Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 16.022
Filter
Add more filters

Publication year range
1.
Cell ; 181(1): 7, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32243798

ABSTRACT

The discovery of the strikingly rapid and robust antidepressant effects of r/s-ketamine for the treatment of antidepressant-resistant symptoms of depression has led to new insights into the biology of antidepressants and the FDA approval of its s-isomer, Esketamine (Spravato), the first mechanistically new treatment for depression in over 60 years. To view this Bench to Bedside, open or download the PDF.


Subject(s)
Antidepressive Agents , Depressive Disorder, Treatment-Resistant/drug therapy , Drug Approval , Ketamine , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , GABAergic Neurons/drug effects , Humans , Ketamine/pharmacology , Ketamine/therapeutic use , Receptors, AMPA/metabolism
2.
Cell ; 173(1): 166-180.e14, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29502969

ABSTRACT

Brain-wide fluctuations in local field potential oscillations reflect emergent network-level signals that mediate behavior. Cracking the code whereby these oscillations coordinate in time and space (spatiotemporal dynamics) to represent complex behaviors would provide fundamental insights into how the brain signals emotional pathology. Using machine learning, we discover a spatiotemporal dynamic network that predicts the emergence of major depressive disorder (MDD)-related behavioral dysfunction in mice subjected to chronic social defeat stress. Activity patterns in this network originate in prefrontal cortex and ventral striatum, relay through amygdala and ventral tegmental area, and converge in ventral hippocampus. This network is increased by acute threat, and it is also enhanced in three independent models of MDD vulnerability. Finally, we demonstrate that this vulnerability network is biologically distinct from the networks that encode dysfunction after stress. Thus, these findings reveal a convergent mechanism through which MDD vulnerability is mediated in the brain.


Subject(s)
Brain/physiology , Depression/pathology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Depression/physiopathology , Disease Models, Animal , Electric Stimulation , Electrodes, Implanted , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Ketamine/pharmacology , Machine Learning , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Physiological Phenomena/drug effects , Prefrontal Cortex/physiology , Stress, Psychological
3.
Nature ; 622(7984): 802-809, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37853123

ABSTRACT

Ketamine, an N-methyl-D-aspartate receptor (NMDAR) antagonist1, has revolutionized the treatment of depression because of its potent, rapid and sustained antidepressant effects2-4. Although the elimination half-life of ketamine is only 13 min in mice5, its antidepressant activities can last for at least 24 h6-9. This large discrepancy poses an interesting basic biological question and has strong clinical implications. Here we demonstrate that after a single systemic injection, ketamine continues to suppress burst firing and block NMDARs in the lateral habenula (LHb) for up to 24 h. This long inhibition of NMDARs is not due to endocytosis but depends on the use-dependent trapping of ketamine in NMDARs. The rate of untrapping is regulated by neural activity. Harnessing the dynamic equilibrium of ketamine-NMDAR interactions by activating the LHb and opening local NMDARs at different plasma ketamine concentrations, we were able to either shorten or prolong the antidepressant effects of ketamine in vivo. These results provide new insights into the causal mechanisms of the sustained antidepressant effects of ketamine. The ability to modulate the duration of ketamine action based on the biophysical properties of ketamine-NMDAR interactions opens up new opportunities for the therapeutic use of ketamine.


Subject(s)
Antidepressive Agents , Depression , Habenula , Ketamine , Receptors, N-Methyl-D-Aspartate , Animals , Mice , Antidepressive Agents/administration & dosage , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacokinetics , Antidepressive Agents/pharmacology , Depression/drug therapy , Depression/metabolism , Habenula/drug effects , Habenula/metabolism , Half-Life , Ketamine/administration & dosage , Ketamine/metabolism , Ketamine/pharmacokinetics , Ketamine/pharmacology , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Time Factors , Protein Binding
4.
Nature ; 608(7922): 368-373, 2022 08.
Article in English | MEDLINE | ID: mdl-35896744

ABSTRACT

Ketamine is used clinically as an anaesthetic and a fast-acting antidepressant, and recreationally for its dissociative properties, raising concerns of addiction as a possible side effect. Addictive drugs such as cocaine increase the levels of dopamine in the nucleus accumbens. This facilitates synaptic plasticity in the mesolimbic system, which causes behavioural adaptations and eventually drives the transition to compulsion1-4. The addiction liability of ketamine is a matter of much debate, in part because of its complex pharmacology that among several targets includes N-methyl-D-aspartic acid (NMDA) receptor (NMDAR) antagonism5,6. Here we show that ketamine does not induce the synaptic plasticity that is typically observed with addictive drugs in mice, despite eliciting robust dopamine transients in the nucleus accumbens. Ketamine nevertheless supported reinforcement through the disinhibition of dopamine neurons in the ventral tegmental area (VTA). This effect was mediated by NMDAR antagonism in GABA (γ-aminobutyric acid) neurons of the VTA, but was quickly terminated by type-2 dopamine receptors on dopamine neurons. The rapid off-kinetics of the dopamine transients along with the NMDAR antagonism precluded the induction of synaptic plasticity in the VTA and the nucleus accumbens, and did not elicit locomotor sensitization or uncontrolled self-administration. In summary, the dual action of ketamine leads to a unique constellation of dopamine-driven positive reinforcement, but low addiction liability.


Subject(s)
Ketamine , Substance-Related Disorders , Animals , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Ketamine/adverse effects , Ketamine/pharmacology , Mice , Neuronal Plasticity/drug effects , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Reinforcement, Psychology , Self Administration , Substance-Related Disorders/etiology , Substance-Related Disorders/prevention & control , Ventral Tegmental Area/cytology , Ventral Tegmental Area/drug effects
5.
Nature ; 596(7871): 301-305, 2021 08.
Article in English | MEDLINE | ID: mdl-34321660

ABSTRACT

Ketamine is a non-competitive channel blocker of N-methyl-D-aspartate (NMDA) receptors1. A single sub-anaesthetic dose of ketamine produces rapid (within hours) and long-lasting antidepressant effects in patients who are resistant to other antidepressants2,3. Ketamine is a racemic mixture of S- and R-ketamine enantiomers, with S-ketamine isomer being the more active antidepressant4. Here we describe the cryo-electron microscope structures of human GluN1-GluN2A and GluN1-GluN2B NMDA receptors in complex with S-ketamine, glycine and glutamate. Both electron density maps uncovered the binding pocket for S-ketamine in the central vestibule between the channel gate and selectivity filter. Molecular dynamics simulation showed that S-ketamine moves between two distinct locations within the binding pocket. Two amino acids-leucine 642 on GluN2A (homologous to leucine 643 on GluN2B) and asparagine 616 on GluN1-were identified as key residues that form hydrophobic and hydrogen-bond interactions with ketamine, and mutations at these residues reduced the potency of ketamine in blocking NMDA receptor channel activity. These findings show structurally how ketamine binds to and acts on human NMDA receptors, and pave the way for the future development of ketamine-based antidepressants.


Subject(s)
Cryoelectron Microscopy , Ketamine/chemistry , Ketamine/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/ultrastructure , Antidepressive Agents/chemistry , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacology , Asparagine/chemistry , Asparagine/metabolism , Binding Sites , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Glycine/chemistry , Glycine/metabolism , Glycine/pharmacology , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Ketamine/metabolism , Leucine/chemistry , Leucine/metabolism , Molecular Dynamics Simulation , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/ultrastructure , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism
6.
Nature ; 590(7845): 315-319, 2021 02.
Article in English | MEDLINE | ID: mdl-33328636

ABSTRACT

Effective pharmacotherapy for major depressive disorder remains a major challenge, as more than 30% of patients are resistant to the first line of treatment (selective serotonin reuptake inhibitors)1. Sub-anaesthetic doses of ketamine, a non-competitive N-methyl-D-aspartate receptor antagonist2,3, provide rapid and long-lasting antidepressant effects in these patients4-6, but the molecular mechanism of these effects remains unclear7,8. Ketamine has been proposed to exert its antidepressant effects through its metabolite (2R,6R)-hydroxynorketamine ((2R,6R)-HNK)9. The antidepressant effects of ketamine and (2R,6R)-HNK in rodents require activation of the mTORC1 kinase10,11. mTORC1 controls various neuronal functions12, particularly through cap-dependent initiation of mRNA translation via the phosphorylation and inactivation of eukaryotic initiation factor 4E-binding proteins (4E-BPs)13. Here we show that 4E-BP1 and 4E-BP2 are key effectors of the antidepressant activity of ketamine and (2R,6R)-HNK, and that ketamine-induced hippocampal synaptic plasticity depends on 4E-BP2 and, to a lesser extent, 4E-BP1. It has been hypothesized that ketamine activates mTORC1-4E-BP signalling in pyramidal excitatory cells of the cortex8,14. To test this hypothesis, we studied the behavioural response to ketamine and (2R,6R)-HNK in mice lacking 4E-BPs in either excitatory or inhibitory neurons. The antidepressant activity of the drugs is mediated by 4E-BP2 in excitatory neurons, and 4E-BP1 and 4E-BP2 in inhibitory neurons. Notably, genetic deletion of 4E-BP2 in inhibitory neurons induced a reduction in baseline immobility in the forced swim test, mimicking an antidepressant effect. Deletion of 4E-BP2 specifically in inhibitory neurons also prevented the ketamine-induced increase in hippocampal excitatory neurotransmission, and this effect concurred with the inability of ketamine to induce a long-lasting decrease in inhibitory neurotransmission. Overall, our data show that 4E-BPs are central to the antidepressant activity of ketamine.


Subject(s)
Antidepressive Agents/pharmacology , Eukaryotic Initiation Factor-4E/metabolism , Ketamine/pharmacology , Neurons/drug effects , Neurons/metabolism , Protein Biosynthesis/drug effects , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Depressive Disorder, Major/drug therapy , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Inhibitory Postsynaptic Potentials/drug effects , Interneurons/drug effects , Interneurons/metabolism , Ketamine/analogs & derivatives , Ketamine/metabolism , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mutation , Neural Inhibition/drug effects , Neural Inhibition/genetics , Neurons/classification , Neurons/cytology , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Synaptic Transmission/drug effects
7.
Proc Natl Acad Sci U S A ; 121(22): e2402732121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38768339

ABSTRACT

Ketamine is an N-methyl-D-aspartate (NMDA)-receptor antagonist that produces sedation, analgesia, and dissociation at low doses and profound unconsciousness with antinociception at high doses. At high and low doses, ketamine can generate gamma oscillations (>25 Hz) in the electroencephalogram (EEG). The gamma oscillations are interrupted by slow-delta oscillations (0.1 to 4 Hz) at high doses. Ketamine's primary molecular targets and its oscillatory dynamics have been characterized. However, how the actions of ketamine at the subcellular level give rise to the oscillatory dynamics observed at the network level remains unknown. By developing a biophysical model of cortical circuits, we demonstrate how NMDA-receptor antagonism by ketamine can produce the oscillatory dynamics observed in human EEG recordings and nonhuman primate local field potential recordings. We have identified how impaired NMDA-receptor kinetics can cause disinhibition in neuronal circuits and how a disinhibited interaction between NMDA-receptor-mediated excitation and GABA-receptor-mediated inhibition can produce gamma oscillations at high and low doses, and slow-delta oscillations at high doses. Our work uncovers general mechanisms for generating oscillatory brain dynamics that differs from ones previously reported and provides important insights into ketamine's mechanisms of action as an anesthetic and as a therapy for treatment-resistant depression.


Subject(s)
Ketamine , Receptors, N-Methyl-D-Aspartate , Ketamine/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Humans , Kinetics , Electroencephalography , Excitatory Amino Acid Antagonists/pharmacology , Models, Neurological
8.
Annu Rev Med ; 75: 129-143, 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-37729028

ABSTRACT

Major depressive disorder (MDD) is a leading cause of suicide in the world. Monoamine-based antidepressant drugs are a primary line of treatment for this mental disorder, although the delayed response and incomplete efficacy in some patients highlight the need for improved therapeutic approaches. Over the past two decades, ketamine has shown rapid onset with sustained (up to several days) antidepressant effects in patients whose MDD has not responded to conventional antidepressant drugs. Recent preclinical studies have started to elucidate the underlying mechanisms of ketamine's antidepressant properties. Herein, we describe and compare recent clinical and preclinical findings to provide a broad perspective of the relevant mechanisms for the antidepressant action of ketamine.


Subject(s)
Depressive Disorder, Major , Ketamine , Humans , Ketamine/therapeutic use , Depression/drug therapy , Depressive Disorder, Major/drug therapy , Antidepressive Agents/therapeutic use , Amines/therapeutic use
9.
N Engl J Med ; 388(25): 2315-2325, 2023 Jun 22.
Article in English | MEDLINE | ID: mdl-37224232

ABSTRACT

BACKGROUND: Electroconvulsive therapy (ECT) and subanesthetic intravenous ketamine are both currently used for treatment-resistant major depression, but the comparative effectiveness of the two treatments remains uncertain. METHODS: We conducted an open-label, randomized, noninferiority trial involving patients referred to ECT clinics for treatment-resistant major depression. Patients with treatment-resistant major depression without psychosis were recruited and assigned in a 1:1 ratio to receive ketamine or ECT. During an initial 3-week treatment phase, patients received either ECT three times per week or ketamine (0.5 mg per kilogram of body weight over 40 minutes) twice per week. The primary outcome was a response to treatment (i.e., a decrease of ≥50% from baseline in the score on the 16-item Quick Inventory of Depressive Symptomatology-Self-Report; scores range from 0 to 27, with higher scores indicating greater depression). The noninferiority margin was -10 percentage points. Secondary outcomes included scores on memory tests and patient-reported quality of life. After the initial treatment phase, the patients who had a response were followed over a 6-month period. RESULTS: A total of 403 patients underwent randomization at five clinical sites; 200 patients were assigned to the ketamine group and 203 to the ECT group. After 38 patients had withdrawn before initiation of the assigned treatment, ketamine was administered to 195 patients and ECT to 170 patients. A total of 55.4% of the patients in the ketamine group and 41.2% of those in the ECT group had a response (difference, 14.2 percentage points; 95% confidence interval, 3.9 to 24.2; P<0.001 for the noninferiority of ketamine to ECT). ECT appeared to be associated with a decrease in memory recall after 3 weeks of treatment (mean [±SE] decrease in the T-score for delayed recall on the Hopkins Verbal Learning Test-Revised, -0.9±1.1 in the ketamine group vs. -9.7±1.2 in the ECT group; scores range from -300 to 200, with higher scores indicating better function) with gradual recovery during follow-up. Improvement in patient-reported quality-of-life was similar in the two trial groups. ECT was associated with musculoskeletal adverse effects, whereas ketamine was associated with dissociation. CONCLUSIONS: Ketamine was noninferior to ECT as therapy for treatment-resistant major depression without psychosis. (Funded by the Patient-Centered Outcomes Research Institute; ELEKT-D ClinicalTrials.gov number, NCT03113968.).


Subject(s)
Antidepressive Agents , Depressive Disorder, Treatment-Resistant , Electroconvulsive Therapy , Ketamine , Humans , Depressive Disorder, Major/diagnosis , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/therapy , Electroconvulsive Therapy/adverse effects , Ketamine/administration & dosage , Ketamine/adverse effects , Ketamine/therapeutic use , Quality of Life , Treatment Outcome , Antidepressive Agents/administration & dosage , Antidepressive Agents/adverse effects , Antidepressive Agents/therapeutic use , Depressive Disorder, Treatment-Resistant/diagnosis , Depressive Disorder, Treatment-Resistant/drug therapy , Depressive Disorder, Treatment-Resistant/therapy , Administration, Intravenous , Psychotic Disorders
10.
N Engl J Med ; 389(14): 1298-1309, 2023 Oct 05.
Article in English | MEDLINE | ID: mdl-37792613

ABSTRACT

BACKGROUND: In treatment-resistant depression, commonly defined as a lack of response to two or more consecutive treatments during the current depressive episode, the percentage of patients with remission is low and the percentage with relapse is high. The efficacy and safety of esketamine nasal spray as compared with extended-release quetiapine augmentation therapy, both in combination with ongoing treatment with a selective serotonin reuptake inhibitor (SSRI) or a serotonin-norepinephrine reuptake inhibitor (SNRI), in patients with treatment-resistant depression are unknown. METHODS: In an open-label, single-blind (with raters unaware of group assignments), multicenter, phase 3b, randomized, active-controlled trial, we assigned patients, in a 1:1 ratio, to receive flexible doses (according to the summary of product characteristics) of esketamine nasal spray (esketamine group) or extended-release quetiapine (quetiapine group), both in combination with an SSRI or SNRI. The primary end point was remission, defined as a score of 10 or less on the Montgomery-Åsberg Depression Rating Scale (MADRS), at week 8 (scores range from 0 to 60, with higher scores indicating more severe depression). The key secondary end point was no relapse through week 32 after remission at week 8. All patients were included in the analysis; patients who discontinued the trial treatment were considered as having had an unfavorable outcome (i.e., they were grouped with patients who did not have remission or who had a relapse). Analyses of the primary and key secondary end points were adjusted for age and number of treatment failures. RESULTS: Overall, 336 patients were assigned to the esketamine group and 340 to the quetiapine group. More patients in the esketamine group than in the quetiapine group had remission at week 8 (91 of 336 patients [27.1%] vs. 60 of 340 patients [17.6%]; P = 0.003) and had no relapse through week 32 after remission at week 8 (73 of 336 patients [21.7%] vs. 48 of 340 patients [14.1%]). Over 32 weeks of follow-up, the percentage of patients with remission, the percentage of patients with a treatment response, and the change in the MADRS score from baseline favored esketamine nasal spray. The adverse events were consistent with the established safety profiles of the trial treatments. CONCLUSIONS: In patients with treatment-resistant depression, esketamine nasal spray plus an SSRI or SNRI was superior to extended-release quetiapine plus an SSRI or SNRI with respect to remission at week 8. (Funded by Janssen EMEA; ESCAPE-TRD ClinicalTrials.gov number, NCT04338321.).


Subject(s)
Antidepressive Agents , Depressive Disorder, Treatment-Resistant , Ketamine , Quetiapine Fumarate , Selective Serotonin Reuptake Inhibitors , Serotonin and Noradrenaline Reuptake Inhibitors , Humans , Antidepressive Agents/administration & dosage , Antidepressive Agents/adverse effects , Antidepressive Agents/therapeutic use , Delayed-Action Preparations , Depression/drug therapy , Drug Therapy, Combination , Nasal Sprays , Quetiapine Fumarate/administration & dosage , Quetiapine Fumarate/adverse effects , Quetiapine Fumarate/therapeutic use , Recurrence , Selective Serotonin Reuptake Inhibitors/administration & dosage , Selective Serotonin Reuptake Inhibitors/adverse effects , Selective Serotonin Reuptake Inhibitors/therapeutic use , Serotonin and Noradrenaline Reuptake Inhibitors/administration & dosage , Serotonin and Noradrenaline Reuptake Inhibitors/adverse effects , Serotonin and Noradrenaline Reuptake Inhibitors/therapeutic use , Single-Blind Method , Treatment Outcome , Ketamine/administration & dosage , Ketamine/adverse effects , Ketamine/therapeutic use , Depressive Disorder, Treatment-Resistant/drug therapy
11.
PLoS Biol ; 21(2): e3002013, 2023 02.
Article in English | MEDLINE | ID: mdl-36802356

ABSTRACT

Substantial progress in the field of neuroscience has been made from anaesthetized preparations. Ketamine is one of the most used drugs in electrophysiology studies, but how ketamine affects neuronal responses is poorly understood. Here, we used in vivo electrophysiology and computational modelling to study how the auditory cortex of bats responds to vocalisations under anaesthesia and in wakefulness. In wakefulness, acoustic context increases neuronal discrimination of natural sounds. Neuron models predicted that ketamine affects the contextual discrimination of sounds regardless of the type of context heard by the animals (echolocation or communication sounds). However, empirical evidence showed that the predicted effect of ketamine occurs only if the acoustic context consists of low-pitched sounds (e.g., communication calls in bats). Using the empirical data, we updated the naïve models to show that differential effects of ketamine on cortical responses can be mediated by unbalanced changes in the firing rate of feedforward inputs to cortex, and changes in the depression of thalamo-cortical synaptic receptors. Combined, our findings obtained in vivo and in silico reveal the effects and mechanisms by which ketamine affects cortical responses to vocalisations.


Subject(s)
Anesthesia , Auditory Cortex , Chiroptera , Ketamine , Animals , Auditory Cortex/physiology , Acoustic Stimulation , Ketamine/pharmacology , Chiroptera/physiology , Neurons/physiology , Auditory Perception/physiology
12.
Nature ; 586(7827): 87-94, 2020 10.
Article in English | MEDLINE | ID: mdl-32939091

ABSTRACT

Advanced imaging methods now allow cell-type-specific recording of neural activity across the mammalian brain, potentially enabling the exploration of how brain-wide dynamical patterns give rise to complex behavioural states1-12. Dissociation is an altered behavioural state in which the integrity of experience is disrupted, resulting in reproducible cognitive phenomena including the dissociation of stimulus detection from stimulus-related affective responses. Dissociation can occur as a result of trauma, epilepsy or dissociative drug use13,14, but despite its substantial basic and clinical importance, the underlying neurophysiology of this state is unknown. Here we establish such a dissociation-like state in mice, induced by precisely-dosed administration of ketamine or phencyclidine. Large-scale imaging of neural activity revealed that these dissociative agents elicited a 1-3-Hz rhythm in layer 5 neurons of the retrosplenial cortex. Electrophysiological recording with four simultaneously deployed high-density probes revealed rhythmic coupling of the retrosplenial cortex with anatomically connected components of thalamus circuitry, but uncoupling from most other brain regions was observed-including a notable inverse correlation with frontally projecting thalamic nuclei. In testing for causal significance, we found that rhythmic optogenetic activation of retrosplenial cortex layer 5 neurons recapitulated dissociation-like behavioural effects. Local retrosplenial hyperpolarization-activated cyclic-nucleotide-gated potassium channel 1 (HCN1) pacemakers were required for systemic ketamine to induce this rhythm and to elicit dissociation-like behavioural effects. In a patient with focal epilepsy, simultaneous intracranial stereoencephalography recordings from across the brain revealed a similarly localized rhythm in the homologous deep posteromedial cortex that was temporally correlated with pre-seizure self-reported dissociation, and local brief electrical stimulation of this region elicited dissociative experiences. These results identify the molecular, cellular and physiological properties of a conserved deep posteromedial cortical rhythm that underlies states of dissociation.


Subject(s)
Brain Waves/physiology , Cerebral Cortex/physiology , Dissociative Disorders/physiopathology , Action Potentials/drug effects , Animals , Behavior/drug effects , Brain Waves/drug effects , Cerebral Cortex/cytology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/drug effects , Dissociative Disorders/diagnostic imaging , Electrophysiology , Female , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Ketamine/pharmacology , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Optogenetics , Self Report , Thalamus/cytology , Thalamus/diagnostic imaging , Thalamus/drug effects , Thalamus/physiology
13.
Proc Natl Acad Sci U S A ; 120(49): e2305772120, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38011560

ABSTRACT

Ketamine has emerged as a transformative and mechanistically novel pharmacotherapy for depression. Its rapid onset of action, efficacy for treatment-resistant symptoms, and protection against relapse distinguish it from prior antidepressants. Its discovery emerged from a reconceptualization of the neurobiology of depression and, in turn, insights from the elaboration of its mechanisms of action inform studies of the pathophysiology of depression and related disorders. It has been 25 y since we first presented our ketamine findings in depression. Thus, it is timely for this review to consider what we have learned from studies of ketamine and to suggest future directions for the optimization of rapid-acting antidepressant treatment.


Subject(s)
Ketamine , Ketamine/pharmacology , Ketamine/therapeutic use , Depression/drug therapy , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use
14.
Proc Natl Acad Sci U S A ; 120(49): e2305776120, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38011563

ABSTRACT

Individuals with a history of early-life stress (ELS) tend to have an altered course of depression and lower treatment response rates. Research suggests that ELS alters brain development, but the molecular changes in the brain following ELS that may mediate altered antidepressant response have not been systematically studied. Sex and gender also impact the risk of depression and treatment response. Here, we leveraged existing RNA sequencing datasets from 1) blood samples from depressed female- and male-identifying patients treated with escitalopram or desvenlafaxine and assessed for treatment response or failure; 2) the nucleus accumbens (NAc) of female and male mice exposed to ELS and/or adult stress; and 3) the NAc of mice after adult stress, antidepressant treatment with imipramine or ketamine, and assessed for treatment response or failure. We find that transcriptomic signatures of adult stress after a history of ELS correspond with transcriptomic signatures of treatment nonresponse, across species and multiple classes of antidepressants. Transcriptomic correspondence with treatment outcome was stronger among females and weaker among males. We next pharmacologically tested these predictions in our mouse model of early-life and adult social defeat stress and treatment with either chronic escitalopram or acute ketamine. Among female mice, the strongest predictor of behavior was an interaction between ELS and ketamine treatment. Among males, however, early experience and treatment were poor predictors of behavior, mirroring our bioinformatic predictions. These studies provide neurobiological evidence for molecular adaptations in the brain related to sex and ELS that contribute to antidepressant treatment response.


Subject(s)
Adverse Childhood Experiences , Ketamine , Humans , Male , Female , Mice , Animals , Depression/drug therapy , Depression/genetics , Escitalopram , Ketamine/pharmacology , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Treatment Outcome , Stress, Psychological/drug therapy , Stress, Psychological/genetics
15.
J Neurosci ; 44(13)2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38378273

ABSTRACT

Patients with chronic pain often develop comorbid depressive symptoms, which makes the pain symptoms more complicated and refractory. However, the underlying mechanisms are poorly known. Here, in a repeated complete Freund's adjuvant (CFA) male mouse model, we reported a specific regulatory role of the paraventricular thalamic nucleus (PVT) glutamatergic neurons, particularly the anterior PVT (PVA) neurons, in mediating chronic pain and depression comorbidity (CDC). Our c-Fos protein staining observed increased PVA neuronal activity in CFA-CDC mice. In wild-type mice, chemogenetic activation of PVA glutamatergic neurons was sufficient to decrease the 50% paw withdrawal thresholds (50% PWTs), while depressive-like behaviors evaluated with immobile time in tail suspension test (TST) and forced swim test (FST) could only be achieved by repeated chemogenetic activation. Chemogenetic inhibition of PVA glutamatergic neurons reversed the decreased 50% PWTs in CFA mice without depressive-like symptoms and the increased TST and FST immobility in CFA-CDC mice. Surprisingly, in CFA-CDC mice, chemogenetically inhibiting PVA glutamatergic neurons failed to reverse the decrease of 50% PWTs, which could be restored by rapid-onset antidepressant S-ketamine. Further behavioral tests in chronic restraint stress mice and CFA pain mice indicated that PVA glutamatergic neuron inhibition and S-ketamine independently alleviate sensory and affective pain. Molecular profiling and pharmacological studies revealed the 5-hydroxytryptamine receptor 1D (Htr1d) in CFA pain-related PVT engram neurons as a potential target for treating CDC. These findings identified novel CDC neuronal and molecular mechanisms in the PVT and provided insight into the complicated pain neuropathology under a comorbid state with depression and related drug development.


Subject(s)
Chronic Pain , Ketamine , Humans , Mice , Male , Animals , Chronic Pain/metabolism , Depression/drug therapy , Thalamus , Neurons/metabolism , Comorbidity
16.
Bioinformatics ; 40(1)2024 01 02.
Article in English | MEDLINE | ID: mdl-38147362

ABSTRACT

MOTIVATION: Up-to-date pathway knowledge is usually presented in scientific publications for human reading, making it difficult to utilize these resources for semantic integration and computational analysis of biological pathways. We here present an approach to mining knowledge graphs by combining manual curation with automated named entity recognition and automated relation extraction. This approach allows us to study pathway-related questions in detail, which we here show using the ketamine pathway, aiming to help improve understanding of the role of gut microbiota in the antidepressant effects of ketamine. RESULTS: The thus devised ketamine pathway 'KetPath' knowledge graph comprises five parts: (i) manually curated pathway facts from images; (ii) recognized named entities in biomedical texts; (iii) identified relations between named entities; (iv) our previously constructed microbiota and pre-/probiotics knowledge bases; and (v) multiple community-accepted public databases. We first assessed the performance of automated extraction of relations between named entities using the specially designed state-of-the-art tool BioKetBERT. The query results show that we can retrieve drug actions, pathway relations, co-occurring entities, and their relations. These results uncover several biological findings, such as various gut microbes leading to increased expression of BDNF, which may contribute to the sustained antidepressant effects of ketamine. We envision that the methods and findings from this research will aid researchers who wish to integrate and query data and knowledge from multiple biomedical databases and literature simultaneously. AVAILABILITY AND IMPLEMENTATION: Data and query protocols are available in the KetPath repository at https://dx.doi.org/10.5281/zenodo.8398941 and https://github.com/tingcosmos/KetPath.


Subject(s)
Gastrointestinal Microbiome , Ketamine , Humans , Ketamine/pharmacology , Databases, Factual , Antidepressive Agents/pharmacology , Neurotransmitter Agents , Data Mining/methods
17.
Mol Psychiatry ; 29(3): 750-759, 2024 03.
Article in English | MEDLINE | ID: mdl-38123725

ABSTRACT

OBJECTIVE: To meta-analyze clinical efficacy and safety of ketamine compared with other anesthetic agents in the course of electroconvulsive therapy (ECT) in major depressive episode (MDE). METHODS: PubMed/MEDLINE, Cochrane Library, Embase, GoogleScholar, and US and European trial registries were searched from inception through May 23, 2023, with no language limits. We included RCTs with (1) a diagnosis of MDE; (2) ECT intervention with ketamine and/or other anesthetic agents; and (3) measures included: depressive symptoms, cognitive performance, remission or response rates, and serious adverse events. Network meta-analysis (NMA) was performed to compare ketamine and 7 other anesthetic agents. Hedges' g standardized mean differences (SMDs) were used for continuous measures, and relative risks (RRs) were used for other binary outcomes using random-effects models. RESULTS: Twenty-two studies were included in the systematic review. A total of 2322 patients from 17 RCTs were included in the NMA. The overall pooled SMD of ketamine, as compared with propofol as a reference group, was -2.21 (95% confidence interval [CI], -3.79 to -0.64) in depressive symptoms, indicating that ketamine had better antidepressant efficacy than propofol. In a sensitivity analysis, however, ketamine-treated patients had a worse outcome in cognitive performance than propofol-treated patients (SMD, -0.18; 95% CI, -0.28 to -0.09). No other statistically significant differences were found. CONCLUSIONS: Ketamine-assisted ECT is tolerable and may be efficacious in improving depressive symptoms, but a relative adverse impact on cognition may be an important clinical consideration. Anesthetic agents should be considered based on patient profiles and/or preferences to improve effectiveness and safety of ECT use.


Subject(s)
Depressive Disorder, Major , Electroconvulsive Therapy , Ketamine , Network Meta-Analysis , Ketamine/therapeutic use , Electroconvulsive Therapy/methods , Humans , Depressive Disorder, Major/therapy , Depressive Disorder, Major/drug therapy , Treatment Outcome , Propofol/therapeutic use , Propofol/adverse effects , Randomized Controlled Trials as Topic , Anesthetics/therapeutic use , Anesthetics/adverse effects , Female , Male
18.
Mol Psychiatry ; 29(4): 1114-1127, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38177353

ABSTRACT

The discovery that subanesthetic doses of (R, S)-ketamine (ketamine) and (S)-ketamine (esketamine) rapidly induce antidepressant effects and promote sustained actions following drug clearance in depressed patients who are treatment-resistant to other therapies has resulted in a paradigm shift in the conceptualization of how rapidly and effectively depression can be treated. Consequently, the mechanism(s) that next generation antidepressants may engage to improve pathophysiology and resultant symptomology are being reconceptualized. Impaired excitatory glutamatergic synapses in mood-regulating circuits are likely a substantial contributor to the pathophysiology of depression. Metaplasticity is the process of regulating future capacity for plasticity by priming neurons with a stimulation that alters later neuronal plasticity responses. Accordingly, the development of treatment modalities that specifically modulate the duration, direction, or magnitude of glutamatergic synaptic plasticity events such as long-term potentiation (LTP), defined here as metaplastogens, may be an effective approach to reverse the pathophysiology underlying depression and improve depression symptoms. We review evidence that the initiating mechanisms of pharmacologically diverse rapid-acting antidepressants (i.e., ketamine mimetics) converge on consistent downstream molecular mediators that facilitate the expression/maintenance of increased synaptic strength and resultant persisting antidepressant effects. Specifically, while the initiating mechanisms of these therapies may differ (e.g., cell type-specificity, N-methyl-D-aspartate receptor (NMDAR) subtype-selective inhibition vs activation, metabotropic glutamate receptor 2/3 antagonism, AMPA receptor potentiation, 5-HT receptor-activating psychedelics, etc.), the sustained therapeutic mechanisms of putative rapid-acting antidepressants will be mediated, in part, by metaplastic effects that converge on consistent molecular mediators to enhance excitatory neurotransmission and altered capacity for synaptic plasticity. We conclude that the convergence of these therapeutic mechanisms provides the opportunity for metaplasticity processes to be harnessed as a druggable plasticity mechanism by next-generation therapeutics. Further, targeting metaplastic mechanisms presents therapeutic advantages including decreased dosing frequency and associated diminished adverse responses by eliminating the requirement for the drug to be continuously present.


Subject(s)
Antidepressive Agents , Ketamine , Neuronal Plasticity , Humans , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Neuronal Plasticity/drug effects , Ketamine/pharmacology , Ketamine/therapeutic use , Animals , Depression/drug therapy , Long-Term Potentiation/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/drug effects , Synapses/metabolism
19.
Mol Psychiatry ; 29(4): 914-928, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38212376

ABSTRACT

We describe evidence for dissociable roles of the medial and lateral orbitofrontal cortex (OFC) in major depressive disorder (MDD) from structure, functional activation, functional connectivity, metabolism, and neurochemical systems. The reward-related medial orbitofrontal cortex has lower connectivity and less reward sensitivity in MDD associated with anhedonia symptoms; and the non-reward related lateral OFC has higher functional connectivity and more sensitivity to non-reward/aversive stimuli in MDD associated with negative bias symptoms. Importantly, we propose that conventional antidepressants act to normalize the hyperactive lateral (but not medial) OFC to reduce negative bias in MDD; while other treatments are needed to operate on the medial OFC to reduce anhedonia, with emerging evidence suggesting that ketamine may act in this way. The orbitofrontal cortex is the key cortical region in emotion and reward, and the current review presents much new evidence about the different ways that the medial and lateral OFC are involved in MDD.


Subject(s)
Anhedonia , Depressive Disorder, Major , Prefrontal Cortex , Humans , Anhedonia/physiology , Antidepressive Agents/therapeutic use , Antidepressive Agents/pharmacology , Depressive Disorder, Major/physiopathology , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/drug therapy , Emotions/physiology , Ketamine/therapeutic use , Ketamine/pharmacology , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Reward
20.
Mol Psychiatry ; 29(5): 1406-1416, 2024 May.
Article in English | MEDLINE | ID: mdl-38388704

ABSTRACT

Chronic social isolation increases the risk of mental health problems, including cognitive impairments and depression. While subanesthetic ketamine is considered effective for cognitive impairments in patients with depression, the neural mechanisms underlying its effects are not well understood. Here we identified unique activation of the anterior insular cortex (aIC) as a characteristic feature in brain-wide regions of mice reared in social isolation and treated with (R)-ketamine, a ketamine enantiomer. Using fiber photometry recording on freely moving mice, we found that social isolation attenuates aIC neuronal activation upon social contact and that (R)-ketamine, but not (S)-ketamine, is able to counteracts this reduction. (R)-ketamine facilitated social cognition in social isolation-reared mice during the social memory test. aIC inactivation offset the effect of (R)-ketamine on social memory. Our results suggest that (R)-ketamine has promising potential as an effective intervention for social cognitive deficits by restoring aIC function.


Subject(s)
Cognitive Dysfunction , Insular Cortex , Ketamine , Social Isolation , Animals , Ketamine/pharmacology , Mice , Male , Insular Cortex/drug effects , Cognitive Dysfunction/drug therapy , Mice, Inbred C57BL , Memory/drug effects , Cognition/drug effects , Social Behavior , Cerebral Cortex/drug effects , Neurons/drug effects , Cognition Disorders/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL