Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Biomedicines ; 10(1)2022 Jan 04.
Article in English | MEDLINE | ID: mdl-35052781

ABSTRACT

The striatum contains several types of neurons including medium spiny projection neurons (MSNs), cholinergic interneurons (ChIs), and fast-spiking interneurons (FSIs). Modulating the activity of these neurons by the dopamine D2 receptor (D2R) can greatly impact motor control and movement disorders. D2R exists in two isoforms: D2L and D2S. Here, we assessed whether alterations in the D2L and D2S expression levels affect neuronal excitability and synaptic function in striatal neurons. We observed that quinpirole inhibited the firing rate of all three types of striatal neurons in wild-type (WT) mice. However, in D2L knockout (KO) mice, quinpirole enhanced the excitability of ChIs, lost influence on spike firing of MSNs, and remained inhibitory effect on spike firing of FSIs. Additionally, we showed mIPSC frequency (but not mIPSC amplitude) was reduced in ChIs from D2L KO mice compared with WT mice, suggesting spontaneous GABA release is reduced at GABAergic terminals onto ChIs in D2L KO mice. Furthermore, we found D2L deficiency resulted in reduced dendritic spine density in ChIs, suggesting D2L activation plays a role in the formation/maintenance of dendritic spines of ChIs. These findings suggest new molecular and cellular mechanisms for causing ChIs abnormality seen in Parkinson's disease or drug-induced dyskinesias.

2.
Am J Psychiatry ; 178(1): 48-64, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32539528

ABSTRACT

OBJECTIVE: Pediatric obsessive-compulsive disorder (OCD) sometimes appears rapidly, even overnight, often after an infection. Pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections, or PANDAS, describes such a situation after infection with Streptococcus pyogenes. PANDAS may result from induced autoimmunity against brain antigens, although this remains unproven. Pilot work suggests that IgG antibodies from children with PANDAS bind to cholinergic interneurons (CINs) in the striatum. CIN deficiency has been independently associated with tics in humans and with repetitive behavioral pathology in mice, making it a plausible locus of pathology. The authors sought to replicate and extend earlier work and to investigate the cellular effects of PANDAS antibodies on cholinergic interneurons. METHODS: Binding of IgG to specific neurons in human and mouse brain slices was evaluated ex vivo after incubation with serum from 27 children with rigorously characterized PANDAS, both at baseline and after intravenous immunoglobulin (IVIG) treatment, and 23 matched control subjects. Binding was correlated with symptom measures. Neural activity after serum incubation was assessed in mouse slices using molecular markers and electrophysiological recording. RESULTS: IgG from children with PANDAS bound to CINs, but not to several other neuron types, more than IgG from control subjects, in three independent cohorts of patients. Post-IVIG serum had reduced IgG binding to CINs, and this reduction correlated with symptom improvement. Baseline PANDAS sera decreased activity of striatal CINs, but not of parvalbumin-expressing GABAergic interneurons, and altered their electrophysiological responses, in acute mouse brain slices. Post-IVIG PANDAS sera and IgG-depleted baseline sera did not alter the activity of striatal CINs. CONCLUSIONS: These findings provide strong evidence for striatal CINs as a critical cellular target that may contribute to pathophysiology in children with rapid-onset OCD symptoms, and perhaps in other conditions.


Subject(s)
Autoantibodies/immunology , Autoimmune Diseases/immunology , Cholinergic Neurons/immunology , Corpus Striatum/immunology , Obsessive-Compulsive Disorder/immunology , Streptococcal Infections/immunology , Animals , Autoimmune Diseases/complications , Case-Control Studies , Child , Child, Preschool , Cholinergic Neurons/physiology , Corpus Striatum/physiopathology , Female , Humans , Immunoglobulin G/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obsessive-Compulsive Disorder/complications , Obsessive-Compulsive Disorder/etiology , Streptococcal Infections/complications
3.
Mol Psychiatry ; 26(9): 5097-5111, 2021 09.
Article in English | MEDLINE | ID: mdl-32488125

ABSTRACT

Both the NMDA receptor (NMDAR) positive allosteric modulator (PAM), and antagonist, can exert rapid antidepressant effects as shown in several animal and human studies. However, how this bidirectional modulation of NMDARs causes similar antidepressant effects remains unknown. Notably, the initial cellular trigger, specific cell-type(s), and subunit(s) of NMDARs mediating the antidepressant-like effects of a PAM or an antagonist have not been identified. Here, we used electrophysiology, microdialysis, and NMR spectroscopy to evaluate the effect of a NMDAR PAM (rapastinel) or NMDAR antagonist, ketamine on NMDAR function and disinhibition-mediated glutamate release. Further, we used cell-type specific knockdown (KD), pharmacological, and behavioral approaches to dissect the cell-type specific role of GluN2B, GluN2A, and dopamine receptor subunits in the actions of NMDAR PAM vs. antagonists. We demonstrate that rapastinel directly enhances NMDAR activity on principal glutamatergic neurons in medial prefrontal cortex (mPFC) without any effect on glutamate efflux, while ketamine blocks NMDAR on GABA interneurons to cause glutamate efflux and indirect activation of excitatory synapses. Behavioral studies using cell-type-specific KD in mPFC demonstrate that NMDAR-GluN2B KD on Camk2a- but not Gad1-expressing neurons blocks the antidepressant effects of rapastinel. In contrast, GluN2B KD on Gad1- but not Camk2a-expressing neurons blocks the actions of ketamine. The results also demonstrate that Drd1-expressing pyramidal neurons in mPFC mediate the rapid antidepressant actions of ketamine and rapastinel. Together, these results demonstrate unique initial cellular triggers as well as converging effects on Drd1-pyramidal cell signaling that underlie the antidepressant actions of NMDAR-positive modulation vs. NMDAR blockade.


Subject(s)
Ketamine , Receptors, N-Methyl-D-Aspartate , Animals , Antidepressive Agents/pharmacology , Humans , Interneurons/metabolism , Ketamine/pharmacology , Prefrontal Cortex/metabolism , Pyramidal Cells/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
4.
Neuropsychopharmacology ; 46(4): 799-808, 2021 03.
Article in English | MEDLINE | ID: mdl-33059355

ABSTRACT

Dysregulation of the glutamatergic system and its receptors in medial prefrontal cortex (mPFC) has been implicated in major depressive disorder. Recent preclinical studies have shown that enhancing NMDA receptor (NMDAR) activity can exert rapid antidepressant-like effects. AGN-241751, an NMDAR positive allosteric modulator (PAM), is currently being tested as an antidepressant in clinical trials, but the mechanism and NMDAR subunit(s) mediating its antidepressant-like effects are unknown. We therefore used molecular, biochemical, and electrophysiological approaches to examine the cell-type-specific role of GluN2B-containing NMDAR in mediating antidepressant-like behavioral effects of AGN-241751. We demonstrate that AGN-241751 exerts antidepressant-like effects and reverses behavioral deficits induced by chronic unpredictable stress in mice. AGN-241751 treatment enhances NMDAR activity of excitatory and parvalbumin-inhibitory neurons in mPFC, activates Akt/mTOR signaling, and increases levels of synaptic proteins crucial for synaptic plasticity in the prefrontal cortex. Furthermore, cell-type-specific knockdown of GluN2B-containing NMDARs in mPFC demonstrates that GluN2B subunits on excitatory, but not inhibitory, neurons are necessary for antidepressant-like effects of AGN-241751. Together, these results demonstrate antidepressant-like actions of the NMDAR PAM AGN-241751 and identify GluN2B on excitatory neurons of mPFC as initial cellular trigger underlying these behavioral effects.


Subject(s)
Depressive Disorder, Major , Receptors, N-Methyl-D-Aspartate , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Depressive Disorder, Major/drug therapy , Mice , Neurons/metabolism , Prefrontal Cortex/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
5.
Neuropsychopharmacology ; 45(10): 1725-1734, 2020 09.
Article in English | MEDLINE | ID: mdl-32396921

ABSTRACT

We previously reported that the serotonergic system is important for the antidepressant-like effects of ketamine, a non-competitive N-methyl-D-aspartate receptor antagonist, which produces rapid and long-lasting antidepressant effects in patients with major depressive disorder (MDD). In particular, selective stimulation of the 5-HT1A receptor in the medial prefrontal cortex (mPFC), as opposed to the somatic 5-HT1A autoreceptor, has been shown to play a critical role in the antidepressant-like actions of ketamine. However, the detailed mechanisms underlying mPFC 5-HT1A receptor-mediated antidepressant-like effects are not fully understood. Here we examined the involvement of the glutamate AMPA receptor and brain-derived neurotrophic factor (BDNF) in the antidepressant-like effects of 5-HT1A receptor activation in the mPFC. The results show that intra-mPFC infusion of the 5-HT1A receptor agonist 8-OH-DPAT induces rapid and long-lasting antidepressant-like effects in the forced swim, novelty-suppressed feeding, female urine sniffing, and chronic unpredictable stress tests. In addition, the results demonstrate that the antidepressant-like effects of intra-mPFC infusion of 8-OH-DPAT are blocked by co-infusion of an AMPA receptor antagonist or an anti-BDNF neutralizing antibody. In addition, mPFC infusion of 8-OH-DPAT increased the phosphorylation of signaling proteins downstream of BDNF, including mTOR, ERK, 4EBP1, and p70S6K. Finally, selective stimulation of the 5-HT1A receptor increased levels of synaptic proteins and synaptic function in the mPFC. Collectively, these results indicate that selective stimulation of 5-HT1A receptor in the mPFC exerts rapid and sustained antidepressant-like effects via activation of AMPA receptor/BDNF/mTOR signaling in mice, which subsequently increase synaptic function in the mPFC, and provide evidence for the 5-HT1A receptor as a target for the treatment of MDD.


Subject(s)
Brain-Derived Neurotrophic Factor , Depressive Disorder, Major , Animals , Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Depression/drug therapy , Depressive Disorder, Major/drug therapy , Female , Humans , Mice , Prefrontal Cortex/metabolism , Receptor, Serotonin, 5-HT1A , Receptors, AMPA
6.
J Clin Invest ; 130(3): 1336-1349, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31743111

ABSTRACT

A single subanesthetic dose of ketamine, an NMDA receptor (NMDAR) antagonist, produces rapid and sustained antidepressant actions in depressed patients, addressing a major unmet need for the treatment of mood disorders. Ketamine produces a rapid increase in extracellular glutamate and synaptic formation in the prefrontal cortex, but the initial cellular trigger that initiates this increase and ketamine's behavioral actions has not been identified. To address this question, we used a combination of viral shRNA and conditional mutation to produce cell-specific knockdown or deletion of a key NMDAR subunit, GluN2B, implicated in the actions of ketamine. The results demonstrated that the antidepressant actions of ketamine were blocked by GluN2B-NMDAR knockdown on GABA (Gad1) interneurons, as well as subtypes expressing somatostatin (Sst) or parvalbumin (Pvalb), but not glutamate principle neurons in the medial prefrontal cortex (mPFC). Further analysis of GABA subtypes showed that cell-specific knockdown or deletion of GluN2B in Sst interneurons blocked or occluded the antidepressant actions of ketamine and revealed sex-specific differences that are associated with excitatory postsynaptic currents on mPFC principle neurons. These findings demonstrate that GluN2B-NMDARs on GABA interneurons are the initial cellular trigger for the rapid antidepressant actions of ketamine and show sex-specific adaptive mechanisms to GluN2B modulation.


Subject(s)
Antidepressive Agents/pharmacology , GABAergic Neurons/metabolism , Interneurons/metabolism , Ketamine/pharmacology , Sex Characteristics , Animals , Female , GABAergic Neurons/pathology , Gene Knockout Techniques , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Interneurons/pathology , Male , Mice , Mice, Transgenic , Parvalbumins/genetics , Parvalbumins/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Somatostatin/genetics , Somatostatin/metabolism
7.
Neurobiol Dis ; 134: 104669, 2020 02.
Article in English | MEDLINE | ID: mdl-31707118

ABSTRACT

Dysfunction of medial prefrontal cortex (mPFC) in association with imbalance of inhibitory and excitatory neurotransmission has been implicated in depression. However, the precise cellular mechanisms underlying this imbalance, particularly for GABAergic transmission in the mPFC, and the link with the rapid acting antidepressant ketamine remains poorly understood. Here we determined the influence of chronic unpredictable stress (CUS), an ethologically validated model of depression, on synaptic markers of GABA neurotransmission, and the influence of a single dose of ketamine on CUS-induced synaptic deficits in mPFC of male rodents. The results demonstrate that CUS decreases GABAergic proteins and the frequency of inhibitory post synaptic currents (IPSCs) of layer V mPFC pyramidal neurons, concomitant with depression-like behaviors. In contrast, a single dose of ketamine can reverse CUS-induced deficits of GABA markers, in conjunction with reversal of CUS-induced depressive-like behaviors. These findings provide further evidence of impairments of GABAergic synapses as key determinants of depressive behavior and highlight ketamine-induced synaptic responses that restore GABA inhibitory, as well as glutamate neurotransmission.


Subject(s)
Antidepressive Agents/administration & dosage , Depression/physiopathology , Ketamine/administration & dosage , Neurons/drug effects , Prefrontal Cortex/drug effects , Stress, Psychological/physiopathology , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/physiology , Animals , Disease Models, Animal , Inhibitory Postsynaptic Potentials/drug effects , Male , Mice, Inbred C57BL , Neurons/physiology , Prefrontal Cortex/physiopathology
8.
Neuropsychopharmacology ; 44(13): 2230-2238, 2019 12.
Article in English | MEDLINE | ID: mdl-31454827

ABSTRACT

Currently available antidepressants have a delayed onset and limited efficacy, highlighting the need for new, rapid and more efficacious agents. Ketamine, an NMDA receptor antagonist, has emerged as a new rapid-acting antidepressant, effective even in treatment resistant patients. However, ketamine induces undesired psychotomimetic and dissociative side effects that limit its clinical use. The d-stereoisomer of methadone (dextromethadone; REL-1017) is a noncompetitive NMDA receptor antagonist with an apparently favorable safety and tolerability profile. The current study examined the rapid and sustained antidepressant actions of d-methadone in several behavioral paradigms, as well as on mTORC1 signaling and synaptic changes in the medial prefrontal cortex (mPFC). A single dose of d-methadone promoted rapid and sustained antidepressant responses in the novelty-suppressed feeding test (NSFT), a measure of anxiety, and in the female urine sniffing test (FUST), a measure of motivation and reward. D-methadone also produced a rapid reversal of the sucrose preference deficit, a measure of anhedonia, in rats exposed to chronic unpredictable stress. D-methadone increased phospho-p70S6 kinase, a downstream target of mTORC1 in the mPFC, and intra-mPFC infusion of the selective mTORC1 inhibitor rapamycin blocked the antidepressant actions of d-methadone in the FUST and NSFT. D-methadone administration also increased levels of the synaptic proteins, PSD95, GluA1, and Synapsin 1 and enhanced synaptic function in the mPFC. Studies in primary cortical cultures show that d-methadone also increases BDNF release, as well as phospho-p70S6 kinase. These findings indicate that d-methadone induces rapid antidepressant actions through mTORC1-mediated synaptic plasticity in the mPFC similar to ketamine.


Subject(s)
Antidepressive Agents/administration & dosage , Mechanistic Target of Rapamycin Complex 1/metabolism , Methadone/administration & dosage , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Dendritic Spines/drug effects , Dendritic Spines/physiology , Excitatory Postsynaptic Potentials/drug effects , Ketamine/administration & dosage , Male , Rats, Sprague-Dawley , Signal Transduction/drug effects
9.
J Clin Invest ; 129(6): 2542-2554, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30990795

ABSTRACT

Preclinical studies demonstrate that rapid acting antidepressants, including ketamine require stimulation of mTORC1 signaling. This pathway is regulated by neuronal activity, endocrine and metabolic signals, notably the amino acid leucine, which activates mTORC1 signaling via binding to the upstream regulator sestrin. Here, we examined the antidepressant actions of NV-5138, a novel highly selective small molecule modulator of sestrin that penetrates the blood brain barrier. The results demonstrate that a single dose of NV-5138 produced rapid and long-lasting antidepressant effects, and rapidly reversed anhedonia caused by chronic stress exposure. The antidepressant actions of NV-5138 required BDNF release as the behavioral responses are blocked by infusion of a BDNF neutralizing antibody into the medial prefrontal cortex (mPFC) or in mice with a knock-in of a BDNF polymorphism that blocks activity dependent BDNF release. NV-5138 administration also rapidly increased synapse number and function in the mPFC, and reversed the synaptic deficits caused by chronic stress. Together, the results demonstrate that NV-5138 produced rapid synaptic and antidepressant behavioral responses via activation of the mTORC1 pathway and BDNF signaling, indicating that pharmacological modulation of sestrin is a novel approach for development of rapid acting antidepressants.


Subject(s)
Antidepressive Agents , Behavior, Animal/drug effects , Heat-Shock Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Synaptic Transmission/drug effects , Animals , Antidepressive Agents/chemistry , Antidepressive Agents/pharmacokinetics , Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Heat-Shock Proteins/genetics , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Synaptic Transmission/genetics
10.
Nat Commun ; 10(1): 223, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30644390

ABSTRACT

Impaired function in the medial prefrontal cortex (mPFC) contributes to depression, and the therapeutic response produced by novel rapid-acting antidepressants such as ketamine are mediated by mPFC activity. The mPFC contains multiple types of pyramidal cells, but it is unclear whether a particular subtype mediates the rapid antidepressant actions of ketamine. Here we tested two major subtypes, Drd1 and Drd2 dopamine receptor expressing pyramidal neurons and found that activating Drd1 expressing pyramidal cells in the mPFC produces rapid and long-lasting antidepressant and anxiolytic responses. In contrast, photostimulation of Drd2 expressing pyramidal cells was ineffective across anxiety-like and depression-like measures. Disruption of Drd1 activity also blocked the rapid antidepressant effects of ketamine. Finally, we demonstrate that stimulation of mPFC Drd1 terminals in the BLA recapitulates the antidepressant effects of somatic stimulation. These findings aid in understanding the cellular target neurons in the mPFC and the downstream circuitry involved in rapid antidepressant responses.


Subject(s)
Antidepressive Agents/pharmacology , Ketamine/pharmacology , Optogenetics , Prefrontal Cortex/drug effects , Receptors, Dopamine D1/metabolism , Animals , Basolateral Nuclear Complex/radiation effects , Dopamine Agonists , Female , Male , Mice, Inbred C57BL , Prefrontal Cortex/metabolism , Receptors, Dopamine D2/metabolism
11.
Proc Natl Acad Sci U S A ; 116(1): 297-302, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30559184

ABSTRACT

Ketamine, a noncompetitive N-methyl-d-aspartate (NMDA) receptor antagonist, produces rapid and long-lasting antidepressant effects in major depressive disorder (MDD) patients. (2R,6R)-Hydroxynorketamine [(2R,6R)-HNK], a metabolite of ketamine, is reported to produce rapid antidepressant effects in rodent models without the side effects of ketamine. Importantly, (2R,6R)-HNK does not block NMDA receptors like ketamine, and the molecular signaling mechanisms for (2R,6R)-HNK remain unknown. Here, we examined the involvement of BDNF/TrkB/mechanistic target of rapamycin complex 1 (mTORC1) signaling in the antidepressant actions of (2R,6R)-HNK. Intramedial prefrontal cortex (intra-mPFC) infusion or systemic (2R,6R)-HNK administration induces rapid and long-lasting antidepressant effects in behavioral tests, identifying the mPFC as a key region for the actions of (2R,6R)-HNK. The antidepressant actions of (2R,6R)-HNK are blocked in mice with a knockin of the BDNF Val66Met allele (which blocks the processing and activity-dependent release of BDNF) or by intra-mPFC microinjection of an anti-BDNF neutralizing antibody. Blockade of L-type voltage-dependent Ca2+ channels (VDCCs), required for activity-dependent BDNF release, also blocks the actions of (2R,6R)-HNK. Intra-mPFC infusion of pharmacological inhibitors of TrkB or mTORC1 signaling, which are downstream of BDNF, also block the actions of (2R,6R)-HNK. Moreover, (2R,6R)-HNK increases synaptic function in the mPFC. These findings indicate that activity-dependent BDNF release and downstream TrkB and mTORC1 signaling, which increase synaptic function in the mPFC, are required for the rapid and long-lasting antidepressant effects of (2R,6R)-HNK, supporting the potential use of this metabolite for the treatment of MDD.


Subject(s)
Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Ketamine/analogs & derivatives , Animals , Cells, Cultured , Depression/drug therapy , Disease Models, Animal , Dose-Response Relationship, Drug , Ketamine/pharmacology , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Neurons/drug effects , Neurons/metabolism , Signal Transduction/drug effects , Synaptic Transmission/drug effects
12.
Neuropsychopharmacology ; 42(6): 1231-1242, 2017 May.
Article in English | MEDLINE | ID: mdl-27634355

ABSTRACT

GLYX-13 is a putative NMDA receptor modulator with glycine-site partial agonist properties that produces rapid antidepressant effects, but without the psychotomimetic side effects of ketamine. Studies were conducted to examine the molecular, cellular, and behavioral actions of GLYX-13 to further characterize the mechanisms underlying the antidepressant actions of this agent. The results demonstrate that a single dose of GLYX-13 rapidly activates the mTORC1 pathway in the prefrontal cortex (PFC), and that infusion of the selective mTORC1 inhibitor rapamycin into the medial PFC (mPFC) blocks the antidepressant behavioral actions of GLYX-13, indicating a requirement for mTORC1 similar to ketamine. The results also demonstrate that GLYX-13 rapidly increases the number and function of spine synapses in the apical dendritic tuft of layer V pyramidal neurons in the mPFC. Notably, GLYX-13 significantly increased the synaptic responses to hypocretin, a measure of thalamocortical synapses, compared with its effects on 5-HT responses, a measure of cortical-cortical responses mediated by the 5-HT2A receptor. Behavioral studies further demonstrate that GLYX-13 does not influence 5-HT2 receptor induced head twitch response or impulsivity in a serial reaction time task (SRTT), whereas ketamine increased responses in both tests. In contrast, both GLYX-13 and ketamine increased attention in the SRTT task, which is linked to hypocretin-thalamocortical responses. The differences in the 5-HT2 receptor synaptic and behavioral responses may be related to the lack of psychotomimetic side effects of GLYX-13 compared with ketamine, whereas regulation of the hypocretin responses may contribute to the therapeutic benefits of both rapid acting antidepressants.


Subject(s)
Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Ketamine/pharmacology , Oligopeptides/pharmacology , Prefrontal Cortex/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Synapses/drug effects , Animals , Antidepressive Agents/administration & dosage , Ketamine/administration & dosage , Male , Mice, Inbred C57BL , Oligopeptides/administration & dosage , Rats , Rats, Sprague-Dawley
13.
Neuropsychopharmacology ; 40(9): 2066-75, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25759300

ABSTRACT

A single sub-anesthetic dose of ketamine, a short-acting NMDA receptor blocker, induces a rapid and prolonged antidepressant effect in treatment-resistant major depression. In animal models, ketamine (24 h) reverses depression-like behaviors and associated deficits in excitatory postsynaptic currents (EPSCs) generated in apical dendritic spines of layer V pyramidal cells of medial prefrontal cortex (mPFC). However, little is known about the effects of ketamine on basal dendrites. The basal dendrites of layer V cells receive an excitatory input from pyramidal cells of the basolateral amygdala (BLA), neurons that are activated by the stress hormone CRF. Here we found that CRF induces EPSCs in PFC layer V cells and that ketamine enhanced this effect through the mammalian target of rapamycin complex 1 synaptogenic pathway; the CRF-induced EPSCs required an intact BLA input and were generated primarily in basal dendrites. In contrast to its detrimental effects on apical dendritic structure and function, chronic stress did not induce a loss of CRF-induced EPSCs in basal dendrites, thereby creating a relative imbalance in favor of amygdala inputs. The effects of ketamine were complex: ketamine enhanced apical EPSC responses in all mPFC subregions, anterior cingulate (AC), prelimbic (PL), and infralimbic (IL) but enhanced CRF-induced EPSCs only in AC and PL-responses were unchanged in IL, a critical area for suppression of stress responses. We propose that by restoring the strength of apical inputs relative to basal amygdala inputs, especially in IL, ketamine would ameliorate the hypothesized disproportional negative influence of the amygdala in chronic stress and major depression.


Subject(s)
Amygdala/physiology , Corticotropin-Releasing Hormone/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Prefrontal Cortex/cytology , Pyramidal Cells/drug effects , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Amygdala/cytology , Amygdala/drug effects , Amygdala/injuries , Animals , Dendrites/drug effects , Excitatory Postsynaptic Potentials/drug effects , In Vitro Techniques , Limbic System/cytology , Limbic System/drug effects , Limbic System/physiology , Male , Neural Pathways/drug effects , Neural Pathways/physiology , Patch-Clamp Techniques , Pyramidal Cells/cytology , Rats , Rats, Sprague-Dawley
14.
Nat Med ; 20(5): 531-5, 2014 May.
Article in English | MEDLINE | ID: mdl-24728411

ABSTRACT

Major depressive disorder (MDD) affects up to 17% of the population, causing profound personal suffering and economic loss. Clinical and preclinical studies have revealed that prolonged stress and MDD are associated with neuronal atrophy of cortical and limbic brain regions, but the molecular mechanisms underlying these morphological alterations have not yet been identified. Here, we show that stress increases levels of REDD1 (regulated in development and DNA damage responses-1), an inhibitor of mTORC1 (mammalian target of rapamycin complex-1; ref. 10), in rat prefrontal cortex (PFC). This is concurrent with a decrease in phosphorylation of signaling targets of mTORC1, which is implicated in protein synthesis-dependent synaptic plasticity. We also found that REDD1 levels are increased in the postmortem PFC of human subjects with MDD relative to matched controls. Mutant mice with a deletion of the gene encoding REDD1 are resilient to the behavioral, synaptic and mTORC1 signaling deficits caused by chronic unpredictable stress, whereas viral-mediated overexpression of REDD1 in rat PFC is sufficient to cause anxiety- and depressive-like behaviors and neuronal atrophy. Taken together, these postmortem and preclinical findings identify REDD1 as a critical mediator of the atrophy of neurons and depressive behavior caused by chronic stress exposure.


Subject(s)
Anxiety Disorders/genetics , Depressive Disorder, Major/genetics , Synapses/pathology , Transcription Factors/genetics , Animals , Anxiety Disorders/etiology , Anxiety Disorders/pathology , Depressive Disorder, Major/etiology , Depressive Disorder, Major/pathology , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neurons/metabolism , Neurons/pathology , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Rats , Signal Transduction , Synapses/genetics , Synapses/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism
15.
Nat Neurosci ; 17(2): 248-53, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24441680

ABSTRACT

Although the prefrontal cortex influences motivated behavior, its role in food intake remains unclear. Here, we demonstrate a role for D1-type dopamine receptor-expressing neurons in the medial prefrontal cortex (mPFC) in the regulation of feeding. Food intake increases activity in D1 neurons of the mPFC in mice, and optogenetic photostimulation of D1 neurons increases feeding. Conversely, inhibition of D1 neurons decreases intake. Stimulation-based mapping of prefrontal D1 neuron projections implicates the medial basolateral amygdala (mBLA) as a downstream target of these afferents. mBLA neurons activated by prefrontal D1 stimulation are CaMKII positive and closely juxtaposed to prefrontal D1 axon terminals. Finally, photostimulating these axons in the mBLA is sufficient to increase feeding, recapitulating the effects of mPFC D1 stimulation. These data describe a new circuit for top-down control of food intake.


Subject(s)
Eating/physiology , Neurons/metabolism , Prefrontal Cortex/cytology , Receptors, Dopamine D1/metabolism , Amygdala/metabolism , Analysis of Variance , Animals , Biophysics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Channelrhodopsins , Eating/genetics , Electric Stimulation , Female , Food Deprivation/physiology , Functional Laterality , Gene Expression Regulation/genetics , In Vitro Techniques , Luminescent Proteins/genetics , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Inhibition/genetics , Neural Inhibition/radiation effects , Neural Pathways/physiology , Optogenetics , Patch-Clamp Techniques , Photic Stimulation/adverse effects , Receptors, Dopamine D1/genetics , Time Factors
16.
Biol Psychiatry ; 74(10): 742-9, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23751205

ABSTRACT

BACKGROUND: Clinical studies report that scopolamine, an acetylcholine muscarinic receptor antagonist, produces rapid antidepressant effects in depressed patients, but the mechanisms underlying the therapeutic response have not been determined. The present study examines the role of the mammalian target of rapamycin complex 1 (mTORC1) and synaptogenesis, which have been implicated in the rapid actions of N-methyl-D-aspartate receptor antagonists. METHODS: The influence of scopolamine on mTORC1 signaling was determined by analysis of the phosphorylated and activated forms of mTORC1 signaling proteins in the prefrontal cortex (PFC). The numbers and function of spine synapses were analyzed by whole cell patch clamp recording and two-photon image analysis of PFC neurons. The actions of scopolamine were examined in the forced swim test in the absence or presence of selective mTORC1 and glutamate receptor inhibitors. RESULTS: The results demonstrate that a single, low dose of scopolamine rapidly increases mTORC1 signaling and the number and function of spine synapses in layer V pyramidal neurons in the PFC. Scopolamine administration also produces an antidepressant response in the forced swim test that is blocked by pretreatment with the mTORC1 inhibitor or by a glutamate alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor antagonist. CONCLUSIONS: Taken together, the results demonstrate that the antidepressant actions of scopolamine require mTORC1 signaling and are associated with increased glutamate transmission, and synaptogenesis, similar to N-methyl-D-aspartate receptor antagonists. These findings provide novel targets for safer and more efficacious rapid-acting antidepressant agents.


Subject(s)
Antidepressive Agents/pharmacology , Multiprotein Complexes/metabolism , Muscarinic Antagonists/pharmacology , Prefrontal Cortex/drug effects , Scopolamine/pharmacology , Stress, Psychological/drug therapy , Synapses/drug effects , TOR Serine-Threonine Kinases/metabolism , Animals , Dendritic Spines/drug effects , Excitatory Postsynaptic Potentials , Male , Mechanistic Target of Rapamycin Complex 1 , Neurons/drug effects , Neurons/physiology , Neurons/ultrastructure , Prefrontal Cortex/metabolism , Prefrontal Cortex/ultrastructure , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Swimming/psychology
17.
Neuropsychopharmacology ; 38(11): 2268-77, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23680942

ABSTRACT

A single dose of the short-acting NMDA antagonist ketamine produces rapid and prolonged antidepressant effects in treatment-resistant patients with major depressive disorder (MDD), which are thought to occur via restoration of synaptic connectivity. However, acute dissociative side effects and eventual fading of antidepressant effects limit widespread clinical use of ketamine. Recent studies in medial prefrontal cortex (mPFC) show that the synaptogenic and antidepressant-like effects of a single standard dose of ketamine in rodents are dependent upon activation of the mammalian target of rapamycin (mTOR) complex 1 (mTORC1) signaling pathway together with inhibitory phosphorylation of glycogen synthase kinase-3 (GSK-3), which relieves its inhibitory in influence on mTOR. Here, we found that the synaptogenic and antidepressant-like effects of a single otherwise subthreshold dose of ketamine were potentiated when given together with a single dose of lithium chloride (a nonselective GSK-3 inhibitor) or a preferential GSK-3ß inhibitor; these effects included rapid activation of the mTORC1 signaling pathway, increased inhibitory phosphorylation of GSK-3ß, increased synaptic spine density/diameter, increased excitatory postsynaptic currents in mPFC layer V pyramidal neurons, and antidepressant responses that persist for up to 1 week in the forced-swim test model of depression. The results demonstrate that low, subthreshold doses of ketamine combined with lithium or a selective GSK-3 inhibitor are equivalent to higher doses of ketamine, indicating the pivotal role of the GSK-3 pathway in modulating the synaptogenic and antidepressant responses to ketamine. The possible mitigation by GSK-3 inhibitors of the eventual fading of ketamine's antidepressant effects remains to be explored.


Subject(s)
Antidepressive Agents/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Ketamine/pharmacology , Lithium Chloride/pharmacology , Synapses/drug effects , Animals , Dendritic Spines/drug effects , Dendritic Spines/ultrastructure , Dose-Response Relationship, Drug , Drug Synergism , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Glycogen Synthase Kinase 3/metabolism , Immobility Response, Tonic/drug effects , Indoles/pharmacology , Male , Maleimides/pharmacology , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/metabolism , Phosphorylation , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Rats , Signal Transduction/drug effects , Synapses/metabolism , TOR Serine-Threonine Kinases/metabolism
18.
Biol Psychiatry ; 71(11): 996-1005, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22036038

ABSTRACT

BACKGROUND: Knock-in mice with the common human brain-derived neurotrophic factor (BDNF) Val66Met polymorphism have impaired trafficking of BDNF messenger RNA to dendrites. It was hypothesized, given evidence that local synapse formation is dependent on dendritic translation of BDNF messenger RNA, that loss-of-function Met allele mice would show synaptic deficits both at baseline and in response to ketamine, an N-methyl-D-aspartate antagonist that stimulates synaptogenesis in prefrontal cortex (PFC). METHODS: Whole-cell recordings from layer V medial PFC pyramidal cells in brain slices were combined with two-photon laser scanning for analysis of wildtype, Val/Met, and Met/Met mice both at baseline and in response to a low dose of ketamine. RESULTS: Val/Met and Met/Met mice were found to have constitutive atrophy of distal apical dendrites and decrements in apically targeted excitatory postsynaptic currents in layer V pyramidal cells of PFC. In addition, spine density and diameter were decreased, indicative of impaired synaptic formation/maturation (synaptogenesis). In Met/Met mice the synaptogenic effect of ketamine was markedly impaired, consistent with the idea that synaptogenesis is dependent on dendritic translation/release of BDNF. In parallel behavioral studies, we found that the antidepressant response to ketamine in the forced swim test was blocked in Met/Met mice. CONCLUSIONS: The results demonstrate that expression of the BDNF Met allele in mice results in basal synaptic deficits and blocks synaptogenic and antidepressant actions of ketamine in PFC, suggesting that the therapeutic response to this drug might be attenuated or blocked in depressed patients who carry the loss of function Met allele.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Excitatory Postsynaptic Potentials , Prefrontal Cortex/metabolism , Pyramidal Cells/metabolism , RNA, Messenger/metabolism , Synapses/metabolism , Alleles , Animals , Dendrites/metabolism , Depressive Disorder, Major/genetics , Depressive Disorder, Major/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Mice , Mice, Transgenic , Patch-Clamp Techniques , Polymorphism, Genetic , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
19.
Neuropharmacology ; 62(1): 35-41, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21907221

ABSTRACT

Currently available medications have significant limitations, most notably low response rate and time lag for treatment response. Recent clinical studies have demonstrated that ketamine, an NMDA receptor antagonist produces a rapid antidepressant response (within hours) and is effective in treatment resistant depressed patients. Molecular and cellular studies in rodent models demonstrate that ketamine rapidly increases synaptogenesis, including increased density and function of spine synapses, in the prefrontal cortex (PFC). Ketamine also produces rapid antidepressant actions in behavioral models of depression, and reverses the deficits in synapse number and behavior resulting from chronic stress exposure. These effects of ketamine are accompanied by stimulation of the mammalian target of rapamycin (mTOR), and increased levels of synaptic proteins. Together these studies indicate that ketamine rapidly reverses the atrophy of spines in the PFC and thereby causes a functional reconnection of neurons that underlies the rapid behavioral responses. These findings identify new targets for rapid acting antidepressants that are safer than ketamine. This article is part of a Special Issue entitled 'Anxiety and Depression'.


Subject(s)
Antidepressive Agents/pharmacology , Ketamine/pharmacology , Neurogenesis/drug effects , Signal Transduction/drug effects , Animals , Antidepressive Agents/therapeutic use , Depression/drug therapy , Depression/pathology , Gene Expression Regulation/drug effects , Humans , Ketamine/therapeutic use , Models, Biological , Neurons/drug effects , Synapses/drug effects
20.
Biol Psychiatry ; 69(8): 754-61, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21292242

ABSTRACT

BACKGROUND: Despite widely reported clinical and preclinical studies of rapid antidepressant actions of glutamate N-methyl-D-aspartate (NMDA) receptor antagonists, there has been very little work examining the effects of these drugs in stress models of depression that require chronic administration of antidepressants or the molecular mechanisms that could account for the rapid responses. METHODS: We used a rat 21-day chronic unpredictable stress (CUS) model to test the rapid actions of NMDA receptor antagonists on depressant-like behavior, neurochemistry, and spine density and synaptic function of prefrontal cortex neurons. RESULTS: The results demonstrate that acute treatment with the noncompetitive NMDA channel blocker ketamine or the selective NMDA receptor 2B antagonist Ro 25-6981 rapidly ameliorates CUS-induced anhedonic and anxiogenic behaviors. We also found that CUS exposure decreases the expression levels of synaptic proteins and spine number and the frequency/amplitude of synaptic currents (excitatory postsynaptic currents) in layer V pyramidal neurons in the prefrontal cortex and that these deficits are rapidly reversed by ketamine. Blockade of the mammalian target of rapamycin protein synthesis cascade abolishes both the behavioral and biochemical effects of ketamine. CONCLUSIONS: The results indicate that the structural and functional deficits resulting from long-term stress exposure, which could contribute to the pathophysiology of depression, are rapidly reversed by NMDA receptor antagonists in a mammalian target of rapamycin dependent manner.


Subject(s)
Behavior, Animal/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Neurons/drug effects , Prefrontal Cortex/drug effects , Stress, Physiological/physiology , Stress, Psychological/physiopathology , Synapses/drug effects , Animals , Behavior, Animal/physiology , Blotting, Western , Choice Behavior/drug effects , Choice Behavior/physiology , Dendritic Spines/drug effects , Dendritic Spines/physiology , Electrophysiology , Ketamine/pharmacology , Neurons/physiology , Phenols/pharmacology , Piperidines/pharmacology , Prefrontal Cortex/physiopathology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , Sirolimus/pharmacology , Synapses/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...