1
|
Mielko J, Pakulska J, Oszczyk A, Lustyk K, Pytka K, Sałaciak K. Beyond surgery: Repurposing anesthetics for treatment of central nervous system disorders. Prog Neuropsychopharmacol Biol Psychiatry 2025; 139:111386. [PMID: 40311741 DOI: 10.1016/j.pnpbp.2025.111386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/11/2025] [Accepted: 04/26/2025] [Indexed: 05/03/2025]
Abstract
The development of new drugs is a complex, expensive, and time-consuming process, often fraught with a high likelihood of failure. Amid these obstacles, drug repurposing, which identifies new therapeutic applications for already existing medications, offers a more economical and time-saving approach, particularly in the challenging field of neurological and psychiatric disorders. This narrative review explores both preclinical and clinical studies to examine the potential of anesthetics such as ketamine, nitrous oxide, isoflurane, sevoflurane, propofol, dexmedetomidine, and sodium oxybate in treating central nervous system disorders. Various research highlights the potential of anesthetics to provide rapid antidepressant effects, enhance learning and memory, improve synaptic plasticity, and offer neuroprotective benefits, demonstrating promise for treating depression, post-traumatic stress disorder, cognitive decline, traumatic brain injury, and neurodegenerative disorders. Anesthetics appear to alleviate symptoms in neurological conditions, likely by modulating GABAergic and glutamatergic pathways. However, challenges such as dose-dependent neurotoxicity, variability in preclinical and clinical outcomes, as well as environmental concerns remain significant issues. Future research is essential to optimize dosing strategies, ensure long-term safety, and gain a deeper understanding of the precise mechanisms of action. The concept of anesthetics' repurposing presents a unique solution to tackle the challenges in neurological and psychiatric therapy by providing a platform for the development of new and improved therapies.
Collapse
Affiliation(s)
- Joana Mielko
- Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Julia Pakulska
- Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Amelia Oszczyk
- Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Klaudia Lustyk
- Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Karolina Pytka
- Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Kinga Sałaciak
- Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
2
|
Williams E, Taujanskaite U, Kamboj SK, Murphy SE, Harmer CJ. Examining memory reconsolidation as a mechanism of nitrous oxide's antidepressant action. Neuropsychopharmacology 2025; 50:609-617. [PMID: 39825109 PMCID: PMC11845508 DOI: 10.1038/s41386-024-02049-0] [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: 09/11/2024] [Revised: 12/13/2024] [Accepted: 12/26/2024] [Indexed: 01/20/2025]
Abstract
There is an ongoing need to identify novel pharmacological agents for the effective treatment of depression. One emerging candidate, which has demonstrated rapid-acting antidepressant effects in treatment-resistant groups, is nitrous oxide (N2O)-a gas commonly used for sedation and pain management in clinical settings and with a range of pharmacological effects, including antagonism of NMDA glutamate receptors. A growing body of evidence suggests that subanaesthetic doses of N2O (50%) can interfere with the reconsolidation of maladaptive memories in healthy participants and across a range of disorders. Negative biases in memory play a key role in the onset, maintenance, and recurrence of depressive episodes, and the disruption of affective memory reconsolidation is one plausible mechanism through which N2O exerts its therapeutic effects. Understanding N2O's mechanisms of action may facilitate future treatment development in depression. In this narrative review, we introduce the evidence supporting an antidepressant profile of N2O and evaluate its clinical use compared to other treatments. With a focus on the specific memory processes that are thought to be disrupted in depression, we consider the effects of N2O on memory reconsolidation and propose a memory-based mechanism of N2O antidepressant action.
Collapse
Affiliation(s)
- Ella Williams
- Department of Psychiatry, Oxford University, Warneford Hospital, Oxford, UK
| | - Ursule Taujanskaite
- Clinical Psychopharmacology Unit, Research Department for Clinical, Educational and Health Psychology, University College London, 1-19 Torrington Place, London, UK
| | - Sunjeev K Kamboj
- Clinical Psychopharmacology Unit, Research Department for Clinical, Educational and Health Psychology, University College London, 1-19 Torrington Place, London, UK
| | - Susannah E Murphy
- Department of Psychiatry, Oxford University, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Catherine J Harmer
- Department of Psychiatry, Oxford University, Warneford Hospital, Oxford, UK.
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK.
| |
Collapse
|
3
|
Vutskits L. Application of Anesthetic Agents in Depression. COTTRELL & PATEL'S NEUROANESTHESIA 2025:503-508. [DOI: 10.1016/b978-0-323-93273-8.00026-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Maji S, Mishra A, Mohapatra D, Mishra BR, Jena M, Srinivasan A, Maiti R. Early augmentation therapy with dextromethorphan in mild to moderate major depressive disorder: A group sequential, response adaptive randomized controlled trial. Psychiatry Res 2024; 342:116257. [PMID: 39551007 DOI: 10.1016/j.psychres.2024.116257] [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: 04/02/2024] [Revised: 10/17/2024] [Accepted: 11/01/2024] [Indexed: 11/19/2024]
Abstract
AIM Therapeutic latency, lack of response, and adverse drug reactions are major challenges in current treatment approaches for major depressive disorder (MDD). Following the success of ketamine, more clinical research on NMDA antagonists is needed for a safe and long-term therapy in MDD. Hence, this study was conducted to evaluate the efficacy and safety of adjunct dextromethorphan to SSRIs in MDD. METHODS In this randomized, double-blind, add-on, placebo-controlled, group sequential design clinical trial, 60 patients with MDD were randomized to receive either adjunct dextromethorphan (30 mg) or adjunct placebo to SSRI for eight weeks. The primary outcome was the change in the MADRS score over eight weeks, and the secondary outcome parameters were response rate, remission rate, change in CGI-S, CGI-I, change in serum BDNF and serum dextromethorphan. RESULTS The results showed a significantly greater reduction in MADRS score from baseline (MD: -3.94; 95 %CI: -5.81 to -2.06; p < 0.001; Cohen's d: 1.05), reduction in CGI-S score (p = 0.002), higher response (p = 0.008) and remission (p = 0.007) rate in the test group compared to the control group. The test group also showed significantly better CGI-I score (p = 0.001) compared to the control group. However, no significant difference was found in the change in serum BDNF (p = 0.751) between the groups. In the test group, serum dextromethorphan levels in all patients were within the therapeutic range. The occurrence of adverse events was comparable in both study groups. CONCLUSION Early augmentation of SSRIs with dextromethorphan (30 mg/day) for the treatment of mild to moderate MDD may improve clinical outcomes significantly in terms of improvement in symptoms, response rate and remission rate. Trial registration ClinicalTrials.gov identifier: NCT05181527.
Collapse
Affiliation(s)
- Shampa Maji
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| | - Archana Mishra
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| | - Debadatta Mohapatra
- Department of Psychiatry, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India.
| | - Biswa Ranjan Mishra
- Department of Psychiatry, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India.
| | - Monalisa Jena
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India.
| | - Anand Srinivasan
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India.
| | - Rituparna Maiti
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India.
| |
Collapse
|
5
|
Freudenberg F, Reif-Leonhard C, Reif A. Advancing past ketamine: emerging glutamatergic compounds for the treatment of depression. Eur Arch Psychiatry Clin Neurosci 2024:10.1007/s00406-024-01875-z. [PMID: 39207462 DOI: 10.1007/s00406-024-01875-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
Changes in glutamatergic neuroplasticity has been proposed as one of the core mechanisms underlying the pathophysiology of depression. In consequence components of the glutamatergic synapse have been explored as potential targets for antidepressant treatment. The rapid antidepressant effect of the NMDA receptor antagonist ketamine and subsequent approval of its S-enantiomer (i.e. esketamine), have set the precedent for investigation into other glutamatergic rapid acting antidepressants (RAADs). In this review, we discuss the potential of the different glutamatergic targets for antidepressant treatment. We describe important clinical outcomes of several key molecules targeting components of the glutamatergic synapse and their applicability as RAADs. Specifically, here we focus on substances beyond (es)ketamine, for which meaningful data from clinical trials are available, including arketamine, esmethadone, nitrous oxide and other glutamate receptor modulators. Molecules only successful in preclinical settings and case reports/series are only marginally discussed. With this review, we aim underscore the critical role of glutamatergic modulation in advancing antidepressant therapy, thereby possibly enhancing clinical outcomes but also to reducing the burden of depression through faster therapeutic effects.
Collapse
Affiliation(s)
- Florian Freudenberg
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt am Main, Germany.
| | - Christine Reif-Leonhard
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt am Main, Germany
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596, Frankfurt Am Main, Germany
| |
Collapse
|
6
|
Conway CR, Palanca BJA, Zeffiro T, Gott BM, Brown F, de Leon V, Barnes L, Nguyen T, Xiong W, Lessov-Schlaggar CN, Espejo G, Mennerick S, Zorumski CF, Nagele P. Nitrous Oxide Alters Functional Connectivity in Medial Limbic Structures in Treatment-Resistant Major Depression. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.12.24311729. [PMID: 39185528 PMCID: PMC11343248 DOI: 10.1101/2024.08.12.24311729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
While nitrous oxide (N2O) has demonstrated antidepressant properties in treatment-resistant major depression (TRD), little is known about neural mechanisms mediating these effects. Employing serial resting-state functional magnetic resonance imaging (rs-fMRI), we compared spatiotemporal effects of inhaled N2O on brain functional connectivity in TRD patients (n=14) and non-depressed healthy controls (n=16, CNTL). Participants received sequential, one-hour inhalations of either 50% N2O/oxygen or air/oxygen (placebo), with sessions separated by at least one month in random cross-over order. BOLD-contrast rs-fMRI scans were acquired at three time points: pre-inhalation, 2 hours post-inhalation, and 24 hours post-inhalation. For the rs-fMRI functional connectivity analyses, five a priori seeds in medial limbic structures targeted cortical networks implicated in major depression - the salience, anterior and posterior default mode, reward, and cingulo-opercular networks - and a nexus in the dorsal paracingulate region previously identified in MDD ("dorsal nexus"). Depression, dissociation, and psychosis assessments were made before and after inhalations. In TRD patients, functional connectivity was reduced in all seeded networks and the voxel-wise global analysis after N2O exposure. N2O progressively decreased connectivity in patients with TRD but increased connectivity in healthy controls. In TRD patients, each seeded network demonstrated post-inhalation functional connectivity reductions in the dorsal paracingulate gyrus ("dorsal nexus"). This study further elucidates neural mechanisms underlying the antidepressant properties of N2O, supporting the notion that N2O specifically alters mood-associated brain regions in the depressed brain state by reducing functional connectivity within these brain networks. The trial was registered at ClinicalTrials.gov (NCT02994433).
Collapse
Affiliation(s)
- Charles R. Conway
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO 63110
| | - Ben Julian A. Palanca
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Thomas Zeffiro
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Britt M. Gott
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - Frank Brown
- Department of Anesthesia and Critical Care, University of Chicago, Chicago, IL 60637
| | - Victoria de Leon
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - Linda Barnes
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - Thomas Nguyen
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Willa Xiong
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | | | - Gemma Espejo
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - Steven Mennerick
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO 63110
| | - Charles F. Zorumski
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO 63110
| | - Peter Nagele
- Department of Anesthesia and Critical Care, University of Chicago, Chicago, IL 60637
| |
Collapse
|
7
|
Brenna CTA, Goldstein BI, Zarate CA, Orser BA. Repurposing General Anesthetic Drugs to Treat Depression: A New Frontier for Anesthesiologists in Neuropsychiatric Care. Anesthesiology 2024; 141:222-237. [PMID: 38856663 DOI: 10.1097/aln.0000000000005037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
During the last 100 years, the role of anesthesiologists in psychiatry has focused primarily on facilitating electroconvulsive therapy and mitigating postoperative delirium and other perioperative neurocognitive disorders. The discovery of the rapid and sustained antidepressant properties of ketamine, and early results suggesting that other general anesthetic drugs (including nitrous oxide, propofol, and isoflurane) have antidepressant properties, has positioned anesthesiologists at a new frontier in the treatment of neuropsychiatric disorders. Moreover, shared interest in understanding the biologic underpinnings of anesthetic drugs as psychotropic agents is eroding traditional academic boundaries between anesthesiology and psychiatry. This article presents a brief overview of anesthetic drugs as novel antidepressants and identifies promising future candidates for the treatment of depression. The authors issue a call to action and outline strategies to foster collaborations between anesthesiologists and psychiatrists as they work toward the common goals of repurposing anesthetic drugs as antidepressants and addressing mood disorders in surgical patients.
Collapse
Affiliation(s)
- Connor T A Brenna
- Department of Anesthesiology & Pain Medicine and Department of Physiology, University of Toronto, Toronto, Canada; Perioperative Brain Health Centre, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Benjamin I Goldstein
- Centre for Addiction and Mental Health, Toronto, Canada; Department of Psychiatry and Department of Pharmacology, University of Toronto, Toronto, Canada
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Beverley A Orser
- Department of Anesthesiology & Pain Medicine and Department of Physiology, University of Toronto, Toronto, Canada; Perioperative Brain Health Centre, Sunnybrook Health Sciences Centre, Toronto, Canada
| |
Collapse
|
8
|
Kronenberg G, Schoretsanitis G, Seifritz E, Olbrich S. The boon and bane of nitrous oxide. Eur Arch Psychiatry Clin Neurosci 2024:10.1007/s00406-024-01801-3. [PMID: 38613686 DOI: 10.1007/s00406-024-01801-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/19/2024] [Indexed: 04/15/2024]
Abstract
Nitrous oxide (N2O) has been known since the end of the eighteenth century. Today, N2O plays a huge role as a greenhouse gas and an ozone-depleting stratospheric molecule. The main sources of anthropogenic N2O emissions are agriculture, fuel combustion, wastewater treatment, and various industrial processes. By contrast, the contribution of medical N2O to the greenhouse effect appears to be small. The recreational and medical uses of N2O gradually diverged over time. N2O has analgesic and anesthetic effects, making it widely used in modern dentistry and surgery. New research has also begun studying N2O's antidepressant actions. N-methyl-D-aspartate (NMDA) antagonism and opioid effects are believed to be the main underlying biochemical mechanisms. At this point, numerous questions remain open and, in particular, the conduct of larger clinical trials will be essential to confirm N2O's use as a rapid-acting antidepressant. The N2O concentration delivered, the duration of a single inhalation, as well as the number of inhalations ultimately required, deserve to be better understood. Finally, the non-medical use of N2O has gained significant attention in recent years. Sudden deaths directly attributed to N2O are primarily due to asphyxia. Heavy, chronic N2O use may result in vitamin B12 deficiency, which, among other things, may cause megaloblastic anemia, venous thrombosis, myeloneuropathy, and skin pigmentation. Helpful biochemical tests include homocysteine and methylmalonic acid. The centerpiece of treatment is complete cessation of N2O use together with parenteral administration of vitamin B12.
Collapse
Affiliation(s)
- Golo Kronenberg
- Department of Psychiatry, Psychotherapy, and Psychosomatics, University Hospital of Psychiatry Zürich, Lenggstrasse 31, P.O. Box 363, 8032, Zurich, Switzerland.
| | - Georgios Schoretsanitis
- Department of Psychiatry, Psychotherapy, and Psychosomatics, University Hospital of Psychiatry Zürich, Lenggstrasse 31, P.O. Box 363, 8032, Zurich, Switzerland
- Department of Psychiatry Research, Zucker Hillside Hospital, Northwell Health, Glen Oaks, New York, USA
- Department of Psychiatry, Donald and Barbara Zucker School of Medicine at Northwell/Hofstra University, Hempstead, New York, USA
| | - Erich Seifritz
- Department of Psychiatry, Psychotherapy, and Psychosomatics, University Hospital of Psychiatry Zürich, Lenggstrasse 31, P.O. Box 363, 8032, Zurich, Switzerland
| | - Sebastian Olbrich
- Department of Psychiatry, Psychotherapy, and Psychosomatics, University Hospital of Psychiatry Zürich, Lenggstrasse 31, P.O. Box 363, 8032, Zurich, Switzerland
| |
Collapse
|
9
|
Ladha KS, Lee J, Mattina GF, Pazmino-Canizares J, Wijeysundera DN, Gholamali Nezhad F, Philip K, Tassone VK, Adamsahib F, Bhat V. Sustained Mood Improvement with Laughing Gas Exposure (SMILE): Study protocol for a randomized placebo-controlled pilot trial of nitrous oxide for treatment-resistant depression. PLoS One 2024; 19:e0297330. [PMID: 38241247 PMCID: PMC10798444 DOI: 10.1371/journal.pone.0297330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 12/12/2023] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND Nitrous oxide has shown potentially as an efficacious intervention for treatment-resistant depression, yet there remains insufficient evidence pertaining to repeated administration of nitrous oxide over time and active placebo-controlled studies with optimal blinding. Thus, we aim to examine the feasibility and preliminary efficacy of a six-week follow up study examining the effects of a 4 week course of weekly administered nitrous oxide as compared to the active placebo, midazolam. METHODS In this randomized, active placebo-controlled, pilot trial, 40 participants with treatment-resistant depression will receive either inhaled nitrous oxide (1 hour at 50% concentration) plus intravenous saline (100mL) or inhaled oxygen (1 hour at 50% concentration) plus intravenous midazolam (0.02 mg/kg in 100mL, up to 2mg) once per week, for 4 consecutive weeks. Participants will be followed up for 6 weeks starting from the first treatment visit. Primary feasibility outcomes include recruitment rate, withdrawal rate, adherence, missing data, and adverse events. The primary exploratory clinical outcome is change in Montgomery-Åsberg Depression Rating Scale (MADRS) score at day 42 of the study. Other exploratory clinical outcomes include remission (defined as MADRS score <10), response (defined as ≥ 50% reduction in MADRS score), and adverse side effects. DISCUSSION This pilot study will provide valuable information regarding the feasibility and preliminary efficacy of repeated nitrous oxide administration over time for treatment-resistant depression. If feasible, this study will inform the design of a future definitive trial of nitrous oxide as an efficacious and fast-acting treatment for treatment-resistant depression. TRIAL REGISTRATION ClinicalTrials.gov NCT04957368. Registered on July 12, 2021.
Collapse
Affiliation(s)
- Karim S. Ladha
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Anesthesia, St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Jiwon Lee
- Department of Anesthesia, St. Michael’s Hospital, Toronto, Ontario, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Duminda N. Wijeysundera
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Anesthesia, St. Michael’s Hospital, Toronto, Ontario, Canada
| | | | - Kaylyssa Philip
- Department of Anesthesia, St. Michael’s Hospital, Toronto, Ontario, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Vanessa K. Tassone
- Interventional Psychiatry Program, St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Fathima Adamsahib
- Interventional Psychiatry Program, St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Venkat Bhat
- Interventional Psychiatry Program, St. Michael’s Hospital, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Neuroscience Research Program, St. Michael’s Hospital, Toronto, Ontario, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
10
|
Wu G, Xu H. A synopsis of multitarget therapeutic effects of anesthetics on depression. Eur J Pharmacol 2023; 957:176032. [PMID: 37660970 DOI: 10.1016/j.ejphar.2023.176032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/05/2023]
Abstract
Depression is a profound mental disorder that dampens the mood and undermines volition, which exhibited an increased incidence over the years. Although drug-based interventions remain the primary approach for depression treatment, the available medications still can't satisfy the patients. In recent years, the newly discovered therapeutic targets such as N-methyl-D-aspartate (NMDA) receptor, α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) receptor, and tyrosine kinase B (TrkB) have brought new breakthroughs in the development of antidepressant drugs. Moreover, it has come to light that certain anesthetics possess pharmacological mechanisms intricately linked to the aforementioned therapeutic targets for depression. At present, numerous preclinical and clinical studies have explored the therapeutic effects of anesthetic drugs such as ketamine, isoflurane, N2O, and propofol, on depression. These investigations suggested that these drugs can swiftly ameliorate patients' depression symptoms and engender long-term effects. In this paper, we provide a comprehensive review of the research progress and potential molecular mechanisms of various anesthetic drugs for depression treatment. By shedding light on this subject, we aim to facilitate the development and clinical implementation of new antidepressant drugs based on anesthetic medications.
Collapse
Affiliation(s)
- Guowei Wu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Hongwei Xu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China.
| |
Collapse
|
11
|
Palanca BJA, Conway CR, Zeffiro T, Gott BM, Nguyen T, Janski A, Jain N, Komen H, Burke BA, Zorumski CF, Nagele P. Persistent Brain Connectivity Changes in Healthy Volunteers Following Nitrous Oxide Inhalation. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:698-704. [PMID: 37881568 PMCID: PMC10593877 DOI: 10.1016/j.bpsgos.2023.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 01/15/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Background Nitrous oxide holds promise in the treatment of major depressive disorder. Its psychotropic effects and NMDA receptor antagonism have led to comparisons with ketamine. Despite longstanding use, persistent effects of nitrous oxide on the brain have not been characterized. Methods Sixteen healthy volunteers were recruited in a double-blind crossover study. In randomized order, individuals underwent a 1-hour inhalation of either 50% nitrous oxide/oxygen or air/oxygen mixtures. At least two 7.5-minute echo-planar resting-state functional magnetic resonance imaging scans were obtained before and at 2 and 24 hours after each inhalation (average 130 min/participant). Using the time series of preprocessed, motion artifact-scrubbed, and nuisance covariate-regressed imaging data, interregional signal correlations were measured and converted to T scores. Hierarchical clustering and linear mixed-effects models were employed. Results Nitrous oxide inhalation produced changes in global brain connectivity that persisted in the occipital cortex at 2 and 24 hours postinhalation (p < .05, false discovery rate-corrected). Analysis of resting-state networks demonstrated robust strengthening of connectivity between regions of the visual network and those of the dorsal attention network, across 2 and 24 hours after inhalation (p < .05, false discovery rate-corrected). Weaker changes in connectivity were found between the visual cortex and regions of the frontoparietal and default mode networks. Parallel analyses following air/oxygen inhalation yielded no significant changes in functional connectivity. Conclusions Nitrous oxide inhalation in healthy volunteers revealed persistent increases in global connectivity between regions of primary visual cortex and dorsal attention network. These findings suggest that nitrous oxide inhalation induces neurophysiological cortical changes that persist for at least 24 hours.
Collapse
Affiliation(s)
- Ben Julian A. Palanca
- Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine in St. Louis, St. Louis, Missouri
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
- Division of Biology and Biomedical Sciences, Washington University School of Medicine in St. Louis, St. Louis, Missouri
- Neuroimaging Labs Research Center, Washington University School of Medicine in St. Louis, St. Louis, Missouri
- Center on Biological Rhythms and Sleep, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Charles R. Conway
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine in St. Louis, St. Louis, Missouri
- Neuroimaging Labs Research Center, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Thomas Zeffiro
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Britt M. Gott
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine in St. Louis, St. Louis, Missouri
- Neuroimaging Labs Research Center, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Thomas Nguyen
- Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Alvin Janski
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Nisha Jain
- Department of Anesthesia and Critical Care, University of Chicago Medicine, Chicago, Illinois
| | - Helga Komen
- Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Broc A. Burke
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado
| | - Charles F. Zorumski
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Peter Nagele
- Department of Anesthesia and Critical Care, University of Chicago Medicine, Chicago, Illinois
| |
Collapse
|
12
|
Qiu Y, Li L, Duan A, Wang M, Xie M, Chen Z, Wang Z. The efficacy and tolerability of inhaled nitrous oxide in major depressive disorder: a systematic review and meta-analysis. Psychopharmacology (Berl) 2023; 240:2033-2043. [PMID: 37608194 DOI: 10.1007/s00213-023-06449-w] [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: 03/10/2023] [Accepted: 08/04/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND Nitrous oxide (N2O) has been initially confirmed by clinical trials to benefit to patients with major depressive disorder (MDD). However, there needs to be a meta-analysis to compare the efficacy and tolerability of N2O in MDD. METHODS PubMed, EMBASE, and Cochrane Library were searched for relevant studies up to Jan 1st, 2023. The meta-analysis mainly compared the outcome of the change in depression severity scores, response, remission, and adverse events in patients with MDD receiving 50% N2O and placebo. RESULTS Four studies with 133 patients were eventually identified. We found that the N2O group and control group showed an overall significant difference in the change in depression severity score for patients at 2 h, 24 h, and 2 weeks or more (2 h, SMD = - 0.64, 95% CI - 0.01 to - 0.28, p < 0.0001) (24 h, SMD = - 0.65, 95% CI - 1.01 to - 0.29, p < 0.0001) (2 weeks, SMD = - 0.76, 95% CI - 1.16 to - 0.36, p < 0.0001). For the response and remission rate, the long-term effect of N2O was also statistically significant (for the response, RR = 2.33, 95% CI 1.23 to 4.44, p = 0.01) (for the remission, RR = 4.68, 95% CI 1.49 to 14.68, p = 0.008). For safety outcomes, patients treated with N2O had higher odds of nausea or vomiting (RR = 10.15, 95% CI 1.96 to 52.59, p = 0.009). CONCLUSION Our study suggested that N2O has a rapid and long-lasting antidepressant effect in patients with MDD. However, the efficacy of lower or titrated concentration of N2O should be further investigated.
Collapse
Affiliation(s)
- Youjia Qiu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Longyuan Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Aojie Duan
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Menghan Wang
- Suzhou Medical College of Soochow USniversity, Suzhou, 215002, Jiangsu Province, China
| | - Minjia Xie
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Zhouqing Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China.
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China.
| |
Collapse
|
13
|
Desmidt T, Dujardin PA, Andersson F, Brizard B, Réméniéras JP, Gissot V, Arlicot N, Barantin L, Espitalier F, Belzung C, Tanti A, Robert G, Bulteau S, Gallet Q, Kazour F, Cognet S, Camus V, El-Hage W, Poupin P, Karim HT. Changes in cerebral connectivity and brain tissue pulsations with the antidepressant response to an equimolar mixture of oxygen and nitrous oxide: an MRI and ultrasound study. Mol Psychiatry 2023; 28:3900-3908. [PMID: 37592013 DOI: 10.1038/s41380-023-02217-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023]
Abstract
Nitrous oxide (N2O) has recently emerged as a potential fast-acting antidepressant but the cerebral mechanisms involved in this effect remain speculative. We hypothesized that the antidepressant response to an Equimolar Mixture of Oxygen and Nitrous Oxide (EMONO) would be associated with changes in cerebral connectivity and brain tissue pulsations (BTP). Thirty participants (20 with a major depressive episode resistant to at least one antidepressant and 10 healthy controls-HC, aged 25-50, only females) were exposed to a 1-h single session of EMONO and followed for 1 week. We defined response as a reduction of at least 50% in the MADRS score 1 week after exposure. Cerebral connectivity of the Anterior Cingulate Cortex (ACC), using ROI-based resting state fMRI, and BTP, using ultrasound Tissue Pulsatility Imaging, were compared before and rapidly after exposure (as well as during exposure for BTP) among HC, non-responders and responders. We conducted analyses to compare group × time, group, and time effects. Nine (45%) depressed participants were considered responders and eleven (55%) non-responders. In responders, we observed a significant reduction in the connectivity of the subgenual ACC with the precuneus. Connectivity of the supracallosal ACC with the mid-cingulate also significantly decreased after exposure in HC and in non-responders. BTP significantly increased in the three groups between baseline and gas exposure, but the increase in BTP within the first 10 min was only significant in responders. We found that a single session of EMONO can rapidly modify the functional connectivity in the subgenual ACC-precuneus, nodes within the default mode network, in depressed participants responders to EMONO. In addition, larger increases in BTP, associated with a significant rise in cerebral blood flow, appear to promote the antidepressant response, possibly by facilitating optimal drug delivery to the brain. Our study identified potential cerebral mechanisms related to the antidepressant response of N2O, as well as potential markers for treatment response with this fast-acting antidepressant.
Collapse
Affiliation(s)
- Thomas Desmidt
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.
- CHU de Tours, Tours, France.
- CIC 1415, CHU de Tours, Inserm, Tours, France.
| | | | | | - Bruno Brizard
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | | | | | - Nicolas Arlicot
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
- CHU de Tours, Tours, France
| | | | - Fabien Espitalier
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
- CHU de Tours, Tours, France
| | | | - Arnaud Tanti
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Gabriel Robert
- Behavior and Basal Ganglia Host Team 4712, University of Rennes 1, Rennes, France Department of Psychiatry, Rennes University Hospital, Guillaume Régnier Hospital Centre, Rennes, France
| | - Samuel Bulteau
- Addictology and Liaison Psychiatry Department, CHU de Nantes, 44000, Nantes, France
| | - Quentin Gallet
- Department of Psychiatry, University Hospital, Angers, France
| | - François Kazour
- Department of Psychiatry, University Hospital, Angers, France
| | | | - Vincent Camus
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
- CHU de Tours, Tours, France
| | - Wissam El-Hage
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
- CHU de Tours, Tours, France
- CIC 1415, CHU de Tours, Inserm, Tours, France
| | | | - Helmet T Karim
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
14
|
Kronenberg G, Müller A, Seifritz E, Olbrich S. A distinct pattern of EEG and ECG changes associated with inhalational nitrous oxide's rapid antidepressant effects. Eur Arch Psychiatry Clin Neurosci 2023; 273:1395-1397. [PMID: 36305920 DOI: 10.1007/s00406-022-01502-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 10/14/2022] [Indexed: 11/03/2022]
Affiliation(s)
- Golo Kronenberg
- Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry Zürich, Lenggstrasse 31, P.O. Box 363, 8032, Zurich, Switzerland
| | - Annette Müller
- Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry Zürich, Lenggstrasse 31, P.O. Box 363, 8032, Zurich, Switzerland
| | - Erich Seifritz
- Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry Zürich, Lenggstrasse 31, P.O. Box 363, 8032, Zurich, Switzerland
| | - Sebastian Olbrich
- Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry Zürich, Lenggstrasse 31, P.O. Box 363, 8032, Zurich, Switzerland.
| |
Collapse
|
15
|
Liu J, Zhao X, Wei X, Yan D, Ou W, Liao M, Ji S, Peng Y, Wu S, Wang M, Ju Y, Zhang L, Li Z, Liu B, Li L, Zhang Y. Empirical evidence for the neurocognitive effect of nitrous oxide as an adjunctive therapy in patients with treatment resistant depression: A randomized controlled study. Psychiatry Res 2023; 326:115326. [PMID: 37390601 DOI: 10.1016/j.psychres.2023.115326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/30/2023] [Accepted: 06/25/2023] [Indexed: 07/02/2023]
Abstract
Nitrous oxide (N2O) has demonstrated an antidepressant effect for treatment-resistant depression (TRD), but no studies investigated the effects of N2O on different cognition domains. This study aimed to test whether N2O would display pro-cognitive effects. We conducted a double-blinded, placebo-controlled, randomized controlled trial, 44 patients with TRD were randomized to N2O group (one-hour inhalation of 50% N2O/50% oxygen) or placebo group (50% air/50% oxygen). Thirty-four patients completed cognitive tests at the pre-treatment phase, 1-week, and 2 weeks post-treatment including subjective cognitive function, processing speed, attention, and executive function. Although the antidepressant effect of N2O was not significant at 1 week, patients still showed better performance of executive function at 1 week after receiving N2O compared with the placebo. Moreover, this significant improvement still existed at 1 week after controlling for the change in depressive symptoms over-time. Additionally, no significant difference was observed in subjective cognitive function, processing speed, and attention between these two groups across the 2-week follow-up period. As the first study investigating the treatment effects of N2O on improving cognitive function in TRD patients, the current study indicated that N2O has a potential pro-cognitive effect on executive function and this effect might be independent from improvements in depressive symptoms.
Collapse
Affiliation(s)
- Jin Liu
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xiaotian Zhao
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xiyu Wei
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Danfeng Yan
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Shanxi Mental Health Center, Taiyuan Fifth People's Hospital, Taiyuan, Shanxi 030045, China
| | - Wenwen Ou
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Mei Liao
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Shanling Ji
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Yan Peng
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Shibin Wu
- Nanning Fifth People's Hospital, Nanning, Guangxi 530028, China
| | - Mi Wang
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Yumeng Ju
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Li Zhang
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zexuan Li
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Bangshan Liu
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.
| | - Lingjiang Li
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.
| | - Yan Zhang
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
16
|
Hassamal S. Chronic stress, neuroinflammation, and depression: an overview of pathophysiological mechanisms and emerging anti-inflammatories. Front Psychiatry 2023; 14:1130989. [PMID: 37252156 PMCID: PMC10213648 DOI: 10.3389/fpsyt.2023.1130989] [Citation(s) in RCA: 125] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 04/21/2023] [Indexed: 05/31/2023] Open
Abstract
In a subset of patients, chronic exposure to stress is an etiological risk factor for neuroinflammation and depression. Neuroinflammation affects up to 27% of patients with MDD and is associated with a more severe, chronic, and treatment-resistant trajectory. Inflammation is not unique to depression and has transdiagnostic effects suggesting a shared etiological risk factor underlying psychopathologies and metabolic disorders. Research supports an association but not necessarily a causation with depression. Putative mechanisms link chronic stress to dysregulation of the HPA axis and immune cell glucocorticoid resistance resulting in hyperactivation of the peripheral immune system. The chronic extracellular release of DAMPs and immune cell DAMP-PRR signaling creates a feed forward loop that accelerates peripheral and central inflammation. Higher plasma levels of inflammatory cytokines, most consistently interleukin IL-1β, IL-6, and TNF-α, are correlated with greater depressive symptomatology. Cytokines sensitize the HPA axis, disrupt the negative feedback loop, and further propagate inflammatory reactions. Peripheral inflammation exacerbates central inflammation (neuroinflammation) through several mechanisms including disruption of the blood-brain barrier, immune cellular trafficking, and activation of glial cells. Activated glial cells release cytokines, chemokines, and reactive oxygen and nitrogen species into the extra-synaptic space dysregulating neurotransmitter systems, imbalancing the excitatory to inhibitory ratio, and disrupting neural circuitry plasticity and adaptation. In particular, microglial activation and toxicity plays a central role in the pathophysiology of neuroinflammation. Magnetic resonance imaging (MRI) studies most consistently show reduced hippocampal volumes. Neural circuitry dysfunction such as hypoactivation between the ventral striatum and the ventromedial prefrontal cortex underlies the melancholic phenotype of depression. Chronic administration of monoamine-based antidepressants counters the inflammatory response, but with a delayed therapeutic onset. Therapeutics targeting cell mediated immunity, generalized and specific inflammatory signaling pathways, and nitro-oxidative stress have enormous potential to advance the treatment landscape. Future clinical trials will need to include immune system perturbations as biomarker outcome measures to facilitate novel antidepressant development. In this overview, we explore the inflammatory correlates of depression and elucidate pathomechanisms to facilitate the development of novel biomarkers and therapeutics.
Collapse
Affiliation(s)
- Sameer Hassamal
- California University of Sciences and Medicine, Colton, CA, United States
- Clinicaltriallink, Los Angeles, CA, United States
- California Neuropsychiatric Institute, Ontario, CA, United States
| |
Collapse
|
17
|
Targeting NMDA Receptors in Emotional Disorders: Their Role in Neuroprotection. Brain Sci 2022; 12:brainsci12101329. [PMID: 36291261 PMCID: PMC9599159 DOI: 10.3390/brainsci12101329] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/28/2022] [Accepted: 09/28/2022] [Indexed: 12/03/2022] Open
Abstract
Excitatory glutamatergic neurotransmission mediated through N-methyl-D-Aspartate (NMDA) receptors (NMDARs) is essential for synaptic plasticity and neuronal survival. While under pathological states, abnormal NMDAR activation is involved in the occurrence and development of psychiatric disorders, which suggests a directional modulation of NMDAR activity that contributes to the remission and treatment of psychiatric disorders. This review thus focuses on the involvement of NMDARs in the pathophysiological processes of psychiatric mood disorders and analyzes the neuroprotective mechanisms of NMDARs. Firstly, we introduce NMDAR-mediated neural signaling pathways in brain function and mood regulation as well as the pathophysiological mechanisms of NMDARs in emotion-related mental disorders such as anxiety and depression. Then, we provide an in-depth summary of current NMDAR modulators that have the potential to be developed into clinical drugs and their pharmacological research achievements in the treatment of anxiety and depression. Based on these findings, drug-targeting for NMDARs might open up novel territory for the development of therapeutic agents for refractory anxiety and depression.
Collapse
|
18
|
Liu H, Kerzner J, Demchenko I, Wijeysundera DN, Kennedy SH, Ladha KS, Bhat V. Nitrous oxide for the treatment of psychiatric disorders: A systematic review of the clinical trial landscape. Acta Psychiatr Scand 2022; 146:126-138. [PMID: 35353901 DOI: 10.1111/acps.13432] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 03/05/2022] [Accepted: 03/26/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To systematically review published research studies and ongoing clinical trials investigating nitrous oxide (N2 O) in psychiatric disorders, providing an up-to-date snapshot of the clinical research landscape. METHODS A comprehensive literature search was conducted for studies published until June 2021 using the OVID databases (MEDLINE, Embase, APA PsycInfo) and the clinical trial registries (ClinicalTrials.gov, ICTRP). RESULTS In total, five relevant published articles were identified, among which four investigated N2 O for depression. One single-dose randomized controlled trial (RCT) for treatment-resistant depression (TRD), one triple crossover RCT comparing 50% vs. 25% N2 O for TRD, and one repeated-dose RCT for major depressive disorder (MDD) suggest that N2 O has preliminary feasibility with rapid-acting effects on symptoms of depression. From the public registries, 10 relevant ongoing clinical trials were identified. They aim to explore the use of N2 O for MDD, post-traumatic stress disorder, bipolar disorder, obsessive-compulsive disorder, and suicidal ideation. To date, the typical treatment protocol parameters were a single session of 50% N2 O delivered for 60 min, although the concentration of 25% is also being explored. Projected enrolment numbers for ongoing trials (M = 55.0) were much higher than sample sizes for published studies (M = 13.0), suggesting that there potentially will be more large-scale RCTs published in the next few years. CONCLUSION Preliminary studies support the feasibility of administering N2 O for depression; however, appropriate blinding is a critical challenge. Larger-scale RCTs with repeated doses of N2 O and follow-up times beyond 1 month are needed to confirm the feasibility, therapeutic efficacy, and sustainability of response.
Collapse
Affiliation(s)
- Helen Liu
- Interventional Psychiatry Program, Mental Health and Addictions Service, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Jaimie Kerzner
- Interventional Psychiatry Program, Mental Health and Addictions Service, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Ilya Demchenko
- Interventional Psychiatry Program, Mental Health and Addictions Service, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Duminda N Wijeysundera
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, Ontario, Canada.,Department of Anesthesia, St. Michael's Hospital, Toronto, Ontario, Canada.,Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Sidney H Kennedy
- Interventional Psychiatry Program, Mental Health and Addictions Service, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Karim S Ladha
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, Ontario, Canada.,Department of Anesthesia, St. Michael's Hospital, Toronto, Ontario, Canada.,Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Venkat Bhat
- Interventional Psychiatry Program, Mental Health and Addictions Service, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
19
|
Elias E, Zhang AY, Manners MT. Novel Pharmacological Approaches to the Treatment of Depression. Life (Basel) 2022; 12:196. [PMID: 35207483 PMCID: PMC8879976 DOI: 10.3390/life12020196] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/19/2022] [Accepted: 01/23/2022] [Indexed: 12/18/2022] Open
Abstract
Major depressive disorder is one of the most prevalent mental health disorders. Monoamine-based antidepressants were the first drugs developed to treat major depressive disorder. More recently, ketamine and other analogues were introduced as fast-acting antidepressants. Unfortunately, currently available therapeutics are inadequate; lack of efficacy, adverse effects, and risks leave patients with limited treatment options. Efforts are now focused on understanding the etiology of depression and identifying novel targets for pharmacological treatment. In this review, we discuss promising novel pharmacological targets for the treatment of major depressive disorder. Targeting receptors including N-methyl-D-aspartate receptors, peroxisome proliferator-activated receptors, G-protein-coupled receptor 39, metabotropic glutamate receptors, galanin and opioid receptors has potential antidepressant effects. Compounds targeting biological processes: inflammation, the hypothalamic-pituitary-adrenal axis, the cholesterol biosynthesis pathway, and gut microbiota have also shown therapeutic potential. Additionally, natural products including plants, herbs, and fatty acids improved depressive symptoms and behaviors. In this review, a brief history of clinically available antidepressants will be provided, with a primary focus on novel pharmaceutical approaches with promising antidepressant effects in preclinical and clinical studies.
Collapse
Affiliation(s)
| | | | - Melissa T. Manners
- Department of Biological Sciences, University of the Sciences, 600 South 43rd Street, Philadelphia, PA 19104, USA; (E.E.); (A.Y.Z.)
| |
Collapse
|
20
|
Demchenko I, Tassone VK, Kennedy SH, Dunlop K, Bhat V. Intrinsic Connectivity Networks of Glutamate-Mediated Antidepressant Response: A Neuroimaging Review. Front Psychiatry 2022; 13:864902. [PMID: 35722550 PMCID: PMC9199367 DOI: 10.3389/fpsyt.2022.864902] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/12/2022] [Indexed: 11/23/2022] Open
Abstract
Conventional monoamine-based pharmacotherapy, considered the first-line treatment for major depressive disorder (MDD), has several challenges, including high rates of non-response. To address these challenges, preclinical and clinical studies have sought to characterize antidepressant response through monoamine-independent mechanisms. One striking example is glutamate, the brain's foremost excitatory neurotransmitter: since the 1990s, studies have consistently reported altered levels of glutamate in MDD, as well as antidepressant effects following molecular targeting of glutamatergic receptors. Therapeutically, this has led to advances in the discovery, testing, and clinical application of a wide array of glutamatergic agents, particularly ketamine. Notably, ketamine has been demonstrated to rapidly improve mood symptoms, unlike monoamine-based interventions, and the neurobiological basis behind this rapid antidepressant response is under active investigation. Advances in brain imaging techniques, including functional magnetic resonance imaging, magnetic resonance spectroscopy, and positron emission tomography, enable the identification of the brain network-based characteristics distinguishing rapid glutamatergic modulation from the effect of slow-acting conventional monoamine-based pharmacology. Here, we review brain imaging studies that examine brain connectivity features associated with rapid antidepressant response in MDD patients treated with glutamatergic pharmacotherapies in contrast with patients treated with slow-acting monoamine-based treatments. Trends in recent brain imaging literature suggest that the activity of brain regions is organized into coherent functionally distinct networks, termed intrinsic connectivity networks (ICNs). We provide an overview of major ICNs implicated in depression and explore how treatment response following glutamatergic modulation alters functional connectivity of limbic, cognitive, and executive nodes within ICNs, with well-characterized anti-anhedonic effects and the enhancement of "top-down" executive control. Alterations within and between the core ICNs could potentially exert downstream effects on the nodes within other brain networks of relevance to MDD that are structurally and functionally interconnected through glutamatergic synapses. Understanding similarities and differences in brain ICNs features underlying treatment response will positively impact the trajectory and outcomes for adults suffering from MDD and will facilitate the development of biomarkers to enable glutamate-based precision therapeutics.
Collapse
Affiliation(s)
- Ilya Demchenko
- Interventional Psychiatry Program, Mental Health and Addictions Service, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Center for Depression and Suicide Studies, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Vanessa K Tassone
- Interventional Psychiatry Program, Mental Health and Addictions Service, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Sidney H Kennedy
- Interventional Psychiatry Program, Mental Health and Addictions Service, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Center for Depression and Suicide Studies, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Katharine Dunlop
- Interventional Psychiatry Program, Mental Health and Addictions Service, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Center for Depression and Suicide Studies, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Venkat Bhat
- Interventional Psychiatry Program, Mental Health and Addictions Service, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Center for Depression and Suicide Studies, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
21
|
Quach DF, de Leon VC, Conway CR. Nitrous Oxide: an emerging novel treatment for treatment-resistant depression. J Neurol Sci 2021; 434:120092. [PMID: 34953347 DOI: 10.1016/j.jns.2021.120092] [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: 06/30/2021] [Revised: 12/03/2021] [Accepted: 12/12/2021] [Indexed: 10/19/2022]
Abstract
Stemming from the results of the historic STAR-D trial, it is evident that a significant subset of individuals (20-25%) with major depressive disorder (MDD) do not respond to conventional antidepressant medications. As a result, an emphasis has been placed on the development of novel therapeutics for MDD over the last two decades. Recently, substantial research efforts have been focused on the use of ketamine as an antidepressant whose mechanism of action is via the N-methyl-d-aspartate (NMDA) receptor. Another potential therapeutic compound of interest is nitrous oxide, which has been utilized for more than a century in multiple fields of medicine for its analgesic and anesthetic properties. Recent clinical studies suggest that nitrous oxide may be effective for treatment-resistant depression. In this review, we will discuss the administration of nitrous oxide as a psychiatric intervention, current use in psychiatry, putative mechanisms of action, and future directions highlighting knowledge gaps and other potential utilities in the field of psychiatry.
Collapse
Affiliation(s)
- Darin F Quach
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Victoria C de Leon
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Charles R Conway
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, United States of America.
| |
Collapse
|