1
|
Scholl JL, Rogers JT, Feng N, Forster GL, Watt MJ, Yaeger JD, Buchanan MW, Lowry CA, Renner KJ. Corticosterone rapidly modulates dorsomedial hypothalamus serotonin and behavior in an estrogen- and progesterone-dependent manner in adult female rats: potential role of organic cation transporter 3 (OCT3). Stress 2025; 28:2457765. [PMID: 39898528 PMCID: PMC11801257 DOI: 10.1080/10253890.2025.2457765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 01/16/2025] [Indexed: 02/04/2025] Open
Abstract
Previous studies have shown that corticosterone rapidly alters extracellular serotonin (5-hydroxytryptamine; 5-HT) concentrations in the dorsomedial hypothalamus (DMH) of adult male rats, suggesting a role for corticosterone actions in the DMH in regulation of physiological and behavioral responses. Whether or not corticosterone also rapidly alters extracellular serotonin concentrations in the DMH of female rats, and the dependence of this effect on ovarian hormones, is not known. To determine the effects of 17β-estradiol (E2), progesterone (P), and corticosterone on extracellular concentrations of serotonin in the DMH, corticosterone and/or P were delivered into the DMH of ovariectomized rats via reverse microdialysis in E2-primed rats. Combined, but not separate, delivery of corticosterone and P into the DMH rapidly and transiently increased extracellular 5-HT concentrations, a result that was dependent upon circulating E2. This effect of corticosterone on DMH 5-HT was replicated by local perfusion of the organic cation transporter 3 (OCT3) competitive inhibitor normetanephrine. Intra-DMH infusions of either corticosterone or normetanephrine also reversibly suppressed lordosis responses in E2 + P-primed females. These results suggest that ovarian hormones in combination with corticosterone modulate OCT3-mediated 5-HT clearance in the DMH, potentially representing an adaptive mechanism that allows sexually receptive females to respond rapidly to acute stressors.
Collapse
Affiliation(s)
- Jamie L. Scholl
- Basic Biomedical Sciences & Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, 414 E. Clark St., Vermillion, SD 57069, USA
| | - Joshua T. Rogers
- Department of Biology & Center for Brain and Behavior Research, University of South Dakota, 414 E. Clark St. Vermillion, SD 57069, USA
| | - Na Feng
- Department of Biology & Center for Brain and Behavior Research, University of South Dakota, 414 E. Clark St. Vermillion, SD 57069, USA
| | - Gina L. Forster
- Basic Biomedical Sciences & Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, 414 E. Clark St., Vermillion, SD 57069, USA
- Department of Anatomy, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Michael J. Watt
- Basic Biomedical Sciences & Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, 414 E. Clark St., Vermillion, SD 57069, USA
- Department of Anatomy, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Jazmine D.W. Yaeger
- Department of Biology & Center for Brain and Behavior Research, University of South Dakota, 414 E. Clark St. Vermillion, SD 57069, USA
| | - Michael W. Buchanan
- Department of Biology & Center for Brain and Behavior Research, University of South Dakota, 414 E. Clark St. Vermillion, SD 57069, USA
| | - Christopher A. Lowry
- Department of Integrative Physiology, Department of Psychology and Neuroscience, Center for Neuroscience, and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Kenneth J. Renner
- Basic Biomedical Sciences & Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, 414 E. Clark St., Vermillion, SD 57069, USA
- Department of Biology & Center for Brain and Behavior Research, University of South Dakota, 414 E. Clark St. Vermillion, SD 57069, USA
| |
Collapse
|
2
|
Honan LE, Fraser-Spears R, Daws LC. Organic cation transporters in psychiatric and substance use disorders. Pharmacol Ther 2024; 253:108574. [PMID: 38072333 PMCID: PMC11052553 DOI: 10.1016/j.pharmthera.2023.108574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/01/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
Psychiatric and substance use disorders inflict major public health burdens worldwide. Their widespread burden is compounded by a dearth of effective treatments, underscoring a dire need to uncover novel therapeutic targets. In this review, we summarize the literature implicating organic cation transporters (OCTs), including three subtypes of OCTs (OCT1, OCT2, and OCT3) and the plasma membrane monoamine transporter (PMAT), in the neurobiology of psychiatric and substance use disorders with an emphasis on mood and anxiety disorders, alcohol use disorder, and psychostimulant use disorder. OCTs transport monoamines with a low affinity but high capacity, situating them to play a central role in regulating monoamine homeostasis. Preclinical evidence discussed here suggests that OCTs may serve as promising targets for treatment of psychiatric and substance use disorders and encourage future research into their therapeutic potential.
Collapse
Affiliation(s)
- Lauren E Honan
- The University of Texas Health Science Center at San Antonio, Department of Cellular & Integrative Physiology, USA
| | - Rheaclare Fraser-Spears
- University of the Incarnate Word, Feik School of Pharmacy, Department of Pharmaceutical Sciences, USA
| | - Lynette C Daws
- The University of Texas Health Science Center at San Antonio, Department of Cellular & Integrative Physiology, USA; The University of Texas Health Science Center at San Antonio, Department of Pharmacology, USA.
| |
Collapse
|
3
|
Andrews PW, Bosyj C, Brenton L, Green L, Gasser PJ, Lowry CA, Pickel VM. All the brain's a stage for serotonin: the forgotten story of serotonin diffusion across cell membranes. Proc Biol Sci 2022; 289:20221565. [PMID: 36321487 PMCID: PMC9627707 DOI: 10.1098/rspb.2022.1565] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/10/2022] [Indexed: 11/05/2022] Open
Abstract
In the conventional model of serotonin neurotransmission, serotonin released by neurons in the midbrain raphe nuclei exerts its actions on forebrain neurons by interacting with a large family of post-synaptic receptors. The actions of serotonin are terminated by active transport of serotonin back into the releasing neuron, which is mediated by the serotonin reuptake transporter (SERT). Because SERT is expressed pre-synaptically and is widely thought to be the only serotonin transporter in the forebrain, the conventional model does not include serotonin transport into post-synaptic neurons. However, a large body of evidence accumulating since the 1970s has shown that serotonin, despite having a positive charge, can cross cell membranes through a diffusion-like process. Multiple low-affinity, high-capacity, sodium-independent transporters, widely expressed in the brain, allow the carrier-mediated diffusion of serotonin into forebrain neurons. The amount of serotonin crossing cell membranes through this mechanism under physiological conditions is considerable. Most prominent textbooks fail to include this alternative method of serotonin uptake in the brain, and even most neuroscientists are unaware of it. This failure has limited our understanding of a key regulator of serotonergic neurotransmission, impeded research on the potential intracellular actions of serotonin in post-synaptic neurons and glial cells, and may have impeded our understanding of the mechanism by which antidepressant medications reduce depressive symptoms.
Collapse
Affiliation(s)
- Paul W. Andrews
- Department of Psychology, Neuroscience and Behaviour, McMaster University, Hamilton, Ontario, Canada
| | - Catherine Bosyj
- Department of Psychology, Neuroscience and Behaviour, McMaster University, Hamilton, Ontario, Canada
| | - Luke Brenton
- Department of Psychology, Neuroscience and Behaviour, McMaster University, Hamilton, Ontario, Canada
| | - Laura Green
- Neuroscience Institute, New York University, New York, NY, USA
| | - Paul J. Gasser
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Christopher A. Lowry
- Department of Integrative Physiology, Center for Neuroscience, and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, USA
| | - Virginia M. Pickel
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
4
|
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are the most commonly prescribed medications for psychiatric disorders, yet they leave the majority of patients without full symptom relief. Therefore, a major research challenge is to identify novel targets for the improved treatment of these disorders. SSRIs act by blocking the serotonin transporter (SERT), the high-affinity, low-capacity, uptake-1 transporter for serotonin. Other classes of antidepressant work by blocking the norepinephrine or dopamine transporters (NET and DAT), the high-affinity, low-capacity uptake-1 transporters for norepinephrine and dopamine, or by blocking combinations of SERT, NET, and DAT. It has been proposed that uptake-2 transporters, which include organic cation transporters (OCTs) and the plasma membrane monoamine transporter (PMAT), undermine the therapeutic utility of uptake-1 acting antidepressants. Uptake-2 transporters for monoamines have low affinity for these neurotransmitters, but a high capacity to transport them. Thus, activity of these transporters may limit the increase of extracellular monoamines thought to be essential for ultimate therapeutic benefit. Here preclinical evidence supporting a role for OCT2, OCT3, and PMAT in behaviors relevant to psychiatric disorders is presented. Importantly, preclinical evidence revealing these transporters as targets for the development of novel therapeutics for psychiatric disorders is discussed.
Collapse
|
5
|
Abstract
Neuromodulators are critical regulators of the brain's signaling processes, and thus they are popular pharmacological targets for psychoactive therapies. It is clear that monoamine uptake mechanisms are complicated and subject to multiple uptake mechanisms. Uptake 1 describes uptake of the monoamine via its designated transporter (SERT for serotonin, NET for norepinephrine, and DAT for dopamine), whereas Uptake 2 details multiple transporter types on neurons and glia taking up different types of modulators, not necessarily specific to the monoamine. While Uptake 1 processes have been well-studied over the past few decades, Uptake 2 mechanisms have remained more difficult to study because of the limitations in methods that have the sensitivity and spatiotemporal resolution to look at the subtleties in uptake profiles. In this chapter we review the different experimental approaches that have yielded important information about Uptake 2 mechanisms in vivo. The techniques (scintillation microspectrophotometry, microdialysis, chronoamperometry, and voltammetry) are described in detail, and pivotal studies associated with each method are highlighted. It is clear from these reviewed works that Uptake 2 processes are critical to consider to advance our understanding of the brain and develop effective neuropsychiatric therapies.
Collapse
|
6
|
Abstract
Doxorubicin is a commonly used anticancer agent that can cause debilitating and irreversible cardiac injury. The initiating mechanisms contributing to this side effect remain unknown, and current preventative strategies offer only modest protection. Using stem-cell-derived cardiomyocytes from patients receiving doxorubicin, we probed the transcriptomic landscape of solute carriers and identified organic cation transporter 3 (OCT3) (SLC22A3) as a critical transporter regulating the cardiac accumulation of doxorubicin. Functional validation studies in heterologous overexpression models confirmed that doxorubicin is transported into cardiomyocytes by OCT3 and that deficiency of OCT3 protected mice from acute and chronic doxorubicin-related changes in cardiovascular function and genetic pathways associated with cardiac damage. To provide proof-of-principle and demonstrate translational relevance of this transport mechanism, we identified several pharmacological inhibitors of OCT3, including nilotinib, and found that pharmacological targeting of OCT3 can also preserve cardiovascular function following treatment with doxorubicin without affecting its plasma levels or antitumor effects in multiple models of leukemia and breast cancer. Finally, we identified a previously unrecognized, OCT3-dependent pathway of doxorubicin-induced cardiotoxicity that results in a downstream signaling cascade involving the calcium-binding proteins S100A8 and S100A9. These collective findings not only shed light on the etiology of doxorubicin-induced cardiotoxicity, but also are of potential translational relevance and provide a rationale for the implementation of a targeted intervention strategy to prevent this debilitating side effect.
Collapse
|
7
|
Sweet DH. Organic Cation Transporter Expression and Function in the CNS. Handb Exp Pharmacol 2021; 266:41-80. [PMID: 33963461 DOI: 10.1007/164_2021_463] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BCSFB) represent major control checkpoints protecting the CNS, by exerting selective control over the movement of organic cations and anions into and out of the CNS compartment. In addition, multiple CNS cell types, e.g., astrocytes, ependymal cells, microglia, contribute to processes that maintain the status quo of the CNS milieu. To fulfill their roles, these barriers and cell types express a multitude of transporter proteins from dozens of different transporter families. Fundamental advances over the past few decades in our knowledge of transporter substrates, expression profiles, and consequences of loss of function are beginning to change basic theories regarding the contribution of various cell types and clearance networks to coordinated neuronal signaling, complex organismal behaviors, and overall CNS homeostasis. In particular, transporters belonging to the Solute Carrier (SLC) superfamily are emerging as major contributors, including the SLC22 organic cation/anion/zwitterion family of transporters (includes OCT1-3 and OCTN1-3), the SLC29 facilitative nucleoside family of transporters (includes PMAT), and the SLC47 multidrug and toxin extrusion family of transporters (includes MATE1-2). These transporters are known to interact with neurotransmitters, antidepressant and anxiolytic agents, and drugs of abuse. Clarifying their contributions to the underlying mechanisms regulating CNS permeation and clearance, as well as the health status of astrocyte, microglial and neuronal cell populations, will drive new levels of understanding as to maintenance of the CNS milieu and approaches to new therapeutics and therapeutic strategies in the treatment of CNS disorders. This chapter highlights organic cation transporters belonging to the SLC superfamily known to be expressed in the CNS, providing an overview of their identification, mechanism of action, CNS expression profile, interaction with neurotransmitters and antidepressant/antipsychotic drugs, and results from behavioral studies conducted in loss of function models (knockout/knockdown).
Collapse
Affiliation(s)
- Douglas H Sweet
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
8
|
Karahoda R, Horackova H, Kastner P, Matthios A, Cerveny L, Kucera R, Kacerovsky M, Duintjer Tebbens J, Bonnin A, Abad C, Staud F. Serotonin homeostasis in the materno-foetal interface at term: Role of transporters (SERT/SLC6A4 and OCT3/SLC22A3) and monoamine oxidase A (MAO-A) in uptake and degradation of serotonin by human and rat term placenta. Acta Physiol (Oxf) 2020; 229:e13478. [PMID: 32311818 DOI: 10.1111/apha.13478] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 04/14/2020] [Accepted: 04/14/2020] [Indexed: 12/18/2022]
Abstract
AIM Serotonin is crucial for proper foetal development, and the placenta has been described as a 'donor' of serotonin for the embryo/foetus. However, in later stages of gestation the foetus produces its own serotonin from maternally-derived tryptophan and placental supply is no longer needed. We propose a novel model of serotonin homeostasis in the term placenta with special focus on the protective role of organic cation transporter 3 (OCT3/SLC22A3). METHODS Dually perfused rat term placenta was employed to quantify serotonin/tryptophan transport and metabolism. Placental membrane vesicles isolated from human term placenta were used to characterize serotonin transporters on both sides of the syncytiotrophoblast. RESULTS We obtained the first evidence that serotonin is massively taken up from the foetal circulation by OCT3. This uptake is concentration-dependent and inhibitable by OCT3 blockers of endogenous (glucocorticoids) or exogenous (pharmaceuticals) origin. Population analyses in rat placenta revealed that foetal sex influences placental extraction of serotonin from foetal circulation. Negligible foetal serotonin levels were detected in maternal-to-foetal serotonin/tryptophan transport and metabolic studies. CONCLUSION We demonstrate that OCT3, localized on the foetus-facing membrane of syncytiotrophoblast, is an essential component of foeto-placental homeostasis of serotonin. Together with serotonin degrading enzyme, monoamine oxidase-A, this offers a protective mechanism against local vasoconstriction effects of serotonin in the placenta. However, this system may be compromised by OCT3 inhibitory molecules, such as glucocorticoids or antidepressants. Our findings open new avenues to explore previously unsuspected/unexplained complications during pregnancy including prenatal glucocorticoid excess and pharmacotherapeutic risks of treating pregnant women with OCT3 inhibitors.
Collapse
Affiliation(s)
- Rona Karahoda
- Faculty of Pharmacy in Hradec Kralove Department of Pharmacology and Toxicology Charles University Hradec Kralove Czech Republic
| | - Hana Horackova
- Faculty of Pharmacy in Hradec Kralove Department of Pharmacology and Toxicology Charles University Hradec Kralove Czech Republic
| | - Petr Kastner
- Faculty of Pharmacy in Hradec Kralove Department of Pharmaceutical Chemistry and Pharmaceutical Analysis Charles University Hradec Kralove Czech Republic
| | - Andreas Matthios
- Faculty of Pharmacy in Hradec Kralove Department of Biophysics and Physical Chemistry Charles University Hradec Kralove Czech Republic
| | - Lukas Cerveny
- Faculty of Pharmacy in Hradec Kralove Department of Pharmacology and Toxicology Charles University Hradec Kralove Czech Republic
| | - Radim Kucera
- Faculty of Pharmacy in Hradec Kralove Department of Pharmaceutical Chemistry and Pharmaceutical Analysis Charles University Hradec Kralove Czech Republic
| | - Marian Kacerovsky
- Department of Gynecology and Obstetrics University Hospital in Hradec Kralove Hradec Kralove Czech Republic
| | - Jurjen Duintjer Tebbens
- Faculty of Pharmacy in Hradec Kralove Department of Biophysics and Physical Chemistry Charles University Hradec Kralove Czech Republic
| | - Alexandre Bonnin
- Department of Physiology and Neuroscience University of Southern California Los Angeles CA USA
| | - Cilia Abad
- Faculty of Pharmacy in Hradec Kralove Department of Pharmacology and Toxicology Charles University Hradec Kralove Czech Republic
| | - Frantisek Staud
- Faculty of Pharmacy in Hradec Kralove Department of Pharmacology and Toxicology Charles University Hradec Kralove Czech Republic
| |
Collapse
|
9
|
Sonobe T, Akiyama T, Du C, Pearson JT. Serotonin uptake via plasma membrane monoamine transporter during myocardial ischemia-reperfusion in the rat heart in vivo. Physiol Rep 2019; 7:e14297. [PMID: 31782271 PMCID: PMC6882957 DOI: 10.14814/phy2.14297] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Serotonin (5-HT) accumulates in the heart during myocardial ischemia and induces deleterious effects on the cardiomyocytes through receptor-dependent and monoamine oxidase-dependent pathways. We aimed to clarify the involvement of extra-neuronal monoamine transporters in the clearance of 5-HT during ischemia and reperfusion in the heart. Using a microdialysis technique in the anesthetized Wistar rat heart, we monitored myocardial interstitial 5-HT and 5-hydroxyindole acetic acid (5-HIAA) concentration by means of electro-chemical detection coupled with high-performance liquid chromatography (HPLC-ECD). Effects of inhibitors of the plasma membrane monoamine transporter (PMAT) and the organic cation transporter 3 (OCT3) (decynium-22 and corticosterone) on the 5-HT and 5-HIAA concentrations during baseline, coronary occlusion, and reperfusion were investigated. Basal dialysate 5-HT concentration were increased by local administration of decynium-22, but not by corticosterone. Addition of fluoxetine, a serotonin transporter (SERT) inhibitor further increased the 5-HT concentration upon during administration of decynium-22. Decynium-22 elevated the background level of 5-HT during coronary occlusion and maintained 5-HT concentration at a high level during reperfusion. Production of 5-HIAA in the early reperfusion was significantly suppressed by decynium-22. These results indicate that PMAT and SERT independently regulate basal level of interstitial 5-HT, and PMAT plays a more important role in the clearance of 5-HT during reperfusion. These data suggest the involvement of PMAT in the monoamine oxidase-dependent deleterious pathway in the heart.
Collapse
Affiliation(s)
- Takashi Sonobe
- Department of Cardiac PhysiologyNational Cerebral and Cardiovascular Center Research InstituteSuitaOsakaJapan
| | - Tsuyoshi Akiyama
- Department of Cardiac PhysiologyNational Cerebral and Cardiovascular Center Research InstituteSuitaOsakaJapan
| | - Cheng‐Kun Du
- Department of Cardiac PhysiologyNational Cerebral and Cardiovascular Center Research InstituteSuitaOsakaJapan
| | - James T. Pearson
- Department of Cardiac PhysiologyNational Cerebral and Cardiovascular Center Research InstituteSuitaOsakaJapan
| |
Collapse
|
10
|
Garbarino VR, Gilman TL, Daws LC, Gould GG. Extreme enhancement or depletion of serotonin transporter function and serotonin availability in autism spectrum disorder. Pharmacol Res 2019; 140:85-99. [PMID: 30009933 PMCID: PMC6345621 DOI: 10.1016/j.phrs.2018.07.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/22/2018] [Accepted: 07/12/2018] [Indexed: 12/15/2022]
Abstract
A variety of human and animal studies support the hypothesis that serotonin (5-hydroxytryptamine or 5-HT) system dysfunction is a contributing factor to the development of autism in some patients. However, many questions remain about how developmental manipulation of various components that influence 5-HT signaling (5-HT synthesis, transport, metabolism) persistently impair social behaviors. This review will summarize key aspects of central 5-HT function important for normal brain development, and review evidence implicating perinatal disruptions in 5-HT signaling in the pathophysiology of autism spectrum disorder. We discuss the importance, and relative dearth, of studies that explore the possible correlation to autism in the interactions between important intrinsic and extrinsic factors that may disrupt 5-HT homeostasis during development. In particular, we focus on exposure to 5-HT transport altering mechanisms such as selective serotonin-reuptake inhibitors or genetic polymorphisms in primary or auxiliary transporters of 5-HT, and how they relate to neurological stores of serotonin and its precursors. A deeper understanding of the many mechanisms by which 5-HT signaling can be disrupted, alone and in concert, may contribute to an improved understanding of the etiologies and heterogeneous nature of this disorder. We postulate that extreme bidirectional perturbations of these factors during development likely compound or synergize to facilitate enduring neurochemical changes resulting in insufficient or excessive 5-HT signaling, that could underlie the persistent behavioral characteristics of autism spectrum disorder.
Collapse
Affiliation(s)
- Valentina R Garbarino
- Department of Cellular and Integrative Physiology, United States; The Sam and Ann Barshop Institute for Longevity and Aging Studies, United States.
| | - T Lee Gilman
- Department of Cellular and Integrative Physiology, United States; Addiction Research, Treatment & Training Center of Excellence, United States.
| | - Lynette C Daws
- Department of Cellular and Integrative Physiology, United States; Addiction Research, Treatment & Training Center of Excellence, United States; Department of Pharmacology, United States.
| | - Georgianna G Gould
- Department of Cellular and Integrative Physiology, United States; Center for Biomedical Neuroscience, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
11
|
Electrophysiological Characterization of Novel Effects of the Uptake-2 Blocker Decynium-22 (D-22) on Dopaminergic Neurons in the Substantia Nigra Pars Compacta. Neuroscience 2019; 396:154-165. [PMID: 30447392 DOI: 10.1016/j.neuroscience.2018.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/23/2018] [Accepted: 11/07/2018] [Indexed: 01/11/2023]
Abstract
Extracellular levels of dopamine (DA) and other monoamines in the brain depend not only on the classic transporters encoded by SLC6A gene family such as DAT, NET and SERT, but also a more recently identified group of low-affinity/high-capacity 'Uptake-2' transporters, mainly OCT3 and PMAT. The most frequently used pharmacological tool in functional studies of Uptake-2 is decynium-22 (D-22) known to block these transporters. However, the effectiveness of this drug in enhancing extracellular DA remains uncertain. Our aim was to test the hypothesis that D-22 increases extracellular levels of DA released from the somatodendritic region of dopaminergic neurons in the substantia nigra pars compacta (SNc) by reducing the OCT3/PMAT-dependent component of DA uptake. Extracellular DA was assessed indirectly, by evoking D2-IPSCs in SNc neurons following stimulated release of this neurotransmitter in midbrain slices obtained from mice. Recordings were conducted after partial inhibition of DAT with nomifensine, and after application of L-DOPA which increased the releasable DA pool. Contrary to our expectations, D-22 reduced, rather than increased, the amplitude of D2-IPSCs. Other effects included inhibition of GABAB-IPSCs and Ih current, and a reduction in firing frequency of nigral neurons. These results show that in addition to the previously known non-specific inhibitory action on α1 adrenoceptors, D-22 exerts additional off-target effects by inhibiting dopaminergic and GABAergic synaptic transmission in the SNc and the spontaneous (pacemaker) activity of nigral neurons. It remains to be established if these novel effects contribute to a reduction in spontaneous locomotor activity reported in previous studies after systemic drug administration.
Collapse
|
12
|
Fraser-Spears R, Krause-Heuer AM, Basiouny M, Mayer FP, Manishimwe R, Wyatt NA, Dobrowolski JC, Roberts MP, Greguric I, Kumar N, Koek W, Sitte HH, Callaghan PD, Fraser BH, Daws LC. Comparative analysis of novel decynium-22 analogs to inhibit transport by the low-affinity, high-capacity monoamine transporters, organic cation transporters 2 and 3, and plasma membrane monoamine transporter. Eur J Pharmacol 2018; 842:351-364. [PMID: 30473490 DOI: 10.1016/j.ejphar.2018.10.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/03/2018] [Accepted: 10/19/2018] [Indexed: 11/30/2022]
Abstract
Growing evidence supports involvement of low-affinity/high-capacity organic cation transporters (OCTs) and plasma membrane monoamine transporter (PMAT) in regulating clearance of monoamines. Currently decynium-22 (D22) is the best pharmacological tool to study these transporters, however it does not readily discriminate among them, underscoring a need to develop compounds with greater selectivity for each of these transporters. We developed seven D22 analogs, and previously reported that some have lower affinity for α1-adrenoceptors than D22 and showed antidepressant-like activity in mice. Here, we extend these findings to determine the affinity of these analogs for OCT2, OCT3 and PMAT, as well as serotonin, norepinephrine and dopamine transporters (SERT, NET and DAT) using a combination of uptake competition with [3H]methyl-4-phenylpyridinium acetate in overexpressed HEK cells and [3H]citalopram, [3H]nisoxetine and [3H]WIN 35428 displacement binding in mouse hippocampal and striatal preparations. Like D22, all analogs showed greater binding affinities for OCT3 than OCT2 and PMAT. However, unlike D22, some analogs also showed modest affinity for SERT and DAT. Dual OCT3/SERT and/or OCT3/DAT actions of certain analogs may help explain their ability to produce antidepressant-like effects in mice and help account for our previous findings that D22 lacks antidepressant-like effects unless SERT function is either genetically or pharmacologically compromised. Though these analogs are not superior than D22 in discriminating among OCTs/PMAT, our findings point to development of compounds with combined ability to inhibit both low-affinity/high-capacity transporters, such as OCT3, and high-affinity/low-capacity transporters, such as SERT, as therapeutics with potentially improved efficacy for treatment of psychiatric disorders.
Collapse
Affiliation(s)
- Rheaclare Fraser-Spears
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, United States; University of the Incarnate Word, Feik School of Pharmacy, Department of Pharmaceutical Sciences, United States
| | - Anwen M Krause-Heuer
- The Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, NSW 2232, Australia
| | - Mohamed Basiouny
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, United States
| | - Felix P Mayer
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090 Vienna, Austria
| | - Retrouvailles Manishimwe
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, United States
| | - Naomi A Wyatt
- The Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, NSW 2232, Australia
| | | | - Maxine P Roberts
- The Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, NSW 2232, Australia
| | - Ivan Greguric
- The Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, NSW 2232, Australia
| | - Naresh Kumar
- University of New South Wales, School of Chemistry, Sydney, NSW 2052, Australia
| | - Wouter Koek
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, United States; Department of Psychiatry, University of Texas Health Science Center at San Antonio, United States
| | - Harald H Sitte
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090 Vienna, Austria
| | - Paul D Callaghan
- The Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, NSW 2232, Australia
| | - Benjamin H Fraser
- The Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, NSW 2232, Australia
| | - Lynette C Daws
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, United States; Department of Pharmacology, University of Texas Health Science Center at San Antonio, United States.
| |
Collapse
|
13
|
Gasser PJ, Lowry CA. Organic cation transporter 3: A cellular mechanism underlying rapid, non-genomic glucocorticoid regulation of monoaminergic neurotransmission, physiology, and behavior. Horm Behav 2018; 104:173-182. [PMID: 29738736 PMCID: PMC7137088 DOI: 10.1016/j.yhbeh.2018.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/02/2018] [Accepted: 05/03/2018] [Indexed: 01/11/2023]
Abstract
Contribution to Special Issue on Fast effects of steroids. Corticosteroid hormones act at intracellular glucocorticoid receptors (GR) and mineralocorticoid receptors (MR) to alter gene expression, leading to diverse physiological and behavioral responses. In addition to these classical genomic effects, corticosteroid hormones also exert rapid actions on physiology and behavior through a variety of non-genomic mechanisms, some of which involve GR or MR, and others of which are independent of these receptors. One such GR-independent mechanism involves corticosteroid-induced inhibition of monoamine transport mediated by "uptake2" transporters, including organic cation transporter 3 (OCT3), a low-affinity, high-capacity transporter for norepinephrine, epinephrine, dopamine, serotonin and histamine. Corticosterone directly and acutely inhibits OCT3-mediated transport. This review describes the studies that initially characterized uptake2 processes in peripheral tissues, and outlines studies that demonstrated OCT3 expression and corticosterone-sensitive monoamine transport in the brain. Evidence is presented supporting the hypothesis that corticosterone can exert rapid, GR-independent actions on neuronal physiology and behavior by inhibiting OCT3-mediated monoamine clearance. Implications of this mechanism for glucocorticoid-monoamine interactions in the context-dependent regulation of behavior are discussed.
Collapse
Affiliation(s)
- Paul J Gasser
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI 53201, USA.
| | - Christopher A Lowry
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Department of Physical Medicine and Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Denver Veterans Affairs Medical Center (VAMC), Denver, CO 80220, USA; Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Denver, CO 80220, USA.
| |
Collapse
|
14
|
Solanki RR, Scholl JL, Watt MJ, Renner KJ, Forster GL. Amphetamine Withdrawal Differentially Increases the Expression of Organic Cation Transporter 3 and Serotonin Transporter in Limbic Brain Regions. J Exp Neurosci 2016; 10:93-100. [PMID: 27478387 PMCID: PMC4957605 DOI: 10.4137/jen.s40231] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/27/2016] [Accepted: 06/28/2016] [Indexed: 12/26/2022] Open
Abstract
Amphetamine withdrawal increases anxiety and stress sensitivity related to blunted ventral hippocampus (vHipp) and enhances the central nucleus of the amygdala (CeA) serotonin responses. Extracellular serotonin levels are regulated by the serotonin transporter (SERT) and organic cation transporter 3 (OCT3), and vHipp OCT3 expression is enhanced during 24 hours of amphetamine withdrawal, while SERT expression is unaltered. Here, we tested whether OCT3 and SERT expression in the CeA is also affected during acute withdrawal to explain opposing regional alterations in limbic serotonergic neurotransmission and if respective changes continued with two weeks of withdrawal. We also determined whether changes in transporter expression were confined to these regions. Male rats received amphetamine or saline for two weeks followed by 24 hours or two weeks of withdrawal, with transporter expression measured using Western immunoblot. OCT3 and SERT expression increased in the CeA at both withdrawal timepoints. In the vHipp, OCT3 expression increased only at 24 hours of withdrawal, with an equivalent pattern seen in the dorsomedial hypothalamus. No changes were evident in any other regions sampled. These regionally specific changes in limbic OCT3 and SERT expression may partially contribute to the serotonergic imbalance and negative affect during amphetamine withdrawal.
Collapse
Affiliation(s)
- Rajeshwari R. Solanki
- Division of Basic Biomedical Sciences, Sanford School of Medicine, Center for Brain and Behavior Research, University of South Dakota, Vermillion, SD, USA
| | - Jamie L. Scholl
- Division of Basic Biomedical Sciences, Sanford School of Medicine, Center for Brain and Behavior Research, University of South Dakota, Vermillion, SD, USA
| | - Michael J. Watt
- Division of Basic Biomedical Sciences, Sanford School of Medicine, Center for Brain and Behavior Research, University of South Dakota, Vermillion, SD, USA
| | - Kenneth J. Renner
- Biology Department, Center for Brain and Behavior Research, University of South Dakota, Vermillion, SD, USA
| | - Gina L. Forster
- Division of Basic Biomedical Sciences, Sanford School of Medicine, Center for Brain and Behavior Research, University of South Dakota, Vermillion, SD, USA
| |
Collapse
|
15
|
Duan H, Hu T, Foti RS, Pan Y, Swaan PW, Wang J. Potent and Selective Inhibition of Plasma Membrane Monoamine Transporter by HIV Protease Inhibitors. Drug Metab Dispos 2015; 43:1773-80. [PMID: 26285765 DOI: 10.1124/dmd.115.064824] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 08/17/2015] [Indexed: 12/13/2022] Open
Abstract
Plasma membrane monoamine transporter (PMAT) is a major uptake-2 monoamine transporter that shares extensive substrate and inhibitor overlap with organic cation transporters 1-3 (OCT1-3). Currently, there are no PMAT-specific inhibitors available that can be used in in vitro and in vivo studies to differentiate between PMAT and OCT activities. In this study, we showed that IDT307 (4-(4-(dimethylamino)phenyl)-1-methylpyridinium iodide), a fluorescent analog of 1-methyl-4-phenylpyridinium (MPP+), is a transportable substrate for PMAT and that IDT307-based fluorescence assay can be used to rapidly identify and characterize PMAT inhibitors. Using the fluorescent substrate-based assays, we analyzed the interactions of eight human immunodeficiency virus (HIV) protease inhibitors (PIs) with human PMAT and OCT1-3 in human embryonic kidney 293 (HEK293) cells stably transfected with individual transporters. Our data revealed that PMAT and OCTs exhibit distinct sensitivity and inhibition patterns toward HIV PIs. PMAT is most sensitive to PI inhibition whereas OCT2 and OCT3 are resistant. OCT1 showed an intermediate sensitivity and a distinct inhibition profile from PMAT. Importantly, lopinavir is a potent PMAT inhibitor and exhibited >120 fold selectivity toward PMAT (IC₅₀ = 1.4 ± 0.2 µM) over OCT1 (IC₅₀ = 174 ± 40 µM). Lopinavir has no inhibitory effect on OCT2 or OCT3 at maximal tested concentrations. Lopinavir also exhibited no or much weaker interactions with uptake-1 monoamine transporters. Together, our results reveal that PMAT and OCTs have distinct specificity exemplified by their differential interaction with HIV PIs. Further, we demonstrate that lopinavir can be used as a selective PMAT inhibitor to differentiate PMAT-mediated monoamine and organic cation transport from those mediated by OCT1-3.
Collapse
Affiliation(s)
- Haichuan Duan
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.D., T.H., J.W.); Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (R.S.F.); and Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland (Y.P., P.W.S.)
| | - Tao Hu
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.D., T.H., J.W.); Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (R.S.F.); and Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland (Y.P., P.W.S.)
| | - Robert S Foti
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.D., T.H., J.W.); Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (R.S.F.); and Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland (Y.P., P.W.S.)
| | - Yongmei Pan
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.D., T.H., J.W.); Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (R.S.F.); and Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland (Y.P., P.W.S.)
| | - Peter W Swaan
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.D., T.H., J.W.); Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (R.S.F.); and Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland (Y.P., P.W.S.)
| | - Joanne Wang
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.D., T.H., J.W.); Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (R.S.F.); and Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland (Y.P., P.W.S.)
| |
Collapse
|
16
|
Marcinkiewcz CA, Devine DP. Modulation of OCT3 expression by stress, and antidepressant-like activity of decynium-22 in an animal model of depression. Pharmacol Biochem Behav 2015; 131:33-41. [PMID: 25597272 DOI: 10.1016/j.pbb.2015.01.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Revised: 01/08/2015] [Accepted: 01/11/2015] [Indexed: 10/24/2022]
Abstract
The organic cation transporter-3 (OCT3) is a glucocorticoid-sensitive uptake mechanism that has been shown to regulate the bioavailability of monoamines in brain regions that are implicated in the pathophysiology of depression. In the present study, the relative impacts of acute stress alone and acute stress with a history of repeated stress (chronic+acute) were evaluated in two strains of rats: the stress-vulnerable Wistar-Kyoto (WKY) strain and the somewhat more stress-resilient Long-Evans (LE) strain. OCT3 mRNA was significantly upregulated in the hippocampus of LE rats 2h after exposure to acute restraint stress, but not in acutely-restrained rats with a history of repeated social defeat stress. WKY rats exhibited a very different pattern. OCT3 mRNA was unaffected by acute restraint stress alone but was robustly upregulated after repeated+acute stress. There was also a corresponding increase in cytosolic OCT3 protein following repeated+acute stress in WKY rats 3h after presentation of the acute stressor. These results are consistent with the hypothesis that altered expression of the OCT3 may play a role in stress coping, and strain differences in regulation of this expression may contribute to differences in physiological and behavioral responses to stress. Furthermore, the OCT3 inhibitor, decynium 22 (1 and 10μg/kg, i.p.) reduced immobility of WKY rats, but not that of LE rats, in the forced swim test, suggesting that blockade of the OCT3 has antidepressant-like effects. Since WKY rats also appear to be resistant to the behavioral effects of traditional antidepressants, this also suggests that OCT3 antagonism may be an alternative therapeutic strategy for the treatment of depression in individuals who do not respond to conventional antidepressants.
Collapse
Affiliation(s)
- C A Marcinkiewcz
- University of Florida, Department of Neuroscience, McKnight Brain Institute, Gainesville, FL 32610-0015, USA
| | - D P Devine
- University of Florida, Department of Neuroscience, McKnight Brain Institute, Gainesville, FL 32610-0015, USA; University of Florida, Department of Psychology, Behavioral and Cognitive Neuroscience Program, Gainesville, FL 32611-2250, USA.
| |
Collapse
|
17
|
Li H, Scholl JL, Tu W, Hassell JE, Watt MJ, Forster GL, Renner KJ. Serotonergic responses to stress are enhanced in the central amygdala and inhibited in the ventral hippocampus during amphetamine withdrawal. Eur J Neurosci 2014; 40:3684-92. [PMID: 25234335 DOI: 10.1111/ejn.12735] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 08/05/2014] [Accepted: 08/20/2014] [Indexed: 12/26/2022]
Abstract
Withdrawal from amphetamine increases anxiety and reduces the ability to cope with stress, which are factors that are believed to contribute to drug relapse. Stress-induced serotonergic transmission in the central nucleus of the amygdala is associated with anxiety states and fear. Conversely, stress-induced increases in ventral hippocampal serotonin (5-HT) levels have been linked to coping mechanisms. The goal of this study was to investigate the neurobiological changes induced by amphetamine that contribute to stress sensitivity during withdrawal. We tested the hypothesis that limbic serotonergic responses to restraint stress would be altered in male Sprague-Dawley rats chronically pretreated with amphetamine (2.5 mg/kg, intraperitoneal) and then subjected to 2 weeks of withdrawal. Amphetamine withdrawal resulted in increased stress-induced behavioral arousal relative to control treatment, suggesting that drug withdrawal induced greater sensitivity to the stressor. When microdialysis was used to determine the effects of restraint on extracellular 5-HT, stress-induced increases in 5-HT levels were abolished in the ventral hippocampus and augmented in the central amygdala during amphetamine withdrawal. Reverse dialysis of the glucocorticoid receptor antagonist mifepristone into the ventral hippocampus blocked the stress-induced increase in 5-HT levels in saline-pretreated rats, suggesting that glucocorticoid receptors mediate stress-induced increases in 5-HT levels in the ventral hippocampus. However, mifepristone had no effect on stress-induced increases in 5-HT levels in the central amygdala, indicating that stress increases 5-HT levels in this region independently of glucocorticoid receptors. During amphetamine withdrawal, the absence of stress-induced increases in ventral hippocampal 5-HT levels combined with enhanced stress-induced serotonergic responses in the central amygdala may contribute to drug relapse by decreasing stress-coping ability and heightening stress responsiveness.
Collapse
Affiliation(s)
- Hao Li
- Department of Biology & Center for Brain and Behavior Research, University of South Dakota, 414 E. Clark St, Vermillion, SD, 57069, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Nguyen HT, Guiard BP, Bacq A, David DJ, David I, Quesseveur G, Gautron S, Sanchez C, Gardier AM. Blockade of the high-affinity noradrenaline transporter (NET) by the selective 5-HT reuptake inhibitor escitalopram: an in vivo microdialysis study in mice. Br J Pharmacol 2014; 168:103-16. [PMID: 22233336 DOI: 10.1111/j.1476-5381.2012.01850.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND AND PURPOSE Escitalopram, the S(+)-enantiomer of citalopram is the most selective 5-HT reuptake inhibitor approved. Although all 5-HT selective reuptake inhibitors (SSRIs) increase extracellular levels of 5-HT ([5-HT](ext)). some also enhance, to a lesser extent, extracellular levels of noradrenaline ([NA](ext)). However, the mechanisms by which SSRIs activate noradrenergic transmission in the brain remain to be determined. EXPERIMENTAL APPROACH This study examined the effects of escitalopram, on both [5-HT](ext) and [NA](ext) in the frontal cortex (FCx) of freely moving wild-type (WT) and mutant mice lacking the 5-HT transporter (SERT(-/-)) by using intracerebral microdialysis. We explored the possibilities that escitalopram enhances [NA](ext), either by a direct mechanism involving the inhibition of the low- or high-affinity noradrenaline transporters, or by an indirect mechanism promoted by [5-HT](ext) elevation. The forced swim test (FST) was used to investigate whether enhancing cortical [5-HT](ext) and/or [NA](ext) affected the antidepressant-like activity of escitalopram. KEY RESULTS In WT mice, a single systemic administration of escitalopram produced a significant increase in cortical [5-HT](ext) and [NA](ext). As expected, escitalopram failed to increase cortical [5-HT](ext) in SERT(-/-) mice, whereas its neurochemical effects on [NA](ext) persisted in these mutants. In WT mice subjected to the FST, escitalopram increased swimming parameters without affecting climbing behaviour. Finally, escitalopram, at relevant concentrations, failed to inhibit cortical noradrenaline and 5-HT uptake mediated by low-affinity monoamine transporters. CONCLUSIONS AND IMPLICATIONS These experiments suggest that escitalopram enhances, although moderately, cortical [NA](ext) in vivo by a direct mechanism involving the inhibition of the high-affinity noradrenaline transporter (NET).
Collapse
Affiliation(s)
- Hai T Nguyen
- Laboratoire de Neuropharmacologie, Faculté de Pharmacie, Université Paris-Sud XI, Châtenay-Malabry, France
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Massmann V, Edemir B, Schlatter E, Al-Monajjed R, Harrach S, Klassen P, Holle SK, Sindic A, Dobrivojevic M, Pavenstädt H, Ciarimboli G. The organic cation transporter 3 (OCT3) as molecular target of psychotropic drugs: transport characteristics and acute regulation of cloned murine OCT3. Pflugers Arch 2013; 466:517-27. [DOI: 10.1007/s00424-013-1335-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 08/05/2013] [Accepted: 08/09/2013] [Indexed: 01/11/2023]
|
20
|
Daws LC, Koek W, Mitchell NC. Revisiting serotonin reuptake inhibitors and the therapeutic potential of "uptake-2" in psychiatric disorders. ACS Chem Neurosci 2013; 4:16-21. [PMID: 23336039 DOI: 10.1021/cn3001872] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 10/25/2012] [Indexed: 01/11/2023] Open
Abstract
Depression is among the most common psychiatric disorders, and in many patients a disorder for which available medications provide suboptimal or no symptom relief. The most commonly prescribed class of antidepressants, the selective serotonin reuptake inhibitors (SSRIs), are thought to act by increasing extracellular serotonin in brain by blocking its uptake via the high-affinity serotonin transporter (SERT). However, the relative lack of therapeutic efficacy of SSRIs has brought into question the utility of increasing extracellular serotonin for the treatment of depression. In this Viewpoint, we discuss why increasing extracellular serotonin should not be written off as a therapeutic strategy. We describe how "uptake-2" transporters may explain the relative lack of therapeutic efficacy of SSRIs, as well as why "uptake-2" transporters might be useful therapeutic targets.
Collapse
Affiliation(s)
- Lynette C. Daws
- Departments of †Physiology, ‡Psychiatry, and §Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229,
United States
| | - Wouter Koek
- Departments of †Physiology, ‡Psychiatry, and §Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229,
United States
| | - Nathan C. Mitchell
- Departments of †Physiology, ‡Psychiatry, and §Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229,
United States
| |
Collapse
|
21
|
Barr JL, Scholl JL, Solanki RR, Watt MJ, Lowry CA, Renner KJ, Forster GL. Influence of chronic amphetamine treatment and acute withdrawal on serotonin synthesis and clearance mechanisms in the rat ventral hippocampus. Eur J Neurosci 2012; 37:479-90. [PMID: 23157166 DOI: 10.1111/ejn.12050] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 10/01/2012] [Accepted: 10/02/2012] [Indexed: 01/11/2023]
Abstract
Amphetamine withdrawal in both humans and rats is associated with increased anxiety states, which are thought to contribute to drug relapse. Serotonin in the ventral hippocampus mediates affective behaviors, and reduced serotonin levels in this region are observed in rat models of high anxiety, including during withdrawal from chronic amphetamine. This goal of this study was to understand the mechanisms by which reduced ventral hippocampus serotonergic neurotransmission occurs during amphetamine withdrawal. Serotonin synthesis (assessed by accumulation of serotonin precursor as a measure of the capacity of in vivo tryptophan hydroxylase activity), expression of serotonergic transporters, and in vivo serotonergic clearance using in vivo microdialysis were assessed in the ventral hippocampus in adult male Sprague Dawley rats at 24 h withdrawal from chronic amphetamine. Overall, results showed that diminished extracellular serotonin at 24 h withdrawal from chronic amphetamine was not accompanied by a change in capacity for serotonin synthesis (in vivo tryptophan hydroxylase activity), or serotonin transporter expression or function in the ventral hippocampus, but instead was associated with increased expression and function of organic cation transporters (low-affinity, high-capacity serotonin transporters). These findings suggest that 24 h withdrawal from chronic amphetamine reduces the availability of extracellular serotonin in the ventral hippocampus by increasing organic cation transporter-mediated serotonin clearance, which may represent a future pharmacological target for reversing anxiety states during drug withdrawal.
Collapse
Affiliation(s)
- Jeffrey L Barr
- Neuroscience Group, Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Hagan CE, McDevitt RA, Liu Y, Furay AR, Neumaier JF. 5-HT(1B) autoreceptor regulation of serotonin transporter activity in synaptosomes. Synapse 2012; 66:1024-34. [PMID: 22961814 DOI: 10.1002/syn.21608] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 08/30/2012] [Indexed: 02/06/2023]
Abstract
Serotonin-1B (5-HT(1B) ) autoreceptors are located in serotonin (5-HT) terminals, along with serotonin transporters (SERT), and play a critical role in autoregulation of serotonergic neurotransmission and are implicated in disorders of serotonergic function, particularly emotional regulation. SERT modulates serotonergic neurotransmission by high-affinity reuptake of 5-HT. Alterations in SERT activity are associated with increased risk for depression and anxiety. Several neurotransmitter receptors are known to regulate SERT K(m) and V(max) , and previous work suggests that 5-HT(1B) autoreceptors may regulate 5-HT reuptake, in addition to modulating 5-HT release and synthesis. We used rotating disk electrode voltammetry to investigate 5-HT(1B) autoreceptor regulation of SERT-mediated 5-HT uptake into synaptosomes. The selective 5-HT(1B) antagonist SB224289 decreased SERT activity in synaptosomes prepared from wild-type but not 5-HT(1B) knockout mice, whereas SERT uptake was enhanced after pretreatment with the selective 5-HT(1B) agonist CP94253. Furthermore, SERT activity varies as a function of 5-HT(1B) receptor expression-specifically, genetic deletion of 5-HT(1B) decreased SERT function, while viral-mediated overexpression of 5-HT(1B) autoreceptors in rat raphe neurons increased SERT activity in rat hippocampal synaptosomes. Considered collectively, these results provide evidence that 5-HT(1B) autoreceptors regulate SERT activity. Because SERT clearance rate varies as a function of 5-HT(1B) autoreceptor expression levels and is modulated by both activation and inhibition of 5-HT(1B) autoreceptors, this dynamic interaction may be an important mechanism of serotonin autoregulation with therapeutic implications.
Collapse
Affiliation(s)
- Catherine E Hagan
- Department of Comparative Medicine and Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, Washington 98195, USA.
| | | | | | | | | |
Collapse
|
23
|
|
24
|
Haenisch B, Hiemke C, Bönisch H. Inhibitory potencies of trimipramine and its main metabolites at human monoamine and organic cation transporters. Psychopharmacology (Berl) 2011; 217:289-95. [PMID: 21484238 DOI: 10.1007/s00213-011-2281-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Accepted: 03/23/2011] [Indexed: 01/11/2023]
Abstract
RATIONALE The antidepressant trimipramine shows an atypical pharmacological profile and its mechanism of action is still obscure. OBJECTIVES The present study investigated whether trimipramine and three of its metabolites interact with targets of other antidepressants, namely, the human monoamine transporters for noradrenaline (hNAT), serotonin (hSERT), and dopamine (hDAT), and with the human organic cation transporters (hOCT1, hOCT2, and hOCT3) which are expressed in the brain and are known to be involved in the uptake of monoamines. METHODS HEK293 cells heterologously expressing the abovementioned transporters were used to determine the inhibition of [(3)H]MPP(+) uptake by trimipramine and its main metabolites. RESULTS At concentrations up to 30 μM, all transporters, except hOCT3, were inhibited by all examined substances. With IC(50) values between 2 and 10 μM, trimipramine inhibited hSERT, hNAT, hOCT1, and hOCT2, whereas clearly higher concentrations were needed for half-maximal inhibition of hDAT. Desmethyl-trimipramine showed about the same potencies as trimipramine, whereas 2-hydroxy-trimipramine was less potent at hNAT, hSERT, and hOCT1. Trimipramine-N-oxide preferentially inhibited hSERT. CONCLUSIONS Neither trimipramine nor its metabolites are highly potent inhibitors of the examined monoamine transporters. However, since at a steady state the sum of the concentrations of the parent compound and its active metabolites is almost two times higher than the plasma concentration of trimipramine and since it is known that tricyclic antidepressants accumulate in the brain (up to tenfold), at least partial inhibition by trimipramine and its metabolites of hSERT and hNAT (but not of hOCT3) may contribute to the antidepressant action of trimipramine.
Collapse
Affiliation(s)
- Britta Haenisch
- Institute of Pharmacology and Toxicology, Biomedical Center, University of Bonn, Sigmund-Freud-Strasse 25, 53127, Bonn, Germany
| | | | | |
Collapse
|
25
|
Hengen N, Lizer MH, Kidd RS. Evaluation of genetic variations in organic cationic transporter 3 in depressed and nondepressed subjects. ISRN PHARMACOLOGY 2011; 2011:161740. [PMID: 22084709 PMCID: PMC3196918 DOI: 10.5402/2011/161740] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/11/2011] [Accepted: 06/29/2011] [Indexed: 01/11/2023]
Abstract
Organic cationic transporter 3 (OCT3, SLS22A3) has only recently emerged as one of the regulators of monoaminergic neurotransmission, which plays a critical role in the pathogenesis of depression and is a potential new antidepressant drug target. OCT3 single-nucleotide polymorphisms (SNPs) have been investigated for their association with psychiatric disorders such as methamphetamine use disorder and obsessive-compulsive disorder in children and adolescents, but not depression. This study was designed to evaluate the allele frequencies of seven OCT3 SNPs in a US Caucasian depressed population and compare these frequencies with a control group of nondepressed subjects. Informed consent and a DNA sample were obtained from 157 subjects and analysis was performed using real-time PCR. Allele and genotype frequencies were compared using a t-test and the Pearson chi-square analysis, respectively. There were no significant differences in OCT3 allele or genotype frequencies between the depressed and non-depressed groups for all seven SNPs evaluated.
Collapse
Affiliation(s)
- Nina Hengen
- Bernard J. Dunn School of Pharmacy, Shenandoah University, 1775 North Sector Court, Winchester, VA 22601, USA
| | | | | |
Collapse
|
26
|
Hagan CE, Schenk JO, Neumaier JF. The contribution of low-affinity transport mechanisms to serotonin clearance in synaptosomes. Synapse 2011; 65:1015-23. [PMID: 21437992 DOI: 10.1002/syn.20929] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 03/01/2011] [Indexed: 01/11/2023]
Abstract
Although many studies assert that the serotonin (5-HT) transporter (SERT) is the predominant mechanism controlling extracellular 5-HT concentrations, accumulating evidence suggests that low affinity, high capacity transport mechanisms may contribute more to 5-HT clearance than previously thought. The goal of this study was to quantify the contributions of SERT relative to other mechanisms in clearing extracellular 5-HT concentrations ranging from 50 nM to 1 μM in synaptosomes prepared from wild-type and SERT knockout mice using rotating disk electrode voltammetry. SERT inhibitors combined with decynium-22 (D-22), a blocker of several low-affinity transporters, blocked all uptake of 5-HT into synaptosomes. We found that SERT is responsible for the majority of synaptosomal uptake only at relatively low 5-HT concentrations, but comprises a diminishing proportion of 5-HT clearance when extracellular 5-HT increases above 100 nM. The effect of D-22 was similar in wild-type and SERT knockout synaptosomes. Thus, there was no evidence of upregulation of low-affinity mechanisms in knockout mice across the concentrations of 5-HT tested. These are surprising results, in light of the prevailing view that SERT is the primary uptake mechanism for extracellular 5-HT at physiological concentrations. We conclude that non-SERT mediated 5-HT uptake is substantial even at modest 5-HT concentrations. These findings, in conjunction with other studies, have important implications for understanding serotonergic disorders and may explain the variable efficacy and stability of patients' responses to antidepressants, such as the selective serotonin reuptake inhibitors.
Collapse
Affiliation(s)
- Catherine E Hagan
- Department of Comparative Medicine, University of Washington, Seattle, Washington 98195, USA.
| | | | | |
Collapse
|
27
|
Daws LC, Gould GG. Ontogeny and regulation of the serotonin transporter: providing insights into human disorders. Pharmacol Ther 2011; 131:61-79. [PMID: 21447358 DOI: 10.1016/j.pharmthera.2011.03.013] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Accepted: 03/11/2011] [Indexed: 12/17/2022]
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) was one of the first neurotransmitters for which a role in development was identified. Pharmacological and gene knockout studies have revealed a critical role for 5-HT in numerous processes, including cell division, neuronal migration, differentiation and synaptogenesis. An excess in brain 5-HT appears to be mechanistically linked to abnormal brain development, which in turn is associated with neurological disorders. Ambient levels of 5-HT are controlled by a vast orchestra of proteins, including a multiplicity of pre- and post-synaptic 5-HT receptors, heteroreceptors, enzymes and transporters. The 5-HT transporter (SERT, 5-HTT) is arguably the most powerful regulator of ambient extracellular 5-HT. SERT is the high-affinity uptake mechanism for 5-HT and exerts tight control over the strength and duration of serotonergic neurotransmission. Perturbation of its expression level or function has been implicated in many diseases, prominent among them are psychiatric disorders. This review synthesizes existing information on the ontogeny of SERT during embryonic and early postnatal development though adolescence, along with factors that influence its expression and function during these critical developmental windows. We integrate this knowledge to emphasize how inappropriate SERT expression or its dysregulation may be linked to the pathophysiology of psychiatric, cardiovascular and gastrointestinal diseases.
Collapse
Affiliation(s)
- Lynette C Daws
- Department of Physiology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MC 7756, San Antonio, TX 78229-3900, USA.
| | | |
Collapse
|
28
|
Repeated swim impairs serotonin clearance via a corticosterone-sensitive mechanism: organic cation transporter 3, the smoking gun. J Neurosci 2010; 30:15185-95. [PMID: 21068324 DOI: 10.1523/jneurosci.2740-10.2010] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Activation of the hypothalamic-pituitary-adrenal (HPA) axis is associated with increased extracellular serotonin (5-HT) in limbic brain regions. The mechanism through which this occurs remains unclear. One way could be via HPA axis-dependent impairment of serotonin transporter (SERT) function, the high-affinity uptake mechanism for 5-HT. Consistent with this idea, we found that 5-HT clearance rate in hippocampus was dramatically reduced in mice exposed to repeated swim, a stimulus known to activate the HPA axis. However, this phenomenon also occurred in mice lacking SERT, ruling out SERT as a mechanism. The organic cation transporter 3 (OCT3) is emerging as an important regulator of brain 5-HT. Moreover, corticosterone, which is released upon HPA axis activation, blocks 5-HT uptake by OCT3. Repeated swim produced a persistent elevation in plasma corticosterone, and, consistent with prolonged blockade by corticosterone, we found that OCT3 expression and function were reduced in these mice. Importantly, this effect of repeated swim to reduce 5-HT clearance rate was corticosterone dependent, as evidenced by its absence in adrenalectomized mice, in which plasma corticosterone levels were essentially undetectable. Behaviorally, mice subjected to repeated swim spent less time immobile in the tail suspension test than control mice, but responded similarly to SERT- and norepinephrine transporter-selective antidepressants. Together, these results show that reduced 5-HT clearance following HPA axis activation is likely mediated, at least in part, by the corticosterone-sensitive OCT3, and that drugs developed to selectively target OCT3 (unlike corticosterone) may be candidates for the development of novel antidepressant medications.
Collapse
|
29
|
Zhu HJ, Appel DI, Gründemann D, Markowitz JS. Interaction of organic cation transporter 3 (SLC22A3) and amphetamine. J Neurochem 2010; 114:142-9. [PMID: 20402963 DOI: 10.1111/j.1471-4159.2010.06738.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The organic cation transporter (OCT) 3 is widely expressed in various organs in humans, and involved in the disposition of many exogenous and endogenous compounds. Several lines of evidence have suggested that OCT3 expressed in the brain plays an important role in the regulation of neurotransmission. Relative to wild-type (WT) animals, Oct3 knockout (KO) mice have displayed altered behavioral and neurochemical responses to psychostimulants such as amphetamine (AMPH) and methamphetamine. In the present study, both in vitro and in vivo approaches were utilized to explore potential mechanisms underlying the disparate neuropharmacological effects observed following AMPH exposure in Oct3 KO mice. In vitro uptake studies conducted in OCT3 transfected cells indicated that dextroamphetamine (d-AMPH) is not a substrate of OCT3. However, OCT3 was determined to be a high-capacity and low-affinity transporter for the neurotransmitters dopamine (DA), norepinephrine (NE), and serotonin (5-HT). Inhibition studies demonstrated that d-AMPH exerts relatively weak inhibitory effects on the OCT3-mediated uptake of DA, NE, 5-HT, and the model OCT3 substrate 4-(4-(dimethylamino)styryl)-N-methylpyridinium iodide. The IC(50) values were determined to be 41.5 +/- 7.5 and 24.1 +/- 7.0 microM for inhibiting DA and 5-HT uptake, respectively, while 50% inhibition of NE and 4-(4-(dimethylamino)styryl)-N-methylpyridinium iodide uptake was not achieved by even the highest concentration of d-AMPH applied (100 microM). Furthermore, the disposition of d-AMPH in various tissues including the brain, liver, heart, kidney, muscle, intestine, spleen, testis, uterus, and plasma were determined in both male and female Oct3 KO and WT mice. No significant difference was observed between either genotypes or sex in all tested organs and tissues. Our findings suggest that OCT3 is not a prominent factor influencing the disposition of d-AMPH. Additionally, based upon the inhibitory potency observed in vitro, d-AMPH is unlikely to inhibit the uptake of monoamines mediated by OCT3 in the brain. Differentiated neuropharmacological effects of AMPHs noted between Oct3 KO and WT mice appear to be due to the absence of Oct3 mediated uptake of neurotransmitters in the KO mice.
Collapse
Affiliation(s)
- Hao-Jie Zhu
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida 32610-0486, USA
| | | | | | | |
Collapse
|
30
|
Feng N, Lowry CA, Lukkes JL, Orchinik M, Forster GL, Renner KJ. Organic cation transporter inhibition increases medial hypothalamic serotonin under basal conditions and during mild restraint. Brain Res 2010; 1326:105-13. [PMID: 20171957 DOI: 10.1016/j.brainres.2010.02.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 02/10/2010] [Accepted: 02/10/2010] [Indexed: 01/01/2023]
Abstract
The dorsomedial hypothalamus (DMH) has been implicated in the coordination of stress responses. Restraint stress or systemic corticosterone (CORT) treatment induces a rapid increase in tissue concentrations of serotonin (5-hydroxytryptamine; 5-HT) in the DMH. Although the mechanism for rapid changes in 5-HT concentrations in the DMH is not clear, earlier results suggest that stress-induced increases in CORT may inhibit 5-HT transport from the extracellular fluid by acting on corticosterone-sensitive organic cation transporters (OCTs). We tested the hypothesis that perfusion of the medial hypothalamus (MH), which includes the DMH, with the OCT blocker decynium 22 (D-22) would potentiate the effects of mild restraint on extracellular 5-HT. Male Sprague-Dawley rats, implanted with a microdialysis probe into the MH, were treated with reverse-dialysis of D-22 (20 microM; 40 min) or vehicle and subjected to either 40 min mild restraint or undisturbed control conditions. Perfusates collected from a separate group of rats were evaluated for the effect of restraint on extracellular CORT concentrations in the MH. Reverse-dialysis of D-22 induced an increase (200%) in extracellular 5-HT concentrations in the MH in undisturbed control rats. Restraint in the absence of D-22 did not significantly affect MH CORT or 5-HT concentrations. However, perfusion of the MH with D-22 during restraint led to an increased magnitude and duration of extracellular 5-HT concentrations, relative to D-22 by itself. These results are consistent with the hypothesis that OCTs in the DMH contribute to the clearance of 5-HT from the extracellular fluid under both baseline conditions and mild restraint.
Collapse
Affiliation(s)
- Na Feng
- Department of Biology, University of South Dakota, Vermillion, SD 57069-2390, USA
| | | | | | | | | | | |
Collapse
|
31
|
Feng N, Telefont M, Kelly KJ, Orchinik M, Forster GL, Renner KJ, Lowry CA. Local perfusion of corticosterone in the rat medial hypothalamus potentiates D-fenfluramine-induced elevations of extracellular 5-HT concentrations. Horm Behav 2009; 56:149-57. [PMID: 19371745 DOI: 10.1016/j.yhbeh.2009.03.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Revised: 03/27/2009] [Accepted: 03/31/2009] [Indexed: 01/11/2023]
Abstract
The dorsomedial hypothalamus (DMH) plays an important role in coordinating physiological and behavioral responses to stress-related stimuli. In vertebrates, DMH serotonin (5-HT) concentrations increase rapidly in response to acute stressors or corticosterone (CORT). Recent studies suggest that CORT inhibits postsynaptic clearance of 5-HT from the extracellular fluid in the DMH by blocking organic cation transporter 3 (OCT3), a polyspecific CORT-sensitive transport protein. Because OCTs are low-affinity, high-capacity transporters, we hypothesized that CORT effects on extracellular 5-HT are most pronounced in the presence of elevated 5-HT release. We predicted that local application of CORT into the DMH would potentiate the effects of d-fenfluramine, a 5-HT-releasing agent, on extracellular 5-HT. These experiments were conducted using in vivo microdialysis in freely-moving male Sprague-Dawley rats implanted with a microdialysis probe into the medial hypothalamus (MH), which includes the DMH. In Experiment 1, rats simultaneously received intraperitoneal (i.p.) injections of 1 mg/kg D-fenfluramine or saline and either 200 ng/mL CORT or dilute ethanol (EtOH) vehicle delivered to the MH by reverse-dialysis for 40 min. In Experiment 2, 5 microM D-fenfluramine and either 200 ng/mL CORT or EtOH vehicle were concurrently delivered to the MH for 40 min using reverse-dialysis. CORT potentiated the increases in extracellular 5-HT concentrations induced by either i.p. or intra-MH administration of D-fenfluramine. Furthermore, CORT and D-fenfluramine interacted to alter home cage behaviors. Our results support the hypothesis that CORT inhibition of OCT3-mediated 5-HT clearance from the extracellular fluid contributes to stress-induced increases in extracellular 5-HT and 5-HT signaling.
Collapse
Affiliation(s)
- Na Feng
- Department of Biology and Neuroscience Group, University of South Dakota, Vermillion, SD 57069, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Lowry CA, Hale MW, Burke KA, Renner KJ, Moore FL. Fluoxetine potentiates the effects of corticotropin-releasing factor on locomotor activity and serotonergic systems in the roughskin newt, Taricha granulosa. Horm Behav 2009; 56:177-84. [PMID: 19409390 DOI: 10.1016/j.yhbeh.2009.04.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Revised: 04/08/2009] [Accepted: 04/23/2009] [Indexed: 11/23/2022]
Abstract
The anxiety- and stress-related neuropeptide corticotropin-releasing factor (CRF) elicits behavioral changes in vertebrates including increases in behavioral arousal and locomotor activity. Intracerebroventricular injections of CRF in an amphibian, the roughskin newt (Taricha granulosa), induces rapid increases in locomotor activity in both intact and hypophysectomized animals. We hypothesized that this CRF-induced increase in locomotor activity involves a central effect of CRF on serotonergic neurons, based on known stimulatory actions of serotonin (5-hydroxytryptamine, 5-HT) on spinal motor neurons and the central pattern generator for locomotor activity in vertebrates. In Experiment 1, we found that neither intracerebroventricular injections of low doses of CRF (25 ng) nor the selective serotonin reuptake inhibitor fluoxetine (10, 100 ng), by themselves, altered locomotor activity. In contrast, newts treated concurrently with CRF and fluoxetine responded with marked increases in locomotor activity. In Experiment 2, we found that increases in locomotor activity following co-administration of CRF (25 ng) and fluoxetine (100 ng) were associated with decreased 5-HT concentrations in a number of forebrain structures involved in regulation of emotional behavior and emotional states, including the ventral striatum, amygdala pars lateralis, and dorsal hypothalamus, measured 37 min after treatment. These results are consistent with the hypothesis that CRF stimulates locomotor activity through activation of serotonergic systems.
Collapse
|
33
|
Gasser PJ, Orchinik M, Raju I, Lowry CA. Distribution of organic cation transporter 3, a corticosterone-sensitive monoamine transporter, in the rat brain. J Comp Neurol 2009; 512:529-55. [PMID: 19025979 DOI: 10.1002/cne.21921] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Organic cation transporter 3 (OCT3) is a high-capacity, low-affinity transporter that mediates bidirectional, sodium-independent transport of dopamine, norepinephrine, epinephrine, serotonin, and histamine. OCT3-mediated transport is directly inhibited by corticosterone, suggesting a potential role for the transporter in mediating some of the effects of stress and glucocorticoids on monoaminergic neurotransmission. To elucidate the importance of OCT3 in clearance of extracellular monoamines in the brain, we used immunohistochemical techniques to describe the distribution of OCT3-like-immunoreactive (OCT3-ir) cells throughout the rostrocaudal extent of adult male rat brains. OCT3-ir cell bodies were widely distributed throughout the brain, with the highest densities observed in the superior and inferior colliculi, islands of Calleja, subiculum, lateral septum, lateral and dorsomedial hypothalamic nuclei, and granule cell layers of the main and accessory olfactory bulbs, the cerebellum, and the retrosplenial granular cortex. OCT3-ir cells and/or fibers were also observed in circumventricular organs, and OCT3-ir ependymal cells were observed in the linings of all cerebral ventricles. The widespread distribution of OCT3-ir cell bodies, including regions receiving dense monoaminergic projections, suggests an important role for this transporter in regulating extracellular concentrations of monoamines in the rat brain and is consistent with the hypothesis that corticosterone-induced inhibition of OCT3-mediated transport may contribute to effects of acute stress or corticosterone on monoaminergic neurotransmission.
Collapse
Affiliation(s)
- Paul J Gasser
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin 53201-1881, USA.
| | | | | | | |
Collapse
|
34
|
Nishida Y, Yoshioka M, Ray CA, Bolduc C, Tanaka H, St-Amand J. Regulation of pituitary gene expression by adrenalectomy. Obesity (Silver Spring) 2009; 17:114-20. [PMID: 19107126 DOI: 10.1038/oby.2008.484] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Excessive secretion of adrenal hormones, such as glucocorticoid and mineralocorticoid, leads to metabolic syndrome, including insulin resistance, obesity, and hypertension. These metabolic abnormalities are ameliorated by adrenalectomy (ADX). To identify pituitary mediators for ADX-induced physiological alterations, such as weight loss and hypotension, we investigated the effect of ADX on the pituitary transcriptome using serial analysis of gene expression (SAGE). SAGE method is based on isolation of short sequence tags, which usually correspond to unique mRNA species. The SAGE libraries were constructed from pituitary gland of intact (n = 51) and ADX (n = 12) mice. Thirty-one transcripts were differentially expressed between intact and ADX. Three transcripts encoding for proopiomelanocortin and three other transcripts involved in regulation of hormone secretion (neuromedin B, proprotein convertase subtilisin/kexin type 2, and IA-2) were induced by ADX. In addition, ADX increased the expression levels of genes encoding for cation extracellular matrix (matrix gamma-carboxyglutamate protein) and transport (solute carrier family 22 member 17). Conversely, ADX downregulated two transcripts involved in mitochondrial oxidative phosphorylation (nicotinamide adenine dinucleotide (NADH) dehydrogenase 3 and cytochrome c oxidase 3). Moreover, ADX significantly modulated the expression levels of one gene with uncharacterized function and 20 novel transcripts. This study reveals alterations of pituitary gene expressions that may be associated with ADX-induced physiological changes including weight loss.
Collapse
Affiliation(s)
- Yuichiro Nishida
- Department of Anatomy and Physiology, Functional Genomics Laboratory, Molecular Endocrinology and Oncology Research Center, Laval University Medical Center, Laval University, Quebec City, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
35
|
Organic cation transporter 3: Keeping the brake on extracellular serotonin in serotonin-transporter-deficient mice. Proc Natl Acad Sci U S A 2008; 105:18976-81. [PMID: 19033200 DOI: 10.1073/pnas.0800466105] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Mood disorders cause much suffering and are the single greatest cause of lost productivity worldwide. Although multiple medications, along with behavioral therapies, have proven effective for some individuals, millions of people lack an effective therapeutic option. A common serotonin (5-HT) transporter (5-HTT/SERT, SLC6A4) polymorphism is believed to confer lower 5-HTT expression in vivo and elevates risk for multiple mood disorders including anxiety, alcoholism, and major depression. Importantly, this variant is also associated with reduced responsiveness to selective 5-HT reuptake inhibitor antidepressants. We hypothesized that a reduced antidepressant response in individuals with a constitutive reduction in 5-HTT expression could arise because of the compensatory expression of other genes that inactivate 5-HT in the brain. A functionally upregulated alternate transporter for 5-HT may prevent extracellular 5-HT from rising to levels sufficiently high enough to trigger the adaptive neurochemical events necessary for therapeutic benefit. Here we demonstrate that expression of the organic cation transporter type 3 (OCT3, SLC22A3), which also transports 5-HT, is upregulated in the brains of mice with constitutively reduced 5-HTT expression. Moreover, the OCT blocker decynium-22 diminishes 5-HT clearance and exerts antidepressant-like effects in these mice but not in WT animals. OCT3 may be an important transporter mediating serotonergic signaling when 5-HTT expression or function is compromised.
Collapse
|
36
|
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: 162] [Impact Index Per Article: 9.5] [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
|
37
|
Vialou V, Balasse L, Callebert J, Launay JM, Giros B, Gautron S. Altered aminergic neurotransmission in the brain of organic cation transporter 3-deficient mice. J Neurochem 2008; 106:1471-82. [PMID: 18513366 DOI: 10.1111/j.1471-4159.2008.05506.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Organic cation transporters (OCTs) are carrier-type polyspecific permeases known to participate in low-affinity extraneuronal catecholamine uptake in peripheral tissues. OCT3 is the OCT subtype most represented in the brain, yet its implication in central aminergic neurotransmission in vivo had not been directly demonstrated. In a detailed immunohistochemistry study, we show that OCT3 is expressed in aminergic pathways in the mouse brain, particularly in dopaminergic neurons of the substantia nigra compacta, non-aminergic neurons of the ventral tegmental area, substantia nigra reticulata (SNr), locus coeruleus, hippocampus and cortex. Although OCT3 was found mainly in neurons, it was also occasionally detected in astrocytes in the SNr, hippocampus and several hypothalamic nuclei. In agreement with this distribution, OCT3/Slc22a3-deficient mice show evidence of altered monoamine neurotransmission in the brain, with decreased intracellular content and increased turnover of aminergic transmitters. The behavioral characterization of these mutants reveal subtle behavioral alterations such as increased sensitivity to psychostimulants and increased levels of anxiety and stress. Altogether our data support a role of OCT3 in the homeostatic regulation of aminergic neurotransmission in the brain.
Collapse
Affiliation(s)
- Vincent Vialou
- Inserm U513, Université Paris VI, Paris, France, and UPMC, Université Paris VI, Paris, France
| | | | | | | | | | | |
Collapse
|
38
|
Rahman Z, Ring RH, Young K, Platt B, Lin Q, Schechter LE, Rosenzweig-Lipson S, Beyer CE. Inhibition of uptake 2 (or extraneuronal monoamine transporter) by normetanephrine potentiates the neurochemical effects of venlafaxine. Brain Res 2008; 1203:68-78. [PMID: 18321472 DOI: 10.1016/j.brainres.2008.01.062] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2007] [Revised: 01/15/2008] [Accepted: 01/21/2008] [Indexed: 01/11/2023]
Abstract
Two distinct norepinephrine (NE) transporter mechanisms (uptake 1 and uptake 2) regulate extracellular NE concentrations. An association has been observed between the gradual improvement in patients treated with antidepressants that inhibit the NE transporter (NET/uptake 1) and increases in urinary normetanephrine, the O-methylated NE metabolite and potent inhibitor of uptake 2. These observations led to the hypothesis that increased levels of normetanephrine, and consequently inhibition of uptake 2, may partly be responsible for the clinical efficacy of some antidepressants. To investigate this hypothesis, we employed microdialysis techniques in the rat frontal cortex to monitor extracellular changes in normetanephrine following chronic administration of the clinically effective antidepressant, venlafaxine (a serotonin (5-HT) and NE reuptake inhibitor). We evaluated the neurochemical effects of inhibiting uptake 2 alone, or in conjunction with venlafaxine, on extracellular levels of NE and 5-HT. Chronic venlafaxine administration (14 days, 10 mg/kg, s.c.) elicited significant increases in cortical NE and 5-HT while producing a non-significant trend to increase cortical levels of normetanephrine. Additional studies revealed that combining normetanephrine with venlafaxine (10 mg/kg, i.p.), at a dose of normetanephrine (10 mg/kg, i.p.) that did not produce changes in extracellular levels of NE on its own, potentiated antidepressant-induced increases in extracellular NE. We also report mouse behavioral data involving the tail suspension test that complement the neurochemical observations. These preclinical findings, taken together, suggest that inhibiting both uptake 1 and uptake 2 via venlafaxine and normetanephrine, respectively, elicits a greater increase in cortical levels of NE than inhibiting either transporter alone.
Collapse
Affiliation(s)
- Zia Rahman
- Wyeth Research, Discovery Neuroscience, Depression and Anxiety Disorders, CN 8000, Princeton, NJ 08543-8000, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Koepsell H, Lips K, Volk C. Polyspecific organic cation transporters: structure, function, physiological roles, and biopharmaceutical implications. Pharm Res 2007. [PMID: 17473959 DOI: 10.1007/s11095‐007‐9254‐z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The body is equipped with broad-specificity transporters for the excretion and distribution of endogeneous organic cations and for the uptake, elimination and distribution of cationic drugs, toxins and environmental waste products. This group of transporters consists of the electrogenic cation transporters OCT1-3 (SLC22A1-3), the cation and carnitine transporters OCTN1 (SLC22A4), OCTN2 (SLC22A5) and OCT6 (SLC22A16), and the proton/cation antiporters MATE1, MATE2-K and MATE2-B. The transporters show broadly overlapping sites of expression in many tissues such as small intestine, liver, kidney, heart, skeletal muscle, placenta, lung, brain, cells of the immune system, and tumors. In epithelial cells they may be located in the basolateral or luminal membranes. Transcellular cation movement in small intestine, kidney and liver is mediated by the combined action of electrogenic OCT-type uptake systems and MATE-type efflux transporters that operate as cation/proton antiporters. Recent data showed that OCT-type transporters participate in the regulation of extracellular concentrations of neurotransmitters in brain, mediate the release of acetylcholine in non-neuronal cholinergic reactions, and are critically involved in the regulation of histamine release from basophils. The recent identification of polymorphisms in human OCTs and OCTNs allows the identification of patients with an increased risk for adverse drug reactions. Transport studies with expressed OCTs will help to optimize pharmacokinetics during development of new drugs.
Collapse
Affiliation(s)
- Hermann Koepsell
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University, Würzburg, Germany.
| | | | | |
Collapse
|
40
|
Koepsell H, Lips K, Volk C. Polyspecific organic cation transporters: structure, function, physiological roles, and biopharmaceutical implications. Pharm Res 2007; 24:1227-51. [PMID: 17473959 DOI: 10.1007/s11095-007-9254-z] [Citation(s) in RCA: 755] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Accepted: 01/26/2007] [Indexed: 12/11/2022]
Abstract
The body is equipped with broad-specificity transporters for the excretion and distribution of endogeneous organic cations and for the uptake, elimination and distribution of cationic drugs, toxins and environmental waste products. This group of transporters consists of the electrogenic cation transporters OCT1-3 (SLC22A1-3), the cation and carnitine transporters OCTN1 (SLC22A4), OCTN2 (SLC22A5) and OCT6 (SLC22A16), and the proton/cation antiporters MATE1, MATE2-K and MATE2-B. The transporters show broadly overlapping sites of expression in many tissues such as small intestine, liver, kidney, heart, skeletal muscle, placenta, lung, brain, cells of the immune system, and tumors. In epithelial cells they may be located in the basolateral or luminal membranes. Transcellular cation movement in small intestine, kidney and liver is mediated by the combined action of electrogenic OCT-type uptake systems and MATE-type efflux transporters that operate as cation/proton antiporters. Recent data showed that OCT-type transporters participate in the regulation of extracellular concentrations of neurotransmitters in brain, mediate the release of acetylcholine in non-neuronal cholinergic reactions, and are critically involved in the regulation of histamine release from basophils. The recent identification of polymorphisms in human OCTs and OCTNs allows the identification of patients with an increased risk for adverse drug reactions. Transport studies with expressed OCTs will help to optimize pharmacokinetics during development of new drugs.
Collapse
Affiliation(s)
- Hermann Koepsell
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University, Würzburg, Germany.
| | | | | |
Collapse
|
41
|
Zhou M, Engel K, Wang J. Evidence for significant contribution of a newly identified monoamine transporter (PMAT) to serotonin uptake in the human brain. Biochem Pharmacol 2007; 73:147-54. [PMID: 17046718 PMCID: PMC1828907 DOI: 10.1016/j.bcp.2006.09.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2006] [Revised: 09/07/2006] [Accepted: 09/08/2006] [Indexed: 01/20/2023]
Abstract
The high affinity serotonin transporter (SERT) constitutes the principal pathway for removal of serotonin (5-HT) from extracellular fluid of brain, but evidence indicates that other transporters may also be involved in this process. We recently reported the cloning of a novel plasma membrane monoamine transporter (PMAT), which is abundantly expressed in the human brain and avidly transports 5-HT [Engel K, Zhou M, Wang J. Identification and characterization of a novel monoamine transporter in the human brain. J Biol Chem 2004;279:50042-9]. In this study, we evaluated whether PMAT contributes to total human brain uptake of 5-HT using a hybrid depletion approach in Xenopus laevis oocytes. We also examined whether PMAT interacts with selective serotonin reuptake inhibitors (SSRIs) using MDCK cells stably expressing recombinant human PMAT. Microinjection of total human brain poly(A)(+) mRNA into oocytes elicited approximately 2.5-3-fold increase in 5-HT uptake. Pre-hybridization of poly(A)(+) mRNA with PMAT or SERT antisense oligonucleotides significantly reduced mRNA-induced 5-HT uptake. An additive inhibitory effect was observed when poly(A)(+) mRNA was co-hybridized with both PMAT and SERT antisense oligonucleotides. In contrast, mRNA-induced 5-HT uptake was not affected by pre-hybridization with sense oligonucleotides. These data suggest that functional transcripts of PMAT are present in the human brain, and the PMAT transporter may be significantly involved in brain uptake of 5-HT. All five tested SSRIs inhibited PMAT with IC(50) values ranging from 11 to 116 microM, which are much greater than clinically encountered concentrations, suggesting that PMAT activity is minimally affected by SSRI therapies.
Collapse
Affiliation(s)
- Mingyan Zhou
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | | | - Joanne Wang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
42
|
Gasser PJ, Lowry CA, Orchinik M. Corticosterone-sensitive monoamine transport in the rat dorsomedial hypothalamus: potential role for organic cation transporter 3 in stress-induced modulation of monoaminergic neurotransmission. J Neurosci 2006; 26:8758-66. [PMID: 16928864 PMCID: PMC6674371 DOI: 10.1523/jneurosci.0570-06.2006] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2006] [Revised: 06/06/2006] [Accepted: 07/18/2006] [Indexed: 12/17/2022] Open
Abstract
Glucocorticoid hormones act within the brain to alter physiological and behavioral responses to stress-related stimuli. Previous studies indicated that acute stressors can increase serotonin [5-hydroxytryptamine (5-HT)] concentrations in the dorsomedial hypothalamus (DMH), a midline hypothalamic structure involved in the integration of physiological and behavioral responses to stress. The current study tests the hypothesis that rapid, stress-induced accumulation of 5-HT is attributable to the inhibition of 5-HT transport via organic cation transporters (OCTs). OCTs are a family of high-capacity, bidirectional, multispecific transporters of organic cations (including 5-HT, dopamine, and norepinephrine) only recently described in brain. In peripheral tissues, organic cation transport via some OCTs is inhibited by corticosterone. We examined the expression and function of OCTs in the periventricular medial hypothalamus of male Sprague Dawley rats using reverse-transcriptase (RT)-PCR, immunohistochemistry, and in vitro transport assays. RT-PCR revealed expression of OCT3 mRNA, but not OCT1 or OCT2 mRNA, in the medial hypothalamus. OCT3-like immunoreactivity was observed in ependymal and glial-like cells in the DMH. Acutely prepared minces of rat medial hypothalamic tissue accumulated the OCT substrates [3H]-histamine and [3H]-N-methyl-4-phenylpyridinium ([3H]-MPP+). Consistent with the pharmacological profile of OCT3, corticosterone, 5-HT, estradiol, and the OCT inhibitor decynium22 dose-dependently inhibited histamine accumulation. Corticosterone and decynium22 also inhibited efflux of [3H]-MPP+ from hypothalamic minces. These data support the hypothesis that corticosterone-induced inhibition of OCT3 mediates stress-induced accumulation of 5-HT in the DMH and suggest that corticosterone may acutely modulate physiological and behavioral responses to stressors by altering serotonergic neurotransmission in this brain region.
Collapse
Affiliation(s)
- Paul J Gasser
- School of Life Sciences, Arizona State University, Tempe, Arizona 85287-4501, USA.
| | | | | |
Collapse
|