101
|
Easton N, Marshall FH, Marsden CA, Fone KCF. Mapping the central effects of methylphenidate in the rat using pharmacological MRI BOLD contrast. Neuropharmacology 2009; 57:653-64. [PMID: 19733553 DOI: 10.1016/j.neuropharm.2009.08.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Revised: 08/10/2009] [Accepted: 08/11/2009] [Indexed: 11/26/2022]
Abstract
Methylphenidate (Ritalin) is a selective dopamine reuptake inhibitor and an effective treatment for attention deficit hyperactivity disorder (ADHD) however the anatomical foci and neuronal circuits involved in these therapeutic benefits are unclear. This study determines the temporal pattern of brain regional activity change produced by systemic administration of a therapeutically relevant dose of methylphenidate in anaesthetised Sprague-Dawley rats using BOLD MRI and a 2.35T Bruker magnet. Following 60 min basal recording separate rats received saline (n = 9) or +/- methylphenidate hydrochloride (2 mg/kg, i.p., n = 9) and BOLD changes were recorded for 90 min using statistical parametric maps. Methylphenidate produced significant positive random BOLD effects in the nucleus accumbens, substantia nigra, entorhinal cortex and medial orbital cortex. Negative random BOLD effects were more widespread and intense, occurring in the motor and somatosensory cortices, caudate putamen, lateral globus pallidus and bed nucleus of the stria terminalis, without accompanying changes in blood pressure or respiratory rate. Methylphenidate-induced negative BOLD in the striatum, and other dopamine terminal areas, may reflect post-synaptic changes produced by blockade of the neuronal dopamine reuptake transporter. While increased positive BOLD in the medial orbital cortex may reflect altered dopamine and/or noradrenaline release indirectly altering striatal activity. The overall pattern of BOLD changes is comparable to that seen in previous studies using guanfacine, amphetamine and atomoxetine, and suggests that although these compounds operate through distinct pharmacological mechanisms the BOLD changes may represent a 'fingerprint pattern' predictive of therapeutic benefit in ADHD.
Collapse
Affiliation(s)
- Neil Easton
- School of Biomedical Sciences, Medical School, Institute of Neuroscience, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | | | | | | |
Collapse
|
102
|
Shibui Y, He XJ, Uchida K, Nakayama H. MPTP-induced neuroblast apoptosis in the subventricular zone is not regulated by dopamine or other monoamine transporters. Neurotoxicology 2009; 30:1036-44. [PMID: 19616025 DOI: 10.1016/j.neuro.2009.07.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Revised: 07/05/2009] [Accepted: 07/07/2009] [Indexed: 11/29/2022]
Abstract
For 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to exert neurotoxicity on dopaminergic neurons, 1-methyl-4-phenylpyridinium (MPP+), a metabolite of MPTP, must be taken up into the dopaminergic neuron via the dopamine transporter (DAT). Previous reports have shown that MPTP also causes neuroblast apoptosis in the subventricular zone (SVZ) of adult mice. The aim of this study is to elucidate the role of DAT and other monoamine transporters including vesicular monoamine transporter 2 (VMAT2), the serotonin transporter (SERT), and the norepinephrine transporter (NET) on the neuroblast apoptosis induced by MPTP administration. There were no DAT-positive neuroblasts in the SVZ, whereas some neuroblasts were immunopositive for VMAT2 and SERT. To examine whether these transporters are involved in MPTP-induced neuroblast apoptosis in the SVZ, terminal deoxynucleotidyl transferase-mediated dUTP endlabeling (TUNEL)-positive cells were semiquantitatively analyzed after the injection of GBR12909 (GBR), a DAT inhibitor; tetrabenazine (TBZ), a VMAT2 inhibitor; fluoxetine (FLU), a SERT inhibitor, or desipramine (DES), a NET inhibitor, prior to MPTP injection. However, the injection of these transporter inhibitors had no influence on the MPTP-induced neuroblast apoptosis in the SVZ. It is likely that neither DAT nor other monoamine transporters are involved in MPTP-induced neuroblast apoptosis. The present findings suggest that the neurotoxicity of MPTP to neuroblasts in the SVZ does not require DAT or other monoamine transporters, and the apoptosis it induces may be executed through other unknown pathways.
Collapse
Affiliation(s)
- Yusuke Shibui
- Department of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-Ku, Tokyo 113-8657, Japan.
| | | | | | | |
Collapse
|
103
|
Fan Y, Huang J, Kieran N, Zhu MY. Effects of transcription factors Phox2 on expression of norepinephrine transporter and dopamine beta-hydroxylase in SK-N-BE(2)C cells. J Neurochem 2009; 110:1502-13. [PMID: 19573018 DOI: 10.1111/j.1471-4159.2009.06260.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Phox2a and Phox2b are two homeodomain proteins that control the differentiation of noradrenergic neurons during embryogenesis. In the present study, we examined the possible effect of Phox2a/2b on the in vitro expression of the norepinephrine transporter (NET) and dopamine beta-hydroxylase (DBH), two important markers of the noradrenergic system. SK-N-BE(2)C cells were transfected with cDNAs or short hairpin RNAs specific to the human Phox2a and Phox2b genes. Transfection of 0.1 to 5 mug of cDNAs of Phox2a or Phox2b significantly increased mRNA and protein levels of NET and DBH in a concentration-dependent manner. As a consequence of the enhanced expression of NET after transfection, there was a parallel increase in the uptake of [(3)H]norepinephrine. Co-transfection of Phox2a and Phox2b did not further increase the expression of noradrenergic markers when compared with transfection of either Phox2a or Phox2b alone. Transfection of shRNAs specific to Phox2a or Phox2b genes significantly reduced mRNA and protein levels of NET and DBH after shutdown of endogenous Phox2, which was accompanied by a decreased [(3)H]norepinephrine uptake. Furthermore, there was an additive effect after cotransfection with both shRNAs specific to Phox2a or Phox2b genes on NET mRNA levels. Finally, the reduced DBH expression caused by the shRNA specific to Phox2a could be reversed by transfection with Phox2b cDNA and vice versa. The present findings verify the determinant role of Phox2a and Phox2b on the expression and function of NET and DBH in vitro. Further clarifying the regulatory role of these two transcription factors on key proteins of the noradrenergic system may open a new avenue for therapeutics of aging-caused dysfunction of the noradrenergic system.
Collapse
Affiliation(s)
- Yan Fan
- Department of Biochemistry and Molecular Biology, School of Medicine, Soochow University Suzhou, China
| | | | | | | |
Collapse
|
104
|
Matthies HJG, Han Q, Shields A, Wright J, Moore JL, Winder DG, Galli A, Blakely RD. Subcellular localization of the antidepressant-sensitive norepinephrine transporter. BMC Neurosci 2009; 10:65. [PMID: 19545450 PMCID: PMC2716352 DOI: 10.1186/1471-2202-10-65] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Accepted: 06/23/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Reuptake of synaptic norepinephrine (NE) via the antidepressant-sensitive NE transporter (NET) supports efficient noradrenergic signaling and presynaptic NE homeostasis. Limited, and somewhat contradictory, information currently describes the axonal transport and localization of NET in neurons. RESULTS We elucidate NET localization in brain and superior cervical ganglion (SCG) neurons, aided by a new NET monoclonal antibody, subcellular immunoisolation techniques and quantitative immunofluorescence approaches. We present evidence that axonal NET extensively colocalizes with syntaxin 1A, and to a limited degree with SCAMP2 and synaptophysin. Intracellular NET in SCG axons and boutons also quantitatively segregates from the vesicular monoamine transporter 2 (VMAT2), findings corroborated by organelle isolation studies. At the surface of SCG boutons, NET resides in both lipid raft and non-lipid raft subdomains and colocalizes with syntaxin 1A. CONCLUSION Our findings support the hypothesis that SCG NET is segregated prior to transport from the cell body from proteins comprising large dense core vesicles. Once localized to presynaptic boutons, NET does not recycle via VMAT2-positive, small dense core vesicles. Finally, once NET reaches presynaptic plasma membranes, the transporter localizes to syntaxin 1A-rich plasma membrane domains, with a portion found in cholera toxin-demarcated lipid rafts. Our findings indicate that activity-dependent insertion of NET into the SCG plasma membrane derives from vesicles distinct from those that deliver NE. Moreover, NET is localized in presynaptic membranes in a manner that can take advantage of regulatory processes targeting lipid raft subdomains.
Collapse
Affiliation(s)
- Heinrich J G Matthies
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | | | | | | | | | | | | | | |
Collapse
|
105
|
Carey MR, Regehr WG. Noradrenergic control of associative synaptic plasticity by selective modulation of instructive signals. Neuron 2009; 62:112-22. [PMID: 19376071 DOI: 10.1016/j.neuron.2009.02.022] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2008] [Revised: 12/10/2008] [Accepted: 02/18/2009] [Indexed: 10/20/2022]
Abstract
Synapses throughout the brain are modified through associative mechanisms in which one input provides an instructive signal for changes in the strength of a second coactivated input. In cerebellar Purkinje cells, climbing fiber synapses provide an instructive signal for plasticity at parallel fiber synapses. Here, we show that noradrenaline activates alpha2-adrenergic receptors to control short-term and long-term associative plasticity of parallel fiber synapses. This regulation of plasticity does not reflect a conventional direct modulation of the postsynaptic Purkinje cell or presynaptic parallel fibers. Instead, noradrenaline reduces associative plasticity by selectively decreasing the probability of release at the climbing fiber synapse, which in turn decreases climbing fiber-evoked dendritic calcium signals. These findings raise the possibility that targeted presynaptic modulation of instructive synapses could provide a general mechanism for dynamic context-dependent modulation of associative plasticity.
Collapse
Affiliation(s)
- Megan R Carey
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
106
|
Zhang H, Li S, Wang M, Vukusic B, Pristupa ZB, Liu F. Regulation of dopamine transporter activity by carboxypeptidase E. Mol Brain 2009; 2:10. [PMID: 19419578 PMCID: PMC2687442 DOI: 10.1186/1756-6606-2-10] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Accepted: 05/06/2009] [Indexed: 12/16/2022] Open
Abstract
Background The dopamine transporter (DAT) plays a critical role in terminating the action of dopamine by rapid reuptake into the presynaptic neuron. Previous studies have revealed that the DAT carboxyl terminus (DAT-CT) can directly interact with other cellular proteins and regulate DAT function and trafficking. Results Here, we have identified that carboxypeptidase E (CPE), a prohormone processing exopeptidase and sorting receptor for the regulated secretory pathway, interacts with the DAT-CT and affects DAT function. Mammalian cell lines coexpressing CPE and DAT exhibited increased DAT-mediated dopamine uptake activity compared to cells expressing DAT alone. Moreover, coexpression of an interfering DAT-CT minigene inhibited the effects of CPE on DAT. Functional changes caused by CPE could be attributed to enhanced DAT expression and subsequent increase in DAT cell surface localization, due to decreased DAT degradation. In addition, CPE association could reduce the phosphorylation state of DAT on serine residues, potentially leading to reduced internalization, thus stabilizing plasmalemmal DAT localization. Conclusion Taken together, our results reveal a novel role for CPE in the regulation of DAT trafficking and DAT-mediated DA uptake, which may provide a novel target in the treatment of dopamine-governed diseases such as drug addiction and obesity.
Collapse
Affiliation(s)
- Heping Zhang
- Department of Neuroscience, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.
| | | | | | | | | | | |
Collapse
|
107
|
Unfaithful neurotransmitter transporters: focus on serotonin uptake and implications for antidepressant efficacy. Pharmacol Ther 2008; 121:89-99. [PMID: 19022290 DOI: 10.1016/j.pharmthera.2008.10.004] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2008] [Accepted: 10/14/2008] [Indexed: 01/11/2023]
Abstract
Biogenic amine transporters for serotonin, norepinephrine and dopamine (SERT, NET and DAT respectively), are the key players terminating transmission of these amines in the central nervous system by their high-affinity uptake. They are also major targets for many antidepressant drugs. Interestingly however, drugs targeted to a specific transporter do not appear to be as clinically efficacious as those that block two or all three of these transporters. A growing body of literature, reviewed here, supports the idea that promiscuity among these transporters (the uptake of multiple amines in addition to their "native" transmitter) may account for improved therapeutic effects of dual and triple uptake blockers. However, even these drugs do not provide effective treatment outcomes for all individuals. An emerging literature suggests that "non-traditional" transporters such as organic cation transporters (OCT) and the plasma membrane monoamine transporter (PMAT) may contribute to the less than hoped for efficacy of currently prescribed uptake inhibitors. OCT and PMAT are capable of clearing biogenic amines from extracellular fluid and may serve to buffer the effects of frontline antidepressants, such as selective serotonin reuptake inhibitors. In addition, polymorphisms that occur in the genes encoding the transporters can lead to variation in transporter expression and function (e.g. the serotonin transporter linked polymorphic region; 5-HTTLPR) and can have profound effects on treatment outcome. This may be accounted for, in part, by compensatory adaptations in other transporters. This review synthesizes the existing literature, focusing on serotonin to illustrate and revive a model for the rationale design of improved antidepressants.
Collapse
|
108
|
The comparative distributions of the monoamine transporters in the rodent, monkey, and human amygdala. Brain Struct Funct 2008; 213:73-91. [PMID: 18283492 PMCID: PMC9741847 DOI: 10.1007/s00429-008-0176-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2007] [Accepted: 01/31/2008] [Indexed: 12/14/2022]
Abstract
The monoamines in the amygdala modulate multiple aspects of emotional processing in the mammalian brain, and organic or pharmacological dysregulation of these systems can result in affective pathologies. Knowledge of the normal distribution of these neurotransmitters, therefore, is central to our understanding of both the normal processes regulated by the amygdala and the pathological conditions associated with monoaminergic dysregulation. The monoaminergic transporters have proven to be accurate and reliable markers of the distributions of their substrates. The purpose of this review was twofold: First, to briefly recount the functional relevance of dopamine, serotonin, and norepinephrine transmission in the amygdala, and second, to describe and compare the distributions of the monoamine transporters in the rodent, monkey, and human brain. The transporters were found to be heterogeneously distributed in the amygdala. The dopamine transporter (DAT) is consistently found to be extremely sparsely distributed, however the various accounts of its subregional topography are inconsistent, making any cross-species comparisons difficult. The serotonin transporter (SERT) had the greatest overall degree of labeling of the three markers, and was characterized by substantial inter-species variability in its relative distribution. The norepinephrine transporter (NET) was shown to possess an intermediate level of labeling, and like the SERT, its distribution is not consistent across the three species. The results of these comparisons indicate that caution should be exercised when using animal models to investigate the complex processes modulated by the monoamines in the amygdala, as their relative contributions to these functions may differ across species.
Collapse
|
109
|
β-adrenergic receptors are differentially expressed in distinct interneuron subtypes in the rat hippocampus. J Comp Neurol 2008; 509:551-65. [DOI: 10.1002/cne.21758] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
110
|
Kim CH, Waldman ID, Blakely RD, Kim KS. Functional gene variation in the human norepinephrine transporter: association with attention deficit hyperactivity disorder. Ann N Y Acad Sci 2008; 1129:256-60. [PMID: 18591486 DOI: 10.1196/annals.1417.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The norepinephrine (NE) transporter (NET) is responsible for the re-uptake of NE into presynaptic nerve terminals, thus critically regulating noradrenergic signaling and homeostasis. Since NE signaling contributes to diverse brain functions, we hypothesize that promoter variation within the human NET gene (solute carrier family 6, member 2; SLC6A2) may impact risk for NE-related disorders, including depression, attention deficit hyperactive disorder (ADHD), and autonomic dysfunction. In support of this, we recently found a functional polymorphism at -3081 position upstream of the transcription initiation site. This polymorphism displayed differential promoter function, which we showed could arise from recruitment of a transcriptional repressor. Further analyses identified Slug and Scratch as candidates involved in repression of SLC6A2 transcription generated by the -3081(T) allele. Moreover, we observed a significant association of the -3081(T) variant with ADHD. Altered transcription of SLC6A2 may therefore represent a novel risk factor for the development of ADHD.
Collapse
Affiliation(s)
- Chun-Hyung Kim
- Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, 115 Mill St., Belmont, MA 02478, USA.
| | | | | | | |
Collapse
|
111
|
Hope SI, Schmipp J, Rossi AH, Bianciotti LG, Vatta MS. Regulation of the neuronal norepinephrine transporter by endothelin-1 and -3 in the rat anterior and posterior hypothalamus. Neurochem Int 2008; 53:207-13. [PMID: 18682267 DOI: 10.1016/j.neuint.2008.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2008] [Revised: 05/27/2008] [Accepted: 07/10/2008] [Indexed: 11/18/2022]
Abstract
We previously reported that endothelin-1 and endothelin-3 modulate norepinephrine neuronal release and tyrosine hydroxylase activity and expression in the hypothalamus. In the present study we sought to establish the role of endothelin-1 and -3 in the regulation of norepinephrine uptake in the anterior and posterior hypothalamus. Results showed that in the anterior hypothalamus endothelin-3 increased neuronal norepinephrine uptake whereas endothelin-1 decreased it. Conversely, in the posterior hypothalamic region both endothelins diminished the neuronal uptake of the amine. Endothelins response was concentration dependent and maintained at all studied times. Endothelins also modified the kinetic and internalization of the NE neuronal transporter. In the anterior hypothalamic region endothelin-3 increased the V(max) and the B(max) whereas endothelin-1 decreased them. However, in the posterior hypothalamic region both endothelins diminished the V(max) as well as B(max). Neither endothelin-1 nor endothelin-3 modified neuronal norepinephrine transporter K(d) in the studied hypothalamic regions. These findings support that in the posterior hypothalamic region both endothelins diminished neuronal norepinephrine transporter activity by reducing the amine transporter expression on the plasmatic membrane. Conversely, in the anterior hypothalamic region endothelin-3 enhanced neuronal norepinephrine transporter activity by increasing the expression of the transporter on the presynaptic membrane, whereas endothelin-1 induced the opposite effect. Present results permit us to conclude that both endothelins play an important role in the regulation of norepinephrine neurotransmission at the presynaptic nerve endings in the hypothalamus.
Collapse
Affiliation(s)
- Sandra I Hope
- Cátedra de Fisiología e Instituto de Química y Metabolismo del Fármaco, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
112
|
Aksenov MY, Aksenova MV, Silvers JM, Mactutus CF, Booze RM. Different effects of selective dopamine uptake inhibitors, GBR 12909 and WIN 35428, on HIV-1 Tat toxicity in rat fetal midbrain neurons. Neurotoxicology 2008; 29:971-7. [PMID: 18606182 DOI: 10.1016/j.neuro.2008.06.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2007] [Revised: 05/19/2008] [Accepted: 06/04/2008] [Indexed: 01/22/2023]
Abstract
Drug abuse is a risk factor for neurological complications in HIV infection. Cocaine has been shown to exacerbate HIV-associated brain pathology and enhance neurotoxicity of HIV-1 Tat and gp120 proteins. In this study, we found that the selective inhibitor of dopamine transporter (DAT) function, 1-[2-[bis(4-fluorophenyl) methoxy]ethyl]-4-(3-phenylpropyl) piperazine (GBR 12909, vanoxerine), but not the selective inhibitors of serotonin and norepinephrine (SERT and NET) transporters, sertraline and nizoxetine, emulated cocaine-mediated enhancement of Tat neurotoxicity in rat fetal midbrain primary cell cultures. Similar to cocaine, the significant increase of Tat toxicity in midbrain cell cultures was observed at micromolar dose (5microM) of GBR 12909. However, different doses of another selective dopamine uptake inhibitor, WIN 35428 did not affect Tat neurotoxicity. The study supports the hypothesis that changes in control of dopamine (DA) homeostasis are important for the cocaine-mediated enhancement of HIV-1 Tat neurotoxicity. Our results also demonstrate that inhibitors of DA uptake, which can bind to different domains of DAT, differ in their ability to mimic synergistic toxicity of cocaine and HIV-1 Tat in the midbrain cell culture.
Collapse
Affiliation(s)
- Michael Y Aksenov
- Program in Behavioral Neuroscience, University of South Carolina, USA.
| | | | | | | | | |
Collapse
|
113
|
Wehrwein EA, Parker LM, Wright AA, Spitsbergen JM, Novotny M, Babankova D, Swain GM, Habecker BA, Kreulen DL. Cardiac norepinephrine transporter protein expression is inversely correlated to chamber norepinephrine content. Am J Physiol Regul Integr Comp Physiol 2008; 295:R857-63. [PMID: 18565836 DOI: 10.1152/ajpregu.00190.2008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The cardiac neuronal norepinephrine (NE) transporter (NET) in sympathetic neurons is responsible for uptake of released NE from the neuroeffector junction. The purpose of this study was to assess the chamber distribution of cardiac NET protein measured using [(3)H]nisoxetine binding in rat heart membranes and to correlate NE content to NET amount. In whole mounts of atria, NET was colocalized in nerve fibers with tyrosine hydroxylase (TH) immunoreactivity. NE content expressed as micrograms NE per gram tissue was lowest in the ventricles; however, NET binding was significantly higher in the left ventricle than the right ventricle and atria (P < 0.05), resulting in a significant negative correlation (r(2) = 0.922; P < 0.05) of NET to NE content. The neurotoxin 6-hydroxydopamine, an NET substrate, reduced NE content more in the ventricles than the atria, demonstrating functional significance of high ventricular NET binding. In summary, there is a ventricular predominance of NET binding that corresponds to a high NE reuptake capacity in the ventricles, yet negatively correlates to tissue NE content.
Collapse
Affiliation(s)
- Erica A Wehrwein
- Dept. of Physiology, Michigan State Univ., East Lansing, MI 48823, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Mlinar B, Mascalchi S, Morini R, Giachi F, Corradetti R. MDMA induces EPSP-Spike potentiation in rat ventral hippocampus in vitro via serotonin and noradrenaline release and coactivation of 5-HT4 and beta1 receptors. Neuropsychopharmacology 2008; 33:1464-75. [PMID: 17653110 DOI: 10.1038/sj.npp.1301512] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
It is well documented that N-methyl-3,4-methylenedioxyamphetamine (MDMA, ecstasy) releases brain serotonin (5-HT; 5-hydroxytryptamine), noradrenaline (NE; norepinephrine), and dopamine, but the consequent effect on brain functioning remains elusive. In this study, we characterized the effects of MDMA on electrically evoked responses in the ventral CA1 region of a rat hippocampal slice preparation. Superfusion with MDMA (10 microM, 30 min) increased the population spike amplitude (PSA) by 48.9+/-31.2% and decreased population spike latency (PSL) by 103+/-139 mus (both: mean+/-SD, n=123; p<0.0001, Wilcoxon test), without affecting field excitatory postsynaptic potential (fEPSP). This effect persisted for at least 1 h after MDMA washout; we have called this EPSP-spike potentiation (ESP) by MDMA, ESP MDMA. Antagonism of GABAergic transmission did not prevent ESP MDMA, suggesting that an increase in excitability of pyramidal cells underlies this MDMA action. Block of serotonin transporter (SERT) with citalopram or 5-HT depletion with (+/-)-p-chlorophenylalanine pretreatment partially inhibited the ESP MDMA. Block of both SERT and NE transporter prevented ESP MDMA, indicating its dependence on release of both 5-HT and NE. ESP MDMA is produced by simultaneous activation of 5-HT4 and beta1 receptors, with a predominant role of 5-HT4 receptors. Block of both 5-HT4 and beta1 receptors revealed an inhibitory component of the MDMA action mediated by 5-HT1A receptor. The concentration range of MDMA which produced ESP MDMA (1-30 microM) corresponds to that commonly reached in human plasma following the ingestion of psychoactive MDMA doses, suggesting that release of both 5-HT and NE, and consequent ESP MDMA may underlie some of the psychoactive effects of MDMA in humans.
Collapse
Affiliation(s)
- Boris Mlinar
- Department of Preclinical and Clinical Pharmacology Mario Aiazzi-Mancini, University of Florence, Florence, Italy.
| | | | | | | | | |
Collapse
|
115
|
Bakuchiol analogs inhibit monoamine transporters and regulate monoaminergic functions. Biochem Pharmacol 2008; 75:1835-47. [PMID: 18329002 DOI: 10.1016/j.bcp.2008.01.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Revised: 01/28/2008] [Accepted: 01/28/2008] [Indexed: 11/24/2022]
Abstract
Monoamine transporters play key roles in controlling monoamine levels and modulating monoamine reuptake. The objective of the present study was to identify monoamine transporter inhibitors from herbal sources. We discovered that bakuchiol analogs isolated from Fructus Psoraleae inhibited monoamine transporter uptake to differing degrees. The bakuchiol analog, Delta3,2-hydroxybakuchiol was the most potent and efficacious reuptake blocker and was thus selected as the candidate target. Monoamine transporter inhibition by Delta3,2-hydroxybakuchiol was more selective for the dopamine transporter (DAT) (IC50=0.58+/-0.1 microM) and norepinephrine transporter (NET) (IC50=0.69+/-0.12 microM) than for the serotonin transporter (SERT) (IC50=312.02+/-56.69 microM). Delta3,2-Hydroxybakuchiol exhibited greater potency (pEC50 for DAT and NET) than bupropion and exhibited similar efficacy (E(max) for DAT and/or NET) to bupropion and GBR12,935. Pharmacokinetically, Delta3,2-hydroxybakuchiol competitively inhibited DAT and NET with partial reversibility and occupied cocaine binding sites. Moreover, Delta3,2-hydroxybakuchiol counteracted 1-methyl-4-phenylpyridinium-induced toxicity in cells expressing DAT with similar efficacy to GBR12,935. In vivo studies showed that Delta3,2-hydroxybakuchiol increased the activity of intact mice and improved the decreased activity of reserpinized mice. In the conditioned place preference test, preference scores in intact mice were unaffected by Delta3,2-hydroxybakuchiol treatment. Bakuchiol analogs, especially Delta3,2-hydroxybakuchiol, are monoamine transporter inhibitors involved in regulating dopaminergic and noradrenergic neurotransmission and may have represented potential pharmacotherapies for disorders such as Parkinson's disease, depression, and cocaine addiction.
Collapse
|
116
|
Moazedi A, Belaran M, Hemmaty A, Rasekh A. The Role of Beta-Adrenergic System on the Enhancement of Spatial Learning Caused by Glucose Injection in Young Male Rats. INT J PHARMACOL 2007. [DOI: 10.3923/ijp.2008.34.39] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
117
|
Amino T, Uchihara T, Tsunekawa H, Takahata K, Shimazu S, Mizusawa H, Orimo S. Myocardial nerve fibers are preserved in MPTP-treated mice, despite cardiac sympathetic dysfunction. Neurosci Res 2007; 60:314-8. [PMID: 18242750 DOI: 10.1016/j.neures.2007.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Revised: 11/25/2007] [Accepted: 11/28/2007] [Indexed: 10/22/2022]
Abstract
Recently, we reported a profound depletion of cardiac sympathetic nerve fibers in Parkinson's disease (PD). This cardiac sympathetic denervation is a characteristic hallmark of PD. Cardiac sympathetic dysfunction was also observed in 1-methyl-4-phenyl-1,2,3,6-tetrahydroxypyridine (MPTP)-treated mice, a model of PD. Although binding assay showed a decreased density of norepinephrine transporter (NET) in the hearts of the mice, their histopathological alterations have not been demonstrated. In this study, we investigated hearts of MPTP-treated mice with immunohistochemical method and Western blot analyses. MPTP-treated mice showed significant decreases in the contents of cardiac noradrenaline and dopamine, suggesting the sympathetic dysfunction. Synaptophysin-, tyrosine hydroxylase- or NET-immunoreactive nerve fibers were abundant in the hearts of control mice and MPTP-treated mice, without apparent differences between the two groups. Western blot analyses also showed no difference in the amounts of these proteins. Myocardial nerve fibers were well preserved in MPTP-treated mice, despite apparent cardiac sympathetic dysfunction.
Collapse
Affiliation(s)
- Takeshi Amino
- Department of Neurology, Tokyo Metropolitan Institute for Neuroscience, 2-6 Musashidai, Fuchu, Tokyo 183-8526, Japan
| | | | | | | | | | | | | |
Collapse
|
118
|
Norrholm SD, Horton DB, Dwoskin LP. The promiscuity of the dopamine transporter: implications for the kinetic analysis of [3H]serotonin uptake in rat hippocampal and striatal synaptosomes. Neuropharmacology 2007; 53:982-9. [PMID: 18022203 DOI: 10.1016/j.neuropharm.2007.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2007] [Revised: 08/23/2007] [Accepted: 10/01/2007] [Indexed: 11/29/2022]
Abstract
Evidence indicates that monoaminergic neurotransmitter transporters are promiscuous, transporting substrates other than their cognate neurotransmitters. For example, serotonin is transported by the dopamine transporter (DAT) under conditions in which serotonin transporter (SERT) activity is eliminated (e.g., pharmacological inhibition). We performed a kinetic analysis of [(3)H]serotonin uptake in rat striatal synaptosomes (expressing DAT and SERT) and hippocampal synaptosomes (expressing SERT, but not DAT). Nonspecific [(3)H]serotonin uptake was defined as the amount of uptake remaining in the presence of fluoxetine (10microM) or paroxetine (0.05microM). In hippocampal synaptosomes, K(m) and V(max) values for [(3)H]serotonin uptake did not differ whether fluoxetine or paroxetine was used to define nonspecific uptake. However, in striatal synaptosomes, both K(m) and V(max) values for [(3)H]serotonin uptake were greater when fluoxetine, rather than paroxetine, was used to define nonspecific uptake. These data suggest that, at the concentrations employed, fluoxetine inhibits serotonin uptake at both DAT and SERT, whereas paroxetine only inhibits serotonin uptake at SERT. Thus, when DAT is inhibited by GBR 12909, kinetic parameters for serotonin uptake via SERT in striatum are not different from those obtained in hippocampus. These findings have important implications regarding the analysis of monoaminergic reuptake in brain regions exhibiting heterogeneous transporter expression.
Collapse
Affiliation(s)
- Seth D Norrholm
- College of Pharmacy, Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536-0082, USA.
| | | | | |
Collapse
|
119
|
Diniz PHC, Silva JH, Gomez MV, Guatimosim C, Gomez RS. Halothane increases non-vesicular [(3)H]dopamine release from brain cortical slices. Cell Mol Neurobiol 2007; 27:757-70. [PMID: 17680357 DOI: 10.1007/s10571-007-9162-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2006] [Accepted: 06/11/2007] [Indexed: 10/23/2022]
Abstract
Experimental data suggest that halothane anesthesia is associated with significant changes in dopamine (DA) concentration in some brain regions but the mechanism of this effect is not well known. Rat brain cortical slices were labeled with [(3)H]DA to further characterize the effects of halothane on the release of this neurotransmitter from the central nervous system. Halothane induced an increase on the release of [(3)H]DA that was dependent on incubation time and anesthetic concentration (0.012, 0.024, 0.048, 0.072 and 0.096 mM). This effect was independent of extracellular or intracellular calcium. In addition, [(3)H]DA release evoked by halothane was not affected by TTX (blocker of voltage-dependent Na(+) channels) or reserpine (a blocker of vesicular monoamine transporter). These data suggest that [(3)H]DA release induced by halothane is non-vesicular and would be mediated by the dopamine transporter (DAT) and norepinephrine transporter (NET). GBR 12909 and nomifensine, inhibitors of DAT, decreased the release of [(3)H]DA evoked by halothane. Nisoxetine, a blocker of NET, reduced the release of [(3)H]DA induced by halothane. In addition, GBR 12909, nisoxetine and, halothane decrease the uptake of [(3)H]DA into rat brain cortical slices. A decrease on halothane-induced release of [(3)H]DA was also observed when the brain cortical slices were incubated at low temperature and low extracellular sodium, which are known to interfere with the carrier-mediated release of the neurotransmitter. Ouabain, a Na(+)/K(+) ATPase pump inhibitor, which induces DA release through reverse transport, decreased [(3)H]DA release induced by halothane. It is suggested that halothane increases [(3)H]DA release in brain cortical slices that is mediated by DAT and NET present in the plasma membrane.
Collapse
Affiliation(s)
- Paulo H C Diniz
- Department of Pharmacology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | | |
Collapse
|
120
|
Zhu J, Bardo MT, Bruntz RC, Stairs DJ, Dwoskin LP. Individual differences in response to novelty predict prefrontal cortex dopamine transporter function and cell surface expression. Eur J Neurosci 2007; 26:717-28. [PMID: 17651428 DOI: 10.1111/j.1460-9568.2007.05690.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Previous research has shown that individual differences in response to novelty predict self-administration and the locomotor response to psychostimulant drugs of abuse. The aim of the present study was to determine if individual differences in response to novelty based on inescapable or free-choice novelty tests predict dopamine transporter (DAT) function and trafficking as well as nicotine-induced modulation of DAT. Results show that the maximal velocity (Vmax) of [3H]dopamine uptake into prefrontal cortex (PFC) synaptosomes correlated negatively with the locomotor response to inescapable novelty. In contrast, Vmax correlated positively with novelty place preference in the free-choice novelty test. The divergent correlations between DAT and the two behavioral phenotypes suggest a differential contribution of DAT in these phenotypes, which are known not to be isomorphic. Furthermore, rats categorized as high responders to inescapable novelty had lower Vmax values, which were accompanied by less DAT expression at the cell surface in PFC compared with low responders, suggesting that inherent individual differences in DAT cellular localization may underlie the differential response to novelty. Compared with the saline control, nicotine increased Vmax and cell surface DAT immunoreactivity in PFC from high responders but not from low responders. Similarly, nicotine increased Vmax and cell surface DAT in PFC in rats classified as low in novelty place preference but not in rats classified as high in novelty place preference. Thus, despite the different behavioral phenotypes, the pharmacological effect of nicotine to increase DAT function and cell surface expression was apparent, such that rats with inherently lower DAT function show a greater sensitivity to the neurochemical effect of nicotine.
Collapse
Affiliation(s)
- Jun Zhu
- College of Pharmacy, University of Kentucky, 725 Rose Street, Lexington, KY 40536-0082, USA
| | | | | | | | | |
Collapse
|
121
|
Tong J, Hornykiewicz O, Furukawa Y, Kish SJ. Marked dissociation between high noradrenaline versus low noradrenaline transporter levels in human nucleus accumbens. J Neurochem 2007; 102:1691-1702. [PMID: 17484728 DOI: 10.1111/j.1471-4159.2007.04636.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We recently identified a noradrenaline-rich caudomedial subdivision of the human nucleus accumbens (NACS), implying a special function for noradrenaline in this basal forebrain area involved in motivation and reward. To establish whether the NACS, as would be expected, contains similarly high levels of other noradrenergic markers, we measured dopamine-beta-hydroxylase (DBH) and noradrenaline transporter in the accumbens and, for comparison, in 23 other brain regions in autopsied human brains by immunoblotting. Although the caudomedial NACS had high DBH levels similar to those in other noradrenaline-rich areas, the noradrenaline transporter concentration was low (only 11% of that in hypothalamus). Within the accumbens, transporter concentration in the caudal portion was only slightly (by 30%) higher than that in the rostral subdivisions despite sharply increasing rostrocaudal gradients of noradrenaline (15-fold) and DBH. In contrast, the rostrocaudal gradient in the accumbens for the serotonin transporter and serotonin were similar (2-fold increase). The caudomedial NACS thus appears to represent the only instance in human brain having a striking mismatch in high levels of a monoamine neurotransmitter versus low levels of its uptake transporter. This suggests that noradrenaline signalling is much less spatially and temporally restricted in the caudomedial accumbens than in other noradrenaline-rich brain areas.
Collapse
Affiliation(s)
- Junchao Tong
- Human Neurochemical Pathology Laboratory, Centre for Addiction and Mental Health, Toronto, Ontario, CanadaCenter for Brain Research, Medical University of Vienna, Spitalgasse, Vienna, AustriaMovement Disorders Research Laboratory, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Oleh Hornykiewicz
- Human Neurochemical Pathology Laboratory, Centre for Addiction and Mental Health, Toronto, Ontario, CanadaCenter for Brain Research, Medical University of Vienna, Spitalgasse, Vienna, AustriaMovement Disorders Research Laboratory, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Yoshiaki Furukawa
- Human Neurochemical Pathology Laboratory, Centre for Addiction and Mental Health, Toronto, Ontario, CanadaCenter for Brain Research, Medical University of Vienna, Spitalgasse, Vienna, AustriaMovement Disorders Research Laboratory, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Stephen J Kish
- Human Neurochemical Pathology Laboratory, Centre for Addiction and Mental Health, Toronto, Ontario, CanadaCenter for Brain Research, Medical University of Vienna, Spitalgasse, Vienna, AustriaMovement Disorders Research Laboratory, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| |
Collapse
|
122
|
Paczkowski FA, Sharpe IA, Dutertre S, Lewis RJ. chi-Conotoxin and tricyclic antidepressant interactions at the norepinephrine transporter define a new transporter model. J Biol Chem 2007; 282:17837-44. [PMID: 17428804 DOI: 10.1074/jbc.m610813200] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Monoamine neurotransmitter transporters for norepinephrine (NE), dopamine and serotonin are important targets for antidepressants and analgesics. The conopeptide chi-MrIA is a noncompetitive and highly selective inhibitor of the NE transporter (NET) and is being developed as a novel intrathecal analgesic. We used site-directed mutagenesis to generate a suite of mutated transporters to identify two amino acids (Leu(469) and Glu(382)) that affected the affinity of chi-MrIA to inhibit [(3)H]NE uptake through human NET. Residues that increased the K(d) of a tricyclic antidepressant (nisoxetine) were also identified (Phe(207), Ser(225), His(296), Thr(381), and Asp(473)). Phe(207), Ser(225), His(296), and Thr(381) also affected the rate of NE transport without affecting NE K(m). In a new model of NET constructed from the bLeuT crystal structure, chi-MrIA-interacting residues were located at the mouth of the transporter near residues affecting the binding of small molecule inhibitors.
Collapse
Affiliation(s)
- Filip A Paczkowski
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | | | | | | |
Collapse
|
123
|
Guo NN, Li BM. Cellular and subcellular distributions of beta1- and beta2-adrenoceptors in the CA1 and CA3 regions of the rat hippocampus. Neuroscience 2007; 146:298-305. [PMID: 17337326 DOI: 10.1016/j.neuroscience.2007.01.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2006] [Accepted: 01/08/2007] [Indexed: 11/16/2022]
Abstract
Beta-adrenoceptors (ARs) in the hippocampus play an important role in regulating synaptic plasticity and memory consolidation. However, little is known about the distributions of beta-ARs in the hippocampus, especially in the cornu ammonis (CA)1 and CA3 regions of Sprague-Dawley rats. Here, we report that beta1- and beta2-ARs in the CA1 and CA3 regions have differential subcellular distributions. Using double immunofluorescence labeling and confocal laser scanning microscopy, we found that almost all of the neuronal nuclei positive cells express beta1- and beta2-ARs, while few glial fibrillary acidic protein positive cells express them. Interestingly, beta1-ARs are predominantly distributed in the cell membrane and cytoplasm, whereas beta2-ARs are predominantly distributed not only in the membrane and cytoplasm, but also in the nucleus. The differential subcellular distribution of beta1- and beta2-ARs may have functional significance.
Collapse
Affiliation(s)
- N-N Guo
- Laboratory of Higher Brain Functions, Institute of Neurobiology, Institutes of Brain Science, Fudan University, 138 Yi Xue Yuan Road, Shanghai 200032, China
| | | |
Collapse
|
124
|
Dvoryanchikov G, Tomchik SM, Chaudhari N. Biogenic amine synthesis and uptake in rodent taste buds. J Comp Neurol 2007; 505:302-13. [PMID: 17879273 DOI: 10.1002/cne.21494] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Although adenosine triphosphate (ATP) is known to be an afferent transmitter in the peripheral taste system, serotonin (5-HT) and norepinephrine (NE) have also been proposed as candidate neurotransmitters and have been detected immunocytochemically in mammalian taste cells. To understand the significance of biogenic amines in taste, we evaluated the ability of taste cells to synthesize, transport, and package 5-HT and NE. We show by reverse transcriptase-polymerase chain reaction and immunofluorescence microscopy that the enzymes for 5-HT synthesis, tryptophan hydroxylase (TPH) and aromatic amino acid decarboxylase (AADC) are expressed in taste cells. In contrast, enzymes necessary for NE synthesis, tyrosine hydroxylase (TH) and dopamine beta-hydroxylase (DBH) are absent. Both TH and DBH are expressed in nerve fibers that penetrate taste buds. Taste buds also robustly express plasma membrane transporters for 5-HT and NE. Within the taste bud NET, a specific NE transporter, is expressed in some presynaptic (type III) and some glial-like (type I) cells but not in receptor (type II) cells. By using enzyme immunoassay, we show uptake of NE, probably through NET in taste epithelium. Proteins involved in inactivating and packaging NE, including catechol-O-methyltransferase (COMT), monoamine oxidase-A (MAO-A), vesicular monoamine transporter (VMAT1,2) and chromogranin A (ChrgA), are also expressed in taste buds. Within the taste bud, ChrgA is found only in presynaptic cells and may account for dense-cored vesicles previously seen in some taste cells. In summary, we postulate that aminergic presynaptic taste cells synthesize only 5-HT, whereas NE (perhaps secreted by sympathetic fibers) may be concentrated and repackaged for secretion.
Collapse
Affiliation(s)
- Gennady Dvoryanchikov
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | | | | |
Collapse
|
125
|
Wersinger C, Jeannotte A, Sidhu A. Attenuation of the norepinephrine transporter activity and trafficking via interactions with α-synuclein. Eur J Neurosci 2006; 24:3141-52. [PMID: 17156375 DOI: 10.1111/j.1460-9568.2006.05181.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Alpha-synuclein (alpha-Syn) has been studied in the context of Parkinson's disease, but its normative role remains elusive. We have shown that alpha-Syn regulates the homeostasis of dopaminergic and serotonergic synapses, through trafficking of the dopamine and serotonin transporter, respectively. In the present study we sought to determine if alpha-Syn could also modulate noradrenergic signaling, by studying its interactions with the norepinephrine transporter (NET). We co-transfected Ltk- cells with increasing amounts of alpha-Syn DNA and a constant amount of NET DNA, and observed a progressive decrease (68%) in [3H]-NE uptake in cells co-transfected with a ratio of 3:1 alpha-Syn:NET DNA. The Kd of transport did not change, but increasing alpha-Syn caused a decrease in the Vmax of the transporter, from 2.27+/-0.14 to 0.89+/-0.15 pmol/min/10(5) cells, with NET expression alone or 4:1 ratio of alpha-Syn:NET transfection, respectively. Decreases in surface biotinylation and [3H]-nisoxetine binding kinetics in intact cells revealed that NET cell surface expression was attenuated in correlation to the amount of alpha-Syn co-transfected into cells. The interaction between NET and alpha-Syn occurred via the NAC domain of alpha-Syn, the region directly responsible for self-aggregation. These findings are the first to show that alpha-Syn has a central role in the homeostasis of noradrenergic neurons. Together with our previous studies on dopamine and serotonin transporters, we propose that a primary physiological role of alpha-Syn may be to regulate the homeostasis of monoamines in synapses, through modulatory interactions of the protein with monoaminergic transporters.
Collapse
Affiliation(s)
- Christophe Wersinger
- Department of Biochemistry, Molecular and Cell Biology, Georgetown University, Washington, DC 20007, USA
| | | | | |
Collapse
|
126
|
McKittrick CR, Abercrombie ED. Catecholamine mapping within nucleus accumbens: differences in basal and amphetamine-stimulated efflux of norepinephrine and dopamine in shell and core. J Neurochem 2006; 100:1247-56. [PMID: 17241132 DOI: 10.1111/j.1471-4159.2006.04300.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The nucleus accumbens is believed to play a critical role in mediating the behavioral responses to rewarding stimuli. Although most studies of the accumbens focus on dopamine, it receives afferents from many other nuclei, including noradrenergic cell groups in the brainstem. We used in vivo microdialysis to measure extracellular levels of both norepinephrine and dopamine in the accumbens shell and core. Regional analysis of shell and core and border regions demonstrated that norepinephrine was high in shell and decreased from medial shell to lateral core, where baseline levels were low or undetectable. Conversely, extracellular dopamine in core was twice the level seen in shell. Both catecholamines increased following a single injection of amphetamine (2 mg/kg, i.p.). The norepinephrine response was greater and long-lasting in shell compared with core. The maximal dopamine response was higher in core than in shell, but the duration of the effect was comparable in both regions. The distinct neurochemical characteristics of shell and core are likely to contribute to the functional heterogeneity of the two subregions. Furthermore, norepinephrine may be involved in many of the functions generally attributed to the accumbens, either directly or indirectly via modulation of extracellular dopamine.
Collapse
Affiliation(s)
- Christina R McKittrick
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, New Jersey, USA.
| | | |
Collapse
|
127
|
Yamashita M, Fukushima S, Shen HW, Hall FS, Uhl GR, Numachi Y, Kobayashi H, Sora I. Norepinephrine transporter blockade can normalize the prepulse inhibition deficits found in dopamine transporter knockout mice. Neuropsychopharmacology 2006; 31:2132-9. [PMID: 16407898 DOI: 10.1038/sj.npp.1301009] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dopamine transporter knockout (DAT KO) mice display deficits in sensorimotor gating that are manifested by reduced prepulse inhibition (PPI) of the acoustic startle reflex. Since PPI deficits may model some of the cognitive dysfunctions identified in certain neuropsychiatric patients, we have studied the effects of transporter blockers on PPI in wild-type and DAT KO mice. Treatments with High dose psychostimulants that block DAT as well as the norepinephrine (NET) and serotonin (SERT) transporters (60 mg/kg cocaine or methylphenidate) significantly impaired PPI in wild-type mice. By contrast, these treatments significantly ameliorated the PPI deficits observed in untreated DAT KO mice. In studies with more selective transport inhibitors, the selective NET inhibitor nisoxetine (10 or 30 mg/kg) also significantly reversed PPI deficits in DAT KO mice. By contrast, while the SERT inhibitor fluoxetine (30 mg/kg) normalized these PPI deficits in DAT KO mice, citalopram (30 or 100 mg/kg) failed to do so. The 'paradoxical' effects of cocaine and methylphenidate in DAT KO mice are thus likely to be mediated, at least in part by the ability of these drugs to block NET, although serotonin systems may also have some role. Together with recent microdialysis data, these results support the hypothesis that prefrontal cortical NET blockade and consequent enhancement of prefrontal cortical extracellular dopamine mediates the reversal of PPI deficits in DAT KO mice.
Collapse
Affiliation(s)
- Motoyasu Yamashita
- Department of Psychobiology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | | | | | | | | | | | | | | |
Collapse
|
128
|
Abstract
The norepinephrine transporter (NET) terminates noradrenergic signalling by rapid re-uptake of neuronally released norepinephrine (NE) into presynaptic terminals. NET exerts a fine regulated control over NE-mediated behavioural and physiological effects including mood, depression, feeding behaviour, cognition, regulation of blood pressure and heart rate. NET is a target of several drugs which are therapeutically used in the treatment or diagnosis of disorders among which depression, attention-deficit hyperactivity disorder and feeding disturbances are the most common. Individual genetic variations in the gene encoding the human NET (hNET), located at chromosome 16q12.2, may contribute to the pathogenesis of those diseases. An increasing number of studies concerning the identification of single nucleotide polymorphisms in the hNET gene and their potential association with disease as well as the functional investigation of naturally occurring or induced amino acid variations in hNET have contributed to a better understanding of NET function, regulation and genetic contribution to disorders. This review will reflect the current knowledge in the field of NET from its initial discovery until now.
Collapse
Affiliation(s)
- H Bönisch
- Department of Pharmacology and Toxicology, University of Bonn, Reuterstr. 2b, 53115 Bonn, Germany.
| | | |
Collapse
|
129
|
Belfield JL, Whittaker C, Cader MZ, Chawla S. Differential effects of Ca2+ and cAMP on transcription mediated by MEF2D and cAMP-response element-binding protein in hippocampal neurons. J Biol Chem 2006; 281:27724-32. [PMID: 16870618 DOI: 10.1074/jbc.m601485200] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In neurons, the second messengers Ca(2+) and cAMP are mediators of transcriptional responses that are important for the development and function of the nervous system. The pro-survival neuronal transcription factors cAMP-response elementbinding protein (CREB) and myocyte enhancer factor-2 (MEF2) both stimulate gene expression in response to activity-dependent increases in the concentration of intracellular Ca(2+) ions. CREB is also activated by increases in intracellular cAMP. Here we have investigated whether the MEF2 family member MEF2D, similar to CREB, is also activated by cAMP in hippocampal neurons. We have shown that, unlike CREB, MEF2D is not activated by agents that increase intracellular cAMP. Moreover, increases in cAMP inhibit Ca(2+)-activated MEF2D-mediated gene expression. We have also shown that cAMP inhibits Ca(2+)-induced nuclear export of the MEF2 co-repressor HDAC5 and prevents Ca(2+)-stimulated nuclear import of the MEF2 co-activator NFAT3/c4. Our results suggest that cAMP interferes with MEF2D-mediated gene expression at multiple levels by antagonizing the derepression of MEF2D by HDAC5 and by inhibiting recruitment of the co-activator NFAT.
Collapse
Affiliation(s)
- Johanna L Belfield
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom
| | | | | | | |
Collapse
|
130
|
Abstract
For many years, the norepinephrine transporter (NET) was considered a 'static' protein that contributed to the termination of the action of norepinephrine in the synapse of noradrenergic neurons. The concept that the NET is dynamically regulated, adjusting noradrenergic transmission by changing its function and/or expression, was considered initially in the mid 1980s. Since that time, a plethora of studies demonstrate that the NET is regulated by several intracellular and extracellular signaling molecules, and that phosphorylation of the NET is a major pathway regulating its cell surface expression and thereby its function. The NET is a target of action of a number of drugs that are used long-term therapeutically or abused chronically. This has driven numerous investigations of how the NET and its function are regulated by long-term exposure to drugs. While repeated exposure to many drugs has been shown to affect NET function and expression, the intracellular mechanisms for these effects remains elusive.
Collapse
Affiliation(s)
- Prashant Mandela
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, USA
| | | |
Collapse
|
131
|
Binda F, Lute BJ, Dipace C, Blakely RD, Galli A. The N-terminus of the norepinephrine transporter regulates the magnitude and selectivity of the transporter-associated leak current. Neuropharmacology 2006; 50:354-61. [PMID: 16289633 DOI: 10.1016/j.neuropharm.2005.09.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2005] [Revised: 09/23/2005] [Accepted: 09/27/2005] [Indexed: 10/25/2022]
Abstract
The norepinephrine (NE) transporter (NET) mediates the removal of NE from synaptic spaces and is a major target for antidepressants, amphetamine and cocaine. Previously, we have shown that syntaxin 1A (SYN 1A) supports human NET (hNET) cell surface expression, that hNET/SYN 1A interactions are direct and mediated by the hNET N-terminus, and that the hNET/SYN 1A association limits substrate-induced hNET-associated currents [Sung, U., Apparsundaram, S., Galli, A., Kahlig, K.M., Savchenko, V., Schroeter, S., Quick, M.W., Blakely, R.D., 2003. A regulated interaction of syntaxin 1A with the antidepressant-sensitive norepinephrine transporter establishes catecholamine clearance capacity. J. Neurosci. 23, 1697-1709]. These data raise the possibility that the hNET N-terminus, and potentially its interaction with SYN 1A, might regulate other hNET conductance states, including the hNET-mediated leak current. Importantly for monoamine transporters, the leak conductance has been shown to play a critical role in regulating cell membrane potential and possibly neuronal excitability [Quick, M.W., 2003. Regulating the conducting states of a mammalian serotonin transporter. Neuron 40, 537-549]. Here we demonstrate that deletion of the binding domain for SYN 1A in the NET N-terminus robustly enhances the NET-mediated leak current as well as its selectivity for Cl- permeation under particular intracellular ionic compositions. In addition, we show that the NET N-terminus coordinates the ability of intracellular Na+ and Cl- to regulate the leak conductance. These data suggest that the NET N-terminus regulates and defines the ionic specificity of the NET-mediated leak current.
Collapse
Affiliation(s)
- Francesca Binda
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 465 21st Ave. South, Nashville, TN 37232-8548, USA
| | | | | | | | | |
Collapse
|
132
|
Amano T, Aoki S, Setsuie R, Sakurai M, Wada K, Noda M. Identification of a novel regulatory mechanism for norepinephrine transporter activity by the IP3 receptor. Eur J Pharmacol 2006; 536:62-8. [PMID: 16554048 DOI: 10.1016/j.ejphar.2006.02.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2005] [Revised: 01/27/2006] [Accepted: 02/21/2006] [Indexed: 10/25/2022]
Abstract
The norepinephrine transporter (NET) plays a crucial role in noradrenergic neurotransmission and is a target of many antidepressants and psychostimulants. Intracellular Ca2+ is reportedly involved in regulating NET activity, but the detailed mechanism is not clear. We employed a norepinephrine uptake assay using SH-SY5Y cells and found that the IP3 receptor inhibitors, 2-aminoethoxydiphenyl borate and xestospongin C, reduced the NET Vmax. These reductions were accompanied by the decreased cell surface expression of NET. Our findings suggest that intracellular Ca2+ mobilized by IP3 receptor is required for the maintenance of NET activity. This adds another pathway involving Ca2+ for the regulation of NET to other known mechanisms providing intracellular Ca2+.
Collapse
Affiliation(s)
- Taiju Amano
- Laboratory of Pathophysiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | | | | | | | | | | |
Collapse
|
133
|
Myers EA, Banihashemi L, Rinaman L. The anxiogenic drug yohimbine activates central viscerosensory circuits in rats. J Comp Neurol 2006; 492:426-41. [PMID: 16228990 DOI: 10.1002/cne.20727] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Systemic administration of the alpha(2)-adrenoceptor antagonist yohimbine (YO) activates the HPA stress axis and promotes anxiety in humans and experimental animals. We propose that visceral malaise contributes to the stressful and anxiogenic effects of systemic YO and that YO recruits brainstem noradrenergic (NA) and peptidergic neurons that relay viscerosensory signals to the hypothalamus and limbic forebrain. To begin testing these hypotheses, the present study explored dose-related effects of YO on food intake, conditioned flavor avoidance (CFA), and Fos immunolabeling in rats. Systemic YO (5.0 mg/kg BW, i.p.) inhibited food intake, supported CFA, and increased Fos immunolabeling in identified NA neurons in the ventrolateral medulla, nucleus of the solitary tract, and locus coeruleus. YO also increased Fos in the majority of corticotropin releasing hormone-positive neurons in the paraventricular nucleus of the hypothalamus. YO administered at 1.0 mg/kg BW did not inhibit food intake, did not support CFA, and did not increase Fos immunolabeling. Retrograde neural tracing demonstrated that neurons activated by YO at 5.0 mg/kg BW included medullary and pontine neurons that project to the central nucleus of the amygdala and to the lateral bed nucleus of the stria terminalis, the latter region receiving comparatively greater input by Fos-positive neurons. We conclude that YO produces anorexigenic and aversive effects that correlate with activation of brainstem viscerosensory inputs to the limbic forebrain. These findings invite continued investigation of how central viscerosensory signaling pathways interact with hypothalamic and limbic regions to influence interrelated physiological and behavioral components of anxiety, stress, and visceral malaise.
Collapse
|
134
|
Schwartz JW, Piston D, DeFelice LJ. Molecular microfluorometry: converting arbitrary fluorescence units into absolute molecular concentrations to study binding kinetics and stoichiometry in transporters. Handb Exp Pharmacol 2006:23-57. [PMID: 16722229 DOI: 10.1007/3-540-29784-7_2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Cotransporters use energy stored in Na+ or H+ gradients to transport neurotransmitters or other substrates against their own gradient. Cotransport is rapid and efficient, and at synapses it helps terminate signaling. Cotransport in norepinephrine (NET), epinephrine (EpiT), dopamine (DAT), and serotonin (SERT) transporters couples downhill Na+ flux to uphill transmitter flux. NETs, for example, attenuate signaling at adrenergic synapses by efficiently clearing NE from the synaptic cleft, thus preparing the synapse for the next signal. Transport inhibition with tricyclic antidepressants prolongs neurotransmitter presence in the synaptic cleft, potentially alleviating symptoms of depression. Transport inhibition with cocaine or amphetamine, which respectively block or replace normal transport, may result in hyperactivity. Little is known about the kinetic interactions of substrates or drugs with transporters, largely because the techniques that have been successful in discovering trans- porter agonists and antagonists do not yield detailed kinetic information. Mechanistic data are for the most part restricted to global parameters, such as Km and Vmax, measured from large populations of transporter molecules averaged over thousands of cells. Three relatively new techniques used in transporter research are electrophysiology, amperometry, and microfluorometry. This review focuses on fluorescence-based methodologies, which--unlike any other technique-permit the simultaneous measurement of binding and transport. Microfluorometry provides unique insights into binding kinetics and transport mechanisms from a quantitative analysis of fluorescence data. Here we demonstrate how to quantify the number of bound substrate molecules, the number of transported substrate molecules, and the kinetics of substrate binding to individual transporters. Although we describe experiments on a specific neurotransmitter transporter, these methods are applicable to other membrane proteins.
Collapse
Affiliation(s)
- J W Schwartz
- Imaging Center, Stowers Institute for Medical Research, 1000 E 50th St., Kansas City, MO 64110, USA
| | | | | |
Collapse
|
135
|
Smith HR, Beveridge TJR, Porrino LJ. Distribution of norepinephrine transporters in the non-human primate brain. Neuroscience 2006; 138:703-14. [PMID: 16427744 DOI: 10.1016/j.neuroscience.2005.11.033] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2005] [Revised: 11/03/2005] [Accepted: 11/20/2005] [Indexed: 11/23/2022]
Abstract
Noradrenergic terminals in the central nervous system are widespread; as such this system plays a role in varying functions such as stress responses, sympathetic regulation, attention, and memory processing, and its dysregulation has been linked to several pathologies. In particular, the norepinephrine transporter is a target in the brain of many therapeutic and abused drugs. We used the selective ligand [(3)H]nisoxetine, therefore, to describe autoradiographically the normal regional distribution of the norepinephrine transporter in the non-human primate central nervous system, thereby providing a baseline to which alterations due to pathological conditions can be compared. The norepinephrine transporter in the monkey brain was distributed heterogeneously, with highest levels occurring in the locus coeruleus complex and raphe nuclei, and moderate binding density in the hypothalamus, midline thalamic nuclei, bed nucleus of the stria terminalis, central nucleus of the amygdala, and brainstem nuclei such as the dorsal motor nucleus of the vagus and nucleus of the solitary tract. Low levels of binding to the norepinephrine transporter were measured in basolateral amygdala and cortical, hippocampal, and striatal regions. The distribution of the norepinephrine transporter in the non-human primate brain was comparable overall to that described in other species, however disparities exist between the rodent and the monkey in brain regions that play a role in such critical processes as memory and learning. The differences in such areas point to the possibility of important functional differences in noradrenergic information processing across species, and suggest the use of caution in applying findings made in the rodent to the human condition.
Collapse
Affiliation(s)
- H R Smith
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | | | | |
Collapse
|
136
|
Keller NR, Diedrich A, Appalsamy M, Miller LC, Caron MG, McDonald MP, Shelton RC, Blakely RD, Robertson D. Norepinephrine transporter-deficient mice respond to anxiety producing and fearful environments with bradycardia and hypotension. Neuroscience 2006; 139:931-46. [PMID: 16515844 DOI: 10.1016/j.neuroscience.2006.01.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2005] [Revised: 11/18/2005] [Accepted: 01/15/2006] [Indexed: 10/24/2022]
Abstract
The study of anxiety and fear involves complex interrelationships between psychiatry and the autonomic nervous system. Altered noradrenergic signaling is linked to certain types of depression and anxiety disorders, and treatment often includes specific transporter blockade. The norepinephrine transporter is crucial in limiting catecholaminergic signaling. Norepinephrine transporter-deficient mice have increased circulating catecholamines and elevated heart rate and blood pressure. We hypothesized, therefore, that reduced norepinephrine clearance would heighten the autonomic cardiovascular response to anxiety and fear. In separate experiments, norepinephrine transporter-deficient (norepinephrine transporter-/-) mice underwent tactile startle and trace fear conditioning to measure hemodynamic responses. A dramatic tachycardia was observed in norepinephrine transporter-/- mice compared with controls following both airpuff or footshock stimuli, and pressure changes were also greater. Interestingly, in contrast to normally elevated home cage levels in norepinephrine transporter-deficient mice, prestimulus heart rate and blood pressure were actually higher in norepinephrine transporter+/+ animals throughout behavioral testing. Upon placement in the behavioral chamber, norepinephrine transporter-deficient mice demonstrated a notable bradycardia and depressor effect that was more pronounced in females. Power spectral analysis indicated an increase in low frequency oscillations of heart rate variability; in mice, suggesting increased parasympathetic tone. Finally, norepinephrine transporter-/- mice exhibited sexual dimorphism in freeze behavior, which was greatest in females. Therefore, while reduced catecholamine clearance amplifies immediate cardiovascular responses to anxiety- or fear-inducing stimuli in norepinephrine transporter-/- mice, norepinephrine transporter deficiency apparently prevents protracted hemodynamic escalation in a fearful environment. Conceivably, chronic norepinephrine transporter blockade with transporter-specific drugs might attenuate recognition of autonomic and somatic distress signals in individuals with anxiety disorders, possibly lessening their behavioral reactivity, and reducing the cardiovascular risk factors associated with persistent emotional arousal.
Collapse
Affiliation(s)
- N R Keller
- Autonomic Dysfunction Center, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, AA3228 MCN, Nashville, TN 37232-2195, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Hillman KL, Knudson CA, Carr PA, Doze VA, Porter JE. Adrenergic receptor characterization of CA1 hippocampal neurons using real time single cell RT-PCR. ACTA ACUST UNITED AC 2005; 139:267-76. [PMID: 16005103 DOI: 10.1016/j.molbrainres.2005.05.033] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2004] [Revised: 05/14/2005] [Accepted: 05/24/2005] [Indexed: 11/17/2022]
Abstract
The CA1 region of the rat hippocampus exhibits both alpha and beta adrenergic receptor (AR) responses, however, the specific AR subtypes involved and the neuronal expression patterns for these receptors are not well understood. We have employed single cell real time RT-PCR in conjunction with cell-specific immunohistochemical markers to determine the AR expression patterns for hippocampal neurons located in CA1, a region often implicated in learning and memory processes. Cytoplasmic samples were taken from 55 individual cells located in stratum oriens, pyramidale, or radiatum and reverse transcribed. All successfully amplified pyramidal neuron samples (n = 17) expressed mRNA for the beta2AR, with four cells additionally expressing mRNA for the beta1AR subtype. Positive interneurons from stratum oriens (n = 10) and stratum radiatum (n = 8) expressed mRNA for the alpha1A and/or alpha(1B)AR (n = 9/18) only when coexpressing transcripts for somatostatin. Interneurons containing neuropeptide Y or cholecystokinin (n = 9/18) were not positive for any of the nine AR subtypes, suggesting that CA1 interneuron AR expression is limited to a subset of somatostatin-positive cells. These findings suggest that only a select number of AR subtypes are transcriptionally expressed in CA1 and that these receptors are selective to specific neuronal cell types.
Collapse
Affiliation(s)
- Kristin L Hillman
- Department of Pharmacology, Physiology, and Therapeutics, School of Medicine and Health Sciences, University of North Dakota, 501 North Columbia Road, Grand Forks, ND 58203, USA
| | | | | | | | | |
Collapse
|
138
|
Huynh TT, Pacak K, Brouwers FM, Abu-Asab MS, Worrell RA, Walther MM, Elkahloun AG, Goldstein DS, Cleary S, Eisenhofer G. Different expression of catecholamine transporters in phaeochromocytomas from patients with von Hippel-Lindau syndrome and multiple endocrine neoplasia type 2. Eur J Endocrinol 2005; 153:551-63. [PMID: 16189177 PMCID: PMC2288736 DOI: 10.1530/eje.1.01987] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Phaeochromocytomas in patients with multiple endocrine neoplasia type 2 (MEN 2) produce adrenaline, whereas those with von Hippel-Lindau (VHL) syndrome do not. This study assessed whether these distinctions relate to differences in expression of the transporters responsible for uptake and storage of catecholamines - the noradrenaline transporter and the vesicular monoamine transporters (VMAT 1 and VMAT 2). METHODS Tumour tissue and plasma samples were obtained from 31 patients with hereditary phaeochromocytoma - 18 with VHL syndrome and 13 with MEN 2. We used quantitative PCR, Western blotting, electron microscopy, immunohistochemistry and measurements of plasma and tumour catecholamines to assess differences in expression of the transporters in noradrenaline-producing vs adrenaline-producing hereditary tumours. These differences were compared with those in a further group of 26 patients with non-syndromic phaeochromocytoma. RESULTS Adrenaline-producing phaeochromocytomas in MEN 2 patients expressed more noradrenaline transporter mRNA and protein than noradrenaline-producing tumours in VHL patients. In contrast, there was greater expression of VMAT 1 in VHL than MEN 2 tumours, while expression of VMAT 2 did not differ significantly. These differences were associated with larger numbers of storage vesicles and higher tissue contents of catecholamines in MEN 2 than in VHL tumours. Differences in expression of the noradrenaline transporter were weaker, and those of VMAT 1 and VMAT 2 stronger, in noradrenaline and adrenaline-producing non-syndromic than in hereditary tumours. CONCLUSIONS The findings show that, in addition to differences in catecholamine biosynthesis, phaeochromocytomas in MEN 2 and VHL syndrome also differ in expression of the transporters responsible for uptake and vesicular storage of catecholamines.
Collapse
Affiliation(s)
- Thanh-Truc Huynh
- Clinical Neurocardiology Section, National Institute of Neurological Disorders and Stroke, Reproductive Biology and Medicine Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Zhu MY, Wang WP, Iyo AH, Ordway GA, Kim KS. Age-associated changes in mRNA levels of Phox2, norepinephrine transporter and dopamine beta-hydroxylase in the locus coeruleus and adrenal glands of rats. J Neurochem 2005; 94:828-38. [PMID: 16033425 PMCID: PMC2923405 DOI: 10.1111/j.1471-4159.2005.03245.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Age-related changes in the gene expression of the transcription factors, Phox2a and 2b, and two marker proteins, norepinephrine transporter (NET) and dopamine beta-hydroxylase (DBH), of noradrenergic neurons were characterized in the locus coeruleus (LC) and adrenal glands using in situ hybridization. Analysis of changes was performed in rats that were 1-23 months of age. Compared to 1-month-old rats, there was a 62% increase of Phox2a messenger RNA (mRNA) in the LC of 3-month-old rats, and a decline of 37% in 23-month-old rats. In contrast, levels of Phox2b mRNA in the LC remained unchanged in 3-month-old rats, but declined to a 30% reduction in 23-month-old rats. Interestingly, mRNA levels of NET in the LC decreased with increasing age to a reduction of 29%, 30% and 43% in 3-, 8- and 23-month-old rats, respectively. Similarly, DBH mRNA in the LC declined with increasing age to a 56% reduction in 23-month-old rats. mRNA levels of Phox2a, Phox2b, NET and DBH in the adrenal medulla of 23-month-old rats were significantly lower than those of 1-month-old rats. Semi-quantitative reverse transcription assays of the same genes yielded data similar to in situ hybridization experiments, with beta-actin mRNA levels being unchanged across the ages. Taken together, these data reveal that reduced Phox2 mRNAs in the LC and adrenal medulla of aging rats are accompanied by a coincidental decline in mRNA levels of NET and DBH and suggest a possible relationship between Phox2 genes and the marker genes in noradrenergic neurons after birth.
Collapse
Affiliation(s)
- Meng-Yang Zhu
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA.
| | | | | | | | | |
Collapse
|
140
|
Jurgens CWD, Boese SJ, King JD, Pyle SJ, Porter JE, Doze VA. Adrenergic receptor modulation of hippocampal CA3 network activity. Epilepsy Res 2005; 66:117-28. [PMID: 16140503 DOI: 10.1016/j.eplepsyres.2005.07.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2005] [Revised: 07/14/2005] [Accepted: 07/27/2005] [Indexed: 10/25/2022]
Abstract
Norepinephrine (NE) has demonstrated proconvulsant and antiepileptic properties; however, the specific pharmacology of these actions has not been clearly established. To address this, we studied the effect of NE on hippocampal CA3 epileptiform activity. Frequency changes of burst discharges in response to NE were biphasic; low concentrations increased the number of bursts, while higher concentrations reduced their frequency, suggesting the involvement of multiple adrenergic receptor (AR) types. This hypothesis was confirmed when, in the presence of betaAR blockade, increasing concentrations of NE caused a monophasic decrease in epileptiform activity. Antagonists selective for alpha1 or alpha2ARs were then used to determine which alphaAR type was involved. While discriminating concentrations of the alpha1AR antagonists prazosin and terazosin had no effect, selective amounts of the alpha2AR antagonists RS79948 and RX821002 significantly reduced the potency of NE in decreasing epileptiform activity. Furthermore, this antiepileptic action of NE persisted when all GABA-mediated inhibition was blocked. This data suggests that, under conditions of impaired GABAergic inhibition, the excitatory and inhibitory effects of NE on hippocampal CA3 epileptiform activity are mediated primarily via beta and alpha2ARs, respectively. Moreover, our results imply that the antiepileptic effect of alpha2AR activation in CA3 is not dependent on the GABAergic system.
Collapse
Affiliation(s)
- Chris W D Jurgens
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota School of Medicine and Health Sciences, 501 N. Columbia Road, Grand Forks, ND 58203, USA
| | | | | | | | | | | |
Collapse
|
141
|
Sung U, Jennings JL, Link AJ, Blakely RD. Proteomic analysis of human norepinephrine transporter complexes reveals associations with protein phosphatase 2A anchoring subunit and 14-3-3 proteins. Biochem Biophys Res Commun 2005; 333:671-8. [PMID: 15963952 DOI: 10.1016/j.bbrc.2005.05.165] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2005] [Accepted: 05/25/2005] [Indexed: 11/29/2022]
Abstract
The norepinephrine transporter (NET) terminates noradrenergic signals by clearing released NE at synapses. NET regulation by receptors and intracellular signaling pathways is supported by a growing list of associated proteins including syntaxin1A, protein phosphatase 2A (PP2A) catalytic subunit (PP2A-C), PICK1, and Hic-5. In the present study, we sought evidence for additional partnerships by mass spectrometry-based analysis of proteins co-immunoprecipitated with human NET (hNET) stably expressed in a mouse noradrenergic neuroblastoma cell line. Our initial proteomic analyses reveal multiple peptides derived from hNET, peptides arising from the mouse PP2A anchoring subunit (PP2A-Ar) and peptides derived from 14-3-3 proteins. We verified physical association of NET with PP2A-Ar via co-immunoprecipitation studies using mouse vas deferens extracts and with 14-3-3 via a fusion pull-down approach, implicating specifically the hNET NH2-terminus for interactions. The transporter complexes described likely support mechanisms regulating transporter activity, localization, and trafficking.
Collapse
Affiliation(s)
- Uhna Sung
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232-8548, USA
| | | | | | | |
Collapse
|
142
|
Cleary S, Brouwers FM, Eisenhofer G, Pacak K, Christie DL, Lipski J, McNeil AR, Phillips JK. Expression of the noradrenaline transporter and phenylethanolamine N-methyltransferase in normal human adrenal gland and phaeochromocytoma. Cell Tissue Res 2005; 322:443-53. [PMID: 16047163 DOI: 10.1007/s00441-005-0026-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2005] [Accepted: 05/30/2005] [Indexed: 11/30/2022]
Abstract
Expression of the noradrenaline transporter (NAT) was examined in normal human adrenal medulla and phaeochromocytoma by using immunohistochemistry and confocal microscopy. The enzymes tyrosine hydroxylase (TH) and phenylethanolamine N-methyltransferase (PNMT) were used as catecholamine biosynthetic markers and chromogranin A (CGA) as a marker for secretory granules. Catecholamine content was measured by using high performance liquid chromatography (HPLC). In normal human adrenal medulla (n=5), all chromaffin cells demonstrated strong TH, PNMT and NAT immunoreactivity. NAT was co-localized with PNMT and was located within the cytoplasm with a punctate appearance. Human phaeochromocytomas demonstrated strong TH expression (n=20 samples tested) but variable NAT and PNMT expression (n=24). NAT immunoreactivity ranged from absent (n=3) to weak (n=10) and strong (n=11) and, in some cases, occupied an apparent nuclear location. Unlike the expression seen in normal human adrenal medullary tissue, NAT expression was not consistently co-localized with PNMT. PNMT also showed highly variable expression that was poorly correlated with tumour adrenaline content. Immunoreactivity for CGA was colocalized with NAT within the cytoplasm of normal human chromaffin cells (n=4). This co-localization was not consistent in phaeochromocytoma tumour cells (n=7). The altered pattern of expression for both NAT and PNMT in phaeochromocytoma indicates a significant disruption in the regulation and possibly in the function of these proteins in adrenal medullary tumours.
Collapse
Affiliation(s)
- Susannah Cleary
- Division of Health Science, Murdoch University, Perth, Western Australia, 6150, Australia
| | | | | | | | | | | | | | | |
Collapse
|
143
|
Dawson PA, Steane SE, Markovich D. Impaired memory and olfactory performance in NaSi-1 sulphate transporter deficient mice. Behav Brain Res 2005; 159:15-20. [PMID: 15794992 DOI: 10.1016/j.bbr.2004.09.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2004] [Accepted: 09/29/2004] [Indexed: 10/26/2022]
Abstract
In the present study, NaSi-1 sulphate transporter knock-out (Nas1-/-) mice, an animal model of hyposulphataemia, were examined for spatial memory and learning in a Morris water maze, and for olfactory function in a cookie test. The Nas1-/- mice displayed significantly (P<0.05) increased latencies to find an escape platform in the reversal learning trials at 2 days but not 1 day after the last acquisition trial in a Morris water maze test, suggesting that Nas1-/- mice may have proactive memory interference. While the wild-type (Nas1+/+) mice showed a significant (P<0.02) decrease in time to locate a hidden food reward over four trials after overnight fasting, Nas1-/- mice did not change their performance, resulting in significantly (P<0.05) higher latencies when compared to their Nas1+/+ littermates. There were no significant differences between Nas1-/- and Nas1+/+ mice in the cookie test after moderate food deprivation. In addition, both Nas1-/- and Nas1+/+ mice displayed similar escape latencies in the acquisition phase of the Morris water maze test, suggesting that learning, motivation, vision and motor skills required for the task may not be affected in Nas1-/- mice. This is the first study to demonstrate an impairment in memory and olfactory performance in the hyposulphataemic Nas1-/- mouse.
Collapse
Affiliation(s)
- Paul Anthony Dawson
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Qld. 4072, Australia
| | | | | |
Collapse
|
144
|
Jurgens CWD, Rau KE, Knudson CA, King JD, Carr PA, Porter JE, Doze VA. Beta1 adrenergic receptor-mediated enhancement of hippocampal CA3 network activity. J Pharmacol Exp Ther 2005; 314:552-60. [PMID: 15908512 DOI: 10.1124/jpet.105.085332] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Norepinephrine is an endogenous neurotransmitter distributed throughout the mammalian brain. In higher cortical structures such as the hippocampus, norepinephrine, via beta adrenergic receptor (AR) activation, has been shown to reinforce the cognitive processes of attention and memory. In this study, we investigated the effect of beta1AR activation on hippocampal cornu ammonis 3 (CA3) network activity. AR expression was first determined using immunocytochemistry with antibodies against beta1ARs, which were found to be exceptionally dense in hippocampal CA3 pyramidal neurons. CA3 network activity was then examined in vitro using field potential recordings in rat brain slices. The selective betaAR agonist isoproterenol caused an enhancement of hippocampal CA3 network activity, as measured by an increase in frequency of spontaneous burst discharges recorded in the CA3 region. In the presence of alphaAR blockade, concentration-response curves for isoproterenol, norepinephrine, and epinephrine suggested that a beta1AR was involved in this response, and the rank order of potency was isoproterenol > norepinephrine = epinephrine. Finally, equilibrium dissociation constants (pK(b)) of subtype-selective betaAR antagonists were functionally determined to characterize the AR subtype modulating hippocampal CA3 activity. The selective beta1AR antagonists atenolol and metoprolol blocked isoproterenol-induced enhancement, with apparent K(b) values of 85 +/- 36 and 3.9 +/- 1.7 nM, respectively. In contrast, the selective beta2AR antagonists ICI-118,551 and butoxamine inhibited isoproterenol-mediated enhancement with apparent low affinities (K(b) of 222 +/- 61 and 9268 +/- 512 nM, respectively). Together, this pharmacological profile of subtype-selective betaAR antagonists indicates that in this model, beta1AR activation is responsible for the enhanced hippocampal CA3 network activity initiated by isoproterenol.
Collapse
Affiliation(s)
- Chris W D Jurgens
- Department of Pharmacology, Physiology and Therapeutics, School of Medicine and Health Sciences, University of North Dakota, 501 North Columbia Rd., Grand Forks, ND 58202-9037, USA
| | | | | | | | | | | | | |
Collapse
|
145
|
Gobert A, Billiras R, Cistarelli L, Millan MJ. Quantification and pharmacological characterization of dialysate levels of noradrenaline in the striatum of freely-moving rats: release from adrenergic terminals and modulation by alpha2-autoreceptors. J Neurosci Methods 2005; 140:141-52. [PMID: 15589344 DOI: 10.1016/j.jneumeth.2004.04.040] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2003] [Accepted: 04/28/2004] [Indexed: 10/26/2022]
Abstract
Information concerning striatal levels of noradrenaline (NA) remains inconsistent. Here we have addressed this issue using a sensitive method of HPLC coupled to amperometric detection. The NA reuptake-inhibitor, reboxetine, selectively elevated levels of NA versus dopamine (DA), and NA levels were also selectively elevated by the alpha2-adrenoceptor (AR) antagonist, atipamezole. The actions of atipamezole were mimicked by the preferential alpha2A-AR antagonist, BRL44408, while JO-1 and prazosin, preferential antagonists at alpha2C-ARs, caused less marked elevations in NA levels. In contrast to antagonists, the alpha2-AR agonist, S18616, decreased NA levels and likewise suppressed those of DA. Unilateral lesions of the substantia nigra with 6-hydroxydopamine depleted DA levels without affecting those of NA. Further, the D3/D2 receptor agonist, quinelorane, decreased levels of DA without modifying those of NA. However, the D3/D2 receptor antagonists, haloperidol and raclopride, and the DA reuptake-inhibitor, GBR12935, elevated levels of both DA and NA. Levels of 5-HT (but not of NA or DA) were increased only by the 5-HT reuptake-inhibitor, citalopram. They were decreased by S18616 and prazosin, reflecting the inhibitory and excitatory influence of alpha2- and alpha1-ARs, respectively, upon serotonergic pathways. In conclusion, NA in the striatum is derived from adrenergic terminals. Its release is subject to tonic, inhibitory control by alpha2-ARs, possibly involving both alpha2A- and alpha2C-AR subtypes, though their respective contribution requires clarification. A role of dopaminergic terminals in the reuptake of NA likely explains the elevation in its levels elicited by DA reuptake-inhibitors and D3/D2 receptor antagonists.
Collapse
Affiliation(s)
- Alain Gobert
- Psychopharmacology Department, Institut de Recherches Servier, Centre de Recherches de Croissy, 125 chemin de Ronde, Croissy/Seine 78290, France.
| | | | | | | |
Collapse
|
146
|
Mash DC, Ouyang Q, Qin Y, Pablo J. Norepinephrine transporter immunoblotting and radioligand binding in cocaine abusers. J Neurosci Methods 2005; 143:79-85. [PMID: 15763139 DOI: 10.1016/j.jneumeth.2004.09.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The norepinephrine transporter (NET) is a membrane protein responsible for transporting extracellular norepinephrine. The cocaine and tricyclic antidepressant-sensitive NET belongs to a family of sodium and chloride coupled transporters that include the monoamines dopamine and serotonin and the amino acids GABA and glycine. The regional distribution of the NET has been defined by synaptosomal uptake of norepinephrine and by autoradiographic approaches in rodent and primate brain. However, the NET has not been well characterized in the human brain due to the overall low abundance of protein expressed in axon terminals. Recently, immunolocalization studies have been used to identify the regional distribution of the cytoplasmic NET epitope in rodent brain. We report here on the characteristics of drug interactions with the native NET protein in human postmortem brain. Antisera raised against a 17-amino acid peptide from the N-terminus of the hNET recognized an 80 kDa species in human cerebral cortex. Chronic exposure to cocaine upregulated NET protein expression and [3H]nisoxetine binding sites in the insular cortex from brains of cocaine addicts. These results demonstrate that immunologic and radioligand binding approaches afford specific labeling of the native transport protein in postmortem human brain.
Collapse
Affiliation(s)
- Deborah C Mash
- Department of Neurology (D4-5), School of Medicine, University of Miami, 1501 N.W. 9th Avenue, Miami, FL 33136, USA.
| | | | | | | |
Collapse
|
147
|
Daws LC, Montañez S, Owens WA, Gould GG, Frazer A, Toney GM, Gerhardt GA. Transport mechanisms governing serotonin clearance in vivo revealed by high-speed chronoamperometry. J Neurosci Methods 2005; 143:49-62. [PMID: 15763136 DOI: 10.1016/j.jneumeth.2004.09.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
High-speed chronoamperometry was used to determine the kinetics of clearance of exogenously applied serotonin (5-HT) in the dorsal raphe nucleus (DRN), dentate gyrus, CA3 region of the hippocampus or corpus callosum of anesthetized rats. Maximal velocity (Vmax) for 5-HT clearance was greatest in the DRN > dentate gyrus > CA3 > corpus callosum. Apparent affinity (K(T)) of the serotonin transporter (5-HTT) was similar in DRN and CA3 but greater in dentate gyrus and corpus callosum. A 90% loss of norepinephrine transporters (NET) produced by 6-hydroxydopamine pretreatment, resulted in a two-fold reduction in Vmax and a 30% decrease in K(T) in the dentate gyrus, but no change in kinetic parameters in the CA3 region. Pretreatment with 5,7-dihydroxytryptamine that resulted in a 90% reduction in 5-HTT density, modestly reduced Vmax in dentate gyrus but not in CA3. The same treatment had no effect on K(T) in the dentate gyrus but increased K(T) two-fold in the CA3. Neurotoxin treatments had no effect on 5-HT clearance in the corpus callosum. In hippocampal regions of intact rats, local application of the selective serotonin reuptake inhibitor, fluvoxamine, inhibited 5-HT clearance most robustly when the extracellular concentration of 5-HT was less than the K(T) value. By contrast, the NET antagonist, desipramine, significantly inhibited 5-HT clearance when extracellular concentrations of 5-HT were greater than the K(T) value. These data indicate that hippocampal uptake of 5-HT may be mediated by two processes, one with high affinity but low capacity (i.e. the 5-HTT) and the other with low affinity but a high capacity (i.e. the NET). These data show for the first time in the whole animal that 5-HT clearance in brain is regionally distinct with regard to rate and affinity.
Collapse
Affiliation(s)
- Lynette C Daws
- Department of Psychiatry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900, USA.
| | | | | | | | | | | | | |
Collapse
|
148
|
Gale SD, Perkel DJ. Properties of dopamine release and uptake in the songbird basal ganglia. J Neurophysiol 2004; 93:1871-9. [PMID: 15548618 DOI: 10.1152/jn.01053.2004] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Vocal learning in songbirds requires a basal ganglia circuit termed the anterior forebrain pathway (AFP). The AFP is not required for song production, and its role in song learning is not well understood. Like the mammalian striatum, the striatal component of the AFP, Area X, receives dense dopaminergic innervation from the midbrain. Since dopamine (DA) clearly plays a crucial role in basal ganglia-mediated motor control and learning in mammals, it seems likely that DA signaling contributes importantly to the functions of Area X as well. In this study, we used voltammetric methods to detect subsecond changes in extracellular DA concentration to gain better understanding of the properties and regulation of DA release and uptake in Area X. We electrically stimulated Ca(2+)- and action potential-dependent release of an electroactive substance in Area X brain slices and identified the substance as DA by the voltammetric waveform, electrode selectivity, and neurochemical and pharmacological evidence. As in the mammalian striatum, DA release in Area X is depressed by autoinhibition, and the lifetime of extracellular DA is strongly constrained by monoamine transporters. These results add to the known physiological similarities of the mammalian and songbird striatum and support further use of voltammetry in songbirds to investigate the role of basal ganglia DA in motor learning.
Collapse
Affiliation(s)
- Samuel D Gale
- Graduate Program in Neurobiology and Behavior, University of Washington, Seattle, WA 98195, USA.
| | | |
Collapse
|
149
|
Beveridge TJR, Smith HR, Nader MA, Porrino LJ. Functional effects of cocaine self-administration in primate brain regions regulating cardiovascular function. Neurosci Lett 2004; 370:201-5. [PMID: 15488323 DOI: 10.1016/j.neulet.2004.08.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2004] [Revised: 08/11/2004] [Accepted: 08/11/2004] [Indexed: 11/23/2022]
Abstract
Cocaine abuse is associated with autonomic dysregulation, such as altered blood pressure and heart rate. Both central and peripheral mechanisms have been implicated in mediating these changes, however, to date, no study has examined functional changes in activity within central autonomic-associated brain regions in response to cocaine in non-human primates. The aim of the present study was to measure local cerebral glucose utilization, in selected autonomic brain regions, in rhesus monkeys that had self-administered cocaine (0.3 mg/kg/infusion) for 5 days (initial) or 100 days (chronic). Measurements were compared with control monkeys, in which responding was maintained by food reinforcement. In general, decreased rates of glucose utilization were observed in targeted areas following both 5 and 100 days of cocaine self-administration compared to control values. However, after initial stages of cocaine exposure, significant reductions in the forebrain were restricted to the bed nucleus of stria terminalis and in the brainstem to the nucleus tractus solitarius and dorsomotor nucleus of the vagus nerve. The pattern of significantly altered functional activity induced by chronic 100-day cocaine self-administration extended within the forebrain to include the paraventricular hypothalamic nucleus, and in the brainstem to include additional autonomic-related nuclei, the nucleus ambiguus and locus coeruleus. These results suggest that even at the initial stages of cocaine self-administration, functional changes in activity occur in autonomic and reward-related brain regions. These alterations progress with prolonged cocaine exposure, and therefore may be involved in mediating changes in central autonomic control and the neuroadaptations reported to result from chronic drug abuse.
Collapse
Affiliation(s)
- Thomas J R Beveridge
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston Salem, NC 27157, USA.
| | | | | | | |
Collapse
|
150
|
Keller NR, Diedrich A, Appalsamy M, Tuntrakool S, Lonce S, Finney C, Caron MG, Robertson D. Norepinephrine transporter-deficient mice exhibit excessive tachycardia and elevated blood pressure with wakefulness and activity. Circulation 2004; 110:1191-6. [PMID: 15337696 DOI: 10.1161/01.cir.0000141804.90845.e6] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Norepinephrine (NE) is a primary neurotransmitter of central autonomic regulation and sympathetic nerve conduction, and the norepinephrine transporter (NET) is crucial in limiting catecholaminergic signaling. NET is sensitive to antidepressants, cocaine, and amphetamine. NET blockade often is associated with cardiovascular side effects, and NET deficiency is linked to tachycardia in familial orthostatic intolerance. METHODS AND RESULTS We telemetrically monitored NET-deficient (NET(-/-)) mice to determine the cardiovascular effects of reduced NE reuptake. Mean arterial pressure was elevated in resting NET(-/-) mice compared with NET(+/+) controls (103+/-0.6 versus 99+/-0.4 mm Hg; P<0.01), and corresponding pressures increased to 122+/-0.3 and 116+/-0.3 mm Hg (P<0.0001) with activity. Heart rate was also greater in resting NET(-/-) mice (565+/-5 versus 551+/-3 bpm; P<0.05), and genotypic differences were highly significant during the active phase (640+/-5 versus 607+/-3 bpm; P<0.0001). Conversely, the respiratory rate of resting NET(-/-) mice was dramatically reduced, whereas increases after the day/night shift surpassed those of controls. Plasma catecholamines in NET(-/-) and NET(+/+) mice were as follows: NE, 69+/-8 and 32+/-7; dihydroxyphenylglycol, 2+0.4 and 17+/-3; epinephrine, 15+/-3 and 4+/-0.6; and dopamine, 13+/-4 and 4+/-1 pmol/mL. Catechols in urine, brain, and heart also were determined. CONCLUSIONS Resting mean arterial pressure and heart rate are maintained at nearly normal levels in NET-deficient mice, most likely as a result of increased central sympathoinhibition. However, sympathetic activation with wakefulness and activity apparently overwhelms central modulation, amplifying peripheral catecholaminergic signaling, particularly in the heart.
Collapse
Affiliation(s)
- Nancy R Keller
- Autonomic Dysfunction Center, Department of Medicine, Nashville, TN 37232-2195, USA
| | | | | | | | | | | | | | | |
Collapse
|