Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cells ; 11(15)2022 08 08.
Article in English | MEDLINE | ID: mdl-35954298

ABSTRACT

Selective serotonin reuptake inhibitors (SSRIs) are less efficacious in treating depression in children than in adults. SSRIs block serotonin uptake via the high-affinity, low-capacity serotonin transporter. However, the low-affinity, high-capacity organic cation transporter 3 (OCT3) and plasma membrane monoamine transporter (PMAT) are emerging as important players in serotonin uptake. We hypothesized that OCT3 and/or PMAT are functionally upregulated in juveniles, thereby buffering SSRIs' ability to enhance serotonergic neurotransmission. Unlike in adult mice, we found the OCT/PMAT blocker, decynium-22, to have standalone antidepressant-like effects in juveniles. Using in vivo high-speed chronoamperometry, we found that juveniles clear serotonin from the CA3 region of the hippocampus ~2-fold faster than adult mice. Cell density did not differ between ages, suggesting that faster serotonin clearance in juveniles is unrelated to faster diffusion through the extracellular matrix. Western blot and immunohistochemistry showed that juvenile mice have modestly greater expression of PMAT than adults, whereas OCT3 expression in the CA3 region of the hippocampus was similar between ages. Together, these data suggest that faster serotonin clearance and antidepressant-like effects of decynium-22 in juvenile mice may be due to functionally upregulated PMAT. Faster serotonin clearance via PMAT in juveniles may contribute to reduced therapeutic efficacy of SSRIs in children relative to adults.


Subject(s)
Antidepressive Agents , Serotonin , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Cell Membrane/metabolism , Hippocampus/metabolism , Mice , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology
2.
Eur J Pharmacol ; 883: 173285, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32697958

ABSTRACT

Depression is a major health problem for which most patients are not effectively treated. This underscores a need to identify new targets for the development of antidepressants with improved efficacy. Studies have shown that blockade of low-affinity/high-capacity transporters, such as organic cation transporters (OCTs) and the plasma membrane monoamine transporter (PMAT), with decynium-22 can produce antidepressant-like effects and inhibit serotonin clearance in brain when the serotonin transporter is pharmacologically or genetically compromised. In vitro studies show that OCTs/PMAT are also capable of norepinephrine transport, raising the possibility that decynium-22 might enhance the antidepressant-like effects of norepinephrine transporter inhibitors. Using in vivo electrochemistry, we show that local administration of decynium-22 into dentate gyrus of hippocampus enhanced the ability of the norepinephrine transporter blocker, desipramine, but not the dual norepinephrine/serotonin transporter blocker venlafaxine, to inhibit norepinephrine clearance. In parallel, systemic administration of decynium-22 (0.32 mg/kg) enhanced the antidepressant-like effects of desipramine (32 mg/kg), but not those of venlafaxine, in the tail suspension test, underscoring the heterogeneous response of mice to antidepressants, including those that share similar mechanisms of action. Systemic administration of normetanephrine, a potent blocker of OCT3, failed to potentiate the antidepressant-like effects of desipramine, suggesting that the actions of decynium-22 to augment the antidepressant-like effects of desipramine are likely mediated by another OCT isoform and/or PMAT. Taken together with existing literature, concurrent blockade of OCTs and/or PMAT merits further investigation as an adjunctive therapeutic for desipramine-like antidepressant drugs.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacology , Antidepressive Agents/pharmacology , Dentate Gyrus/drug effects , Depression/drug therapy , Desipramine/pharmacology , Norepinephrine/metabolism , Organic Cation Transport Proteins/antagonists & inhibitors , Quinolines/pharmacology , Serotonin and Noradrenaline Reuptake Inhibitors/pharmacology , Venlafaxine Hydrochloride/pharmacology , Animals , Behavior, Animal/drug effects , Dentate Gyrus/metabolism , Dentate Gyrus/physiopathology , Depression/metabolism , Depression/physiopathology , Depression/psychology , Disease Models, Animal , Locomotion/drug effects , Male , Mice, Inbred C57BL , Organic Cation Transport Proteins/metabolism
3.
Exp Neurol ; 314: 1-8, 2019 04.
Article in English | MEDLINE | ID: mdl-30605624

ABSTRACT

Exposure to acute intermittent hypoxia (AIH) induces a progressive increase of sympathetic nerve activity (SNA) that reflects a form of neuroplasticity known as sympathetic long-term facilitation (sLTF). Our recent findings indicate that activity of neurons in the hypothalamic paraventricular nucleus (PVN) contributes to AIH-induced sLTF, but neither the intra-PVN distribution nor the neurochemical identity of AIH responsive neurons has been determined. Here, awake rats were exposed to 10 cycles of AIH and c-Fos immunohistochemistry was performed to identify transcriptionally activated neurons in rostral, middle and caudal planes of the PVN. Effects of graded intensities of AIH were investigated in separate groups of rats (n = 6/group) in which inspired oxygen (O2) was reduced every 6 min from 21% to nadirs of 10%, 8% or 6%. All intensities of AIH failed to increase c-Fos counts in the caudally located lateral parvocellular region of the PVN. c-Fos counts increased in the dorsal parvocellular and central magnocellular regions, but significance was achieved only with AIH to 6% O2 (P < 0.002). By contrast, graded intensities of AIH induced graded c-Fos activation in the stress-related medial parvocellular (MP) region. Focusing on AIH exposure to 8% O2, experiments next investigated the stress-regulatory neuropeptide content of AIH-activated MP neurons. Tissue sections immunostained for corticotropin-releasing hormone (CRH) or arginine vasopressin (AVP) revealed a significantly greater number of neurons stained for CRH than AVP (P < 0.0001), though AIH induced expression of c-Fos in a similar fraction (~14%) of each neurochemical class. Amongst AIH-activated MP neurons, ~30% stained for CRH while only ~2% stained for AVP. Most AIH-activated CRH neurons (~82%) were distributed in the rostral one-half of the PVN. Results indicate that AIH recruits CRH, but not AVP, neurons in rostral to middle levels of the MP region of PVN, and raise the possibility that these CRH neurons may be a substrate for AIH-induced sLTF neuroplasticity.


Subject(s)
Hypoxia/physiopathology , Neuronal Plasticity , Paraventricular Hypothalamic Nucleus/physiopathology , Sympathetic Nervous System/physiopathology , Animals , Arginine Vasopressin/metabolism , Arterial Pressure , Cell Count , Corticotropin-Releasing Hormone/metabolism , Heart Rate , Long-Term Potentiation , Male , Neuropeptides/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley
4.
Front Behav Neurosci ; 13: 271, 2019.
Article in English | MEDLINE | ID: mdl-31920580

ABSTRACT

Stress-related neuropsychiatric (e.g., anxiety, depression) and cardiovascular diseases are frequently comorbid, though discerning the directionality of their association has been challenging. One of the most controllable risk factors for cardiovascular disease is salt intake. Though high salt intake is implicated in neuropsychiatric diseases, its direct neurobehavioral effects have seldom been explored. We reported that elevated salt intake in mice augments neuroinflammation, particularly after an acute stressor. Here, we explored how high salt consumption affected behavioral responses of mice to mildly arousing environmental and social tests, then assessed levels of the stress-related hormone corticosterone. Unexpectedly, anxiety-related behaviors in the elevated plus maze, open field, and marble burying test were unaffected by increased salt intake. However, nest building was diminished in mice consuming high salt, and voluntary social interaction was elevated, suggesting reduced engagement in ethologically-relevant behaviors that promote survival by attenuating threat exposure. Moreover, we observed significant positive correlations between social preference and subsequent corticosterone only in mice consuming increased salt, as well as negative correlations between open arm exploration in the elevated plus maze and corticosterone selectively in mice in the highest salt condition. Thus, heightened salt consumption reduces behavioral inhibition under relatively low-threat conditions, and enhances circulating corticosterone proportional to specific behavioral shifts. Considering the adverse health consequences of high salt intake, combined with evidence that increased salt consumption impairs inhibition of context-inappropriate behaviors, we suggest that prolonged high salt intake likely promulgates maladaptive behavioral and cardiovascular responses to perceived stressors that may explain some of the prevalent comorbidity between cardiovascular and neuropsychiatric diseases.

5.
Int J Neuropsychopharmacol ; 22(2): 137-142, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30535261

ABSTRACT

High dietary salt intake increases risk of stress-related neuropsychiatric disorders. Here, we explored the contribution of high dietary salt intake-induced neuroinflammation in key stress-responsive brain regions, the hypothalamic paraventricular nucleus and basolateral amygdala, in promoting exaggerated neuronal activation and coping behaviors in response to acute psychogenic stress. Mice that underwent high dietary salt intake exhibited increased active stress coping behaviors during and after an acute swim stress, and these were reduced by concurrent administration of minocycline, an inhibitor of microglial activation, without affecting body fluid hyperosmolality caused by high dietary salt intake. Moreover, minocycline attenuated high dietary salt intake-induced increases of paraventricular nucleus tumor necrosis factor-α, activated microglia (ionized calcium-binding adaptor molecule 1), and acute swim stress-induced neuronal activation (c-Fos). In the basolateral amygdala, similar effects were observed on ionized calcium-binding adaptor molecule 1+ and c-Fos+ counts, but not tumor necrosis factor-α levels. These data indicate that high dietary salt intake promotes neuroinflammation, increasing recruitment of neurons in key stress-associated brain regions and augmenting behavioral hyper-responsivity to acute psychological stress.


Subject(s)
Adaptation, Psychological , Anti-Inflammatory Agents/pharmacology , Basolateral Nuclear Complex , Brain , Inflammation , Microglia , Minocycline/pharmacology , Paraventricular Hypothalamic Nucleus , Sodium Chloride, Dietary/adverse effects , Stress, Psychological , Adaptation, Psychological/drug effects , Animals , Basolateral Nuclear Complex/drug effects , Basolateral Nuclear Complex/immunology , Basolateral Nuclear Complex/metabolism , Behavior, Animal/drug effects , Brain/drug effects , Brain/immunology , Brain/physiopathology , Disease Models, Animal , Inflammation/complications , Inflammation/drug therapy , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/immunology , Paraventricular Hypothalamic Nucleus/metabolism , Stress, Psychological/drug therapy , Stress, Psychological/immunology , Stress, Psychological/physiopathology
6.
Behav Pharmacol ; 29(4): 365-369, 2018 06.
Article in English | MEDLINE | ID: mdl-29035921

ABSTRACT

In humans, chronic treatment with selective serotonin reuptake inhibitors (SSRIs) decreases anxiety, unlike acute treatment, which can increase anxiety. Although this biphasic pattern is observed clinically, preclinical demonstrations are rare. In an animal model of antidepressant-induced anxiolytic effects, the novelty-induced hypophagia (NIH) test, a single administration of the SSRI citalopram reportedly elicited anxiogenic-like effects, whereas three administrations over 24 h were sufficient to produce anxiolytic-like effects. Extending these findings, the present study examined the effects of acute and repeated escitalopram in a similar NIH test in a commonly used mouse strain (i.e. C57BL/6J), analyzing results with a method (i.e. survival analysis) that can model the skewed distribution of latencies to consume food and that can deal with censored data (i.e. when consumption does not occur during the test). Saline-treated mice showed robust NIH. Acute escitalopram enhanced NIH, but did so only at a dose (i.e. 32 mg/kg) that similarly enhanced hypophagia in a familiar environment. The effects of escitalopram on NIH did not significantly change after repeated (three times) administration over 24 h. Additional studies are necessary to delineate the conditions under which rapid reversal of SSRI-induced anxiety can be modeled in animals using the NIH test.


Subject(s)
Anxiety/drug therapy , Behavior, Animal/drug effects , Citalopram/pharmacology , Animals , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Anxiety Disorders/drug therapy , Citalopram/metabolism , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Selective Serotonin Reuptake Inhibitors/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology
7.
J Pharmacol Exp Ther ; 360(1): 84-94, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27831486

ABSTRACT

Depression is a major public health concern with symptoms that are often poorly controlled by treatment with common antidepressants. This problem is compounded in juveniles and adolescents, because therapeutic options are limited to selective serotonin reuptake inhibitors (SSRIs). Moreover, therapeutic benefits of SSRIs are often especially limited in certain subpopulations of depressed patients, including children and carriers of low-expressing serotonin transporter (SERT) gene variants. Tricyclic antidepressants (TCAs) offer an alternative to SSRIs; however, how age and SERT expression influence antidepressant response to TCAs is not understood. We investigated the relation between antidepressant-like response to the TCA desipramine using the tail suspension test and saturation binding of [3H]nisoxetine to the norepinephrine transporter (NET), the primary target of desipramine, in juvenile (21 days postnatal [P21]), adolescent (P28), and adult (P90) wild-type (SERT+/+) mice. To model carriers of low-expressing SERT gene variants, we used mice with reduced SERT expression (SERT+/-) or lacking SERT (SERT-/-). The potency and maximal antidepressant-like effect of desipramine was greater in P21 mice than in P90 mice and was SERT genotype independent. NET expression decreased with age in the locus coeruleus and increased with age in several terminal regions (e.g., the cornu ammonis CA1 and CA3 regions of the hippocampus). Binding affinity of [3H]nisoxetine did not vary as a function of age or SERT genotype. These data show age-dependent shifts for desipramine to produce antidepressant-like effects that correlate with NET expression in the locus coeruleus and suggest that drugs with NET-blocking activity may be an effective alternative to SSRIs in juveniles.


Subject(s)
Antidepressive Agents/pharmacology , Desipramine/pharmacology , Gene Expression Regulation/drug effects , Mutation , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Aging/metabolism , Animals , Antidepressive Agents/therapeutic use , Desipramine/therapeutic use , Female , Genotype , Hindlimb Suspension , Hippocampus/drug effects , Hippocampus/metabolism , Immobility Response, Tonic/drug effects , Locus Coeruleus/drug effects , Locus Coeruleus/metabolism , Male , Mice
8.
J Pharmacol Exp Ther ; 358(2): 271-81, 2016 08.
Article in English | MEDLINE | ID: mdl-27288483

ABSTRACT

Depression is a disabling affective disorder for which the majority of patients are not effectively treated. This problem is exacerbated in children and adolescents for whom only two antidepressants are approved, both of which are selective serotonin reuptake inhibitor (SSRIs). Unfortunately SSRIs are often less effective in juveniles than in adults; however, the mechanism(s) underlying age-dependent responses to SSRIs is unknown. To this end, we compared the antidepressant-like response to the SSRI escitalopram using the tail suspension test and saturation binding of [(3)H]citalopram to the serotonin transporter (SERT), the primary target of SSRIs, in juvenile [postnatal day (P)21], adolescent (P28), and adult (P90) wild-type (SERT+/+) mice. In addition, to model individuals carrying low-expressing SERT variants, we studied mice with reduced SERT expression (SERT+/-) or lacking SERT (SERT-/-). Maximal antidepressant-like effects were less in P21 mice relative to P90 mice. This was especially apparent in SERT+/- mice. However, the potency for escitalopram to produce antidepressant-like effects in SERT+/+ and SERT+/- mice was greater in P21 and P28 mice than in adults. SERT expression increased with age in terminal regions and decreased with age in cell body regions. Binding affinity values did not change as a function of age or genotype. As expected, in SERT-/- mice escitalopram produced no behavioral effects, and there was no specific [(3)H]citalopram binding. These data reveal age- and genotype-dependent shifts in the dose-response for escitalopram to produce antidepressant-like effects, which vary with SERT expression, and may contribute to the limited therapeutic response to SSRIs in juveniles and adolescents.


Subject(s)
Antidepressive Agents/pharmacology , Citalopram/pharmacology , Gene Expression Regulation/drug effects , Mutation , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Aging/metabolism , Animals , Behavior, Animal/drug effects , Female , Genotype , Hindlimb Suspension , Hippocampus/drug effects , Hippocampus/metabolism , Immobility Response, Tonic/drug effects , Male , Mice , Raphe Nuclei/drug effects , Raphe Nuclei/metabolism , Serotonin/metabolism
9.
Genes Brain Behav ; 14(7): 543-549, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26250357

ABSTRACT

Monoamine uptake inhibitors are common treatments for depression; however, the therapeutic efficacy of these drugs varies widely. Two factors that are commonly linked to clinical outcome are age and serotonin transporter (SERT) genotype. Mouse models provide powerful tools to study consequences of age and genotype on antidepressant-like efficacy; however, to date, systematic studies of this nature are lacking. Here, we used the tail suspension test (TST), a preclinical assay for antidepressant efficacy, to gain insight into age and SERT genotype dependency of immobility time in the TST under control conditions (saline injection) and in response to the tricyclic antidepressant, desipramine (DMI). Immobility after saline injection in juvenile, adolescent, adult, mature adult and middle-aged mice (postnatal days 21, 28, 90, 210 and 300, respectively) significantly increased with age; however, the rate of increase was slower for SERT null (-/-) mice than for wild-type (+/+) or heterozygote (+/-) mice. Desipramine reduced immobility across ages and SERT genotypes. Middle-aged, but not adult, SERT(-/-) mice were significantly more sensitive to DMI than age-matched SERT(+/+) or SERT(+/-) mice. Desipramine was less potent in middle-aged SERT(+/+) and SERT(+/-) mice than in adult SERT(+/+) or SERT(+/-) mice. Regardless of age, DMI's maximal effects were greater in SERT(-/-) mice than in SERT(+/+) or SERT(+/-) mice. These results show that immobility time in the TST varies as a function of age and SERT genotype, underscoring the utility of the TST as a potential model to examine age- and SERT genotype-dependent influences on antidepressant response.


Subject(s)
Antidepressive Agents, Tricyclic/pharmacology , Desipramine/pharmacology , Genotype , Motor Activity/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Stress, Psychological/genetics , Age Factors , Animals , Antidepressive Agents, Tricyclic/therapeutic use , Desipramine/therapeutic use , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Stress, Psychological/drug therapy , Stress, Psychological/physiopathology
10.
Front Pharmacol ; 4: 131, 2013.
Article in English | MEDLINE | ID: mdl-24191152

ABSTRACT

Depression is a major health problem for which most patients are not effectively treated. This problem is further compounded in children and adolescents where only two antidepressants [both selective serotonin reuptake inhibitors (SSRIs)] are currently approved for clinical use. Mouse models provide tools to identify mechanisms that might account for poor treatment response to antidepressants. However, there are few studies in adolescent mice and none in juvenile mice. The tail suspension test (TST) is commonly used to assay for antidepressant-like effects of drugs in adult mice. Here we show that the TST can also be used to assay antidepressant-like effects of drugs in C57Bl/6 mice aged 21 (juvenile) and 28 (adolescent) days post-partum (P). We found that the magnitude of antidepressant-like response to the SSRI escitalopram was less in P21 mice than in P28 or adult mice. The smaller antidepressant response of juveniles was not related to either maximal binding (B max) or affinity (K d) for [(3)H]citalopram binding to the serotonin transporter (SERT) in hippocampus, which did not vary significantly among ages. Magnitude of antidepressant-like response to the tricyclic desipramine was similar among ages, as were B max and K d values for [(3)H]nisoxetine binding to the norepinephrine transporter in hippocampus. Together, these findings suggest that juvenile mice are less responsive to the antidepressant-like effects of escitalopram than adults, but that this effect is not due to delayed maturation of SERT in hippocampus. Showing that the TST is a relevant behavioral assay of antidepressant-like activity in juvenile and adolescent mice sets the stage for future studies of the mechanisms underlying the antidepressant response in these young populations.

11.
J Neurosci ; 33(25): 10534-43, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23785165

ABSTRACT

Mood disorders cause much suffering and lost productivity worldwide, compounded by the fact that many patients are not effectively treated by currently available medications. The most commonly prescribed antidepressant drugs are the selective serotonin (5-HT) reuptake inhibitors (SSRIs), which act by blocking the high-affinity 5-HT transporter (SERT). The increase in extracellular 5-HT produced by SSRIs is thought to be critical to initiate downstream events needed for therapeutic effects. A potential explanation for their limited therapeutic efficacy is the recently characterized presence of low-affinity, high-capacity transporters for 5-HT in brain [i.e., organic cation transporters (OCTs) and plasma membrane monoamine transporter], which may limit the ability of SSRIs to increase extracellular 5-HT. Decynium-22 (D-22) is a blocker of these transporters, and using this compound we uncovered a significant role for OCTs in 5-HT uptake in mice genetically modified to have reduced or no SERT expression (Baganz et al., 2008). This raised the possibility that pharmacological inactivation of D-22-sensitive transporters might enhance the neurochemical and behavioral effects of SSRIs. Here we show that in wild-type mice D-22 enhances the effects of the SSRI fluvoxamine to inhibit 5-HT clearance and to produce antidepressant-like activity. This antidepressant-like activity of D-22 was attenuated in OCT3 KO mice, whereas the effect of D-22 to inhibit 5-HT clearance in the CA3 region of hippocampus persisted. Our findings point to OCT3, as well as other D-22-sensitive transporters, as novel targets for new antidepressant drugs with improved therapeutic potential.


Subject(s)
Antidepressive Agents/pharmacology , Depression/drug therapy , Quinolines/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Blood-Brain Barrier , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Drug Synergism , Electrophysiological Phenomena , Fluvoxamine/pharmacology , Hindlimb Suspension , Hippocampus , Mice , Mice, Inbred C57BL , Mice, Knockout , Microinjections , Neurotransmitter Transport Proteins/antagonists & inhibitors , Neurotransmitter Transport Proteins/metabolism , Octamer Transcription Factor-3/genetics , Quinolines/pharmacokinetics , Serotonin/metabolism , Serotonin Syndrome/psychology , Spectrophotometry, Ultraviolet
12.
ACS Chem Neurosci ; 4(1): 16-21, 2013 Jan 16.
Article in English | MEDLINE | ID: mdl-23336039

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.


Subject(s)
Antidepressive Agents/therapeutic use , Depressive Disorder, Major/drug therapy , Selective Serotonin Reuptake Inhibitors/therapeutic use , Depressive Disorder, Major/etiology , Humans , Models, Biological , Organic Cation Transport Proteins/drug effects , Serotonin/physiology , Serotonin Plasma Membrane Transport Proteins/drug effects , Serotonin Plasma Membrane Transport Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...