.5. These results extend previous ligand-binding data to significantly earlier ages and suggest that early developmental events in both neural and non-neural tissues may be modulated by opioid receptors. Several examples of possible autocrine and paracrine loops of opioid peptide and receptor expression have been identified, suggesting a role for these local circuits in developmental processes.
Collapse
MESH Headings
- Animals
- Central Nervous System/chemistry
- Central Nervous System/embryology
- Female
- Ganglia, Spinal/chemistry
- Ganglia, Spinal/embryology
- Ganglia, Sympathetic/chemistry
- Ganglia, Sympathetic/embryology
- Gene Expression Regulation, Developmental
- In Situ Hybridization
- Intestines/chemistry
- Intestines/embryology
- Mice
- Mice, Inbred C57BL
- Pregnancy
- RNA, Messenger/analysis
- Receptors, Opioid/genetics
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, mu/genetics
- Trigeminal Ganglion/chemistry
- Trigeminal Ganglion/embryology
Collapse
Affiliation(s)
- Y Zhu
- Department of Neuroscience and Cell Biology, University of Medicine and Dentistry of New Jersey-Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA
| | | | | |
Collapse
|
96
|
Tryoen-Toth P, Gavériaux-Ruff C, Maderspach K, Labourdette G. Regulation of kappa-opioid receptor mRNA level by cyclic AMP and growth factors in cultured rat glial cells. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1998; 55:141-50. [PMID: 9645969 DOI: 10.1016/s0169-328x(97)00373-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The mRNA of the kappa-opioid receptor (KOR) has been found recently in cultured astrocytes and in microglia. By using RT-PCR and Southern hybridization, we confirmed these observations and, in addition, we observed that KOR mRNA was expressed in oligodendrocytes and in the precursors of astrocytes and oligodendrocytes. KOR mRNA level was the highest in the immature astrocytes and decreased with their maturation. Very few data are available on the regulation of KOR level by extracellular signals. Therefore, we examined the effect of three growth factors known to be present in the adult brain, basic fibroblast growth factor (bFGF), platelet-derived growth factor (PDGF-BB) and leukemia inhibitory factor (LIF) and of two cyclic AMP (cAMP) generating systems, the cAMP analog, 8-(4-chlorophenylthio)-cAMP and forskolin, on this level. It was found that in astrocytes, KOR mRNA level decreased dramatically under the effect of cAMP and less under the effect of bFGF while it did not change significantly after LIF treatment. In oligodendrocytes, it also decreased with cAMP, but increased under the effect of bFGF and PDGF-BB. In microglia, a decrease was observed with cAMP and lipopolysaccharides (LPS), the most used activators of macrophages. These results shed new evidence on the expression of opioid receptor mRNA in the glial cells of the rat CNS. The regulation of KOR mRNA level under the effect of extracellular signals suggests that opioids take part in dynamic processes in glial cells, possibly related to glial-neuron communication.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Astrocytes/drug effects
- Astrocytes/metabolism
- Becaplermin
- Blotting, Southern
- Brain/cytology
- Cell Communication
- Cells, Cultured
- Colforsin/pharmacology
- Cyclic AMP/analogs & derivatives
- Cyclic AMP/pharmacology
- Cyclic AMP/physiology
- Fibroblast Growth Factor 2/pharmacology
- Gene Expression Regulation/drug effects
- Growth Inhibitors/pharmacology
- Interleukin-6
- Leukemia Inhibitory Factor
- Lipopolysaccharides/pharmacology
- Lymphokines/pharmacology
- Microglia/drug effects
- Microglia/metabolism
- Nerve Tissue Proteins/biosynthesis
- Nerve Tissue Proteins/genetics
- Neuroglia/drug effects
- Neuroglia/metabolism
- Oligodendroglia/drug effects
- Oligodendroglia/metabolism
- Opioid Peptides/physiology
- Platelet-Derived Growth Factor/pharmacology
- Polymerase Chain Reaction
- Proto-Oncogene Proteins c-sis
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Rats
- Rats, Wistar
- Receptors, Opioid, kappa/biosynthesis
- Receptors, Opioid, kappa/genetics
- Stem Cells/drug effects
- Stem Cells/metabolism
- Thionucleotides/pharmacology
Collapse
Affiliation(s)
- P Tryoen-Toth
- Laboratoire de neurobiologie du Développement et de la Régénération, UPR 132 CNRS, Strasbourg, France
| | | | | | | |
Collapse
|
97
|
Stiene-Martin A, Zhou R, Hauser KF. Regional, developmental, and cell cycle-dependent differences in ?, ?, and ?-opioid receptor expression among cultured mouse astrocytes. Glia 1998. [DOI: 10.1002/(sici)1098-1136(199803)22:3<249::aid-glia4>3.0.co;2-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
98
|
Leslie FM, Chen Y, Winzer-Serhan UH. Opioid receptor and peptide mRNA expression in proliferative zones of fetal rat central nervous system. Can J Physiol Pharmacol 1998. [DOI: 10.1139/y98-028] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
There is increasing evidence to suggest that opioid peptides may have widespread effects as regulators of growth. To evaluate the hypothesis that endogenous opioids control cellular proliferation during neural development, we have used in situ hybridization to examine opioid peptide and receptor mRNA expression in neuroepithelial zones of fetal rat brain and spinal cord. Our data show that proenkephalin mRNA is widely expressed in forebrain germinal zones and choroid plexus during the second half of gestation. In contrast, prodynorphin mRNA expression is restricted to the periventricular region of the ventral spinal cord. Little µ or delta receptor mRNA expression was detected in any regions of neuronal proliferation prior to birth. However, kappa receptor mRNA is widely expressed in hindbrain germinal zones during the 3rd week of gestation. Our present findings support the hypothesis that endogenous opioids may regulate proliferation of both neuronal and non-neuronal cells during central nervous system development. Given the segregated expression of proenkephalin mRNA in forebrain neuroepithelium and kappa receptor mRNA within hindbrain, different opioid mechanisms may regulate cell division in rostral and caudal brain regions.Key words: enkephalin, dynorphin, ontogeny, neurogenesis.
Collapse
|
99
|
Stiene-Martin A, Zhou R, Hauser KF. Regional, developmental, and cell cycle-dependent differences in mu, delta, and kappa-opioid receptor expression among cultured mouse astrocytes. Glia 1998; 22:249-59. [PMID: 9482211 PMCID: PMC4319791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
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.
Collapse
MESH Headings
- 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/chemistry
- Astrocytes/cytology
- Astrocytes/metabolism
- Brain Chemistry/physiology
- Calcium/metabolism
- Cell Cycle/physiology
- 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/biosynthesis
- Receptors, Opioid/immunology
- Receptors, Opioid, delta/analysis
- Receptors, Opioid, delta/biosynthesis
- Receptors, Opioid, delta/immunology
- Receptors, Opioid, kappa/analysis
- Receptors, Opioid, kappa/biosynthesis
- Receptors, Opioid, kappa/immunology
- Receptors, Opioid, mu/analysis
- Receptors, Opioid, mu/biosynthesis
- Receptors, Opioid, mu/immunology
Collapse
Affiliation(s)
- A Stiene-Martin
- Department of Clinical Science, University of Kentucky Medical Center, Lexington 40536-0084, USA
| | | | | |
Collapse
|
100
|
Knapp PE, Maderspach K, Hauser KF. Endogenous opioid system in developing normal and jimpy oligodendrocytes: mu and kappa opioid receptors mediate differential mitogenic and growth responses. Glia 1998; 22:189-201. [PMID: 9537839 DOI: 10.1002/(sici)1098-1136(199802)22:2<189::aid-glia10>3.0.co;2-u] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The early development of both neurons and neuroglia may be modulated by signaling through opioid mediated pathways. Neurons and astroglia not only express specific types of opiate receptors, but also respond functionally to opioids with altered rates of proliferation and growth. The present study was undertaken to determine if opioids also modulate development of the other major CNS macroglial cell, the oligodendrocyte (OL). Using well-characterized polyclonal antibodies specific for delta-, kappa-, and mu-opiate receptors, OLs grown in vitro were shown to express mu-receptors at a very immature stage prior to expression of kappa-receptors. This developmentally regulated sequence differs from the pattern of expression in neurons and astroglia. delta-receptors are apparently absent from cultured OLs. OLs also have physiologic responses to selective mu- and kappa-receptor agonists and antagonists. Exposure of relatively immature O4+ OLs to the mu-receptor agonist PL017 [H-Tyr-Pro-Phe(N-Me)-D-Pro-NH2] resulted in a significant enhancement in the rate of DNA synthesis. This effect, which was not observed in more mature MBP+ OLs, was entirely blocked by the antagonist naloxone. Although the kappa-receptor pathway appeared to be uninvolved in controlling proliferation, the kappa-receptor antagonist nor-binaltorphimine significantly increased the size of myelin-like membranes produced by the cultured OLs. Interestingly, OLs derived from the jimpy mouse, a mutant characterized by an almost complete lack of CNS myelin and premature death of OLs, were found to be deficient in kappa-opiate receptors. Our findings clearly show that OLs not only express specific opiate receptors, but also respond to changes in their level of stimulation in ways that could profoundly impact nervous system morphology and function. If opiate receptors are expressed by OLs in vivo, their pharmacological manipulation might provide a novel pathway for modulating OL and myelin production both during development and in demyelinated conditions.
Collapse
MESH Headings
- Animals
- Animals, Newborn/physiology
- Antimetabolites
- Bromodeoxyuridine
- Cell Size
- Cells, Cultured
- Endorphins/pharmacology
- Endorphins/physiology
- Immunohistochemistry
- Mice
- Mice, Jimpy
- Mitogens/pharmacology
- Oligodendroglia/drug effects
- Oligodendroglia/metabolism
- Oligodendroglia/ultrastructure
- Receptors, Opioid/drug effects
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/biosynthesis
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/biosynthesis
Collapse
Affiliation(s)
- P E Knapp
- Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington 40536-0084, USA.
| | | | | |
Collapse
|