1
|
Graf S, Dörl G, Milz C, Kathofer M, Stöhrmann P, Gomola D, Briem E, Schlosser G, Mayerweg A, Semmelweis-Tomits J, Hoti A, Eggerstorfer B, Schmidt C, Crone J, Rujescu D, Spies M, Lanzenberger R, Spurny-Dworak B. Morphological correlates of anxiety-related experiences during a ketamine infusion. World J Biol Psychiatry 2024; 25:537-546. [PMID: 39394769 DOI: 10.1080/15622975.2024.2402261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 10/14/2024]
Abstract
OBJECTIVES Ketamine exerts rapid antidepressant effects by enhancing neuroplasticity, particularly in the amygdala and hippocampus-regions involved in fear processing and learning. While the role of ketamine's dissociative effects in its antidepressant response is debated, anxiety experienced during infusion has been negatively correlated with treatment outcomes. METHODS In this single-blind, placebo-controlled study, a subset of 17 healthy volunteers (6 males, 23.12 ± 1.9 years) received intravenously a placebo in the first and 0.5 mg/kg racemic ketamine in the second session. Anxiety-related experiences were assessed by the 5D-ASC score obtained post-infusion, structural magnetic resonance imaging scans were acquired 4 h post-infusion. An anxiety-score was obtained from the 5D-ASC. Relation between post-placebo amygdala volume, hippocampal volume, and its subfields with the anxiety-score were assessed using linear regression models. RESULTS Results showed a statistically significant negative relation between hippocampal head volume and the anxiety score (β = -0.733, p = 0.006), with trending negative association for each subfield's head and the score. CONCLUSION These findings suggest that anxiety-related experiences during ketamine infusion may be mediated by the hippocampus, with smaller hippocampal volumes leading to more anxiety-related experiences. Thus, hippocampal subfield volumes may be used as a predictor for anxiety-related events during ketamine use and might predict treatment outcome in future approaches.
Collapse
Affiliation(s)
- S Graf
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - G Dörl
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - C Milz
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - M Kathofer
- Vienna Cognitive Science Hub, University of Vienna, Vienna, Austria
| | - P Stöhrmann
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - D Gomola
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - E Briem
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - G Schlosser
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - A Mayerweg
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - J Semmelweis-Tomits
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - A Hoti
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - B Eggerstorfer
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - C Schmidt
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - J Crone
- Vienna Cognitive Science Hub, University of Vienna, Vienna, Austria
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
- Faculty of Psychology, University of Vienna, Vienna, Austria
| | - D Rujescu
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - M Spies
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - R Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - B Spurny-Dworak
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| |
Collapse
|
2
|
Meiering MS, Weigner D, Gärtner M, Carstens L, Keicher C, Hertrampf R, Beckmann CF, Mennes M, Wunder A, Weigand A, Grimm S. Functional activity and connectivity signatures of ketamine and lamotrigine during negative emotional processing: a double-blind randomized controlled fMRI study. Transl Psychiatry 2024; 14:436. [PMID: 39402015 PMCID: PMC11479267 DOI: 10.1038/s41398-024-03120-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/17/2024] Open
Abstract
Ketamine is a highly effective antidepressant (AD) that targets the glutamatergic system and exerts profound effects on brain circuits during negative emotional processing. Interestingly, the effects of ketamine on brain measures are sensitive to modulation by pretreatment with lamotrigine, which inhibits glutamate release. Examining the antagonistic effects of ketamine and lamotrigine on glutamate transmission holds promise to identify effects of ketamine that are mediated through changes in the glutamatergic system. Investigating this modulation in relation to both the acute and sustained effects of ketamine on functional activity and connectivity during negative emotional processing should therefore provide novel insights. 75 healthy subjects were investigated in a double-blind, single-dose, randomized, placebo-controlled, parallel-group study with three treatment conditions (ketamine, lamotrigine pre-treatment, placebo). Participants completed an emotional face viewing task during ketamine infusion and 24 h later. Acute ketamine administration decreased hippocampal and Default Mode Network (DMN) activity and increased fronto-limbic coupling during negative emotional processing. Furthermore, while lamotrigine abolished the ketamine-induced increase in functional connectivity, it had no acute effect on activity. Sustained (24 h later) effects of ketamine were only found for functional activity, with a significant reduction in the posterior DMN. This effect was blocked by pretreatment with lamotrigine. Our results suggest that both the acute increases in fronto-limbic coupling and the delayed decrease in posterior DMN activity, but not the attenuated limbic and DMN recruitment after ketamine, are mediated by altered glutamatergic transmission.
Collapse
Affiliation(s)
- Marvin S Meiering
- Medical School Berlin, Berlin, Germany.
- Department of Education and Psychology, Freie Universität Berlin, Berlin, Germany.
| | - David Weigner
- Medical School Berlin, Berlin, Germany
- Department of Education and Psychology, Freie Universität Berlin, Berlin, Germany
| | | | | | | | | | | | | | - Andreas Wunder
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | | | - Simone Grimm
- Medical School Berlin, Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universitiät Zu Berlin, Berlin, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland
| |
Collapse
|
3
|
Juneja K, Afroze S, Goti Z, Sahu S, Asawa S, Bhuchakra HP, Natarajan B. Beyond therapeutic potential: a systematic investigation of ketamine misuse in patients with depressive disorders. DISCOVER MENTAL HEALTH 2024; 4:23. [PMID: 38951348 PMCID: PMC11217219 DOI: 10.1007/s44192-024-00077-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 06/17/2024] [Indexed: 07/03/2024]
Abstract
Ketamine, a pharmacological agent that acts as an antagonist of the N-methyl-D-aspartate (NMDA) receptor, has garnered considerable interest because of its notable and expeditious antidepressant properties observed in individuals diagnosed with major depressive disorder (MDD) who exhibit resistance to conventional therapeutic interventions. A comprehensive and rigorous systematic review was undertaken to evaluate the prevalence of ketamine abuse undergoing ketamine treatment for depressive disorders. A comprehensive search was conducted across the electronic databases to identify pertinent studies published between 2021 and 2023. The present investigation incorporated a comprehensive range of studies encompassing the abuse or misuse of ketamine, including case reports, observational studies, and clinical trials. Data extraction and quality assessment were conducted in accordance with predetermined criteria. The findings of this systematic review demonstrate the importance of monitoring and addressing ketamine abuse in patients receiving ketamine treatment for depressive disorders like MDD. The wide range of reported prevalence rates highlights the need for standardized criteria and measures for defining and assessing ketamine abuse. This study presents a significant contribution to the field by introducing a novel screening questionnaire and assessment algorithm designed to identify and evaluate ketamine misuse among major depressive disorder (MDD) patients undergoing ketamine treatment. This innovative tool holds the potential to enhance clinical practice by providing healthcare professionals with a standardized approach to promptly detect and address ketamine misuse. The integration of this screening tool into routine care protocols can facilitate more effective monitoring and management of ketamine misuse in this population, ultimately leading to improved patient outcomes and safety.
Collapse
Affiliation(s)
| | - Sabah Afroze
- Shadan Hospital and Institute of Medical Sciences, Hyderabad, India
| | - Zeel Goti
- Government Medical College, Surat, India
| | | | | | | | | |
Collapse
|
4
|
Martens MAG, Zghoul T, Watson E, Rieger SW, Capitão LP, Harmer CJ. Acute neural effects of the mood stabiliser lamotrigine on emotional processing in healthy volunteers: a randomised control trial. Transl Psychiatry 2024; 14:211. [PMID: 38802372 PMCID: PMC11130123 DOI: 10.1038/s41398-024-02944-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024] Open
Abstract
Lamotrigine is an effective mood stabiliser, largely used for the management and prevention of depression in bipolar disorder. The neuropsychological mechanisms by which lamotrigine acts to relieve symptoms as well as its neural effects on emotional processing remain unclear. The primary objective of this current study was to investigate the impact of an acute dose of lamotrigine on the neural response to a well-characterised fMRI task probing implicit emotional processing relevant to negative bias. 31 healthy participants were administered either a single dose of lamotrigine (300 mg, n = 14) or placebo (n = 17) in a randomized, double-blind design. Inside the 3 T MRI scanner, participants completed a covert emotional faces gender discrimination task. Brain activations showing significant group differences were identified using voxel-wise general linear model (GLM) nonparametric permutation testing, with threshold free cluster enhancement (TFCE) and a family wise error (FWE)-corrected cluster significance threshold of p < 0.05. Participants receiving lamotrigine were more accurate at identifying the gender of fearful (but not happy or angry) faces. A network of regions associated with emotional processing, including amygdala, insula, and the anterior cingulate cortex (ACC), was significantly less activated in the lamotrigine group compared to the placebo group across emotional facial expressions. A single dose of lamotrigine reduced activation in limbic areas in response to faces with both positive and negative expressions, suggesting a valence-independent effect. However, at a behavioural level lamotrigine appeared to reduce the distracting effect of fear on face discrimination. Such effects may be relevant to the mood stabilisation effects of lamotrigine.
Collapse
Affiliation(s)
- Marieke A G Martens
- Department of Psychiatry, University of Oxford, Oxford, UK.
- Oxford Health NHS Foundation Trust, Oxford, UK.
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK.
| | - Tarek Zghoul
- Department of Psychiatry, University of Oxford, Oxford, UK
- Oxford Health NHS Foundation Trust, Oxford, UK
| | - Evelyn Watson
- Department of Psychiatry, University of Oxford, Oxford, UK
- Institute of Sport Exercise and Health, Faculty of Medical Sciences, University College London, London, UK
- Institute of Cognitive Neuroscience, Faculty of Brain Sciences, University College London, London, UK
| | - Sebastian W Rieger
- Department of Psychiatry, University of Oxford, Oxford, UK
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| | - Liliana P Capitão
- Psychology Research Centre (CIPsi), School of Psychology, University of Minho, Braga, Portugal
| | - Catherine J Harmer
- Department of Psychiatry, University of Oxford, Oxford, UK
- Oxford Health NHS Foundation Trust, Oxford, UK
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| |
Collapse
|
5
|
Oemisch M, Seo H. Therapeutic doses of ketamine acutely attenuate the aversive effect of losses during decision-making. eLife 2024; 12:RP87529. [PMID: 38700991 PMCID: PMC11068354 DOI: 10.7554/elife.87529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024] Open
Abstract
The discovery of rapid-acting antidepressant, ketamine has opened a pathway to a new generation of treatments for depression, and inspired neuroscientific investigation based on a new perspective that non-adaptive changes in the intrinsic excitatory and inhibitory circuitry might underlie the pathophysiology of depression. Nevertheless, it still remains largely unknown how the hypothesized molecular and synaptic levels of changes in the circuitry might mediate behavioral and neuropsychological changes underlying depression, and how ketamine might restore adaptive behavior. Here, we used computational models to analyze behavioral changes induced by therapeutic doses of ketamine, while rhesus macaques were iteratively making decisions based on gains and losses of tokens. When administered intramuscularly or intranasally, ketamine reduced the aversiveness of undesirable outcomes such as losses of tokens without significantly affecting the evaluation of gains, behavioral perseveration, motivation, and other cognitive aspects of learning such as temporal credit assignment and time scales of choice and outcome memory. Ketamine's potentially antidepressant effect was separable from other side effects such as fixation errors, which unlike outcome evaluation, was readily countered with strong motivation to avoid errors. We discuss how the acute effect of ketamine to reduce the initial impact of negative events could potentially mediate longer-term antidepressant effects through mitigating the cumulative effect of those events produced by slowly decaying memory, and how the disruption-resistant affective memory might pose challenges in treating depression. Our study also invites future investigations on ketamine's antidepressant action over diverse mood states and with affective events exerting their impacts at diverse time scales.
Collapse
Affiliation(s)
- Mariann Oemisch
- Department of Neuroscience, Yale School of MedicineNew HavenUnited States
| | - Hyojung Seo
- Department of Psychiatry and Neuroscience, Yale School of MedicineNew HavenUnited States
| |
Collapse
|
6
|
Evans JW, Graves MC, Nugent AC, Zarate CA. Hippocampal volume changes after (R,S)-ketamine administration in patients with major depressive disorder and healthy volunteers. Sci Rep 2024; 14:4538. [PMID: 38402253 PMCID: PMC10894199 DOI: 10.1038/s41598-024-54370-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 02/12/2024] [Indexed: 02/26/2024] Open
Abstract
The hippocampus and amygdala have been implicated in the pathophysiology and treatment of major depressive disorder (MDD). Preclinical models suggest that stress-related changes in these regions can be reversed by antidepressants, including ketamine. Clinical studies have identified reduced volumes in MDD that are thought to be potentiated by early life stress and worsened by repeated depressive episodes. This study used 3T and 7T structural magnetic resonance imaging data to examine longitudinal changes in hippocampal and amygdalar subfield volumes associated with ketamine treatment. Data were drawn from a previous double-blind, placebo-controlled, crossover trial of healthy volunteers (HVs) unmedicated individuals with treatment-resistant depression (TRD) (3T: 18 HV, 26 TRD, 7T: 17 HV, 30 TRD) who were scanned at baseline and twice following either a 40 min IV ketamine (0.5 mg/kg) or saline infusion (acute: 1-2 days, interim: 9-10 days post infusion). No baseline differences were noted between the two groups. At 10 days post-infusion, a slight increase was observed between ketamine and placebo scans in whole left amygdalar volume in individuals with TRD. No other differences were found between individuals with TRD and HVs at either field strength. These findings shed light on the timing of ketamine's effects on cortical structures.
Collapse
Affiliation(s)
- Jennifer W Evans
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr., Bldg 10, Rm 7-3335, Bethesda, MD, 20814, USA.
| | - Morgan C Graves
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr., Bldg 10, Rm 7-3335, Bethesda, MD, 20814, USA
| | - Allison C Nugent
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr., Bldg 10, Rm 7-3335, Bethesda, MD, 20814, USA
- MEG Core, NIMH, Bethesda, MD, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr., Bldg 10, Rm 7-3335, Bethesda, MD, 20814, USA
| |
Collapse
|
7
|
Ali DN, Ali HM, Lopez MR, Kang S, Choi DS. Astrocytic GABAergic Regulation in Alcohol Use and Major Depressive Disorders. Cells 2024; 13:318. [PMID: 38391931 PMCID: PMC10887002 DOI: 10.3390/cells13040318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/26/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
Gamma-aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the central nervous system (CNS). Most GABAergic neurons synthesize GABA from glutamate and release it in the synaptic cleft in the CNS. However, astrocytes can also synthesize and release GABA, activating GABA receptors in the neighboring neurons in physiological and pathological conditions. As the primary homeostatic glial cells in the brain, astrocytes play a crucial role in regulating GABA homeostasis and synaptic neurotransmission. Accumulating evidence demonstrates that astrocytic GABA dysregulation is implicated in psychiatric disorders, including alcohol use disorder (AUD) and major depressive disorder (MDD), the most prevalent co-occurring psychiatric disorders. Several current medications and emerging pharmacological agents targeting GABA levels are in clinical trials for treating AUD and MDD. This review offers a concise summary of the role of astrocytic GABA regulation in AUD and MDD. We also provide an overview of the current understanding and areas of debate regarding the mechanisms by which astrocytes regulate GABA in the CNS and their potential significance in the molecular basis of AUD and MDD, paving the way toward future research directions and potential therapeutic target areas within this field.
Collapse
Affiliation(s)
- Dina N. Ali
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
| | - Hossam M. Ali
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
| | - Matthew R. Lopez
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
| | - Shinwoo Kang
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
- Neuroscience Program, Rochester, MN 55905, USA
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| |
Collapse
|
8
|
Rothärmel M, Mekaoui L, Kazour F, Herrero M, Beetz-Lobono EM, Lengvenyte A, Holtzmann J, Raynaud P, Cuenca M, Bulteau S, de Maricourt P, Husson T, Olié E, Gohier B, Sauvaget A, Gaillard R, Richieri R, Szekely D, Samalin L, Guillin O, Moulier V, El-Hage W, Laurin A, Berkovitch L. Esketamine-induced post-traumatic stress disorder flashbacks during treatment-resistant depression indication: is it just a side effect? MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.09.24300998. [PMID: 38293161 PMCID: PMC10827260 DOI: 10.1101/2024.01.09.24300998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Background Posttraumatic stress disorder (PTSD) is a severe and frequent affection that is highly comorbid to major depressive disorder. Comorbid PTSD and depression are usually treatment-resistant, with a high risk of functional impairment and suicide. Esketamine nasal spray is a recent validated treatment for treatment-resistant depression (TRD), but its efficacy on comorbid TRD-PTSD remains insufficiently documented. In particular, flashbacks can occur during esketamine administration and their influence on clinical outcomes is unknown. Objectives Our main objective was to describe esketamine-induced traumatic flashbacks and their impact on clinical trajectories within a sample of patients with comorbid TRD-PTSD. Methods We retrospectively collected clinical data of patients receiving esketamine nasal spray for TRD with comorbid PTSD who experienced at least one flashback of their trauma during esketamine sessions across 11 psychiatric departments. Results Between February 2020 and March 2023, 22 adult patients with TRD met inclusion criteria. In sixteen patients (72.7%) flashbacks disappeared as the sessions progressed. In six patients (27.3%), esketamine treatment was stopped because of persistent flashbacks. When esketamine was continued, clinical response was observed both for depression and PTSD (depression response rate: 45.5% and remission rate: 22.7%; PTSD response rate: 45.5% and remission: 18.2%). Limitations The retrospective design of the study and the absence of a comparator group are the main limitations of our study. Conclusions Our results suggest that the occurrence of esketamine-induced traumatic flashbacks does not hinder clinical response. On the contrary, when managed appropriately and combined with targeted psychotherapy, it could even contribute to positive outcomes.
Collapse
Affiliation(s)
- Maud Rothärmel
- University Department of Psychiatry, Therapeutic Centre of Excellence, Institute of Psychiatry – Rouvray Hospital Centre, Sotteville-lès-Rouen, France
| | - Lila Mekaoui
- Mental and Brain Illness Clinic, Sainte-Anne Hospital, GHU Paris – Psychiatry and Neurosciences, Paris, France
| | - François Kazour
- Department of Psychiatry and Addictology, CHU Angers, Angers, France
| | - Morgane Herrero
- Department of Psychiatry, CHU Saint Etienne, Saint Etienne, France
| | | | - Aiste Lengvenyte
- Department of Emergency Psychiatry and Post-Acute Care, CHU Montpellier, France; IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Jérôme Holtzmann
- Service de Psychiatrie de l’Adulte, Centre Expert Dépression Résistante FondaMental, CHU de Grenoble, hôpital Nord, Grenoble, France
| | | | - Macarena Cuenca
- University Department of Psychiatry, Pôle Hospitalo-Universitaire Psychiatrie Paris 15, Groupe Hospitalier Universitaire Paris, Paris, France
| | - Samuel Bulteau
- Nantes Université, CHU Nantes, INSERM, MethodS in Patients-centered outcomes and HEalth Research, SPHERE, F-44000 Nantes, France
| | - Pierre de Maricourt
- University Department of Psychiatry, Pôle Hospitalo-Universitaire Psychiatrie Paris 15, Groupe Hospitalier Universitaire Paris, Paris, France
| | - Thomas Husson
- University Department of Psychiatry, Therapeutic Centre of Excellence, Institute of Psychiatry – Rouvray Hospital Centre, Sotteville-lès-Rouen, France
| | - Emilie Olié
- Department of Emergency Psychiatry and Post-Acute Care, CHU Montpellier, France; IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Bénédicte Gohier
- Department of Psychiatry and Addictology, CHU Angers, Angers, France
| | - Anne Sauvaget
- Nantes Université, CHU Nantes, INSERM, MethodS in Patients-centered outcomes and HEalth Research, SPHERE, F-44000 Nantes, France
| | - Raphaël Gaillard
- University Department of Psychiatry, Pôle Hospitalo-Universitaire Psychiatrie Paris 15, Groupe Hospitalier Universitaire Paris, Paris, France
| | - Raphaëlle Richieri
- Service Universitaire de santé mentale et physique, Centre Expert Dépression Résistante, CHU Sainte-Marguerite, APHM, Université Aix-Marseille, Marseille, France; Aix Marseille Univ, CNRS, Centrale Marseille, Institut Fresnel, Marseille, France
| | - David Szekely
- Centre Hospitalier Princesse Grace, Service de psychiatrie, Principauté de Monaco
| | - Ludovic Samalin
- Department of Psychiatry, CHU Clermont-Ferrand, University of Clermont Auvergne, CNRS, Clermont Auvergne INP, Institut Pascal (UMR 6602), Clermont-Ferrand, France
| | - Olivier Guillin
- University Department of Psychiatry, Therapeutic Centre of Excellence, Institute of Psychiatry – Rouvray Hospital Centre, Sotteville-lès-Rouen, France
- CHU Rouen, Normandy University, Rouen, France
| | - Virginie Moulier
- University Department of Psychiatry, Therapeutic Centre of Excellence, Institute of Psychiatry – Rouvray Hospital Centre, Sotteville-lès-Rouen, France
- Unité de Recherche Clinique (URC), EPS Ville Evrard, Neuilly-sur-Marne, France
| | - Wissam El-Hage
- Centre Régional de Psychotraumatologie, Centre Hospitalier Régional
- Universitaire (CHRU) de Tours, Tours, France, 3 INSERM U1253 Imagerie et Cerveau (iBrain), Tours, France
| | - Andrew Laurin
- Nantes Université, CHU Nantes, INSERM, MethodS in Patients-centered outcomes and HEalth Research, SPHERE, F-44000 Nantes, France
| | - Lucie Berkovitch
- University Department of Psychiatry, Pôle Hospitalo-Universitaire Psychiatrie Paris 15, Groupe Hospitalier Universitaire Paris, Paris, France
- Paris Cité University, Paris, France
- Department of Psychiatry, Yale University School of Medicine, 40 Temple Street, New Haven, CT, 06511, United States
- Saclay CEA Centre, Neurospin, Gif-Sur-Yvette Cedex, France
| |
Collapse
|
9
|
Hack LM, Zhang X, Heifets BD, Suppes T, van Roessel PJ, Yesavage JA, Gray NJ, Hilton R, Bertrand C, Rodriguez CI, Deisseroth K, Knutson B, Williams LM. Ketamine's acute effects on negative brain states are mediated through distinct altered states of consciousness in humans. Nat Commun 2023; 14:6631. [PMID: 37857620 PMCID: PMC10587184 DOI: 10.1038/s41467-023-42141-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 09/27/2023] [Indexed: 10/21/2023] Open
Abstract
Ketamine commonly and rapidly induces dissociative and other altered states of consciousness (ASCs) in humans. However, the neural mechanisms that contribute to these experiences remain unknown. We used functional neuroimaging to engage key regions of the brain's affective circuits during acute ketamine-induced ASCs within a randomized, multi-modal, placebo-controlled design examining placebo, 0.05 mg/kg ketamine, and 0.5 mg/kg ketamine in nonclinical adult participants (NCT03475277). Licensed clinicians monitored infusions for safety. Linear mixed effects models, analysis of variance, t-tests, and mediation models were used for statistical analyses. Our design enabled us to test our pre-specified primary and secondary endpoints, which were met: effects of ketamine across dose conditions on (1) emotional task-evoked brain activity, and (2) sub-components of dissociation and other ASCs. With this design, we also could disentangle which ketamine-induced affective brain states are dependent upon specific aspects of ASCs. Differently valenced ketamine-induced ASCs mediated opposing effects on right anterior insula activity. Participants experiencing relatively higher depersonalization induced by 0.5 mg/kg of ketamine showed relief from negative brain states (reduced task-evoked right anterior insula activity, 0.39 SD). In contrast, participants experiencing dissociative amnesia showed an exacerbation of insula activity (0.32 SD). These results in nonclinical participants may shed light on the mechanisms by which specific dissociative states predict response to ketamine in depressed individuals.
Collapse
Affiliation(s)
- Laura M Hack
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Sierra-Pacific Mental Illness Research, Education and Clinical Center (MIRECC), Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Xue Zhang
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Boris D Heifets
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Trisha Suppes
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Peter J van Roessel
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Sierra-Pacific Mental Illness Research, Education and Clinical Center (MIRECC), Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Jerome A Yesavage
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Sierra-Pacific Mental Illness Research, Education and Clinical Center (MIRECC), Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Nancy J Gray
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Rachel Hilton
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Claire Bertrand
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Carolyn I Rodriguez
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Karl Deisseroth
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Brian Knutson
- Department of Psychology, Stanford University, Stanford, CA, USA
| | - Leanne M Williams
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA.
- Sierra-Pacific Mental Illness Research, Education and Clinical Center (MIRECC), Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
10
|
Sen ZD, Chand T, Danyeli LV, Kumar VJ, Colic L, Li M, Yemisken M, Javaheripour N, Refisch A, Opel N, Macharadze T, Kretzschmar M, Ozkan E, Deliano M, Walter M. The effect of ketamine on affective modulation of the startle reflex and its resting-state brain correlates. Sci Rep 2023; 13:13323. [PMID: 37587171 PMCID: PMC10432502 DOI: 10.1038/s41598-023-40099-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 08/04/2023] [Indexed: 08/18/2023] Open
Abstract
Ketamine is a rapid-acting antidepressant that also influences neural reactivity to affective stimuli. However, the effect of ketamine on behavioral affective reactivity is yet to be elucidated. The affect-modulated startle reflex paradigm (AMSR) allows examining the valence-specific aspects of behavioral affective reactivity. We hypothesized that ketamine alters the modulation of the startle reflex during processing of unpleasant and pleasant stimuli and weakens the resting-state functional connectivity (rsFC) within the modulatory pathway, namely between the centromedial nucleus of the amygdala and nucleus reticularis pontis caudalis. In a randomized, double-blind, placebo-controlled, cross-over study, thirty-two healthy male participants underwent ultra-high field resting-state functional magnetic resonance imaging at 7 T before and 24 h after placebo and S-ketamine infusions. Participants completed the AMSR task at baseline and one day after each infusion. In contrast to our hypothesis, ketamine infusion did not impact startle potentiation during processing of unpleasant stimuli but resulted in diminished startle attenuation during processing of pleasant stimuli. This diminishment significantly correlated with end-of-infusion plasma levels of ketamine and norketamine. Furthermore, ketamine induced a decrease in rsFC within the modulatory startle reflex pathway. The results of this first study on the effect of ketamine on the AMSR suggest that ketamine might attenuate the motivational significance of pleasant stimuli in healthy participants one day after infusion.
Collapse
Affiliation(s)
- Zümrüt Duygu Sen
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743, Jena, Germany
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany
- Department of Psychiatry and Psychotherapy, University Tübingen, Tübingen, Germany
- German Center for Mental Health (DZPG), Halle-Jena-Magdeburg Site, Jena, Germany
| | - Tara Chand
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743, Jena, Germany
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany
- Department of Clinical Psychology, Friedrich Schiller University Jena, Am Steiger 3-1, 07743, Jena, Germany
- Jindal Institute of Behavioural Sciences, O. P. Jindal Global University (Sonipat), Haryana, India
| | - Lena Vera Danyeli
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743, Jena, Germany
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany
- Department of Psychiatry and Psychotherapy, University Tübingen, Tübingen, Germany
| | | | - Lejla Colic
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743, Jena, Germany
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany
- German Center for Mental Health (DZPG), Halle-Jena-Magdeburg Site, Jena, Germany
| | - Meng Li
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743, Jena, Germany
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany
- German Center for Mental Health (DZPG), Halle-Jena-Magdeburg Site, Jena, Germany
| | - Merve Yemisken
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany
| | - Nooshin Javaheripour
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743, Jena, Germany
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany
| | - Alexander Refisch
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743, Jena, Germany
| | - Nils Opel
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743, Jena, Germany
- German Center for Mental Health (DZPG), Halle-Jena-Magdeburg Site, Jena, Germany
| | - Tamar Macharadze
- Department of Anesthesiology and Intensive Care Medicine, Medical Faculty, Otto-Von-Guericke-Universität Magdeburg, Magdeburg, Germany
- Department Systems Physiology of Learning, Leibniz Institute for Neurobiology, Magdeburg, Germany
- Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Moritz Kretzschmar
- Department of Anesthesiology and Intensive Care Medicine, Medical Faculty, Otto-Von-Guericke-Universität Magdeburg, Magdeburg, Germany
| | - Esra Ozkan
- Koç University Research Center for Translational Medicine, Istanbul, Turkey
| | - Matthias Deliano
- Center for Behavioral Brain Sciences, Magdeburg, Germany.
- Leibniz Institute for Neurobiology, Magdeburg, Combinatorial NeuroImaging Core Facility, Brenneckestraße 6, 39118, Magdeburg, Germany.
| | - Martin Walter
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743, Jena, Germany.
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany.
- Department of Psychiatry and Psychotherapy, University Tübingen, Tübingen, Germany.
- German Center for Mental Health (DZPG), Halle-Jena-Magdeburg Site, Jena, Germany.
- Max Planck Institute for Biological Cybernetics, Tübingen, Germany.
- Center for Behavioral Brain Sciences, Magdeburg, Germany.
- Leibniz Institute for Neurobiology, Magdeburg, Germany.
| |
Collapse
|
11
|
Lullau APM, Haga EMW, Ronold EH, Dwyer GE. Antidepressant mechanisms of ketamine: a review of actions with relevance to treatment-resistance and neuroprogression. Front Neurosci 2023; 17:1223145. [PMID: 37614344 PMCID: PMC10442706 DOI: 10.3389/fnins.2023.1223145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/12/2023] [Indexed: 08/25/2023] Open
Abstract
Concurrent with recent insights into the neuroprogressive nature of depression, ketamine shows promise in interfering with several neuroprogressive factors, and has been suggested to reverse neuropathological patterns seen in depression. These insights come at a time of great need for novel approaches, as prevalence is rising and current treatment options remain inadequate for a large number of people. The rapidly growing literature on ketamine's antidepressant potential has yielded multiple proposed mechanisms of action, many of which have implications for recently elucidated aspects of depressive pathology. This review aims to provide the reader with an understanding of neuroprogressive aspects of depressive pathology and how ketamine is suggested to act on it. Literature was identified through PubMed and Google Scholar, and the reference lists of retrieved articles. When reviewing the evidence of depressive pathology, a picture emerges of four elements interacting with each other to facilitate progressive worsening, namely stress, inflammation, neurotoxicity and neurodegeneration. Ketamine acts on all of these levels of pathology, with rapid and potent reductions of depressive symptoms. Converging evidence suggests that ketamine works to increase stress resilience and reverse stress-induced dysfunction, modulate systemic inflammation and neuroinflammation, attenuate neurotoxic processes and glial dysfunction, and facilitate synaptogenesis rather than neurodegeneration. Still, much remains to be revealed about ketamine's antidepressant mechanisms of action, and research is lacking on the durability of effect. The findings discussed herein calls for more longitudinal approaches when determining efficacy and its relation to neuroprogressive factors, and could provide relevant considerations for clinical implementation.
Collapse
Affiliation(s)
- August P. M. Lullau
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Emily M. W. Haga
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Eivind H. Ronold
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Gerard E. Dwyer
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
- NORMENT Centre of Excellence, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
12
|
Doss MK, de Wit H, Gallo DA. The acute effects of psychoactive drugs on emotional episodic memory encoding, consolidation, and retrieval: A comprehensive review. Neurosci Biobehav Rev 2023; 150:105188. [PMID: 37085021 PMCID: PMC10247427 DOI: 10.1016/j.neubiorev.2023.105188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 04/23/2023]
Abstract
Psychoactive drugs modulate learning and emotional processes in ways that could impact their recreational and medical use. Recent work has revealed how drugs impact different stages of processing emotional episodic memories, specifically encoding (forming memories), consolidation (stabilizing memories), and retrieval (accessing memories). Drugs administered before encoding may preferentially impair (e.g., GABAA sedatives including alcohol and benzodiazepines, Δ9-tetrahydrocannabinol or THC, ketamine), enhance (e.g., dextroamphetamine and dextromethamphetamine), or both impair and enhance (i.e., ± 3,4-methylenedioxymethylamphetamine or MDMA) emotionally negative and positive compared to neutral memories. GABAA sedatives administered immediately post-encoding (during consolidation) can preferentially enhance emotional memories, though this selectivity may decline or even reverse (i.e., preferential enhancement of neutral memories) as the delay between encoding and retrieval increases. Finally, retrieving memories under the effects of THC, dextroamphetamine, MDMA, and perhaps GABAA sedatives distorts memory, with potentially greater selectively for emotional (especially positive) memories. We review these effects, propose neural mechanisms, discuss methodological considerations for future work, and speculate how drug effects on emotional episodic memory may contribute to drug use and abuse.
Collapse
Affiliation(s)
- Manoj K Doss
- Department of Psychiatry and Behavioral Sciences, Center for Psychedelic & Consciousness Research, USA.
| | - Harriet de Wit
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, USA
| | - David A Gallo
- Department of Psychology, University of Chicago, USA
| |
Collapse
|
13
|
Alijanpour S, Rezayof A. Activation of ventral hippocampal CB1 receptors inhibits ketamine-induced anxiogenic-like behavior: Alteration of BDNF/c-Fos levels in the mouse hippocampus. Brain Res 2023; 1810:148378. [PMID: 37121426 DOI: 10.1016/j.brainres.2023.148378] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/15/2023] [Accepted: 04/25/2023] [Indexed: 05/02/2023]
Abstract
Considering the increasing usage of ketamine as a recreational drug with hallucinogenic properties and also scarce studies about receptor systems responsible for its effects, in the present study we aimed to investigate whether the activation of the ventral hippocampal (VH) CB1 cannabinoid receptors affects the anxiety-like behaviors induced by ketamine. Also, the levels of BDNF and c-Fos proteins in the mouse hippocampus were measured following the treatments. For this purpose, male NMRI mice were cannulated bilaterally in the VH with a stereotaxic apparatus. Anxiety properties and protein changes were measured using elevated plus-maze (EPM) and western blotting respectively. The results revealed that intraperitoneal (i.p.) administration of ketamine (5-20 mg/kg) significantly decreased the percentage of open arm time (%OAT) and open arm entry (%OAE) in the EPM with no alteration in the locomotor activity suggesting an anxiogenic-like behavior to ketamine. Furthermore, ketamine administration (10 mg/kg, i.p.) increased BDNF and c-Fos levels in the hippocampus. Interestingly, activation of the VH CB1 receptors by ACPA (0.5-4 ng/mouse) inhibited the anxiogenic-like behaviors produced by ketamine, whereas the microinjection of the same doses of ACPA into VH by itself had no effect on the EPM parameters. Hippocampal levels of BDNF and c-Fos decreased after treatment with combined ketamine with ACPA. These results suggest the therapeutic potency of cannabinoid receptor agonists for ketamine-induced anxiogenic-related responses. This effect might be at least partially mediated by the alteration of BDNF and c-Fos signaling in the hippocampus.
Collapse
Affiliation(s)
- Sakineh Alijanpour
- Department of Biology, Faculty of Science, Gonbad Kavous University, Gonbad Kavous, Iran.
| | - Ameneh Rezayof
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
14
|
The Altered States Database: Psychometric data from a systematic literature review. Sci Data 2022; 9:720. [PMID: 36418335 PMCID: PMC9684144 DOI: 10.1038/s41597-022-01822-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 11/01/2022] [Indexed: 11/26/2022] Open
Abstract
In this paper, we present the development of the Altered States Database (ASDB), an open-science project based on a systematic literature review. The ASDB contains psychometric questionnaire data on subjective experiences of altered states of consciousness (ASC) induced by pharmacological and non-pharmacological methods. The systematic review follows the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Scientific journal articles were identified through PubMed and Web of Science. We included studies that examined ASC using the following validated questionnaires: Altered States of Consciousness Rating Scale (APZ, 5D-ASC, 11-ASC), Phenomenology of Consciousness Inventory (PCI), Hallucinogen Rating Scale (HRS), or Mystical Experience Questionnaire (MEQ30). The systematic review resulted in the inclusion of a total of 165 journal articles, whereof questionnaire data was extracted and is now available on the Open Science Framework (OSF) website (https://osf.io/8mbru) and on the ASDB website (http://alteredstatesdb.org), where questionnaire data can be easily retrieved and visualized. This data allows the calculation of comparable psychometric values of ASC experiences and of dose-response relationships of substances inducing ASC. Measurement(s) | Psychometric questionnaire data | Technology Type(s) | Systematic literature review (PRISMA) | Sample Characteristic - Organism | Human |
Collapse
|
15
|
Vogt KM, Pryor KO. Anesthesia and the neurobiology of fear and posttraumatic stress disorder. Curr Opin Anaesthesiol 2022; 35:593-599. [PMID: 35993581 PMCID: PMC9469898 DOI: 10.1097/aco.0000000000001176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
PURPOSE OF REVIEW Dysfunction of fear memory systems underlie a cluster of clinically important and highly prevalent psychological morbidities seen in perioperative and critical care patients, most archetypally posttraumatic stress disorder (PTSD). Several sedative-hypnotics and analgesics are known to modulate fear systems, and it is theoretically plausible that clinical decisions of the anesthesiologist could impact psychological outcomes. This review aims to provide a focused synthesis of relevant literature from multiple fields of research. RECENT FINDINGS There is evidence in some contexts that unconscious fear memory systems are less sensitive to anesthetics than are conscious memory systems. Opiates may suppress the activation of fear systems and have benefit in the prevention of PTSD following trauma. There is inconsistent evidence that the use of propofol and benzodiazepines for sedation following trauma may potentiate the development of PTSD relative to other drugs. The benefits of ketamine seen in the treatment of major depression are not clearly replicated in PTSD-cluster psychopathologies, and its effects on fear processes are complex. SUMMARY There are multiple theoretical mechanisms by which anesthetic drugs can modulate fear systems and clinically important fear-based psychopathologies. The current state of research provides some evidence to support further hypothesis investigation. However, the absence of effectiveness studies and the inconsistent signals from smaller studies provide insufficient evidence to currently offer firm clinical guidance.
Collapse
Affiliation(s)
- Keith M. Vogt
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, School of Medicine
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh
- Center for the Neural Basis of Cognition
- Clinical and Translational Science Institute, University of Pittsburgh
| | - Kane O. Pryor
- Department of Anesthesiology, Weill Cornell Medicine
| |
Collapse
|
16
|
Zhang F, Wang C, Lan X, Li W, Fu L, Ye Y, Liu H, Wu K, Zhou Y, Ning Y. The functional connectivity of the middle frontal cortex predicts ketamine’s outcome in major depressive disorder. Front Neurosci 2022; 16:956056. [PMID: 36188452 PMCID: PMC9521309 DOI: 10.3389/fnins.2022.956056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 08/15/2022] [Indexed: 11/24/2022] Open
Abstract
Background Ketamine, a robust antidepressant, has promising potential in the treatment of major depressive disorder (MDD). However, it does not work for all MDD patients, and the mechanism underlying its anti-depressive effects is unclear. Researchers have explored the mechanisms of ketamine action in MDD patients through MRI, a technique that measures brain activity intuitively. Notably, many MRI results were inconsistent because they selected different brain regions as seeds, particularly with respect to functional connectivity (FC) analysis. To eliminate the influence of prior seeds as much as possible, we used the significantly different results in degree centrality (DC) analysis as seeds to explore the FC changes in MDD patients to identify an imaging biomarker of ketamine’s effect. Methods Forty-four MDD patients and 45 healthy controls (HCs) were included in the study. Patients, aged 18–65, received six intravenous ketamine injections over 12 days. Depressive symptoms were estimated and MRI scans were performed at baseline and the day after the sixth infusion. We estimated FC differences between responders, non-responders and HCs using the region that showed significant differences between responders and non-responders in DC analysis as the seed. The correlation between the MADRS changes and zFC values was performed, and the potential of zFC values to be a neuroimaging biomarker was explored using the receiver operating characteristic curve. Result Compared with non-responders, responders had significantly decreased DC values in the right middle frontal gyrus (MFG). In the analysis of FC using the region that showed significant differences in DC as a seed, there was a significant difference in the region of the right supplementary motor area (SMA) among responders, non-responders, and HCs. This region also overlapped with the bilateral median cingulate gyrus. In post hoc analysis, responders had higher FC than non-responders and HCs, and non-responders had lower FC than HCs. Importantly, the FC between the MFG and SMA (overlapping bilateral median cingulate gyrus) was correlated with the improvement of symptoms, which was estimated by the Mongomery-Asberg Depression Scale (MADRS). FC has the potential to be an imaging biomarker that can predict the ketamine effect in MDD patients according to the receiver operating characteristic curve analysis. Conclusion Our results revealed that FC between the SMG and SMA and mACC was highly correlated with depressive symptoms and has the potential to be a neuroimaging biomarker to predict the effect of ketamine in MDD.
Collapse
Affiliation(s)
- Fan Zhang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Chengyu Wang
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Xiaofeng Lan
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Weicheng Li
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Ling Fu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yanxiang Ye
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Haiyan Liu
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Kai Wu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzho, China
| | - Yanling Zhou
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yuping Ning
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
- *Correspondence: Yuping Ning,
| |
Collapse
|
17
|
Zhornitsky S, Tourjman V, Pelletier J, Assaf R, Li CSR, Potvin S. Acute effects of ketamine and esketamine on cognition in healthy subjects: A meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry 2022; 118:110575. [PMID: 35568275 DOI: 10.1016/j.pnpbp.2022.110575] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/04/2022] [Accepted: 05/09/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Impairment in cognition is frequently associated with acute ketamine administration. However, some questions remain unanswered as to which deficits are most prominent and what variables modulate these effects. METHODS A literature search yielded 56 experimental studies of acute ketamine administration that assessed cognition in 1041 healthy volunteers. A multivariate meta-analysis was performed, and effect sizes were estimated for eleven cognitive domains: attention, executive function, response inhibition, social cognition, speed of processing, verbal / language, verbal learning, verbal memory, visual learning & memory, visuospatial abilities, and working memory. RESULTS There were small-to-moderate impairments across all cognitive domains. Deficits in verbal learning / memory were most prominent, whereas response inhibition was the least affected. Meta-regression analysis revealed that the negative effects of ketamine on cognition are dependent on infusion dose and plasma level, but unaffected by enantiomer type, route of administration, sex or age. A publication bias was observed. DISCUSSION Acute ketamine broadly impairs cognition across all domains among healthy individuals. Verbal learning and memory figures most prominently in cognitive impairment elicited by acute ketamine administration.
Collapse
Affiliation(s)
- Simon Zhornitsky
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Valérie Tourjman
- Department of Psychiatry, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada; Centre de recherche de l'Institut Universitaire en Santé Mentale de Montréal, Montreal, QC, Canada
| | - Julie Pelletier
- Department of Psychiatry, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Roxane Assaf
- Department of Psychiatry, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada; Centre de recherche de l'Institut Universitaire en Santé Mentale de Montréal, Montreal, QC, Canada
| | - Chiang-Shan R Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA; Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA
| | - Stéphane Potvin
- Department of Psychiatry, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada; Centre de recherche de l'Institut Universitaire en Santé Mentale de Montréal, Montreal, QC, Canada.
| |
Collapse
|
18
|
Optimized Clinical Strategies for Treatment-Resistant Depression: Integrating Ketamine Protocols with Trauma- and Attachment-Informed Psychotherapy. PSYCH 2022. [DOI: 10.3390/psych4010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Strategically timed trauma- and attachment-informed psychotherapy to address underlying emotional wounds, paired with ketamine administered in precision-calibrated doses to ensure high-entropy brain states, may be key to improving the quality and duration of ketamine’s therapeutic efficacy for treatment-resistant depression. This approach optimizes the opportunities for change created by ketamine’s known effects as a rapid antidepressant that stimulates synaptogenesis, normalizes neural connectivity and coherence, enhances neuroplasticity, reduces inflammation, and induces high-entropy brain states with associated subjective psychedelic experiences. Ketamine, a non-selective N-methyl-D-aspartate (NMDA) receptor antagonist is a safe, effective, fast-acting dissociative anesthetic that, as a standalone treatment, also exhibits rapid sustained antidepressant effects, even in many patients with treatment-resistant depression. A prior history of developmental trauma and attachment injuries are known primary factors in the etiology of treatment resistance in depression and other mental disorders. Thus, the adjunct of targeted psychotherapy attuned to trauma and attachment injuries may enhance and prolong ketamine efficacy and provide an opportunity for lasting therapeutic change. Psychotherapy engagement during repeated ketamine sessions for patient safety and integration of altered states, paired with separate individualized psychotherapy-only sessions timed 24–48 h post ketamine induction, takes advantage of peak ketamine-induced dendritic spine growth in the prefrontal cortex and limbic system, and normalized network connectivity across brain structures. This strategically timed paired-session approach also exploits the therapeutic potential created by precision-calibrated ketamine-linked high-entropy brain states and associated psychedelic experiences that are posited to disrupt overly rigid maladaptive thoughts, behaviors, and disturbing memories associated with treatment-resistant depression; paired sessions also support integration of the felt sense of happiness and connectivity associated with psychedelic experiences.
Collapse
|
19
|
Kotoula V, Webster T, Stone J, Mehta MA. Resting-state connectivity studies as a marker of the acute and delayed effects of subanaesthetic ketamine administration in healthy and depressed individuals: A systematic review. Brain Neurosci Adv 2021; 5:23982128211055426. [PMID: 34805548 PMCID: PMC8597064 DOI: 10.1177/23982128211055426] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 09/24/2021] [Indexed: 11/15/2022] Open
Abstract
Acute ketamine administration has been widely used in neuroimaging research to mimic psychosis-like symptoms. Within the last two decades, ketamine has also emerged as a potent, fast-acting antidepressant. The delayed effects of the drug, observed 2–48 h after a single infusion, are associated with marked improvements in depressive symptoms. At the systems’ level, several studies have investigated the acute ketamine effects on brain activity and connectivity; however, several questions remain unanswered around the brain changes that accompany the drug’s antidepressant effects and how these changes relate to the brain areas that appear with altered function and connectivity in depression. This review aims to address some of these questions by focusing on resting-state brain connectivity. We summarise the studies that have examined connectivity changes in treatment-naïve, depressed individuals and those studies that have looked at the acute and delayed effects of ketamine in healthy and depressed volunteers. We conclude that brain areas that are important for emotional regulation and reward processing appear with altered connectivity in depression whereas the default mode network presents with increased connectivity in depressed individuals compared to healthy controls. This finding, however, is not as prominent as the literature often assumes. Acute ketamine administration causes an increase in brain connectivity in healthy volunteers. The delayed effects of ketamine on brain connectivity vary in direction and appear to be consistent with the drug normalising the changes observed in depression. The limited number of studies however, as well as the different approaches for resting-state connectivity analysis make it very difficult to draw firm conclusions and highlight the importance of data sharing and larger future studies.
Collapse
Affiliation(s)
- Vasileia Kotoula
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | | | | | - Mitul A Mehta
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
20
|
Van Hedger K, Mayo LM, Bershad AK, Madray R, de Wit H. Effects of Acute Drug Administration on Emotion: A Review of Pharmacological MRI Studies. CURRENT ADDICTION REPORTS 2021; 8:181-193. [PMID: 34631363 DOI: 10.1007/s40429-021-00362-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Purpose of review Many drug users claim to use drugs to cope with negative emotions, which may, in turn, result in persistent emotional blunting or anhedonia even when they are not using drugs. The purpose of this review is to describe the ways acute administration of psychoactive drugs impacts brain regions during emotion-related tasks, as a first step in understanding how drugs influence emotion processing in the brain. Recent findings Drugs have varying effects on neural responses to emotional stimuli. In general, alcohol, analgesics, and psychedelics reduce neural reactivity to negative emotional stimuli in the amygdala and other brain regions. Other drugs produce mixed effects: Stimulants such as caffeine and modafinil increase brain activation while viewing emotional stimuli, whereas MDMA decreases activation during presentation of negative images. The effects of cannabinoids (cannabidiol and THC) are mixed. There are also inconsistent findings on the associations between neural responses to emotional stimuli and subjective drug effects. Summary Consistent with the notion that individuals might use drugs non-medically to diminish the experience of negative emotions, several drugs of abuse decrease neural responses to negative stimuli in limbic brain regions. These neural actions may underlie the reported 'emotional blunting' of drugs, which may contribute to drug-seeking behavior. Future work is needed to examine these limbic responses in relation to self-reports of changes in affect, both during acute administration and after extended drug use.
Collapse
Affiliation(s)
| | - Leah M Mayo
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| | - Anya K Bershad
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Racheal Madray
- Undergraduate Program in Neuroscience, Western University, London, ON, Canada
| | - Harriet de Wit
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, USA
| |
Collapse
|
21
|
Therapeutic potential of ketamine for alcohol use disorder. Neurosci Biobehav Rev 2021; 126:573-589. [PMID: 33989669 DOI: 10.1016/j.neubiorev.2021.05.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 04/02/2021] [Accepted: 05/09/2021] [Indexed: 12/12/2022]
Abstract
Excessive alcohol consumption is involved in 1/10 of deaths of U.S. working-age adults and costs the country around $250,000,000 yearly. While Alcohol Use Disorder (AUD) pathology is complex and involves multiple neurotransmitter systems, changes in synaptic plasticity, hippocampal neurogenesis, and neural connectivity have been implicated in the behavioral characteristics of AUD. Depressed mood and stress are major determinants of relapse in AUD, and there is significant comorbidity between AUD, depression, and stress disorders, suggesting potential for overlap in their treatments. Disulfiram, naltrexone, and acamprosate are current pharmacotherapies for AUD, but these treatments have limitations, highlighting the need for novel therapeutics. Ketamine is a N-methyl-D-Aspartate receptor antagonist, historically used in anesthesia, but also affects other neurotransmitters systems, synaptic plasticity, neurogenesis, and neural connectivity. Currently under investigation for treating AUDs and other Substance Use Disorders (SUDs), ketamine has strong support for efficacy in treating clinical depression, recently receiving FDA approval. Ketamine's effect in treating depression and stress disorders, such as PTSD, and preliminary evidence for treating SUDs further suggests a role for treating AUDs. This review explores the behavioral and neural evidence for treating AUDs with ketamine and clinical data on ketamine therapy for AUDs and SUDs.
Collapse
|
22
|
Alexander L, Jelen LA, Mehta MA, Young AH. The anterior cingulate cortex as a key locus of ketamine's antidepressant action. Neurosci Biobehav Rev 2021; 127:531-554. [PMID: 33984391 DOI: 10.1016/j.neubiorev.2021.05.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 12/30/2022]
Abstract
The subdivisions of the anterior cingulate cortex (ACC) - including subgenual, perigenual and dorsal zones - are implicated in the etiology, pathogenesis and treatment of major depression. We review an emerging body of evidence which suggests that changes in ACC activity are critically important in mediating the antidepressant effects of ketamine, the prototypical member of an emerging class of rapidly acting antidepressants. Infusions of ketamine induce acute (over minutes) and post-acute (over hours to days) modulations in subgenual and perigenual activity, and importantly, these changes can correlate with antidepressant efficacy. The subgenual and dorsal zones of the ACC have been specifically implicated in ketamine's anti-anhedonic effects. We emphasize the synergistic relationship between neuroimaging studies in humans and brain manipulations in animals to understand the causal relationship between changes in brain activity and therapeutic efficacy. We conclude with circuit-based perspectives on ketamine's action: first, related to ACC function in a central network mediating affective pain, and second, related to its role as the anterior node of the default mode network.
Collapse
Affiliation(s)
- Laith Alexander
- Department of Psychological Medicine, School of Academic Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; St Thomas' Hospital, London, United Kingdom.
| | - Luke A Jelen
- Department of Psychological Medicine, School of Academic Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - Mitul A Mehta
- Department of Psychological Medicine, School of Academic Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Allan H Young
- Department of Psychological Medicine, School of Academic Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; South London and Maudsley NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
23
|
Chírico MTT, Guedes MR, Vieira LG, Reis TO, Dos Santos AM, Souza ABF, Ribeiro IML, Noronha SISR, Nogueira KO, Oliveira LAM, Gomes FAR, Silva FC, Chianca-Jr DA, Bezerra FS, de Menezes RCA. Lasting effects of ketamine and isoflurane administration on anxiety- and panic-like behavioral responses in Wistar rats. Life Sci 2021; 276:119423. [PMID: 33785344 DOI: 10.1016/j.lfs.2021.119423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/18/2021] [Accepted: 03/20/2021] [Indexed: 10/21/2022]
Abstract
In clinical and laboratory practice, the use of anesthetics is essential in order to perform surgeries. Anesthetics, besides causing sedation and muscle relaxation, promote several physiological outcomes, such as psychotomimetic alterations, increased heart rate, and blood pressure. However, studies depicting the behavioral effect induced by ketamine and isoflurane are conflicting. In the present study, we assessed the behavioral effects precipitated by ketamine and isoflurane administration. We have also evaluated the ketamine effect on cell cytotoxicity and viability in an amygdalar neuronal primary cell culture. Ketamine (80 mg/kg) caused an anxiogenic effect in rats exposed to the elevated T-maze test (ETM) 2 and 7 days after ketamine administration. Ketamine (40 and 80 mg/kg) administration also decreased panic-like behavior in the ETM. In the light/dark test, ketamine had an anxiogenic effect. Isoflurane did not change animal behavior on the ETM. Neither ketamine nor isoflurane changed the spontaneous locomotor activity in the open field test. However, isoflurane-treated animals explored less frequently the OF central area seven days after treatment. Neither anesthetic caused oxidative damage in the liver. Ketamine also reduced cellular metabolism and led to neuronal death in amygdalar primary cell cultures. Thus, our work provides evidence that ketamine and isoflurane induce pronounced long lasting anxiety-related behaviors in male rats.
Collapse
Affiliation(s)
- Máira Tereza Talma Chírico
- Department of Biological Sciences, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil
| | - Mariana Reis Guedes
- Department of Biological Sciences, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil
| | - Lucas Gabriel Vieira
- Department of Biological Sciences, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil
| | - Thayane Oliveira Reis
- Department of Biological Sciences, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil
| | - Aline Maria Dos Santos
- Department of Biological Sciences, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil
| | - Ana Beatriz Farias Souza
- Department of Biological Sciences, Laboratory of Experimental Pathophysiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil
| | - Iara Mariana Léllis Ribeiro
- Department of Biological Sciences, Laboratory of Biomaterials and Experimental Pathology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil
| | - Sylvana I S R Noronha
- Department of Biological Sciences, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil
| | - Katiane O Nogueira
- Department of Biological Sciences, Laboratory of Biomaterials and Experimental Pathology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil.
| | - Laser Antonio Machado Oliveira
- Department of Biological Sciences, Laboratory of Biomaterials and Experimental Pathology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil.
| | - Fabiana Aparecida Rodrigues Gomes
- Department of Biological Sciences, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil
| | - Fernanda Cacilda Silva
- Department of Biological Sciences, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil.
| | - Deoclécio Alves Chianca-Jr
- Department of Biological Sciences, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil.
| | - Frank Silva Bezerra
- Department of Biological Sciences, Laboratory of Experimental Pathophysiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil.
| | - Rodrigo Cunha Alvim de Menezes
- Department of Biological Sciences, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil.
| |
Collapse
|
24
|
Muscat SA, Hartelius G, Crouch CR, Morin KW. An Integrative Approach to Ketamine Therapy May Enhance Multiple Dimensions of Efficacy: Improving Therapeutic Outcomes With Treatment Resistant Depression. Front Psychiatry 2021; 12:710338. [PMID: 34899408 PMCID: PMC8653702 DOI: 10.3389/fpsyt.2021.710338] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 10/21/2021] [Indexed: 12/31/2022] Open
Abstract
Research over the last two decades has established ketamine as a safe, effective, fast-acting, and sustained antidepressant that significantly reduces adverse symptoms associated with depression, even in patients who are treatment resistant. Much of this research has evolved within the framework of several independent branches of scientific inquiry: in addition to the study of ketamine is a non-selective NMDAR antagonist with rapid antidepressant effects, it has also been found effective as a psychoplastogen that stimulates synaptogenesis and increases neuroplasticity, as a powerful anti-inflammatory that may improve inflammation-related depressive symptoms, as a substance that induces beneficial high entropy brain states, and as a subjectively impactful psychedelic agent. Each branch of inquiry has generated independent evidence of ketamine's efficacy but has advanced without substantive coordination or communication with other lines of inquiry. Integrative research that considers these branches of research together may lead toward a better understanding of ketamine's effects and improved treatment protocols and clinical outcomes. Such an overview can inform more comprehensive patient care through: (a) informed patient psychoeducation that encompasses all of ketamine's mechanisms of action; (b) calibration of optimal dosage to ensure induction and maintenance of high entropy brain states during each ketamine session utilizing EEG measurement; (c) Improved management of emergence side effects through proper care for set and setting; (d) inclusion of pre-selected appropriate music to enhance the emotional experience; (e) increased monitoring of ketamine effects on cortical activity, inter-hemispheric imbalance, and inflammation-related levels of cytokines to further improvements in ketamine protocols; and (f) appropriate timing of any adjunctive psychotherapy sessions to coincide with peak neurogenesis at 24-48 h post ketamine treatment.
Collapse
Affiliation(s)
- Sherry-Anne Muscat
- Youth Forensic Psychiatry, Alberta Hospital, Alberta Health Services, Edmonton, AB, Canada.,Integral and Transpersonal Psychology, California Institute of Integral Studies, San Francisco, CA, United States
| | - Glenn Hartelius
- Integral and Transpersonal Psychology, California Institute of Integral Studies, San Francisco, CA, United States
| | - Courtenay Richards Crouch
- Integral and Transpersonal Psychology, California Institute of Integral Studies, San Francisco, CA, United States
| | - Kevin W Morin
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada.,Acute Adult Psychiatry, Alberta Hospital, Alberta Health Services, Edmonton, AB, Canada
| |
Collapse
|
25
|
Haarsma J, Harmer CJ, Tamm S. A continuum hypothesis of psychotomimetic rapid antidepressants. Brain Neurosci Adv 2021; 5:23982128211007772. [PMID: 34017922 PMCID: PMC8114748 DOI: 10.1177/23982128211007772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/08/2021] [Indexed: 01/10/2023] Open
Abstract
Ketamine, classical psychedelics and sleep deprivation are associated with rapid effects on depression. Interestingly, these interventions also have common psychotomimetic actions, mirroring aspects of psychosis such as an altered sense of self, perceptual distortions and distorted thinking. This raises the question whether these interventions might be acute antidepressants through the same mechanisms that underlie some of their psychotomimetic effects. That is, perhaps some symptoms of depression can be understood as occupying the opposite end of a spectrum where elements of psychosis can be found on the other side. This review aims at reviewing the evidence underlying a proposed continuum hypothesis of psychotomimetic rapid antidepressants, suggesting that a range of psychotomimetic interventions are also acute antidepressants as well as trying to explain these common features in a hierarchical predictive coding framework, where we hypothesise that these interventions share a common mechanism by increasing the flexibility of prior expectations. Neurobiological mechanisms at play and the role of different neuromodulatory systems affected by these interventions and their role in controlling the precision of prior expectations and new sensory evidence will be reviewed. The proposed hypothesis will also be discussed in relation to other existing theories of antidepressants. We also suggest a number of novel experiments to test the hypothesis and highlight research areas that could provide further insights, in the hope to better understand the acute antidepressant properties of these interventions.
Collapse
Affiliation(s)
- Joost Haarsma
- Wellcome Centre for Human Neuroimaging, University College London, London, UK
| | - Catherine J Harmer
- Department of Psychiatry and Oxford Health NHS Foundation Trust, Warneford Hospital, University of Oxford, Oxford, UK
| | - Sandra Tamm
- Department of Psychiatry and Oxford Health NHS Foundation Trust, Warneford Hospital, University of Oxford, Oxford, UK
- Stress Research Institute, Department of Psychology, Stockholm University, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
26
|
Godlewska BR, Harmer CJ. Cognitive neuropsychological theory of antidepressant action: a modern-day approach to depression and its treatment. Psychopharmacology (Berl) 2021; 238:1265-1278. [PMID: 31938879 PMCID: PMC8062380 DOI: 10.1007/s00213-019-05448-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 12/27/2019] [Indexed: 12/12/2022]
Abstract
Depression is a leading cause of disability worldwide and improving its treatment is a core research priority for future programmes. A change in the view of psychological and biological processes, from seeing them as separate to complementing one another, has introduced new perspectives on pathological mechanisms of depression and treatment mode of action. This review presents a theoretical model that incorporated this novel approach, the cognitive neuropsychological hypothesis of antidepressant action. This model proposes that antidepressant treatments decrease the negative bias in the processing of emotionally salient information early in the course of antidepressant treatment, which leads to the clinically significant mood improvement later in treatment. The paper discusses the role of negative affective biases in the development of depression and response to antidepressant treatments. It also discusses whether the model can be applied to other antidepressant interventions and its potential translational value, including treatment choice, prediction of response and drug development.
Collapse
Affiliation(s)
- Beata R Godlewska
- Department of Psychiatry, Psychopharmacology Research Unit, University Department of Psychiatry (PPRU), University of Oxford, Oxford, UK.
- Department of Psychiatry, Psychopharmacology and Emotion Research Laboratory (PERL), University of Oxford, Oxford, UK.
- Oxford Health Foundation Trust, University Department of Psychiatry, Warneford Hospital, Oxford, OX3 7JX, UK.
| | - Catherine J Harmer
- Department of Psychiatry, Psychopharmacology and Emotion Research Laboratory (PERL), University of Oxford, Oxford, UK
- Oxford Health Foundation Trust, University Department of Psychiatry, Warneford Hospital, Oxford, OX3 7JX, UK
| |
Collapse
|
27
|
Abstract
An intranasal formulation of esketamine, the S enantiomer of ketamine, in conjunction with an oral antidepressant, has been approved by the FDA for treating treatment-resistant major depressive disorder (TRD) in 2019, almost 50 years after it was approved as an intravenous anesthetic. In contrast to traditional antidepressants, ketamine shows a rapid (within 2 h) and sustained (∼7 days) antidepressant effect and has significant positive effects on antisuicidal ideation. Ketamine's antidepressant mechanism is predominantly mediated by the N-methyl-d-aspartate receptor (NMDA) receptor, although NMDA-independent mechanisms are not ruled out. At the neurocircuitry level, ketamine affects the brain's reward and mood circuitry located in the corticomesolimbic structures involving the hippocampus, nucleus accumbens, and prefrontal cortex. Repurposing of ketamine for treating TRD provided a new understanding of the pathophysiology of depression, a paradigm shift from monoamine to glutamatergic neurotransmission, thus making it a unique tool to investigate the brain and its complex neurocircuitries.
Collapse
Affiliation(s)
- Joydip Das
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas 77204, United States
| |
Collapse
|
28
|
Osborne MT, Shin LM, Mehta NN, Pitman RK, Fayad ZA, Tawakol A. Disentangling the Links Between Psychosocial Stress and Cardiovascular Disease. Circ Cardiovasc Imaging 2020; 13:e010931. [PMID: 32791843 DOI: 10.1161/circimaging.120.010931] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Stress is a pervasive component of the human experience. While often considered an adversity to be ignored, chronic stress has important pathological consequences, including cardiovascular disease (CVD). Stress also increases the prevalence and severity of several CVD risk factors, including hypertension, diabetes mellitus, and obesity. Yet even after adjustment, stress' attributable CVD risk is similar to those risk factors, suggesting it is a particularly potent contributor. Nevertheless, there has been insufficient study of mechanisms linking stress to CVD or of methods to attenuate stress' pathological impact. This review covers the current concepts of how stress impacts CVD and emerging approaches to mitigate stress-attributable CVD risk.
Collapse
Affiliation(s)
- Michael T Osborne
- Cardiology Division (M.T.O., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston, MA.,Cardiovascular Imaging Research Center, Departments of Medicine and Imaging (M.T.O., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Lisa M Shin
- Department of Psychiatry (L.M.S., R.K.P.), Massachusetts General Hospital and Harvard Medical School, Boston, MA.,Department of Psychology, Tufts University, Boston, MA (L.M.S.)
| | - Nehal N Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, Bethesda, MD (N.N.M.)
| | - Roger K Pitman
- Department of Psychiatry (L.M.S., R.K.P.), Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Zahi A Fayad
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY (Z.A.F.)
| | - Ahmed Tawakol
- Cardiology Division (M.T.O., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston, MA.,Cardiovascular Imaging Research Center, Departments of Medicine and Imaging (M.T.O., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
29
|
Zhou YL, Wu FC, Liu WJ, Zheng W, Wang CY, Zhan YN, Lan XF, Ning YP. Volumetric changes in subcortical structures following repeated ketamine treatment in patients with major depressive disorder: a longitudinal analysis. Transl Psychiatry 2020; 10:264. [PMID: 32747631 PMCID: PMC7400625 DOI: 10.1038/s41398-020-00945-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/07/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Abnormal subcortical structures have been associated with major depressive disorder (MDD) and could be reversed by antidepressant treatment. To date no study has examined the relationship between subcortical volumes and repeated ketamine treatment. The current study investigated volume changes in specific subcortical structures and hippocampal subfields after six ketamine infusions. Forty-four patients with MDD received six subanesthetic dose infusions of ketamine. Depressive symptoms were assessed and magnetic resonance imaging scans were performed before and after six ketamine infusions. FreeSurfer software was used to process the T1 images and analyze the volumes of the subcortical regions and hippocampal subfields. After six ketamine infusions, increases were observed in the volumes of the left amygdala; the right hippocampus; the cornu ammonis 4 body, granule cell and molecular layer of the dentate gyrus body in the left hippocampus; and the cornu ammonis 4 head and molecular layer head in the right hippocampus. Positive correlations were found between symptom improvement and the pretreatment volumes of the right thalamus (r = 0.501; P = 0.001) and left subiculum head of the hippocampus (r = 0.471; P = 0.002), and changes in the volumes of the left amygdala (r = -0.452; P = 0.003) and the left cornu ammonis 4 body (r = -0.537; P < 0.001). Our findings provided evidence for critical roles of the amygdala and specific hippocampal subfields in the antidepressant effect of repeated ketamine treatment. Relatively larger volumes in right thalamus and left subiculum head in the hippocampus can predict a superior clinical outcome of ketamine treatment in MDD patients.
Collapse
Affiliation(s)
- Yan-Ling Zhou
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Feng-Chun Wu
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Wei-Jian Liu
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Wei Zheng
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Cheng-Yu Wang
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yan-Ni Zhan
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Xiao-Feng Lan
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yu-Ping Ning
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China. .,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China. .,The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
30
|
McMillan R, Muthukumaraswamy SD. The neurophysiology of ketamine: an integrative review. Rev Neurosci 2020; 31:457-503. [DOI: 10.1515/revneuro-2019-0090] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 01/26/2020] [Indexed: 12/13/2022]
Abstract
AbstractThe drug ketamine has been extensively studied due to its use in anaesthesia, as a model of psychosis and, most recently, its antidepressant properties. Understanding the physiology of ketamine is complex due to its rich pharmacology with multiple potential sites at clinically relevant doses. In this review of the neurophysiology of ketamine, we focus on the acute effects of ketamine in the resting brain. We ascend through spatial scales starting with a complete review of the pharmacology of ketamine and then cover its effects on in vitro and in vivo electrophysiology. We then summarise and critically evaluate studies using EEG/MEG and neuroimaging measures (MRI and PET), integrating across scales where possible. While a complicated and, at times, confusing picture of ketamine’s effects are revealed, we stress that much of this might be caused by use of different species, doses, and analytical methodologies and suggest strategies that future work could use to answer these problems.
Collapse
Affiliation(s)
- Rebecca McMillan
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Suresh D. Muthukumaraswamy
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
31
|
Manduca JD, Thériault RK, Williams OOF, Rasmussen DJ, Perreault ML. Transient Dose-dependent Effects of Ketamine on Neural Oscillatory Activity in Wistar-Kyoto Rats. Neuroscience 2020; 441:161-175. [PMID: 32417341 DOI: 10.1016/j.neuroscience.2020.05.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/16/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022]
Abstract
Ketamine is a promising therapeutic for treatment-resistant depression (TRD) but is associated with an array of short-term psychomimetic side-effects. These disparate drug effects may be elicited through the modulation of neural circuit activity. The purpose of this study was to therefore delineate dose- and time-dependent changes in ketamine-induced neural oscillatory patterns in regions of the brain implicated in depression. Wistar-Kyoto rats were used as a model system to study these aspects of TRD neuropathology whereas Wistar rats were used as a control strain. Animals received a low (10 mg/kg) or high (30 mg/kg) dose of ketamine and temporal changes in neural oscillatory activity recorded from the prefrontal cortex (PFC), cingulate cortex (Cg), and nucleus accumbens (NAc) for ninety minutes. Effects of each dose of ketamine on immobility in the forced swim test were also evaluated. High dose ketamine induced a transient increase in theta power in the PFC and Cg, as well as a dose-dependent increase in gamma power in these regions 10-min, but not 90-min, post-administration. In contrast, only low dose ketamine normalized innate deficits in fast gamma coherence between the NAc-Cg and PFC-Cg, an effect that persisted at 90-min post-injection. These low dose ketamine-induced oscillatory alterations were accompanied by a reduction in immobility time in the forced swim test. These results show that ketamine induces time-dependent effects on neural oscillations at specific frequencies. These drug-induced changes may differentially contribute to the psychomimetic and therapeutic effects of the drug.
Collapse
Affiliation(s)
- Joshua D Manduca
- Department of Molecular and Cellular Biology, University of Guelph (ON), Canada
| | - Rachel-Karson Thériault
- Department of Molecular and Cellular Biology, University of Guelph (ON), Canada; Collaborative Neuroscience Program, University of Guelph (ON), Canada
| | - Olivia O F Williams
- Department of Molecular and Cellular Biology, University of Guelph (ON), Canada
| | - Duncan J Rasmussen
- Department of Molecular and Cellular Biology, University of Guelph (ON), Canada
| | - Melissa L Perreault
- Department of Molecular and Cellular Biology, University of Guelph (ON), Canada; Collaborative Neuroscience Program, University of Guelph (ON), Canada.
| |
Collapse
|
32
|
Averill LA, Fouda S, Murrough JW, Abdallah CG. Chronic stress pathology and ketamine-induced alterations in functional connectivity in major depressive disorder: An abridged review of the clinical evidence. ADVANCES IN PHARMACOLOGY 2020; 89:163-194. [PMID: 32616206 DOI: 10.1016/bs.apha.2020.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A paradigm shift in the conceptualization of the neurobiology of depression and the serendipitous discovery of ketamine's rapid-acting antidepressant (RAAD) effects has ushered in a new era of innovative research and novel drug development. Since the initial discovery of ketamine's RAAD effects, multiple studies have supported its short-term efficacy for fast-tracked improvements in treatment-resistant depression. Evidence from MRI studies have repeatedly demonstrated functional connectivity alterations in stress- and trauma-related disorders suggesting this may be a viable biomarker of chronic stress pathology (CSP). Human mechanistic studies further support this by coupling functional connectivity to ketamine's RAAD effects including connectivity to glutamate neurotransmission, ketamine to normalized connectivity, and these advantageous normalizations to symptom improvement/ketamine response. This review provides an abridged discussion of the suspected neurobiological underpinnings of ketamine's RAAD effects, highlighting ketamine-induced alterations in prefrontal, striatal, and anterior cingulate cortex functional connectivity in major depressive disorder. We present a model of CSP underscoring the role of synaptic loss and dysconnectivity and discuss how ketamine may be used both as (1) a treatment to restore and normalize these stress-induced neural alterations and (2) a tool to study potential biomarkers of CSP and treatment response. We conclude by noting challenges and future directions including heterogeneity, sex differences, the role of early life stress, and the need for proliferation of new methods, paradigms, and tools that will optimize signal and allow analyses at different levels of complexity, according to the needs of the question at hand, perhaps by thinking hierarchically about both clinical and biological phenotypes.
Collapse
Affiliation(s)
- Lynnette A Averill
- Clinical Neurosciences Division, US Department of Veterans Affairs, West Haven, CT, United States; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States.
| | - Samar Fouda
- Clinical Neurosciences Division, US Department of Veterans Affairs, West Haven, CT, United States; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - James W Murrough
- Department of Psychiatry, Depression and Anxiety Center for Discovery and Treatment, Icahn School of Medicine of Mount Sinai, New York, NY, United States; Department of Neuroscience, Icahn School of Medicine of Mount Sinai, New York, NY, United States
| | - Chadi G Abdallah
- Clinical Neurosciences Division, US Department of Veterans Affairs, West Haven, CT, United States; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
33
|
Peng FZ, Fan J, Ge TT, Liu QQ, Li BJ. Rapid anti-depressant-like effects of ketamine and other candidates: Molecular and cellular mechanisms. Cell Prolif 2020; 53:e12804. [PMID: 32266752 PMCID: PMC7260066 DOI: 10.1111/cpr.12804] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 12/27/2022] Open
Abstract
Major depressive disorder takes at least 3 weeks for clinical anti‐depressants, such as serotonin selective reuptake inhibitors, to take effect, and only one‐third of patients remit. Ketamine, a kind of anaesthetic, can alleviate symptoms of major depressive disorder patients in a short time and is reported to be effective to treatment‐resistant depression patients. The rapid and strong anti‐depressant‐like effects of ketamine cause wide concern. In addition to ketamine, caloric restriction and sleep deprivation also elicit similar rapid anti‐depressant‐like effects. However, mechanisms about the rapid anti‐depressant‐like effects remain unclear. Elucidating the mechanisms of rapid anti‐depressant effects is the key to finding new therapeutic targets and developing therapeutic patterns. Therefore, in this review we summarize potential molecular and cellular mechanisms of rapid anti‐depressant‐like effects based on the pre‐clinical and clinical evidence, trying to provide new insight into future therapy.
Collapse
Affiliation(s)
- Fan Zhen Peng
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| | - Jie Fan
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| | - Tong Tong Ge
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| | - Qian Qian Liu
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| | - Bing Jin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
34
|
Al Shweiki MR, Oeckl P, Steinacker P, Barschke P, Pryce C, Dorner-Ciossek C, Schönfeldt-Lecuona C, Hengerer B, Otto M. S-ketamine induces acute changes in the proteome of the mouse amygdala. J Proteomics 2020; 216:103679. [PMID: 32032757 DOI: 10.1016/j.jprot.2020.103679] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 01/21/2020] [Accepted: 02/02/2020] [Indexed: 12/28/2022]
Abstract
Current understanding of the molecular mechanisms underlying ketamine's antidepressant effect remains largely incomplete. Recent imaging studies provide evidence for ketamine effects on amygdalo-hippocampal. This study in mice aimed to investigate acute proteomic changes after ketamine administration in various brain regions including amygdala and hippocampus. One hour after administration of s-ketamine, the brain-region tissues of interest were dissected out and analyzed using label-free shotgun proteomics. The deep proteomic analysis of amygdala and hippocampus identified 89,526 peptides corresponding to 8000 proteins. The analysis revealed a pronounced proteomic signature of the acute ketamine effect in the amygdala. We anticipate that this proteomic dataset will improve understanding of the mechanism of action of ketamine and identification of new drug targets. SIGNIFICANCE: Major depressive disorder (MDD) is the leading cause of global disability and it presents a significant challenge to human health. S-ketamine has been proposed as a rapid acting antidepressant and, indeed, the FDA recently approved it for treatment of resistant MDD. However, the mechanism of action of s-ketamine as an antidepressant is still elusive. In this context, we investigated the short-term proteomic changes after ketamine administration in mouse brain regions previously related to ketamine effects such as amygdala and hippocampus. We anticipate that this proteomic dataset will provide highly useful information to improve our understanding of the mechanism of action of ketamine and identification of new drug targets.
Collapse
Affiliation(s)
| | - Patrick Oeckl
- Department of Neurology, Ulm University, Ulm, Germany
| | | | | | - Christopher Pryce
- Preclinical Laboratory for Translational Research into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| | - Cornelia Dorner-Ciossek
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | | | - Bastian Hengerer
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Markus Otto
- Department of Neurology, Ulm University, Ulm, Germany.
| |
Collapse
|
35
|
Loureiro JRA, Leaver A, Vasavada M, Sahib AK, Kubicki A, Joshi S, Woods RP, Wade B, Congdon E, Espinoza R, Narr KL. Modulation of amygdala reactivity following rapidly acting interventions for major depression. Hum Brain Mapp 2020; 41:1699-1710. [PMID: 32115848 PMCID: PMC7268016 DOI: 10.1002/hbm.24895] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/26/2019] [Accepted: 11/30/2019] [Indexed: 12/28/2022] Open
Abstract
Electroconvulsive therapy (ECT) and ketamine treatment both induce rapidly acting antidepressant effects in patients with major depressive disorder unresponsive to standard treatments, yet their specific impact on emotion processing is unknown. Here, we examined the neural underpinnings of emotion processing within and across patients (N = 44) receiving either ECT (N = 17, mean age: 36.8, 11.0 SD) or repeated subanesthetic (0.5 mg/kg) intravenous ketamine therapy (N = 27, mean age: 37.3, 10.8 SD) using a naturalistic study design. MRI and clinical data were collected before (TP1) and after treatment (TP2); healthy controls (N = 31, mean age: 34.5, 13.5 SD) completed one MRI session (TP1). An fMRI face-matching task probed negative- and positive-valence systems. Whole-brain analysis, comparing neurofunctional changes within and across treatment groups, targeted brain regions involved in emotional facial processing, and included regions-of-interest analysis of amygdala responsivity. Main findings revealed a decrease in amygdalar reactivity after both ECT and ketamine for positive and negative emotional face processing (p < .05 family wise-error (FWE) corrected). Subthreshold changes were observed between treatments within the dorsolateral prefrontal cortex and insula (p < .005, uncorrected). BOLD change for positive faces in the inferior parietal cortex significantly correlated with overall symptom improvement, and BOLD change in frontal regions correlated with anxiety for negative faces, and anhedonia for positive faces (p < .05 FWE corrected). Both serial ketamine and ECT treatment modulate amygdala response, while more subtle treatment-specific changes occur in the larger functional network. Findings point to both common and differential mechanistic upstream systems-level effects relating to fast-acting antidepressant response, and symptoms of anxiety and anhedonia, for the processing of emotionally valenced stimuli.
Collapse
Affiliation(s)
- Joana R. A. Loureiro
- Department of NeurologyAhamason‐Lovelace Brain Mapping CenterLos AngelesCalifornia
| | - Amber Leaver
- Northwestern University Clinical and Translational Sciences Institute (NUCATS)ChicagoIllinois
| | - Megha Vasavada
- Department of NeurologyAhamason‐Lovelace Brain Mapping CenterLos AngelesCalifornia
| | - Ashish K. Sahib
- Department of NeurologyAhamason‐Lovelace Brain Mapping CenterLos AngelesCalifornia
| | - Antoni Kubicki
- Department of NeurologyAhamason‐Lovelace Brain Mapping CenterLos AngelesCalifornia
| | - Shantanu Joshi
- Department of NeurologyAhamason‐Lovelace Brain Mapping CenterLos AngelesCalifornia
| | - Roger P. Woods
- Department of NeurologyAhamason‐Lovelace Brain Mapping CenterLos AngelesCalifornia
| | - Benjamin Wade
- Department of NeurologyAhamason‐Lovelace Brain Mapping CenterLos AngelesCalifornia
| | - Eliza Congdon
- Department of Psychiatry and Biobehavioral SciencesUniversity of California Los AngelesLos AngelesCalifornia
| | - Randall Espinoza
- Department of Psychiatry and Biobehavioral SciencesUniversity of California Los AngelesLos AngelesCalifornia
| | - Katherine L. Narr
- Department of NeurologyAhamason‐Lovelace Brain Mapping CenterLos AngelesCalifornia
- Department of Psychiatry and Biobehavioral SciencesUniversity of California Los AngelesLos AngelesCalifornia
| |
Collapse
|
36
|
Resting-state brain functional connectivity in patients with chronic pain who responded to subanesthetic-dose ketamine. Sci Rep 2019; 9:12912. [PMID: 31501482 PMCID: PMC6733873 DOI: 10.1038/s41598-019-49360-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 08/23/2019] [Indexed: 11/30/2022] Open
Abstract
Ketamine has been used to treat chronic pain; however, it is still unknown as to what types of chronic pain is ketamine effective against. To identify the effect of administration of subanesthetic-dose ketamine in patients with chronic pain and to clarify the mechanism of the effect, we retrospectively investigated brain functional connectivity using resting-state functional magnetic resonance imaging (rs-fMRI). Patients were divided into responders (Group R: ≥50% improvement on Numerical Rating Scale) and non-responders (Group NR). We compared the differences in terms of brain functional connectivity by seed-to-voxel correlation analysis. Two-sample t-test revealed significant lower connectivity between the medial prefrontal cortex (mPFC) and precuneus in Group R. We also found a significant negative correlation between the improvement rate and functional connectivity strength between the mPFC and precuneus. These findings suggest that subanesthetic-dose ketamine is effective in patients with chronic pain whose brain functional connectivity between the mPFC and precuneus is low. We believe that the current study explored for the first time the correlation between brain functional connectivity and the effect of subanesthetic-dose ketamine for chronic pain and indicated the possibility of use of the predictive marker in pharmacological treatment of chronic pain.
Collapse
|
37
|
Ionescu DF, Felicione JM, Gosai A, Cusin C, Shin P, Shapero BG, Deckersbach T. Ketamine-Associated Brain Changes: A Review of the Neuroimaging Literature. Harv Rev Psychiatry 2019; 26:320-339. [PMID: 29465479 PMCID: PMC6102096 DOI: 10.1097/hrp.0000000000000179] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Major depressive disorder (MDD) is one of the most prevalent conditions in psychiatry. Patients who do not respond to traditional monoaminergic antidepressant treatments have an especially difficult-to-treat type of MDD termed treatment-resistant depression. Subanesthetic doses of ketamine-a glutamatergic modulator-have shown great promise for rapidly treating patients with the most severe forms of depression. As such, ketamine represents a promising probe for understanding the pathophysiology of depression and treatment response. Through neuroimaging, ketamine's mechanism may be elucidated in humans. Here, we review 47 articles of ketamine's effects as revealed by neuroimaging studies. Some important brain areas emerge, especially the subgenual anterior cingulate cortex. Furthermore, ketamine may decrease the ability to self-monitor, may increase emotional blunting, and may increase activity in reward processing. Further studies are needed, however, to elucidate ketamine's mechanism of antidepressant action.
Collapse
Affiliation(s)
- Dawn F. Ionescu
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | | | - Aishwarya Gosai
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
| | - Cristina Cusin
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Philip Shin
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
| | - Benjamin G. Shapero
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Thilo Deckersbach
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Athinoula A. Martinos Center for Biomedical Imaging, Boston, MA
| |
Collapse
|
38
|
Duman RS, Sanacora G, Krystal JH. Altered Connectivity in Depression: GABA and Glutamate Neurotransmitter Deficits and Reversal by Novel Treatments. Neuron 2019; 102:75-90. [PMID: 30946828 PMCID: PMC6450409 DOI: 10.1016/j.neuron.2019.03.013] [Citation(s) in RCA: 538] [Impact Index Per Article: 107.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/04/2019] [Accepted: 03/07/2019] [Indexed: 12/12/2022]
Abstract
The mechanisms underlying the pathophysiology and treatment of depression and stress-related disorders remain unclear, but studies in depressed patients and rodent models are beginning to yield promising insights. These studies demonstrate that depression and chronic stress exposure cause atrophy of neurons in cortical and limbic brain regions implicated in depression, and brain imaging studies demonstrate altered connectivity and network function in the brains of depressed patients. Studies of the neurobiological basis of the these alterations have focused on both the principle, excitatory glutamate neurons, as well as inhibitory GABA interneurons. They demonstrate structural, functional, and neurochemical deficits in both major neuronal types that could lead to degradation of signal integrity in cortical and hippocampal regions. The molecular mechanisms underlying these changes have not been identified but are thought to be related to stress induced excitotoxic effects in combination with elevated adrenal glucocorticoids and inflammatory cytokines as well as other environmental factors. Transcriptomic studies are beginning to demonstrate important sex differences and, together with genomic studies, are starting to reveal mechanistic domains of overlap and uniqueness with regards to risk and pathophysiological mechanisms with schizophrenia and bipolar disorder. These studies also implicate GABA and glutamate dysfunction as well as immunologic mechanisms. While current antidepressants have significant time lag and efficacy limitations, new rapid-acting agents that target the glutamate and GABA systems address these issues and offer superior therapeutic interventions for this widespread and debilitating disorder.
Collapse
Affiliation(s)
- Ronald S Duman
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06508, USA.
| | - Gerard Sanacora
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06508, USA
| | - John H Krystal
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06508, USA
| |
Collapse
|
39
|
Gärtner M, Aust S, Bajbouj M, Fan Y, Wingenfeld K, Otte C, Heuser-Collier I, Böker H, Hättenschwiler J, Seifritz E, Grimm S, Scheidegger M. Functional connectivity between prefrontal cortex and subgenual cingulate predicts antidepressant effects of ketamine. Eur Neuropsychopharmacol 2019; 29:501-508. [PMID: 30819549 DOI: 10.1016/j.euroneuro.2019.02.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 02/12/2019] [Accepted: 02/14/2019] [Indexed: 12/28/2022]
Abstract
Converging evidence suggests that a single sub-anesthetic dose of ketamine can produce strong and rapid antidepressant effects in patients that do not respond to standard treatment. Despite a considerable amount of research investigating ketamine's mechanisms of action, the exact neuronal targets conveying the antidepressant effects have not been identified yet. Preclinical studies suggest that molecular changes induced by ketamine bring forward large-scale network reconfigurations that might relate to ketamine's antidepressant properties. In this prospective two-site study we measured resting state fMRI in 24 depressed patients prior to, and 24 h after a single sub-anesthetic dose of ketamine. We analyzed functional connectivity (FC) at baseline and after ketamine and focused our analysis on baseline FC and FC changes directly linked to symptom reduction in order to identify neuronal targets that predict individual clinical responses to ketamine. Our results show that FC increases after ketamine between right lateral prefrontal cortex (PFC) and subgenual anterior cingulate cortex (sgACC) are positively linked to treatment response. Furthermore, low baseline FC between these regions predicts treatment outcome. We conclude that PFC-sgACC connectivity may represent a promising biomarker with both predictive and explanatory power.
Collapse
Affiliation(s)
- Matti Gärtner
- Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; MSB - Medical School Berlin, Calandrellistraße 1-9, 12247 Berlin, Germany.
| | - Sabine Aust
- Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Malek Bajbouj
- Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Yan Fan
- Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Katja Wingenfeld
- Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Christian Otte
- Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Isabella Heuser-Collier
- Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Heinz Böker
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Lenggstrasse 31, 8032 Zurich, Switzerland
| | - Josef Hättenschwiler
- Center for Anxiety and Depression (ZADZ), Riesbachstrasse 61, 8008 Zurich, Switzerland
| | - Erich Seifritz
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Lenggstrasse 31, 8032 Zurich, Switzerland
| | - Simone Grimm
- Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; MSB - Medical School Berlin, Calandrellistraße 1-9, 12247 Berlin, Germany; Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Lenggstrasse 31, 8032 Zurich, Switzerland
| | - Milan Scheidegger
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Lenggstrasse 31, 8032 Zurich, Switzerland
| |
Collapse
|
40
|
Reed JL, Nugent AC, Furey ML, Szczepanik JE, Evans JW, Zarate CA. Effects of Ketamine on Brain Activity During Emotional Processing: Differential Findings in Depressed Versus Healthy Control Participants. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2019; 4:610-618. [PMID: 30826253 DOI: 10.1016/j.bpsc.2019.01.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/04/2019] [Accepted: 01/07/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND In the search for novel treatments for depression, ketamine has emerged as a unique agent with rapid antidepressant effects. Experimental tasks involving emotional processing can be used during functional magnetic resonance imaging scanning to investigate ketamine's effects on brain function in major depressive disorder (MDD). This study examined ketamine's effects on functional magnetic resonance imaging activity during an emotional processing task. METHODS A total of 33 individuals with treatment-resistant MDD and 24 healthy control participants (HCs) took part in this double-blind, placebo-controlled crossover study. Participants received ketamine and placebo infusions 2 weeks apart, and functional magnetic resonance imaging scans were conducted at baseline and 2 days after each infusion. Blood oxygen level-dependent signal was measured during an emotional processing task, and a linear mixed-effects model was used to analyze differences in activation among group, drug, and task-specific factors. RESULTS A group-by-drug interaction was observed in several brain regions, including a right frontal cluster extending into the anterior cingulate cortex and insula. Participants with MDD had greater activity than HCs after placebo infusion but showed lower activity after ketamine infusion, which was similar to the activity in HCs after placebo. A group-by-drug-by-task condition interaction was also found, which showed further differences that varied between implicit and explicit emotional conditions. CONCLUSIONS The main results indicate that ketamine had differential effects on brain activity in participants with MDD versus HCs. The pattern of activation in participants with MDD after ketamine infusion resembled the activation in HCs after placebo infusion, suggesting a normalization of function during emotional processing. The findings contribute to a better understanding of ketamine's actions in the brain.
Collapse
Affiliation(s)
- Jessica L Reed
- Section on Neurobiology and Treatment of Mood Disorders, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland.
| | - Allison C Nugent
- Section on Neurobiology and Treatment of Mood Disorders, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Maura L Furey
- Section on Neurobiology and Treatment of Mood Disorders, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; Janssen Pharmaceuticals of Johnson & Johnson, San Diego, California
| | - Joanna E Szczepanik
- Section on Neurobiology and Treatment of Mood Disorders, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Jennifer W Evans
- Section on Neurobiology and Treatment of Mood Disorders, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Carlos A Zarate
- Section on Neurobiology and Treatment of Mood Disorders, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
41
|
Godlewska BR. Cognitive neuropsychological theory: Reconciliation of psychological and biological approaches for depression. Pharmacol Ther 2018; 197:38-51. [PMID: 30578809 DOI: 10.1016/j.pharmthera.2018.12.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
New antidepressants and individualized approaches to treatment, matching specific therapies to individual patients, are urgently needed. For this, a better understanding of processes underpinning the development of depressive symptoms and response to medications are required. The cognitive neuropsychological model offers a novel approach uniquely combining biological and psychological approaches to explain how antidepressants exert their effect, why there is a delay in the onset of their clinical effect, and how changes in emotional processing are an essential step for a clinical antidepressant effect to take place. The paper presents the model and its underpinnings in the form of research in both healthy and depressed individuals, as well as the potential for its practical use.
Collapse
Affiliation(s)
- Beata R Godlewska
- Psychopharmacology Research Unit, University Department of Psychiatry (PPRU), University of Oxford, Oxford, UK.
| |
Collapse
|
42
|
Haaf M, Leicht G, Curic S, Mulert C. Glutamatergic Deficits in Schizophrenia - Biomarkers and Pharmacological Interventions within the Ketamine Model. Curr Pharm Biotechnol 2018; 19:293-307. [PMID: 29929462 PMCID: PMC6142413 DOI: 10.2174/1389201019666180620112528] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/12/2018] [Accepted: 05/26/2018] [Indexed: 11/30/2022]
Abstract
Background: The observation that N-methyl-D-aspartate glutamate receptor (NMDAR) antagonists such as ketamine transiently induce schizophrenia-like positive, negative and cognitive symptoms has led to a paradigm shift from dopaminergic to glutamatergic dysfunction in pharmacological models of schizophrenia. NMDAR hypofunction can explain many schizophrenia symptoms directly due to excitatory-to-inhibitory (E/I) imbalance, but also dopaminergic dysfunction itself. However, so far no new drug targeting the NMDAR has been successfully approved. In the search for possible biomarkers it is interesting that ketamine-induced psychopathological changes in healthy participants were accompanied by altered electro-(EEG), magnetoencephalographic (MEG) and functional magnetic resonance imaging (fMRI) signals. Methods: We systematically searched PubMed/Medline and Web of Knowledge databases (January 2006 to July 2017) to identify EEG/MEG and fMRI studies of the ketamine model of schizophrenia with human subjects. The search strategy identified 209 citations of which 46 articles met specified eligibility criteria. Results: In EEG/MEG studies, ketamine induced changes of event-related potentials, such as the P300 potential and the mismatch negativity, similar to alterations observed in schizophrenia patients. In fMRI studies, alterations of activation were observed in different brain regions, most prominently within the anterior cingulate cortex and limbic structures as well as task-relevant brain regions. These alterations were accompanied by changes in functional connectivity, indicating a balance shift of the underlying brain networks. Pharmacological treatments did alter ketamine-induced changes in EEG/MEG and fMRI studies to different extents. Conclusion: This review highlights the potential applicability of the ketamine model for schizophrenia drug development by offering the possibility to assess the effect of pharmacological agents on schizophrenia-like symptoms and to find relevant neurophysiological and neuroimaging biomarkers.
Collapse
Affiliation(s)
- Moritz Haaf
- Psychiatry Neuroimaging Branch (PNB), Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gregor Leicht
- Psychiatry Neuroimaging Branch (PNB), Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stjepan Curic
- Psychiatry Neuroimaging Branch (PNB), Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Mulert
- Psychiatry Neuroimaging Branch (PNB), Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Psychiatry and Psychotherapy, UKGM, Justus-Liebig University Giessen, Giessen, Germany
| |
Collapse
|
43
|
Evans JW, Szczepanik J, Brutsché N, Park LT, Nugent AC, Zarate CA. Default Mode Connectivity in Major Depressive Disorder Measured Up to 10 Days After Ketamine Administration. Biol Psychiatry 2018; 84:582-590. [PMID: 29580569 PMCID: PMC6093808 DOI: 10.1016/j.biopsych.2018.01.027] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/05/2018] [Accepted: 01/23/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND The symptoms of major depressive disorder (MDD) are rapidly alleviated by administration of a single dose of the glutamatergic modulator ketamine. However, few studies have investigated the potential sustained neural effects of this agent beyond immediate infusion. This study used functional magnetic resonance imaging to examine the effect of a single ketamine infusion on the resting state default mode network (DMN) at 2 and 10 days after a single ketamine infusion in unmedicated subjects with MDD as well as healthy control subjects (HCs). METHODS Data were drawn from a double-blind, placebo-controlled crossover study of 58 participants (33 with MDD and 25 HCs) who received an intravenous infusion of either ketamine hydrochloride (0.5 mg/kg) or placebo on 2 separate test days spaced 2 weeks apart. Eight minutes of functional magnetic resonance imaging resting state data was acquired at baseline and at about 2 and 10 days after both infusions. The DMN was defined using seed-based correlation and was compared across groups and scans. RESULTS In subjects with MDD, connectivity between the insula and the DMN was normalized compared with HCs 2 days postketamine infusion. This change was reversed after 10 days and did not appear in either of the placebo scans. Group-specific connectivity differences in drug response were observed, most notably in the insula in subjects with MDD and in the thalamus in HCs. CONCLUSIONS Connectivity changes in the insula in subjects with MDD suggest that ketamine may normalize the interaction between the DMN and salience networks, supporting the triple network dysfunction model of MDD.
Collapse
Affiliation(s)
- Jennifer W Evans
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland.
| | - Joanna Szczepanik
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Nancy Brutsché
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Lawrence T Park
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Allison C Nugent
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
44
|
Individual differences in tendencies to attention-deficit/hyperactivity disorder and emotionality: empirical evidence in young healthy adults from Germany and China. ACTA ACUST UNITED AC 2018; 11:167-182. [DOI: 10.1007/s12402-018-0266-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 08/27/2018] [Indexed: 11/26/2022]
|
45
|
Miskowiak KW, Macoveanu J, Jørgensen MB, Ott CV, Støttrup MM, Jensen HM, Jørgensen A, Harmer CJ, Paulson OB, Siebner HR, Kessing LV. Effect of electroconvulsive therapy on neural response to affective pictures: A randomized, sham-controlled fMRI study. Eur Neuropsychopharmacol 2018; 28:915-924. [PMID: 29891215 DOI: 10.1016/j.euroneuro.2018.05.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 05/17/2018] [Accepted: 05/21/2018] [Indexed: 12/23/2022]
Abstract
Electroconvulsive therapy (ECT) is the most effective treatment for severe depression but its neurocognitive mechanisms are unclear. This randomized, sham-controlled functional magnetic resonance imaging (fMRI) study explored the effects of a single ECT on neural response to affective pictures. Twenty-seven patients with major depressive disorder were randomized to a single active ECT (N = 15) or sham (N = 12) session in a double-blind, parallel-group design. On the following day, patients underwent fMRI during which they viewed pleasant, unpleasant and neutral pictures and performed a free recall test after the scan. Mood symptoms were assessed before ECT/sham and at the time of fMRI. Subsequently, all patients continued active ECT as usual. Mood symptoms were reassessed after six active ECT sessions. A single ECT vs. sham session reduced neural response to unpleasant vs. pleasant pictures in the medial prefrontal cortex, a region showing greater response in the more depressed patients. This effect occurred in the absence of between-group differences in picture recall, mood symptoms or concomitant medication. In conclusion, modulation of medial prefrontal hyper-activity during encoding of negative affective information may be a common mechanism of distinct biological depression treatments.
Collapse
Affiliation(s)
- K W Miskowiak
- Psychiatric Centre Copenhagen, Copenhagen University Hospital, Rigshospitalet Dep. 6233, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Department of Psychology, University of Copenhagen, Øster Farimagsgade 2A, Copenhagen, Denmark.
| | - J Macoveanu
- Psychiatric Centre Copenhagen, Copenhagen University Hospital, Rigshospitalet Dep. 6233, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Kettegård Alle 30, Hvidovre, Denmark; Center for Integrated Molecular Brain Imaging, Rigshospitalet, Blegdamsvej 9, Copenhagen, Denmark
| | - M B Jørgensen
- Psychiatric Centre Copenhagen, Copenhagen University Hospital, Rigshospitalet Dep. 6233, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - C V Ott
- Psychiatric Centre Copenhagen, Copenhagen University Hospital, Rigshospitalet Dep. 6233, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - M M Støttrup
- Psychiatric Centre Copenhagen, Copenhagen University Hospital, Rigshospitalet Dep. 6233, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - H M Jensen
- Psychiatric Centre Copenhagen, Digevej 110, Amager, Denmark
| | - A Jørgensen
- Psychiatric Centre Copenhagen, Copenhagen University Hospital, Rigshospitalet Dep. 6233, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - C J Harmer
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
| | - O B Paulson
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Kettegård Alle 30, Hvidovre, Denmark; Center for Integrated Molecular Brain Imaging, Rigshospitalet, Blegdamsvej 9, Copenhagen, Denmark; Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, Copenhagen, Denmark
| | - H R Siebner
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Kettegård Alle 30, Hvidovre, Denmark; Center for Integrated Molecular Brain Imaging, Rigshospitalet, Blegdamsvej 9, Copenhagen, Denmark; Department of Neurology, Copenhagen University Hospital Bispebjerg, Bispebjerg Bakke 23, Denmark
| | - L V Kessing
- Psychiatric Centre Copenhagen, Copenhagen University Hospital, Rigshospitalet Dep. 6233, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| |
Collapse
|
46
|
Reed JL, Nugent AC, Furey ML, Szczepanik JE, Evans JW, Zarate CA. Ketamine normalizes brain activity during emotionally valenced attentional processing in depression. NEUROIMAGE-CLINICAL 2018; 20:92-101. [PMID: 30094160 PMCID: PMC6070691 DOI: 10.1016/j.nicl.2018.07.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 05/15/2018] [Accepted: 07/04/2018] [Indexed: 01/01/2023]
Abstract
Background An urgent need exists for faster-acting pharmacological treatments in major depressive disorder (MDD). The glutamatergic modulator ketamine has been shown to have rapid antidepressant effects, but much remains unknown about its mechanism of action. Functional MRI (fMRI) can be used to investigate how ketamine impacts brain activity during cognitive and emotional processing. Methods This double-blind, placebo-controlled, crossover study of 33 unmedicated participants with MDD and 26 healthy controls (HCs) examined how ketamine affected fMRI activation during an attentional bias dot probe task with emotional face stimuli across multiple time points. A whole brain analysis was conducted to find regions with differential activation associated with group, drug session, or dot probe task-specific factors (emotional valence and congruency of stimuli). Results A drug session by group interaction was observed in several brain regions, such that ketamine had opposite effects on brain activation in MDD versus HC participants. Additionally, there was a similar finding related to emotional valence (a drug session by group by emotion interaction) in a large cluster in the anterior cingulate and medial frontal cortex. Conclusions The findings show a pattern of brain activity in MDD participants following ketamine infusion that is similar to activity observed in HCs after placebo. This suggests that ketamine may act as an antidepressant by normalizing brain function during emotionally valenced attentional processing. Clinical trial NCT#00088699: https://www.clinicaltrials.gov/ct2/show/NCT00088699 The effects of ketamine versus placebo on brain activation were studied using fMRI. MDD and healthy participants were tested on an fMRI emotion-based attentional task. Ketamine had opposite effects on brain activity in MDD versus healthy participants. In MDD, brain activity post-ketamine was similar to healthy controls post-placebo. These findings suggest that ketamine may act by normalizing brain function.
Collapse
Affiliation(s)
- Jessica L Reed
- Experimental Therapeutics & Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States.
| | - Allison C Nugent
- Experimental Therapeutics & Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States.
| | - Maura L Furey
- Experimental Therapeutics & Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States; Janssen Pharmaceuticals of Johnson and Johnson Inc., San Diego, CA, United States.
| | - Joanna E Szczepanik
- Experimental Therapeutics & Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States.
| | - Jennifer W Evans
- Experimental Therapeutics & Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States.
| | - Carlos A Zarate
- Experimental Therapeutics & Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
47
|
Schenberg EE. Psychedelic-Assisted Psychotherapy: A Paradigm Shift in Psychiatric Research and Development. Front Pharmacol 2018; 9:733. [PMID: 30026698 PMCID: PMC6041963 DOI: 10.3389/fphar.2018.00733] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 06/18/2018] [Indexed: 12/20/2022] Open
Abstract
Mental disorders are rising while development of novel psychiatric medications is declining. This stall in innovation has also been linked with intense debates on the current diagnostics and explanations for mental disorders, together constituting a paradigmatic crisis. A radical innovation is psychedelic-assisted psychotherapy (PAP): professionally supervised use of ketamine, MDMA, psilocybin, LSD and ibogaine as part of elaborated psychotherapy programs. Clinical results so far have shown safety and efficacy, even for “treatment resistant” conditions, and thus deserve increasing attention from medical, psychological and psychiatric professionals. But more than novel treatments, the PAP model also has important consequences for the diagnostics and explanation axis of the psychiatric crisis, challenging the discrete nosological entities and advancing novel explanations for mental disorders and their treatment, in a model considerate of social and cultural factors, including adversities, trauma, and the therapeutic potential of some non-ordinary states of consciousness.
Collapse
|
48
|
Cohen SP, Bhatia A, Buvanendran A, Schwenk ES, Wasan AD, Hurley RW, Viscusi ER, Narouze S, Davis FN, Ritchie EC, Lubenow TR, Hooten WM. Consensus Guidelines on the Use of Intravenous Ketamine Infusions for Chronic Pain From the American Society of Regional Anesthesia and Pain Medicine, the American Academy of Pain Medicine, and the American Society of Anesthesiologists. Reg Anesth Pain Med 2018; 43:521-546. [PMID: 29870458 PMCID: PMC6023575 DOI: 10.1097/aap.0000000000000808] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND Over the past 2 decades, the use of intravenous ketamine infusions as a treatment for chronic pain has increased dramatically, with wide variation in patient selection, dosing, and monitoring. This has led to a chorus of calls from various sources for the development of consensus guidelines. METHODS In November 2016, the charge for developing consensus guidelines was approved by the boards of directors of the American Society of Regional Anesthesia and Pain Medicine and, shortly thereafter, the American Academy of Pain Medicine. In late 2017, the completed document was sent to the American Society of Anesthesiologists' Committees on Pain Medicine and Standards and Practice Parameters, after which additional modifications were made. Panel members were selected by the committee chair and both boards of directors based on their expertise in evaluating clinical trials, past research experience, and clinical experience in developing protocols and treating patients with ketamine. Questions were developed and refined by the committee, and the groups responsible for addressing each question consisted of modules composed of 3 to 5 panel members in addition to the committee chair. Once a preliminary consensus was achieved, sections were sent to the entire panel, and further revisions were made. In addition to consensus guidelines, a comprehensive narrative review was performed, which formed part of the basis for guidelines. RESULTS Guidelines were prepared for the following areas: indications; contraindications; whether there was evidence for a dose-response relationship, or a minimum or therapeutic dose range; whether oral ketamine or another N-methyl-D-aspartate receptor antagonist was a reasonable treatment option as a follow-up to infusions; preinfusion testing requirements; settings and personnel necessary to administer and monitor treatment; the use of preemptive and rescue medications to address adverse effects; and what constitutes a positive treatment response. The group was able to reach consensus on all questions. CONCLUSIONS Evidence supports the use of ketamine for chronic pain, but the level of evidence varies by condition and dose range. Most studies evaluating the efficacy of ketamine were small and uncontrolled and were either unblinded or ineffectively blinded. Adverse effects were few and the rate of serious adverse effects was similar to placebo in most studies, with higher dosages and more frequent infusions associated with greater risks. Larger studies, evaluating a wider variety of conditions, are needed to better quantify efficacy, improve patient selection, refine the therapeutic dose range, determine the effectiveness of nonintravenous ketamine alternatives, and develop a greater understanding of the long-term risks of repeated treatments.
Collapse
Affiliation(s)
- Steven P. Cohen
- From the Departments of Anesthesiology & Critical Care Medicine, Neurology, and Physical Medicine & Rehabilitation, Johns Hopkins School of Medicine; and
- Uniformed Services University of the Health Sciences, Bethesda, MD
| | - Anuj Bhatia
- Department of Anesthesiology, University of Toronto, Toronto Western Hospital, Toronto, Ontario, Canada
| | | | - Eric S. Schwenk
- Department of Anesthesiology, Jefferson Medical College, Philadelphia; and
| | - Ajay D. Wasan
- Departments of Anesthesiology and Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - Robert W. Hurley
- Departments of Anesthesiology and Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| | - Eugene R. Viscusi
- Department of Anesthesiology, Jefferson Medical College, Philadelphia; and
| | - Samer Narouze
- Departments of Anesthesiology and Neurosurgery, Western Reserve Hospital, Akron, OH
| | - Fred N. Davis
- Procare Pain Solutions and
- Department of Anesthesiology, Michigan State University College of Human Medicine, Grand Rapids, MI
| | - Elspeth C. Ritchie
- Department of Psychiatry, Uniformed Services University of the Health Sciences, Georgetown University School of Medicine, Bethesda, MD; and
- Howard University College of Medicine, Washington, DC; and
| | | | - William M. Hooten
- Departments of Anesthesiology and Psychiatry, Mayo College of Medicine, Rochester, MN
| |
Collapse
|
49
|
Mueller F, Musso F, London M, de Boer P, Zacharias N, Winterer G. Pharmacological fMRI: Effects of subanesthetic ketamine on resting-state functional connectivity in the default mode network, salience network, dorsal attention network and executive control network. NEUROIMAGE-CLINICAL 2018; 19:745-757. [PMID: 30003027 PMCID: PMC6040604 DOI: 10.1016/j.nicl.2018.05.037] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/13/2018] [Accepted: 05/30/2018] [Indexed: 11/26/2022]
Abstract
Background Subanesthetic dosages of the NMDAR antagonist, S-Ketamine, can cause changes in behavior in healthy subjects, which are similar to the state acute psychosis and are relevant in translational schizophrenia research. Functional magnetic resonance imaging (fMRI) can be used for non-hypothesis-driven analysis of brain connectivity. The correlation between clinical behavioral scores and neuroimaging can help to characterize ketamine effects on healthy brains in resting state. Method seventeen healthy, male subjects (mean: 27.42 years, SD: 4.42) were administered an infusion with S-Ketamine (initial bolus 1 mg/kg and continuous infusion of 0.015625 mg/kg/min with dosage reduction −10%/10 min) or saline in a randomized, double-blind, cross-over study. During infusion, resting state connectivity was measured and analyzed with a seed-to-voxel fMRI analysis approach. The seed regions were located in the posterior cingulate cortex, intraparietal sulcus, dorsolateral prefrontal cortex and fronto-insular cortex. Receiver operating characteristics (ROC) were calculated to assess the accuracy of the ketamine-induced functional connectivity changes. Bivariate Pearson correlation was used for correlation testing of functional connectivity changes with changes of clinical scores (PANSS, 5D-ASC). Results In the executive network (ECN), ketamine significantly increases the functional connectivity with parts of the anterior cingulum and superior frontal gyrus, but no significant correlations with clinical symptoms were found. Decreased connectivity between the salience network (SN) and the calcarine fissure was found, which is significantly correlated with negative symptoms (PANSS) (R2 > 0.4). Conclusion Decreased ketamine-induced functional connectivity in the salience network may qualify as accurate and highly predictive biomarkers for ketamine induced negative symptoms. All seed regions showed ketamine induced changes of functional connectivity Signficant changes of functional connectivity were found in the salience and executive control network PANSS and 5D-ASC scores are highly sensitive and specific to differentiate between placebo and ketamine condition A significant correlation between salience network- visual cortex connectivity and negative symptom scores was found
Collapse
Affiliation(s)
- Felix Mueller
- Experimental and Clinical Research Center (ECRC), Dept. of Anesthesiology and Intensive Care Medicine, Charité - University Medicine Berlin, Germany.
| | - Francesco Musso
- Department of Psychiatry, Heinrich-Heine University, Düsseldorf, Germany
| | - Markus London
- Janssen-Cilag GmbH, Early Development and Clinical Pharmacology, Neuss, Germany
| | - Peter de Boer
- Janssen Pharmaceutica, Johnson & Johnson Pharmaceutical Research and Development, Beerse, Belgium
| | - Norman Zacharias
- Experimental and Clinical Research Center (ECRC), Dept. of Anesthesiology and Intensive Care Medicine, Charité - University Medicine Berlin, Germany
| | - Georg Winterer
- Experimental and Clinical Research Center (ECRC), Dept. of Anesthesiology and Intensive Care Medicine, Charité - University Medicine Berlin, Germany; Pharmaimage Biomarker Solutions GmbH, Berlin, Germany; Pharmaimage Biomarker Solutions Inc., Boston, USA
| |
Collapse
|
50
|
Li CSR, Zhang S, Hung CC, Chen CM, Duann JR, Lin CP, Lee TSH. Depression in chronic ketamine users: Sex differences and neural bases. Psychiatry Res 2017; 269:1-8. [PMID: 28892733 PMCID: PMC5634929 DOI: 10.1016/j.pscychresns.2017.09.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/16/2017] [Accepted: 09/01/2017] [Indexed: 01/10/2023]
Abstract
Chronic ketamine use leads to cognitive and affective deficits including depression. Here, we examined sex differences and neural bases of depression in chronic ketamine users. Compared to non-drug using healthy controls (HC), ketamine-using females but not males showed increased depression score as assessed by the Center of Epidemiological Studies Depression Scale (CES-D). We evaluated resting state functional connectivity (rsFC) of the subgenual anterior cingulate cortex (sgACC), a prefrontal structure consistently implicated in the pathogenesis of depression. Compared to HC, ketamine users (KU) did not demonstrate significant changes in sgACC connectivities at a corrected threshold. However, in KU, a linear regression against CES-D score showed less sgACC connectivity to the orbitofrontal cortex (OFC) with increasing depression severity. Examined separately, male and female KU showed higher sgACC connectivity to bilateral superior temporal gyrus and dorsomedial prefrontal cortex (dmPFC), respectively, in correlation with depression. The linear correlation of sgACC-OFC and sgACC-dmPFC connectivity with depression was significantly different in slope between KU and HC. These findings highlighted changes in rsFC of the sgACC as associated with depression and sex differences in these changes in chronic ketamine users.
Collapse
Affiliation(s)
- Chiang-Shan R Li
- Department of Psychiatry, Yale University, New Haven, CT, USA; Department of Neuroscience, Yale University, New Haven, CT, USA; Beijing Huilongguan Hospital, Beijing, China.
| | - Sheng Zhang
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Chia-Chun Hung
- Bali Psychiatric Center, Ministry of Health and Welfare, Taiwan
| | - Chun-Ming Chen
- Department of Radiology, China Medical University Hospital, Taichung, Taiwan
| | - Jeng-Ren Duann
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan; Institute for Neural Computation, University of California San Diego, La Jolla, CA, USA
| | - Ching-Po Lin
- Institute of Neuroscience, National Yang Ming University, Taipei, Taiwan
| | - Tony Szu-Hsien Lee
- Department of Health Promotion and Health Education, National Taiwan Normal University, Taipei, Taiwan.
| |
Collapse
|