151
|
Savić Vujović K, Jotić A, Medić B, Srebro D, Vujović A, Žujović J, Opanković A, Vučković S. Ketamine, an Old-New Drug: Uses and Abuses. Pharmaceuticals (Basel) 2023; 17:16. [PMID: 38276001 PMCID: PMC10820504 DOI: 10.3390/ph17010016] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 01/27/2024] Open
Abstract
Ketamine as an old-new drug has a variety of clinical implications. In the last 30 years, ketamine has become popular for acute use in humans. Ketamine in standard doses is principally utilized for the induction and maintenance of surgical procedures. Besides its use in anesthesia and analgesia, recent studies have shown that ketamine has found a place in the treatment of asthma, epilepsy, depression, bipolar affective disorders, alcohol and heroin addiction. Ketamine primarily functions as a noncompetitive antagonist targeting the N-methyl-D-aspartate (NMDA) receptor, but its mechanism of action is complex. It is generally regarded as safe, with low doses and short-term use typically not leading to significant adverse effects. Also, ketamine is known as a powerful psychostimulant. During the past decade, ketamine has been one of the commonly abused drugs.
Collapse
Affiliation(s)
- Katarina Savić Vujović
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (B.M.); (D.S.); (S.V.)
| | - Ana Jotić
- Clinic for Otorhinolaryngology and Maxillofacial Surgery, University Clinical Center of Serbia, Pasterova 2, 11000 Belgrade, Serbia;
| | - Branislava Medić
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (B.M.); (D.S.); (S.V.)
| | - Dragana Srebro
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (B.M.); (D.S.); (S.V.)
| | | | - Janko Žujović
- Clinical Centre of Montenegro, Centre for Abdominal Surgery, 81000 Podgorica, Montenegro;
| | - Ana Opanković
- Clinical Centre of Serbia, Clinic for Psychiatry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Sonja Vučković
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (B.M.); (D.S.); (S.V.)
| |
Collapse
|
152
|
Moaddel R, Farmer CA, Yavi M, Kadriu B, Zhu M, Fan J, Chen Q, Lehrmann E, Fantoni G, De S, Mazucanti CH, Acevedo-Diaz EE, Yuan P, Gould TD, Park LT, Egan JM, Ferrucci L, Zarate CA. Cerebrospinal fluid exploratory proteomics and ketamine metabolite pharmacokinetics in human volunteers after ketamine infusion. iScience 2023; 26:108527. [PMID: 38162029 PMCID: PMC10755719 DOI: 10.1016/j.isci.2023.108527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/13/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024] Open
Abstract
Ketamine is a treatment for both refractory depression and chronic pain syndromes. In order to explore ketamine's potential mechanism of action and whether ketamine or its metabolites cross the blood brain barrier, we examined the pharmacokinetics of ketamine and its metabolites-norketamine (NK), dehydronorketamine (DHNK), and hydroxynorketamines (HNKs)-in cerebrospinal fluid (CSF) and plasma, as well as in an exploratory proteomic analysis in the CSF of nine healthy volunteers who received ketamine intravenously (0.5 mg/kg IV). We found that ketamine, NK, and (2R,6R;2S,6S)-HNK readily crossed the blood brain barrier. Additionally, 354 proteins were altered in the CSF in at least two consecutive timepoints (p < 0.01). Proteins in the classes of tyrosine kinases, cellular adhesion molecules, and growth factors, including insulin, were most affected, suggesting an interplay of altered neurotransmission, neuroplasticity, neurogenesis, synaptogenesis, and neural network functions following ketamine administration.
Collapse
Affiliation(s)
- Ruin Moaddel
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Cristan A. Farmer
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Mani Yavi
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Bashkim Kadriu
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Min Zhu
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jinshui Fan
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Qinghua Chen
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Elin Lehrmann
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Giovanna Fantoni
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Supriyo De
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Caio H. Mazucanti
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Elia E. Acevedo-Diaz
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Peixiong Yuan
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Todd D. Gould
- Departments of Psychiatry, Pharmacology, and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Veterans Affairs Maryland Health Care System, Baltimore, MD 21201, USA
| | - Lawrence T. Park
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Josephine M. Egan
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Luigi Ferrucci
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Carlos A. Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
153
|
Rodrigues T, Bressan GN, Krum BN, Soares FAA, Fachinetto R. Influence of the dose of ketamine used on schizophrenia-like symptoms in mice: A correlation study with TH, GAD 67, and PPAR-γ. Pharmacol Biochem Behav 2023; 233:173658. [PMID: 37804866 DOI: 10.1016/j.pbb.2023.173658] [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: 07/14/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Schizophrenia is a chronic, debilitating mental illness that has not yet been completely understood. In this study, we aimed to investigate the effects of different doses of ketamine, a non-competitive NMDA receptor antagonist, on the positive- and negative-like symptoms of schizophrenia. We also explored whether these effects are related to changes in the immunoreactivity of GAD67, TH, and PPAR-γ in brain structures. To conduct the study, male mice received ketamine (20-40 mg/kg) or its vehicle (0.9 % NaCl) intraperitoneally for 14 consecutive days. We quantified stereotyped behavior, the time of immobility in the forced swimming test (FST), and locomotor activity after 7 or 14 days. In addition, we performed ex vivo analysis of the immunoreactivity of GAD, TH, and PPAR-γ, in brain tissues after 14 days. The results showed that ketamine administration for 14 days increased the grooming time in the nose region at all tested doses. It also increased immobility in the FST at 30 mg/kg doses and decreased the number of rearing cycles during stereotyped behavior at 40 mg/kg. These behavioral effects were not associated with changes in locomotor activity. We did not observe any significant alterations regarding the immunoreactivity of brain proteins. However, we found that GAD and TH were positively correlated with the number of rearing during the stereotyped behavior at doses of 20 and 30 mg/kg ketamine, respectively. GAD was positively correlated with the number of rearing in the open field test at a dose of 20 mg/kg. TH was inversely correlated with immobility time in the FST at a dose of 30 mg/kg. PPAR-γ was inversely correlated with the number of bouts of stereotyped behavior at a dose of 40 mg/kg of ketamine. In conclusion, the behavioral alterations induced by ketamine in positive-like symptoms were reproduced with all doses tested and appear to depend on the modulatory effects of TH, GAD, and PPAR-γ. Conversely, negative-like symptoms were associated with a specific dose of ketamine.
Collapse
Affiliation(s)
- Talita Rodrigues
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Getulio Nicola Bressan
- Programa de Pós-Graduação em Ciências Biológicas, Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil
| | - Bárbara Nunes Krum
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Félix Alexandre Antunes Soares
- Programa de Pós-Graduação em Ciências Biológicas, Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil
| | - Roselei Fachinetto
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil.
| |
Collapse
|
154
|
Doan J, Defaix C, Mendez-David I, Gardier AM, Colle R, Corruble E, McGowan JC, David DJ, Guilloux JP, Tritschler L. Intrahippocampal injection of a selective blocker of NMDA receptors containing the GluN2B subunit, Ro25-6981, increases glutamate neurotransmission and induces antidepressant-like effects. Fundam Clin Pharmacol 2023; 37:1119-1128. [PMID: 37161789 DOI: 10.1111/fcp.12917] [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: 11/23/2022] [Revised: 04/07/2023] [Accepted: 05/09/2023] [Indexed: 05/11/2023]
Abstract
Major depressive disorder (MDD) is a serious public health problem, as it is the most common psychiatric disorder worldwide. Antidepressant drugs increase adult hippocampal neurogenesis, which is required to induce some behavioral effects of antidepressants. Adult-born granule cells in the dentate gyrus (DG) and the glutamate receptors subunits 2 (GluN2B) subunit of N-methyl-D-aspartate (NMDA) ionotropic receptors play an important role in these effects. However, the precise neurochemical role of the GluN2B subunit of the NMDA receptor on adult-born GCs for antidepressant-like effects has yet to be elucidated. The present study aims to explore the contribution of the GluN2B-containing NMDA receptors in the ventral dentate gyrus (vDG) to the antidepressant drug treatment using a pharmacological approach. Thus, (αR)-(4-hydroxyphenyl)-(βS)-methyl-4-(phenylmethyl)-1-piperidinepropanol (Ro25-6981), a selective antagonist of the GluN2B subunit, was acutely administered locally into the ventral DG (vDG, 1 μg each side) following a chronic fluoxetine (18 mg/kg/day) treatment-known to increase adult hippocampal neurogenesis-in a mouse model of anxiety/depression. Responses in a neurogenesis-dependent task, the novelty suppressed feeding (NSF), and neurochemical consequences on extracellular glutamate and gamma-aminobutyric acid (GABA) levels in the vDG were measured. Here, we show a rapid-acting antidepressant-like effect of local Ro25-6981 administration in the NSF independent of fluoxetine treatment. Furthermore, we revealed a fluoxetine-independent increase in the glutamatergic transmission in the vDG. Our results suggest behavioral and neurochemical effects of GluN2B subunit independent of serotonin reuptake inhibition.
Collapse
Affiliation(s)
- Julie Doan
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018 CESP, INSERM MOODS Team, Orsay, France
| | - Céline Defaix
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018 CESP, INSERM MOODS Team, Orsay, France
| | - Indira Mendez-David
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018 CESP, INSERM MOODS Team, Orsay, France
| | - Alain M Gardier
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018 CESP, INSERM MOODS Team, Orsay, France
| | - Romain Colle
- Université Paris-Saclay, Faculté de Médecine, UMR 1018 CESP, INSERM MOODS Team, Le Kremlin Bicêtre, France
- Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Emmanuelle Corruble
- Université Paris-Saclay, Faculté de Médecine, UMR 1018 CESP, INSERM MOODS Team, Le Kremlin Bicêtre, France
- Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Josephine C McGowan
- Doctoral Program in Neurobiology and Behavior, Columbia University, New York, New York, USA
| | - Denis J David
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018 CESP, INSERM MOODS Team, Orsay, France
| | - Jean-Philippe Guilloux
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018 CESP, INSERM MOODS Team, Orsay, France
| | - Laurent Tritschler
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018 CESP, INSERM MOODS Team, Orsay, France
| |
Collapse
|
155
|
Vazquez-Juarez E, Srivastava I, Lindskog M. The effect of ketamine on synaptic mistuning induced by impaired glutamate reuptake. Neuropsychopharmacology 2023; 48:1859-1868. [PMID: 37301901 PMCID: PMC10584870 DOI: 10.1038/s41386-023-01617-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/05/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023]
Abstract
Mistuning of synaptic transmission has been proposed to underlie many psychiatric disorders, with decreased reuptake of the excitatory neurotransmitter glutamate as one contributing factor. Synaptic tuning occurs through several diverging and converging forms of plasticity. By recording evoked field postsynaptic potentials in the CA1 area in hippocampal slices, we found that inhibiting glutamate transporters using DL-TBOA causes retuning of synaptic transmission, resulting in a new steady state with reduced synaptic strength and a lower threshold for inducing long-term synaptic potentiation (LTP). Moreover, a similar reduced threshold for LTP was observed in a rat model of depression with decreased levels of glutamate transporters. Most importantly, we found that the antidepressant ketamine counteracts the effects of increased glutamate on the various steps involved in synaptic retuning. We, therefore, propose that ketamine's mechanism of action as an antidepressant is to restore adequate synaptic tuning.
Collapse
Affiliation(s)
- Erika Vazquez-Juarez
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Ipsit Srivastava
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden
- Department of Medical Cell Biology, Uppsala University, 751 24, Uppsala, Sweden
| | - Maria Lindskog
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden.
- Department of Medical Cell Biology, Uppsala University, 751 24, Uppsala, Sweden.
| |
Collapse
|
156
|
Réus GZ, Manosso LM, Quevedo J, Carvalho AF. Major depressive disorder as a neuro-immune disorder: Origin, mechanisms, and therapeutic opportunities. Neurosci Biobehav Rev 2023; 155:105425. [PMID: 37852343 DOI: 10.1016/j.neubiorev.2023.105425] [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: 06/20/2023] [Revised: 08/16/2023] [Accepted: 10/12/2023] [Indexed: 10/20/2023]
Abstract
Notwithstanding advances in understanding the pathophysiology of major depressive disorder (MDD), no single mechanism can explain all facets of this disorder. An expanding body of evidence indicates a putative role for the inflammatory response. Several meta-analyses showed an increase in systemic peripheral inflammatory markers in individuals with MDD. Numerous conditions and circumstances in the modern world may promote chronic systemic inflammation through mechanisms, including alterations in the gut microbiota. Peripheral cytokines may reach the brain and contribute to neuroinflammation through cellular, humoral, and neural pathways. On the other hand, antidepressant drugs may decrease peripheral levels of inflammatory markers. Anti-inflammatory drugs and nutritional strategies that reduce inflammation also could improve depressive symptoms. The present study provides a critical review of recent advances in the role of inflammation in the pathophysiology of MDD. Furthermore, this review discusses the role of glial cells and the main drivers of changes associated with neuroinflammation. Finally, we highlight possible novel neurotherapeutic targets for MDD that could exert antidepressant effects by modulating inflammation.
Collapse
Affiliation(s)
- Gislaine Z Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| | - Luana M Manosso
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil; Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - André F Carvalho
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| |
Collapse
|
157
|
Zhu Y, Feng W, Kong Q, Sheng F, Li Z, Xu W, Li Q, Han Y, Wu X, Jia C, Guo J, Zhao Y. Evaluating the effects of S-ketamine on postoperative delirium in elderly patients following total hip or knee arthroplasty under intraspinal anesthesia: a single-center randomized, double-blind, placebo-controlled, pragmatic study protocol. Front Aging Neurosci 2023; 15:1298661. [PMID: 38099265 PMCID: PMC10720081 DOI: 10.3389/fnagi.2023.1298661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/14/2023] [Indexed: 12/17/2023] Open
Abstract
Introduction Postoperative delirium (POD) is an acute, transient brain disorder associated with decreased postoperative quality of life, dementia, neurocognitive changes, and mortality. A small number of trials have explored the role of S-ketamine in the treatment of POD due to its neuroprotective effects. Surprisingly, these trials have failed to yield supportive results. However, heterogeneity in delirium assessment methodologies, sample sizes, and outcome settings as well as deficiencies in S-ketamine use methods make the evidence provided by these studies less persuasive. Given the severe impact of POD on the health of elderly patients and the potential for S-ketamine to prevent it, we believe that designing a large sample size, and rigorous randomized controlled trial for further evaluation is necessary. Methods This is a single-center, randomized, double-blind, placebo-controlled, pragmatic study. Subjects undergoing total hip or knee arthroplasty will be randomized in a 1:1 ratio to intervention (n = 186) and placebo (n = 186) groups. This trial aims to explore the potential role of S-ketamine in the prevention of POD. Its primary outcome is the incidence of POD within 3 postoperative days. Secondary outcomes include the number of POD episodes, the onset and duration of POD, the severity and subtype of POD, pain scores and opioid consumption, sleep quality, clinical outcomes, and safety outcomes. Discussion To our knowledge, this is the first pragmatic study that proposes to use S-ketamine to prevent POD. We reviewed a large body of literature to identify potential preoperative confounding variables that may bias associations between the intervention and primary outcome. We will use advanced statistical methods to correct potential confounding variables, improving the test's power and external validity of test results. Of note, the patient population included in this trial will undergo intraspinal anesthesia. Although large, multicenter, randomized controlled studies have found no considerable difference in the effects of regional and general anesthesia on POD, patients receiving intraspinal anesthesia have less exposure to at-risk drugs, such as sevoflurane, propofol, and benzodiazepines, than patients receiving general anesthesia. At-risk drugs have been shown to negatively interfere with the neuroprotective effects of S-ketamine, which may be the reason for the failure of a large number of previous studies. There is currently a lack of randomized controlled studies evaluating S-ketamine for POD prevention, and our trial helps to fill a gap in this area.Trial registration: http://www.chictr.org.cn, identifier ChiCTR2300075796.
Collapse
Affiliation(s)
- Youzhuang Zhu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Wei Feng
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Qinghan Kong
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Fang Sheng
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Zhichao Li
- Department of Anesthesiology, Cancer Hospital Chinese Academy of Medical Science, Beijing, China
| | - Weilong Xu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Qun Li
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Yan Han
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xiuyun Wu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Changxin Jia
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Jie Guo
- Department of Anesthesiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yang Zhao
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
158
|
Wang YJ, Zan GY, Xu C, Li XP, Shu X, Yao SY, Xu XS, Qiu X, Chen Y, Jin K, Zhou QX, Ye JY, Wang Y, Xu L, Chen Z, Liu JG. The claustrum-prelimbic cortex circuit through dynorphin/κ-opioid receptor signaling underlies depression-like behaviors associated with social stress etiology. Nat Commun 2023; 14:7903. [PMID: 38036497 PMCID: PMC10689794 DOI: 10.1038/s41467-023-43636-x] [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: 05/04/2023] [Accepted: 11/15/2023] [Indexed: 12/02/2023] Open
Abstract
Ample evidence has suggested the stress etiology of depression, but the underlying mechanism is not fully understood yet. Here, we report that chronic social defeat stress (CSDS) attenuates the excitatory output of the claustrum (CLA) to the prelimbic cortex (PL) through the dynorphin/κ-opioid receptor (KOR) signaling, being critical for depression-related behaviors in male mice. The CSDS preferentially impairs the excitatory output from the CLA onto the parvalbumin (PV) of the PL, leading to PL micronetwork dysfunction by disinhibiting pyramidal neurons (PNs). Optogenetic activation or inhibition of this circuit suppresses or promotes depressive-like behaviors, which is reversed by chemogenetic inhibition or activation of the PV neurons. Notably, manipulating the dynorphin/KOR signaling in the CLA-PL projecting terminals controls depressive-like behaviors that is suppressed or promoted by optogenetic activation or inhibition of CLA-PL circuit. Thus, this study reveals both mechanism of the stress etiology of depression and possibly therapeutic interventions by targeting CLA-PL circuit.
Collapse
Affiliation(s)
- Yu-Jun Wang
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19 A Yuquan Road, 100049, Beijing, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Gui-Ying Zan
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19 A Yuquan Road, 100049, Beijing, China
| | - Cenglin Xu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xue-Ping Li
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Xuelian Shu
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19 A Yuquan Road, 100049, Beijing, China
| | - Song-Yu Yao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiao-Shan Xu
- Laboratory of Learning and Memory, Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Kunming, 650223, China
| | - Xiaoyun Qiu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yexiang Chen
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurobiology of Zhejiang Province, Hangzhou, 310053, China
| | - Kai Jin
- Laboratory of Learning and Memory, Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Kunming, 650223, China
| | - Qi-Xin Zhou
- Laboratory of Learning and Memory, Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Kunming, 650223, China
| | - Jia-Yu Ye
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurobiology of Zhejiang Province, Hangzhou, 310053, China
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lin Xu
- Laboratory of Learning and Memory, Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Kunming, 650223, China.
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Jing-Gen Liu
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, No. 19 A Yuquan Road, 100049, Beijing, China.
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurobiology of Zhejiang Province, Hangzhou, 310053, China.
| |
Collapse
|
159
|
Huang J, Huang W, Yi J, Deng Y, Li R, Chen J, Shi J, Qiu Y, Wang T, Chen X, Zhang X, Xiang AP. Mesenchymal stromal cells alleviate depressive and anxiety-like behaviors via a lung vagal-to-brain axis in male mice. Nat Commun 2023; 14:7406. [PMID: 37973914 PMCID: PMC10654509 DOI: 10.1038/s41467-023-43150-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 11/01/2023] [Indexed: 11/19/2023] Open
Abstract
Major depressive disorder (MDD) is one of the most common and disabling mental disorders, and current strategies remain inadequate. Although mesenchymal stromal cells (MSCs) have shown beneficial effects in experimental models of depression, underlying mechanisms remain elusive. Here, using murine depression models, we demonstrated that MSCs could alleviate depressive and anxiety-like behaviors not due to a reduction in proinflammatory cytokines, but rather activation of dorsal raphe nucleus (DRN) 5-hydroxytryptamine (5-HT) neurons. Mechanistically, peripheral delivery of MSCs activated pulmonary innervating vagal sensory neurons, which projected to the nucleus tractus solitarius, inducing the release of 5-HT in DRN. Furthermore, MSC-secreted brain-derived neurotrophic factor activated lung sensory neurons through tropomyosin receptor kinase B (TrkB), and inhalation of a TrkB agonist also achieved significant therapeutic effects in male mice. This study reveals a role of peripheral MSCs in regulating central nervous system function and demonstrates a potential "lung vagal-to-brain axis" strategy for MDD.
Collapse
Affiliation(s)
- Jing Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Weijun Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Junzhe Yi
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Yiwen Deng
- Key Laboratory of Medical Transformation of Jiujiang, Jiujiang University, Jiujiang, Jiangxi, 332005, China
| | - Ruijie Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Jieying Chen
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Jiahao Shi
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Yuan Qiu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Tao Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Xiaoyong Chen
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Xiaoran Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
160
|
Kim M, Kim W, Chung C. The neural basis underlying female vulnerability to depressive disorders. Anim Cells Syst (Seoul) 2023; 27:297-308. [PMID: 38023591 PMCID: PMC10653660 DOI: 10.1080/19768354.2023.2276815] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Depressive disorders are more prevalent and severe in women; however, our knowledge of the underlying factors contributing to female vulnerability to depression remains limited. Additionally, females are notably underrepresented in studies seeking to understand the mechanisms of depression. Various animal models of depression have been devised, but only recently have females been included in research. In this comprehensive review, we aim to describe the sex differences in the prevalence, pathophysiology, and responses to drug treatment in patients with depression. Subsequently, we highlight animal models of depression in which both sexes have been studied, in the pursuit of identifying models that accurately reflect female vulnerability to depression. We also introduce explanations for the neural basis of sex differences in depression. Notably, the medial prefrontal cortex and the nucleus accumbens have exhibited sex differences in previous studies. Furthermore, other brain circuits involving the dopaminergic center (ventral tegmental area) and the serotonergic center (dorsal raphe nucleus), along with their respective projections, have shown sex differences in relation to depression. In conclusion, our review covers the critical aspects of sex differences in depression, with a specific focus on female vulnerability in humans and its representation in animal models, including the potential underlying mechanisms. Employing suitable animal models that effectively represent female vulnerability would benefit our understanding of the sex-dependent pathophysiology of depression.
Collapse
Affiliation(s)
- Minsoo Kim
- Department of Biological Sciences, Konkuk University, Seoul, South Korea
| | - Woonhee Kim
- Department of Biological Sciences, Konkuk University, Seoul, South Korea
| | - ChiHye Chung
- Department of Biological Sciences, Konkuk University, Seoul, South Korea
| |
Collapse
|
161
|
Vecera CM, C. Courtes A, Jones G, Soares JC, Machado-Vieira R. Pharmacotherapies Targeting GABA-Glutamate Neurotransmission for Treatment-Resistant Depression. Pharmaceuticals (Basel) 2023; 16:1572. [PMID: 38004437 PMCID: PMC10675154 DOI: 10.3390/ph16111572] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/22/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Treatment-resistant depression (TRD) is a term used to describe a particular type of major depressive disorder (MDD). There is no consensus about what defines TRD, with various studies describing between 1 and 4 failures of antidepressant therapies, with or without electroconvulsive therapy (ECT). That is why TRD is such a growing concern among clinicians and researchers, and it explains the necessity for investigating novel therapeutic targets beyond conventional monoamine pathways. An imbalance between two primary central nervous system (CNS) neurotransmitters, L-glutamate and γ-aminobutyric acid (GABA), has emerged as having a key role in the pathophysiology of TRD. In this review, we provide an evaluation and comprehensive review of investigational antidepressants targeting these two systems, accessing their levels of available evidence, mechanisms of action, and safety profiles. N-methyl-D-aspartate (NMDA) receptor antagonism has shown the most promise amongst the glutamatergic targets, with ketamine and esketamine (Spravato) robustly generating responses across trials. Two specific NMDA-glycine site modulators, D-cycloserine (DCS) and apimostinel, have also generated promising initial safety and efficacy profiles, warranting further investigation. Combination dextromethorphan-bupropion (AXS-05/Auvelity) displays a unique mechanism of action and demonstrated positive results in particular applicability in subpopulations with cognitive dysfunction. Currently, the most promising GABA modulators appear to be synthetic neurosteroid analogs with positive GABAA receptor modulation (such as brexanolone). Overall, advances in the last decade provide exciting perspectives for those who do not improve with conventional therapies. Of the compounds reviewed here, three are approved by the Food and Drug Administration (FDA): esketamine (Spravato) for TRD, Auvelity (dextromethorphan-bupropion) for major depressive disorder (MDD), and brexanolone (Zulresso) for post-partum depression (PPD). Notably, some concerns have arisen with esketamine and brexanolone, which will be detailed in this study.
Collapse
Affiliation(s)
- Courtney M. Vecera
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center, Houston, TX 77054, USA
| | - Alan C. Courtes
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center, Houston, TX 77054, USA
| | - Gregory Jones
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center, Houston, TX 77054, USA
| | - Jair C. Soares
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center, Houston, TX 77054, USA
| | - Rodrigo Machado-Vieira
- John S. Dunn Behavioral Sciences Center at UTHealth Houston, 5615 H.Mark Crosswell Jr St, Houston, TX 77021, USA
| |
Collapse
|
162
|
Petzi M, Singh S, Trappenberg T, Nunes A. Mechanisms of Sustained Increases in γ Power Post-Ketamine in a Computational Model of the Hippocampal CA3: Implications for Ketamine's Antidepressant Mechanism of Action. Brain Sci 2023; 13:1562. [PMID: 38002522 PMCID: PMC10670117 DOI: 10.3390/brainsci13111562] [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: 09/30/2023] [Revised: 10/31/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
Subanaesthetic doses of ketamine increase γ oscillation power in neural activity measured using electroencephalography (EEG), and this effect lasts several hours after ketamine administration. The mechanisms underlying this effect are unknown. Using a computational model of the hippocampal cornu ammonis 3 (CA3) network, which is known to reproduce ketamine's acute effects on γ power, we simulated the plasticity of glutamatergic synapses in pyramidal cells to test which of the following hypotheses would best explain this sustained γ power: the direct inhibition hypothesis, which proposes that increased γ power post-ketamine administration may be caused by the potentiation of recurrent collateral synapses, and the disinhibition hypothesis, which proposes that potentiation affects synapses from both recurrent and external inputs. Our results suggest that the strengthening of external connections to pyramidal cells is able to account for the sustained γ power increase observed post-ketamine by increasing the overall activity of and synchrony between pyramidal cells. The strengthening of recurrent pyramidal weights, however, would cause an additional phase shifted voltage increase that ultimately reduces γ power due to partial cancellation. Our results therefore favor the disinhibition hypothesis for explaining sustained γ oscillations after ketamine administration.
Collapse
Affiliation(s)
- Maximilian Petzi
- Faculty of Computer Science, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.P.); (T.T.)
| | - Selena Singh
- Department of Psychology, Neuroscience & Behaviour, McMaster University, Hamilton, ON L8S 4L6, Canada;
| | - Thomas Trappenberg
- Faculty of Computer Science, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.P.); (T.T.)
| | - Abraham Nunes
- Faculty of Computer Science, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.P.); (T.T.)
- Department of Psychiatry, Dalhousie University, Halifax, NS B3H 4K3, Canada
| |
Collapse
|
163
|
Paolini M, Fortaner-Uyà L, Lorenzi C, Spadini S, Maccario M, Zanardi R, Colombo C, Poletti S, Benedetti F. Association between NTRK2 Polymorphisms, Hippocampal Volumes and Treatment Resistance in Major Depressive Disorder. Genes (Basel) 2023; 14:2037. [PMID: 38002980 PMCID: PMC10671548 DOI: 10.3390/genes14112037] [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: 09/30/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
Despite the increasing availability of antidepressant drugs, a high rate of patients with major depression (MDD) does not respond to pharmacological treatments. Brain-derived neurotrophic factor (BDNF)-tyrosine receptor kinase B (TrkB) signaling is thought to influence antidepressant efficacy and hippocampal volumes, robust predictors of treatment resistance. We therefore hypothesized the possible role of BDNF and neurotrophic receptor tyrosine kinase 2 (NTRK2)-related polymorphisms in affecting both hippocampal volumes and treatment resistance in MDD. A total of 121 MDD inpatients underwent 3T structural MRI scanning and blood sampling to obtain genotype information. General linear models and binary logistic regressions were employed to test the effect of genetic variations related to BDNF and NTRK2 on bilateral hippocampal volumes and treatment resistance, respectively. Finally, the possible mediating role of hippocampal volumes on the relationship between genetic markers and treatment response was investigated. A significant association between one NTRK2 polymorphism with hippocampal volumes and antidepressant response was found, with significant indirect effects. Our results highlight a possible mechanistic explanation of antidepressant action, possibly contributing to the understanding of MDD pathophysiology.
Collapse
Affiliation(s)
- Marco Paolini
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Lidia Fortaner-Uyà
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Cristina Lorenzi
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Sara Spadini
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Melania Maccario
- Mood Disorders Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Faculty of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Raffaella Zanardi
- Mood Disorders Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Cristina Colombo
- Mood Disorders Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Faculty of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Sara Poletti
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Faculty of Psychology, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Francesco Benedetti
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Faculty of Psychology, Vita-Salute San Raffaele University, 20132 Milan, Italy
| |
Collapse
|
164
|
Mrozek W, Socha J, Sidorowicz K, Skrok A, Syrytczyk A, Piątkowska-Chmiel I, Herbet M. Pathogenesis and treatment of depression: Role of diet in prevention and therapy. Nutrition 2023; 115:112143. [PMID: 37562078 PMCID: PMC10299949 DOI: 10.1016/j.nut.2023.112143] [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: 01/30/2023] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 08/12/2023]
Abstract
In recent years, there has been a significant increase in depression, which is related to, among other things, the COVID-19 pandemic. Depression can be fatal if not treated or if treated inappropriately. Depression is the leading cause of suicide attempts. The disease is multifactorial, and pharmacotherapy often fails to bring satisfactory results. Therefore, increasingly more importance is attached to the natural healing substances and nutrients in food, which can significantly affect the therapy process and prevention of depressive disorders. A proper diet is vital to preventing depression and can be a valuable addition to psychological and pharmacologic treatment. An inadequate diet may reduce the effectiveness of antidepressants or increase their side effects, leading to life-threatening symptoms. This study aimed to review the literature on the pathogenesis of the development and treatment of depression, with particular emphasis on dietary supplements and the role of nutrition in the prevention and treatment of depressive disorders.
Collapse
Affiliation(s)
- Weronika Mrozek
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Justyna Socha
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Klara Sidorowicz
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Aleksandra Skrok
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Aleksandra Syrytczyk
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | | | - Mariola Herbet
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland.
| |
Collapse
|
165
|
Bottemanne H, Berkovitch L, Gauld C, Balcerac A, Schmidt L, Mouchabac S, Fossati P. Storm on predictive brain: A neurocomputational account of ketamine antidepressant effect. Neurosci Biobehav Rev 2023; 154:105410. [PMID: 37793581 DOI: 10.1016/j.neubiorev.2023.105410] [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: 04/22/2023] [Revised: 08/24/2023] [Accepted: 09/26/2023] [Indexed: 10/06/2023]
Abstract
For the past decade, ketamine, an N-methyl-D-aspartate receptor (NMDAr) antagonist, has been considered a promising treatment for major depressive disorder (MDD). Unlike the delayed effect of monoaminergic treatment, ketamine may produce fast-acting antidepressant effects hours after a single administration at subanesthetic dose. Along with these antidepressant effects, it may also induce transient dissociative (disturbing of the sense of self and reality) symptoms during acute administration which resolve within hours. To understand ketamine's rapid-acting antidepressant effect, several biological hypotheses have been explored, but despite these promising avenues, there is a lack of model to understand the timeframe of antidepressant and dissociative effects of ketamine. In this article, we propose a neurocomputational account of ketamine's antidepressant and dissociative effects based on the Predictive Processing (PP) theory, a framework for cognitive and sensory processing. PP theory suggests that the brain produces top-down predictions to process incoming sensory signals, and generates bottom-up prediction errors (PEs) which are then used to update predictions. This iterative dynamic neural process would relies on N-methyl-D-aspartate (NMDAr) and α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic receptors (AMPAr), two major component of the glutamatergic signaling. Furthermore, it has been suggested that MDD is characterized by over-rigid predictions which cannot be updated by the PEs, leading to miscalibration of hierarchical inference and self-reinforcing negative feedback loops. Based on former empirical studies using behavioral paradigms, neurophysiological recordings, and computational modeling, we suggest that ketamine impairs top-down predictions by blocking NMDA receptors, and enhances presynaptic glutamate release and PEs, producing transient dissociative symptoms and fast-acting antidepressant effect in hours following acute administration. Moreover, we present data showing that ketamine may enhance a delayed neural plasticity pathways through AMPAr potentiation, triggering a prolonged antidepressant effect up to seven days for unique administration. Taken together, the two sides of antidepressant effects with distinct timeframe could constitute the keystone of antidepressant properties of ketamine. These PP disturbances may also participate to a ketamine-induced time window of mental flexibility, which can be used to improve the psychotherapeutic process. Finally, these proposals could be used as a theoretical framework for future research into fast-acting antidepressants, and combination with existing antidepressant and psychotherapy.
Collapse
Affiliation(s)
- Hugo Bottemanne
- Paris Brain Institute - Institut du Cerveau (ICM), UMR 7225 / UMRS 1127, Sorbonne University / CNRS / INSERM, Paris, France; Sorbonne University, Department of Philosophy, Science Norm Democracy Research Unit, UMR, 8011, Paris, France; Sorbonne University, Department of Psychiatry, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.
| | - Lucie Berkovitch
- Saclay CEA Centre, Neurospin, Gif-Sur-Yvette Cedex, France; Department of Psychiatry, GHU Paris Psychiatrie et Neurosciences, Service Hospitalo-Universitaire, Paris, France
| | - Christophe Gauld
- Department of Child Psychiatry, CHU de Lyon, F-69000 Lyon, France; Institut des Sciences Cognitives Marc Jeannerod, UMR 5229 CNRS & Université Claude Bernard Lyon 1, F-69000 Lyon, France
| | - Alexander Balcerac
- Paris Brain Institute - Institut du Cerveau (ICM), UMR 7225 / UMRS 1127, Sorbonne University / CNRS / INSERM, Paris, France; Sorbonne University, Department of Neurology, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Liane Schmidt
- Paris Brain Institute - Institut du Cerveau (ICM), UMR 7225 / UMRS 1127, Sorbonne University / CNRS / INSERM, Paris, France
| | - Stephane Mouchabac
- Paris Brain Institute - Institut du Cerveau (ICM), UMR 7225 / UMRS 1127, Sorbonne University / CNRS / INSERM, Paris, France; Sorbonne University, Department of Psychiatry, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Philippe Fossati
- Paris Brain Institute - Institut du Cerveau (ICM), UMR 7225 / UMRS 1127, Sorbonne University / CNRS / INSERM, Paris, France; Sorbonne University, Department of Philosophy, Science Norm Democracy Research Unit, UMR, 8011, Paris, France
| |
Collapse
|
166
|
Gupta A, Laha JK. Growing Utilization of Radical Chemistry in the Synthesis of Pharmaceuticals. CHEM REC 2023; 23:e202300207. [PMID: 37565381 DOI: 10.1002/tcr.202300207] [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: 06/15/2023] [Revised: 07/19/2023] [Indexed: 08/12/2023]
Abstract
Our current unhealthy lifestyle and the exponential surge in the population getting affected by a variety of diseases have made pharmaceuticals or drugs an imperative part of life, making the development of innovative strategies for drug discovery or the introduction of refined, cost-effective and modern technologies for the synthesis of clinically used drugs, a need of the hour. Ever since their discovery, free radicals and radical cations or anions as reactive intermediates have captivated the chemists, resulting in an exceptional utilization of these moieties throughout the field of chemical synthesis, owing to their unprecedented and widespread reactivity. Sticking with the idea of not judging the book by its cover, despite the conventional thought process of radicals being unstable and difficult to control entities, scientists and academicians around the globe have done an appreciable amount of work utilizing both persistent as well as transient radicals for a variety of organic transformations, exemplifying them with the synthesis of significant biologically active pharmaceutical ingredients. This review truly accounts for the organic radical transformations including radical addition, radical cascade cyclization, radical/radical cross-coupling, coupling with metal-complexes and radical cations coupling with nucleophiles, that offers fascinating and unconventional approaches towards the construction of intricate structural frameworks of marketed APIs with high atom- and step-economy; complementing the otherwise employed traditional methods. This tutorial review presents a comprehensive package of diverse methods utilized for radical generation, featuring their reactivity to form critical bonds in pharmaceutical total synthesis or in building key starting materials or intermediates of their synthetic journey, acknowledging their excellence, downsides and underlying mechanisms, which are otherwise poorly highlighted in the literature. Despite great achievements over the past few decades in this area, many challenges and obstacles are yet to be unraveled to shorten the distance between the academics and the industry, which are all discussed in summary and outlook.
Collapse
Affiliation(s)
- Anjali Gupta
- Department of Pharmaceutical Technology (Process Chemistry), National Institute of Pharmaceutical Education & Research (NIPER) S.A.S. Nagar, Sahibzada Ajit Singh Nagar, Mohali, 160062, India
| | - Joydev K Laha
- Department of Pharmaceutical Technology (Process Chemistry), National Institute of Pharmaceutical Education & Research (NIPER) S.A.S. Nagar, Sahibzada Ajit Singh Nagar, Mohali, 160062, India
| |
Collapse
|
167
|
Coffelt CB, Gibson K, VanLandingham J. Ketamine for Traumatic Assault-Induced Depression: A Case Report. J Trauma Nurs 2023; 30:353-356. [PMID: 37937877 DOI: 10.1097/jtn.0000000000000755] [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: 11/09/2023]
Abstract
BACKGROUND This case report describes the use of ketamine as a rapid, effective treatment of depression in a 68-year-old female patient with no significant medical history of psychiatric disorders. Patients who experience intentional or unintentional traumas are at an increased risk for developing depression or posttraumatic stress disorder, and emerging evidence has supported the use of ketamine as an alternative treatment of depression. CASE PRESENTATION This is the case of a 68-year-old female patient who was assaulted, resulting in multiple stab wounds to both hands and the right upper quadrant. She underwent placement of a chest tube and surgical repair of the liver and was subsequently admitted to the intensive care unit. These events led to the development of severe depression symptoms, as evidenced by a Montgomery-Asberg Depression Rating Scale (MADRS) score of 37. As treatment of her acute depression, the patient received a single intravenous dose of ketamine (0.5 mg/kg) infused over 40 min and was monitored for side effects. The MADRS is a 10-item depression screening tool that assesses symptoms and changes over time. Within 4 hr of receiving ketamine, the patient reported a significant improvement in her mood and her MADRS score decreased to 16, classifying this patient as experiencing mild depression. The patient continued to improve, and 24 hr after receiving ketamine, her MADRS score was 4, indicating remission of her depression symptoms. CONCLUSION This case report aims to provide an account of the potential benefits of ketamine as a rapid treatment of depression in an adult trauma patient.
Collapse
Affiliation(s)
- Caitlyn B Coffelt
- University of Georgia College of Pharmacy, Athens, Georgia (Dr Coffelt); and Northeast Georgia Medical Center, Gainesville, Georgia (Ms Gibson and Dr VanLandingham)
| | | | | |
Collapse
|
168
|
Ladagu AD, Olopade FE, Adejare A, Olopade JO. GluN2A and GluN2B N-Methyl-D-Aspartate Receptor (NMDARs) Subunits: Their Roles and Therapeutic Antagonists in Neurological Diseases. Pharmaceuticals (Basel) 2023; 16:1535. [PMID: 38004401 PMCID: PMC10674917 DOI: 10.3390/ph16111535] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/11/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are ion channels that respond to the neurotransmitter glutamate, playing a crucial role in the permeability of calcium ions and excitatory neurotransmission in the central nervous system (CNS). Composed of various subunits, NMDARs are predominantly formed by two obligatory GluN1 subunits (with eight splice variants) along with regulatory subunits GluN2 (GluN2A-2D) and GluN3 (GluN3A-B). They are widely distributed throughout the CNS and are involved in essential functions such as synaptic transmission, learning, memory, plasticity, and excitotoxicity. The presence of GluN2A and GluN2B subunits is particularly important for cognitive processes and has been strongly implicated in neurodegenerative diseases like Parkinson's disease and Alzheimer's disease. Understanding the roles of GluN2A and GluN2B NMDARs in neuropathologies provides valuable insights into the underlying causes and complexities of major nervous system disorders. This knowledge is vital for the development of selective antagonists targeting GluN2A and GluN2B subunits using pharmacological and molecular methods. Such antagonists represent a promising class of NMDA receptor inhibitors that have the potential to be developed into neuroprotective drugs with optimal therapeutic profiles.
Collapse
Affiliation(s)
- Amany Digal Ladagu
- Department of Veterinary Anatomy, University of Ibadan, Ibadan 200284, Nigeria; (A.D.L.); (J.O.O.)
| | - Funmilayo Eniola Olopade
- Developmental Neurobiology Laboratory, Department of Anatomy, College of Medicine, University of Ibadan, Ibadan 200284, Nigeria
| | - Adeboye Adejare
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, Saint Joseph’s University, Philadelphia, PA 19131, USA
| | - James Olukayode Olopade
- Department of Veterinary Anatomy, University of Ibadan, Ibadan 200284, Nigeria; (A.D.L.); (J.O.O.)
| |
Collapse
|
169
|
Dębowska W, Więdłocha M, Dębowska M, Kownacka Z, Marcinowicz P, Szulc A. Transcranial magnetic stimulation and ketamine: implications for combined treatment in depression. Front Neurosci 2023; 17:1267647. [PMID: 37954877 PMCID: PMC10637948 DOI: 10.3389/fnins.2023.1267647] [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: 07/27/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023] Open
Abstract
Drug-resistant mental disorders, particularly treatment-resistant depression, pose a significant medical and social problem. To address this challenge, modern psychiatry is constantly exploring the use of novel treatment methods, including biological treatments, such as transcranial magnetic stimulation (TMS), and novel rapid-acting antidepressants, such as ketamine. While both TMS and ketamine demonstrate high effectiveness in reducing the severity of depressive symptoms, some patients still do not achieve the desired improvement. Recent literature suggests that combining these two methods may yield even stronger and longer-lasting results. This review aims to consolidate knowledge in this area and elucidate the potential mechanisms of action underlying the increased efficacy of combined treatment, which would provide a foundation for the development and optimization of future treatment protocols.
Collapse
Affiliation(s)
- Weronika Dębowska
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Magdalena Więdłocha
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
- KeyClinic, Warsaw, Poland
| | - Marta Dębowska
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Zuzanna Kownacka
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Marcinowicz
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
- KeyClinic, Warsaw, Poland
| | - Agata Szulc
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
- MindHealth, Warsaw, Poland
| |
Collapse
|
170
|
Harkin EF, Nasrallah G, Le François B, Albert PR. Transcriptional Regulation of the Human 5-HT1A Receptor Gene by Lithium: Role of Deaf1 and GSK3β. Int J Mol Sci 2023; 24:15620. [PMID: 37958600 PMCID: PMC10647674 DOI: 10.3390/ijms242115620] [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: 09/19/2023] [Revised: 10/11/2023] [Accepted: 10/21/2023] [Indexed: 11/15/2023] Open
Abstract
Serotonin 1A (5-HT1A) autoreceptors located on serotonin neurons inhibit their activity, and their upregulation has been implicated in depression, suicide and resistance to antidepressant treatment. Conversely, post-synaptic 5-HT1A heteroreceptors are important for antidepressant response. The transcription factor deformed epidermal autoregulatory factor 1 (Deaf1) acts as a presynaptic repressor and postsynaptic enhancer of 5-HT1A transcription, but the mechanism is unclear. Because Deaf1 interacts with and is phosphorylated by glycogen synthase kinase 3β (GSK3β)-a constitutively active protein kinase that is inhibited by the mood stabilizer lithium at therapeutic concentrations-we investigated the role of GSK3β in Deaf1 regulation of human 5-HT1A transcription. In 5-HT1A promoter-reporter assays, human HEK293 kidney and 5-HT1A-expressing SKN-SH neuroblastoma cells, transfection of Deaf1 reduced 5-HT1A promoter activity by ~45%. To identify potential GSK3β site(s) on Deaf1, point mutations of known and predicted phosphorylation sites on Deaf1 were tested. Deaf1 repressor function was not affected by any of the mutants tested except the Y300F mutant, which augmented Deaf1 repression. Both lithium and the selective GSK3 inhibitors CHIR-99021 and AR-014418 attenuated and reversed Deaf1 repression compared to vector. This inhibition was at concentrations that maximally inhibit GSK3β activity as detected by the GSK3β-sensitive TCF/LEF reporter construct. Our results support the hypothesis that GSK3β regulates the activity of Deaf1 to repress 5-HT1A transcription and provide a potential mechanism for actions of GSK3 inhibitors on behavior.
Collapse
Affiliation(s)
| | | | | | - Paul R. Albert
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa, 451 Smyth Road, Ottawa, ON K1H-8M5, Canada (B.L.F.)
| |
Collapse
|
171
|
Ledesma-Corvi S, Jornet-Plaza J, García-Fuster MJ. Aromatase inhibition and ketamine in rats: sex-differences in antidepressant-like efficacy. Biol Sex Differ 2023; 14:73. [PMID: 37876000 PMCID: PMC10599051 DOI: 10.1186/s13293-023-00560-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/17/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Ketamine has been recently approved to treat resistant depression; however preclinical studies showed sex differences in its efficacy. Sex steroids, such as estrogens and testosterone, both in the periphery and locally in the brain, are regarded as important modulators of these sex differences. Therefore, the present study evaluated how inhibiting the biosynthesis of estrogens with letrozole (an aromatase inhibitor) could affect the observed sex differences in ketamine's antidepressant-like-response. METHODS We performed several consecutive studies in adult Sprague-Dawley rats to evaluate potential sex differences in the antidepressant-like effects of ketamine (5 mg/kg, 7 days, i.p.), letrozole (1 mg/kg, 8 days, i.p.) and their combination (letrozole pre-treatment 3 h before ketamine). Acute and repeated antidepressant-like responses were ascertained in a series of behavioral tests (forced-swim, novelty-suppressed feeding, two-bottle choice for sucrose preference). RESULTS The main results proved clear sex differences in the antidepressant-like response induced by ketamine, which was observed following a repeated paradigm in adult male rats, but rendered inefficacious in female rats. Moreover, decreasing estrogens production with letrozole induced on itself an antidepressant-like response in female rats, while also increased ketamine's response in male rats (i.e., quicker response observed after only a single dose). Interestingly, both the antidepressant-like effects induced by ketamine in male rats or letrozole in female rats persisted over time up to 65 days post-treatment, suggesting long-term sex-directed benefits for these drugs. CONCLUSIONS The present results demonstrated a sex-specific role for aromatase inhibition with letrozole in the antidepressant-like response induced by ketamine in male rats. Moreover, letrozole itself presented as a potential antidepressant for females with persistent effects over time. Clearly, the production of estrogens is key in modulating, in a sex-specific manner, affective-like responses and thus deserve further studies.
Collapse
Affiliation(s)
- Sandra Ledesma-Corvi
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa Km 7.5, 07122, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Jordi Jornet-Plaza
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa Km 7.5, 07122, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - M Julia García-Fuster
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa Km 7.5, 07122, Palma, Spain.
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.
- Department of Medicine, University of the Balearic Islands, Palma, Spain.
| |
Collapse
|
172
|
Schwartz OS, Amminger P, Baune BT, Bedi G, Berk M, Cotton SM, Daglas-Georgiou R, Glozier N, Harrison B, Hermens DF, Jennings E, Lagopoulos J, Loo C, Mallawaarachchi S, Martin D, Phelan B, Read N, Rodgers A, Schmaal L, Somogyi AA, Thurston L, Weller A, Davey CG. The Study of Ketamine for Youth Depression (SKY-D): study protocol for a randomised controlled trial of low-dose ketamine for young people with major depressive disorder. Trials 2023; 24:686. [PMID: 37875938 PMCID: PMC10594918 DOI: 10.1186/s13063-023-07631-3] [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: 09/07/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Existing treatments for young people with severe depression have limited effectiveness. The aim of the Study of Ketamine for Youth Depression (SKY-D) trial is to determine whether a 4-week course of low-dose subcutaneous ketamine is an effective adjunct to treatment-as-usual in young people with major depressive disorder (MDD). METHODS SKY-D is a double-masked, randomised controlled trial funded by the Australian Government's National Health and Medical Research Council (NHMRC). Participants aged between 16 and 25 years (inclusive) with moderate-to-severe MDD will be randomised to receive either low-dose ketamine (intervention) or midazolam (active control) via subcutaneous injection once per week for 4 weeks. The primary outcome is change in depressive symptoms on the Montgomery-Åsberg Depression Rating Scale (MADRS) after 4 weeks of treatment. Further follow-up assessment will occur at 8 and 26 weeks from treatment commencement to determine whether treatment effects are sustained and to investigate safety outcomes. DISCUSSION Results from this trial will be important in determining whether low-dose subcutaneous ketamine is an effective treatment for young people with moderate-to-severe MDD. This will be the largest randomised trial to investigate the effects of ketamine to treat depression in young people. TRIAL REGISTRATION Australian and New Zealand Clinical Trials Registry ID: ACTRN12619000683134. Registered on May 7, 2019. https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=377513 .
Collapse
Affiliation(s)
- Orli S Schwartz
- Department of Psychiatry, University of Melbourne, Melbourne, Australia.
- Orygen, Melbourne, Australia.
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia.
| | - Paul Amminger
- Orygen, Melbourne, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
| | - Bernard T Baune
- Department of Psychiatry, University of Melbourne, Melbourne, Australia
- Department of Psychiatry, University of Münster, Münster, Germany
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
| | - Gillinder Bedi
- Orygen, Melbourne, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
| | - Michael Berk
- Orygen, Melbourne, Australia
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, Australia
| | - Sue M Cotton
- Orygen, Melbourne, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
| | - Rothanthi Daglas-Georgiou
- Orygen, Melbourne, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
| | - Nick Glozier
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Australian Research Council Centre of Excellence for Children and Families Over the Life Course, Sydney, Australia
- Professor Marie Bashir Centre, Royal Prince Alfred Hospital, Sydney, Australia
| | - Ben Harrison
- Department of Psychiatry, University of Melbourne, Melbourne, Australia
| | - Daniel F Hermens
- Thompson Institute, University of the Sunshine Coast, Sunshine Coast, Australia
| | - Emma Jennings
- Orygen, Melbourne, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
| | - Jim Lagopoulos
- Thompson Institute, University of the Sunshine Coast, Sunshine Coast, Australia
- Thompson Brain and Mind Healthcare, Sunshine Coast, Australia
| | - Colleen Loo
- School of Psychiatry, University of New South Wales, Sydney, Australia
- Black Dog Institute, Sydney, Australia
| | - Sumudu Mallawaarachchi
- Orygen, Melbourne, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
| | - Donel Martin
- Black Dog Institute, Sydney, Australia
- School of Clinical Medicine, University of New South Wales, Sydney, Australia
| | - Bethany Phelan
- Orygen, Melbourne, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
| | - Nikki Read
- Orygen, Melbourne, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
| | - Anthony Rodgers
- The George Institute for Global Health, University of New South Wales, Sydney, Australia
| | - Lianne Schmaal
- Orygen, Melbourne, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
| | - Andrew A Somogyi
- School of Biomedicine, University of Adelaide, Adelaide, Australia
| | - Lily Thurston
- Orygen, Melbourne, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
| | - Amber Weller
- Orygen, Melbourne, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
- Victorian Department of Health, Melbourne, Australia
| | - Christopher G Davey
- Department of Psychiatry, University of Melbourne, Melbourne, Australia
- Orygen, Melbourne, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
| |
Collapse
|
173
|
Liu AR, Lin ZJ, Wei M, Tang Y, Zhang H, Peng XG, Li Y, Zheng YF, Tan Z, Zhou LJ, Feng X. The potent analgesia of intrathecal 2R, 6R-HNK via TRPA1 inhibition in LF-PENS-induced chronic primary pain model. J Headache Pain 2023; 24:141. [PMID: 37858040 PMCID: PMC10585932 DOI: 10.1186/s10194-023-01667-1] [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: 04/26/2023] [Accepted: 09/11/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Chronic primary pain (CPP) is an intractable pain of unknown cause with significant emotional distress and/or dysfunction that is a leading factor of disability globally. The lack of a suitable animal model that mimic CPP in humans has frustrated efforts to curb disease progression. 2R, 6R-hydroxynorketamine (2R, 6R-HNK) is the major antidepressant metabolite of ketamine and also exerts antinociceptive action. However, the analgesic mechanism and whether it is effective for CPP are still unknown. METHODS Based on nociplastic pain is evoked by long-term potentiation (LTP)-inducible high- or low-frequency electrical stimulation (HFS/LFS), we wanted to develop a novel CPP mouse model with mood and cognitive comorbidities by noninvasive low-frequency percutaneous electrical nerve stimulation (LF-PENS). Single/repeated 2R, 6R-HNK or other drug was intraperitoneally (i.p.) or intrathecally (i.t.) injected into naïve or CPP mice to investigate their analgesic effect in CPP model. A variety of behavioral tests were used to detect the changes in pain, mood and memory. Immunofluorescent staining, western blot, reverse transcription-quantitative real-time polymerase chain reaction (RT-qPCR) and calcium imaging of in cultured dorsal root ganglia (DRG) neurons by Fluo-8-AM were used to elucidate the role and mechanisms of 2R, 6R-HNK in vivo or in vitro. RESULTS Intrathecal 2R, 6R-HNK, rather than intraperitoneal 2R, 6R-HNK or intrathecal S-Ketamine, successfully mitigated HFS-induced pain. Importantly, intrathecal 2R, 6R-HNK displayed effective relief of bilateral pain hypersensitivity and depressive and cognitive comorbidities in a dose-dependent manner in LF-PENS-induced CPP model. Mechanically, 2R, 6R-HNK markedly attenuated neuronal hyperexcitability and the upregulation of calcitonin gene-related peptide (CGRP), transient receptor potential ankyrin 1 (TRPA1) or vanilloid-1 (TRPV1), and vesicular glutamate transporter-2 (VGLUT2) in peripheral nociceptive pathway. In addition, 2R, 6R-HNK suppressed calcium responses and CGRP overexpression in cultured DRG neurons elicited by the agonists of TRPA1 or/and TRPV1. Strikingly, the inhibitory effects of 2R, 6R-HNK on these pain-related molecules and mechanical allodynia were substantially occluded by TRPA1 antagonist menthol. CONCLUSIONS In the newly designed CPP model, our findings highlighted the potential utility of intrathecal 2R, 6R-HNK for preventing and therapeutic modality of CPP. TRPA1-mediated uprgulation of CGRP and neuronal hyperexcitability in nociceptive pathways may undertake both unique characteristics and solving process of CPP.
Collapse
Affiliation(s)
- An-Ran Liu
- Department of Anesthesiology and Pain Clinic, First Affiliated Hospital of Sun Yat-Sen University, No.58, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China
| | - Zhen-Jia Lin
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University, No.74, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China
| | - Ming Wei
- Department of Anesthesiology and Pain Clinic, First Affiliated Hospital of Sun Yat-Sen University, No.58, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China
| | - Yuan Tang
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University, No.74, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China
| | - Hui Zhang
- Department of Anesthesiology, Guangdong Second Provincial General Hospital, No.466, Mid Xingang Road, Haizhu District, Guangzhou, 510317, China
| | - Xiang-Ge Peng
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University, No.74, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China
| | - Ying Li
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University, No.74, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China
| | - Yu-Fan Zheng
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University, No.74, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China
| | - Zhi Tan
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University, No.74, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China.
| | - Li-Jun Zhou
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University, No.74, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China.
| | - Xia Feng
- Department of Anesthesiology and Pain Clinic, First Affiliated Hospital of Sun Yat-Sen University, No.58, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China.
| |
Collapse
|
174
|
Guo L, Wang S, Tian H, Shang M, Xu J, Wang C. Novel synergistic treatment for depression: involvement of GSK3β-regulated AMPA receptors in the prefrontal cortex of mice. Cereb Cortex 2023; 33:10504-10513. [PMID: 37566915 DOI: 10.1093/cercor/bhad299] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Previous evidence has suggested a vital role of glycogen synthase kinase 3β-mediated α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors trafficking in depression. Considering the antidepressant effect of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors activation in the prefrontal cortex, we hypothesized that glycogen synthase kinase 3β-induced alterations in α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors function in the prefrontal cortex participate in depression. Herein, we confirmed that the levels of phosphorylated glycogen synthase kinase 3β and GluA1, the latter being a subunit of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors, were decreased in the prefrontal cortex of the chronic social defeat stress model mice presenting with depressive-like behaviors. We then found that a glycogen synthase kinase 3β (p.S9A) point mutation downregulated GluA1 and induced depressive-like behaviors in mice, whereas an agonist of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors, PF-4778574 (2 mg/kg) did not reversed the molecular changes. On the other hand, the antidepressant effect of PF-4778574 was dose dependent, and the single administration of PF-4778574 at a lower dose (0.5 mg/kg) or of the glycogen synthase kinase 3β inhibitor SB216763 (5 and 10 mg/kg) did not evoke an antidepressant effect. In contrast, co-treatment with PF-4778574 (0.5 mg/kg) and SB216763 (10 mg/kg) led to antidepressant effects similar to those of PF-4778574 (2 mg/kg). Our results suggest that glycogen synthase kinase 3β-induced α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors dysfunction in the prefrontal cortex is one of the key mechanisms of depression, and the combination of a lower dose of PF-4778574 with SB216763 shows potential as a novel synergistic treatment for depression.
Collapse
Affiliation(s)
- Lei Guo
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Shuzhuo Wang
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Haihua Tian
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
- Department of Psychiatry, Ningbo Kangning Hospital, Ningbo, Zhejiang 315201, PR China
| | - Mengyuan Shang
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Jia Xu
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Chuang Wang
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
| |
Collapse
|
175
|
Fancy F, Rodrigues NB, Di Vincenzo JD, Chau EH, Sethi R, Husain MI, Gill H, Tabassum A, Mckenzie A, Phan L, McIntyre RS, Rosenblat JD. Real-World Effectiveness of Repeated Ketamine Infusions for Treatment-Resistant Bipolar Depression. FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2023; 21:420-429. [PMID: 38694999 PMCID: PMC11058957 DOI: 10.1176/appi.focus.23021022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Background Clinical trials have demonstrated rapid antidepressant effects with intravenous (IV) ketamine for major depressive disorder, with relatively less research specifically for bipolar depression. Herein, we describe the real-world effectiveness of repeated ketamine infusions for treatment-resistant bipolar depression. Methods This study was conducted in a community clinic in Mississauga, Ontario (Canadian Rapid Treatment Centre of Excellence; Braxia Health). In this observational study (NCT04209296), patients with treatment-resistant bipolar I/II depression (n = 66) received four sub-anesthetic doses of IV ketamine (0.5-0.75 mg/kg) over a two-week period. Symptoms of depression, suicidality, anxiety, and functioning were assessed with validated self-report measures. Results Statistically and clinically significant antidepressant effects were observed in the overall sample, as measured by the Quick Inventory for Depression Symptomatology-Self Report-16 (QIDS-SR16) with further reductions in depressive symptoms observed after each subsequent infusion (n = 66; mean QIDS-SR16 reduction of 6.08+/-1.39; p < 0.0001). Significant reductions of suicidal thoughts (QIDS-SR16-Suicide Item) and anxiety (Generalized Anxiety Disorder-7) were also observed with functional improvements on the Sheehan Disability Scale (p < 0.0001 on all measures). Moreover, the response rate (QIDS-SR16 total score decrease ≥50% from baseline) was 35% and remission rate (QIDS-SR16 total score ≤5) was 20% after four infusions. Infusions were generally well tolerated with treatment-emergent hypomania observed in only three patients (4.5%) with zero cases of mania or psychosis. Conclusions Real-world effectiveness of IV ketamine for bipolar depression was observed. Repeated doses were associated with greater symptom reduction and adequate tolerability.Reprinted from Bipolar Disord 2023; 25:99-109, with permission from John Wiley and Sons. Copyright © 2023.
Collapse
Affiliation(s)
- Farhan Fancy
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Nelson B Rodrigues
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Joshua D Di Vincenzo
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Edmond H Chau
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Rickinder Sethi
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Muhammad I Husain
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Hartej Gill
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Aniqa Tabassum
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Andrea Mckenzie
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Lee Phan
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Joshua D Rosenblat
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| |
Collapse
|
176
|
Miao H, Li R, Li W, Wu F, Li H, Luo H. Electroacupuncture attenuates ketamine-induced neuronal injury in the locus coeruleus of rats through modulation of the CAMK II/CREB pathway. Brain Res Bull 2023; 202:110724. [PMID: 37543295 DOI: 10.1016/j.brainresbull.2023.110724] [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: 05/25/2023] [Revised: 07/02/2023] [Accepted: 08/01/2023] [Indexed: 08/07/2023]
Abstract
BACKGROUND Ketamine, despite its efficacy in treating depression, raises concerns regarding safety due to potential abuse, cognitive impairment, and bladder toxicity. Ketamine can affect the locus coeruleus (LC) norepinephrine and attention networks. This study explored the protective effects of electroacupuncture (EA) on the LC of rats exposed to repeated administration of ketamine while investigating the potential role of the Calcium CaM-dependent protein kinase II (CAMK II)/ cAMP response element binding protein (CREB) pathway in mediating EA's impact on ketamine-induced neuronal injury in LC. METHODS Rats were repeatedly injected intraperitoneally with ketamine hydrochloride (50 mg/kg) once daily for seven days. Subsequently, EA was performed at the acupoints "Zusanli" (ST36) and "Sanyinjiao" (SP-6) once daily following ketamine administration. The Morris water maze test was employed to assess behavioral changes in the rats. Neuronal injury was examined using Nissl staining, and the expression of CAMK II, CREB, and phospho-CREB (p-CREB) was evaluated through immunohistochemistry and western blotting. RESULTS EA mitigated the cognitive and exploratory impairments and attenuated neuronal injury in the LC induced by repeated administration of ketamine. The expression of CAMK II and p-CREB proteins in the LC increased following 7 days of ketamine administration. However, EA treatment led to a downregulation of CAMK II and p-CREB expression. CONCLUSION Repeated administration of ketamine in male rats can lead to neuronal injury and neurobehavioral dysfunction. However, EA was found to ameliorate neurodegeneration in the LC and enhance neurobehavioral symptoms. This therapeutic effect of EA may be attributed to its modulation of the CAMKII/CREB pathway, thereby mitigating the aforementioned adverse effects.
Collapse
Affiliation(s)
- Huachun Miao
- Department of Human Anatomy, Wannan Medical College, Wuhu, Anhui Province 241002, China
| | - Runzhi Li
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province 310053, China
| | - Wenjuan Li
- Department of Human Anatomy, Wannan Medical College, Wuhu, Anhui Province 241002, China
| | - Feng Wu
- Department of Human Anatomy, Wannan Medical College, Wuhu, Anhui Province 241002, China
| | - Huaibin Li
- Department of Human Anatomy, Wannan Medical College, Wuhu, Anhui Province 241002, China.
| | - Hong Luo
- The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang Province 311121, China.
| |
Collapse
|
177
|
Can AT, Mitchell JS, Dutton M, Bennett M, Hermens DF, Lagopoulos J. Insights into the neurobiology of suicidality: explicating the role of glutamatergic systems through the lens of ketamine. Psychiatry Clin Neurosci 2023; 77:513-529. [PMID: 37329495 DOI: 10.1111/pcn.13572] [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: 01/19/2023] [Revised: 05/16/2023] [Accepted: 06/08/2023] [Indexed: 06/19/2023]
Abstract
Suicidality is a prevalent mental health condition, and managing suicidal patients is one of the most challenging tasks for health care professionals due to the lack of rapid-acting, effective psychopharmacological treatment options. According to the literature, suicide has neurobiological underpinnings that are not fully understood, and current treatments for suicidal tendencies have considerable limitations. To treat suicidality and prevent suicide, new treatments are required; to achieve this, the neurobiological processes underlying suicidal behavior must be thoroughly investigated. Although multiple neurotransmitter systems, particularly serotonergic systems, have been studied in the past, less has been reported in relation to disruptions in glutamatergic neurotransmission, neuronal plasticity, and neurogenesis that result from stress-related abnormalities of the hypothalamic-pituitary-adrenal system. Informed by the literature, which reports robust antisuicidal and antidepressive properties of subanaesthetic doses of ketamine, this review aims to provide an examination of the neurobiology of suicidality (and relevant mood disorders) with implications of pertinent animal, clinical, and postmortem studies. We discuss dysfunctions in the glutamatergic system, which may play a role in the neuropathology of suicidality and the role of ketamine in restoring synaptic connectivity at the molecular levels.
Collapse
Affiliation(s)
- Adem Tevfik Can
- Thompson Institute, University of the Sunshine Coast, Birtinya, Queensland, Australia
| | - Jules Shamus Mitchell
- Thompson Institute, University of the Sunshine Coast, Birtinya, Queensland, Australia
| | - Megan Dutton
- Thompson Institute, University of the Sunshine Coast, Birtinya, Queensland, Australia
| | - Maxwell Bennett
- Thompson Institute, University of the Sunshine Coast, Birtinya, Queensland, Australia
| | | | - Jim Lagopoulos
- Thompson Institute, University of the Sunshine Coast, Birtinya, Queensland, Australia
| |
Collapse
|
178
|
Chen Y, Yan P, Wei S, Zhu Y, Lai J, Zhou Q. Ketamine metabolite alleviates morphine withdrawal-induced anxiety via modulating nucleus accumbens parvalbumin neurons in male mice. Neurobiol Dis 2023; 186:106279. [PMID: 37661023 DOI: 10.1016/j.nbd.2023.106279] [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: 05/19/2023] [Revised: 07/20/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023] Open
Abstract
Opioid withdrawal generates extremely unpleasant physical symptoms and negative affective states. A rapid relief of opioid withdrawal-induced anxiety has obvious clinical relevance but has been rarely reported. We have shown that injection of ketamine metabolite (2R,6R)-hydroxynorketamine (HNK) leads to a rapid alleviation of anxiety-like behaviors in male mice undergoing chronic morphine withdrawal. Here we investigated the contribution of nucleus accumbens shell (sNAc) parvalbumin (PV)-neurons to this process. Chronic morphine withdrawal was associated with higher intrinsic excitability of sNAc PV-neurons via reduced voltage-dependent potassium currents. Chemogenetic inhibition of sNAc PV-neurons reversed the enhanced excitability of PV-neurons and anxiety-like behaviors in these morphine withdrawal male mice, while activation of sNAc PV-neurons induced anxiety-like behaviors in naive male mice. (2R,6R)-HNK reversed the altered potassium currents and intrinsic excitability of sNAc PV-neurons. Our findings demonstrate an important contribution of sNAc PV-neurons to modulating morphine withdrawal-induced anxiety-like behaviors and rapid relief of anxiety-like behaviors by (2R,6R)-HNK, this newly identified target may have therapeutic potentials in treating opioid addiction and anxiety disorders.
Collapse
Affiliation(s)
- Yuanyuan Chen
- College of Forensic Science, Xi'an Jiaotong University, Xi'an, China; School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Peng Yan
- College of Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Shuguang Wei
- College of Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Yongsheng Zhu
- College of Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Jianghua Lai
- College of Forensic Science, Xi'an Jiaotong University, Xi'an, China.
| | - Qiang Zhou
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China.
| |
Collapse
|
179
|
Xie Z, Xie H, Peng X, Hu J, Chen L, Li X, Qi H, Zeng J, Zeng N. The antidepressant-like effects of Danzhi Xiaoyao San and its active ingredients. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:155015. [PMID: 37597362 DOI: 10.1016/j.phymed.2023.155015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/08/2023] [Accepted: 08/06/2023] [Indexed: 08/21/2023]
Abstract
BACKGROUND Depression is a severe mental illness that endangers human health. Depressed individuals are prone to sleep less and to the loss of appetite for food; their thinking and cognition processes, as well as mood, may even be affected. Danzhi Xiaoyao San (DXS), documented in the Internal Medicine Summary, has been used for hundreds of years in China and is widely applied traditionally to treat liver qi stagnation, liver and spleen blood deficiency, menstrual disorders, and spontaneous and night sweating. DXS can also clear heat and drain the liver. Presently, it is used frequently in the treatment of depression based on its ability to clear the liver and alleviate depression. PURPOSE To summarize clinical and preclinical studies on the antidepressant-like effects of DXS, understand the material basis and mechanisms of these effects, and offer new suggestions and methods for the clinical treatment of depression. METHODS "Danzhi Xiaoyao", "Danzhixiaoyao", "Xiaoyao", "depression" and active ingredients were entered as keywords in PubMed, Google Scholar, CNKI and WANFANG DATA databases in the search for material on DXS and its active ingredients. The PRISMA guidelines were followed in this review process. RESULTS Per clinical reports, DXS has a therapeutic effect on patients with depression but few side effects. DXS and its active ingredients allegedly produce their neuroprotective antidepressant-like effects by modulating monoamine neurotransmitter levels, inhibiting the hypothalamic-pituitary-adrenal (HPA) axis hyperfunction, reducing neuroinflammation and increasing neurotrophic factors. CONCLUSION Overall, DXS influences multiple potential mechanisms to exert its antidepressant-like effects thanks to its multicomponent character. Because depression is not caused by a single mechanism, probing the antidepressant-like effects of DXS could further help understand the pathogenesis of depression and discover new antidepressant drugs.
Collapse
Affiliation(s)
- Zhiqiang Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Hongxiao Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Xi Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Jingwen Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Li Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China; Department of Pharmacy, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Xiangyu Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Hu Qi
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Jiuseng Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China.
| | - Nan Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China.
| |
Collapse
|
180
|
Chaki S, Watanabe M. mGlu2/3 receptor antagonists for depression: overview of underlying mechanisms and clinical development. Eur Arch Psychiatry Clin Neurosci 2023; 273:1451-1462. [PMID: 36715750 DOI: 10.1007/s00406-023-01561-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/17/2023] [Indexed: 01/31/2023]
Abstract
Triggered by the ground-breaking finding that ketamine exerts robust and rapid-acting antidepressant effects in patients with treatment-resistant depression, glutamatergic systems have attracted attention as targets for the development of novel antidepressants. Among glutamatergic systems, group II metabotropic glutamate (mGlu) receptors, consisting of mGlu2 and mGlu3 receptors, are of interest because of their modulatory roles in glutamatergic transmission. Accumulating evidence has indicated that mGlu2/3 receptor antagonists have antidepressant-like effects in rodent models that mirror those of ketamine and that mGlu2/3 receptor antagonists also share underlying mechanisms with ketamine that are responsible for these antidepressant-like actions. Importantly, contrary to their antidepressant-like profile, preclinical studies have revealed that mGlu2/3 receptor antagonists are devoid of ketamine-like adverse effects, such as psychotomimetic-like behavior, abuse potential and neurotoxicity. Despite some discouraging results for an mGlu2/3 receptor antagonist decoglurant (classified as a negative allosteric modulator [NAM]) in patients with major depressive disorder, clinical trials of two mGlu2/3 receptor antagonists, a phase 2 trial of TS-161 (an orthosteric antagonist) and a phase 1 trial of DSP-3456 (a NAM), are presently on-going. mGlu2/3 receptors still hold promise for the development of safer and more efficacious antidepressants.
Collapse
Affiliation(s)
- Shigeyuki Chaki
- Research Headquarters, Taisho Pharmaceutical Co., Ltd, 1-403 Yoshino-cho, Kita-ku, Saitama, Saitama, 331-9530, Japan.
| | - Mai Watanabe
- Taisho Pharmaceutical R&D Inc, 350 Mt. Kemble Avenue, Morristown, NJ, 07960, USA
| |
Collapse
|
181
|
Medeiros GC, Matheson M, Demo I, Reid MJ, Matheson S, Twose C, Smith GS, Gould TD, Zarate CA, Barrett FS, Goes FS. Brain-based correlates of antidepressant response to ketamine: a comprehensive systematic review of neuroimaging studies. Lancet Psychiatry 2023; 10:790-800. [PMID: 37625426 PMCID: PMC11534374 DOI: 10.1016/s2215-0366(23)00183-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/28/2023] [Accepted: 05/12/2023] [Indexed: 08/27/2023]
Abstract
Ketamine is an effective antidepressant, but there is substantial variability in patient response and the precise mechanism of action is unclear. Neuroimaging can provide predictive and mechanistic insights, but findings are limited by small sample sizes. This systematic review covers neuroimaging studies investigating baseline (pre-treatment) and longitudinal (post-treatment) biomarkers of responses to ketamine. All modalities were included. We performed searches of five electronic databases (from inception to April 26, 2022). 69 studies were included (with 1751 participants). There was substantial methodological heterogeneity and no well replicated biomarker. However, we found convergence across some significant results, particularly in longitudinal biomarkers. Response to ketamine was associated with post-treatment increases in gamma power in frontoparietal regions in electrophysiological studies, post-treatment increases in functional connectivity within the prefrontal cortex, and post-treatment increases in the functional activation of the striatum. Although a well replicated neuroimaging biomarker of ketamine response was not identified, there are biomarkers that warrant further investigation.
Collapse
Affiliation(s)
- Gustavo C Medeiros
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Malcolm Matheson
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Isabella Demo
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew J Reid
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Claire Twose
- Welch Medical Library, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gwenn S Smith
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA; Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, NIMH-NIH, Bethesda, MD, USA
| | - Frederick S Barrett
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neuroscience, Department of Psychological and Brain Sciences, and Center for Psychedelic and Consciousness Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fernando S Goes
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
182
|
Kaur T, Sharma K, Groban L. Subanesthetic Ketamine Infusion Reducing Symptoms of Depression in a Patient With End-Stage Heart Failure Enrolled in Hospice Care: A Case Report. J Palliat Med 2023; 26:1435-1438. [PMID: 37327367 DOI: 10.1089/jpm.2022.0430] [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] [Indexed: 06/18/2023] Open
Abstract
Background: The development of major depressive disorder in patients at end of life often goes undiagnosed, as it is difficult to distinguish from preparatory grief and/or hypoactive delirium in this unique patient population. If this preliminary barrier of appropriate diagnosis is overcome, it can be quite difficult to properly select and adjust pharmacological therapy. Many well-established antidepressants take four to five weeks for maximal effectiveness (which may be far too long of a titration period for patients at end of life), have various contraindications to patients' comorbid chronic conditions (particularly patients with cardiovascular disease), or may simply be ineffective. Case: We present a case report of severe treatment-resistant depression in an end-stage heart failure patient enrolled in hospice care. Discussion: We discuss the potential use of a single low-dose intravenous racemic ketamine infusion to reduce end-of-life suffering related to depression, despite the theoretical contraindication of ketamine use in such patients, in part, due to its sympathomimetic secondary effect.
Collapse
Affiliation(s)
- Tejaspreet Kaur
- Hospice and Palliative Medicine Department, Advocate Aurora Health, Milwaukee, Wisconsin, USA
- Zilber Family Hospice of Advocate Aurora Health, Milwaukee, Wisconsin, USA
| | - Kavita Sharma
- Hospice and Palliative Medicine Department, Advocate Aurora Health, Milwaukee, Wisconsin, USA
- Zilber Family Hospice of Advocate Aurora Health, Milwaukee, Wisconsin, USA
| | - Leanne Groban
- Hospice and Palliative Medicine Department, Advocate Aurora Health, Milwaukee, Wisconsin, USA
- Zilber Family Hospice of Advocate Aurora Health, Milwaukee, Wisconsin, USA
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston Salem, North Carolina, USA
| |
Collapse
|
183
|
Kalkman HO. Activation of σ1-Receptors by R-Ketamine May Enhance the Antidepressant Effect of S-Ketamine. Biomedicines 2023; 11:2664. [PMID: 37893038 PMCID: PMC10604479 DOI: 10.3390/biomedicines11102664] [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: 08/29/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 10/29/2023] Open
Abstract
Ketamine is a racemic mixture composed of two enantiomers, S-ketamine and R-ketamine. In preclinical studies, both enantiomers have exhibited antidepressant effects, but these effects are attributed to distinct pharmacological activities. The S-enantiomer acts as an NMDA-channel blocker and as an opioid μ-receptor agonist, whereas the R-enantiomer binds to σ1-receptors and is believed to act as an agonist. As racemate, ketamine potentially triggers four biochemical pathways involving the AGC-kinases, PKA, Akt (PKB), PKC and RSK that ultimately lead to inhibitory phosphorylation of GSK3β in microglia. In patients with major depressive disorder, S-ketamine administered as a nasal spray has shown clear antidepressant activity. However, when compared to intravenously infused racemic ketamine, the response rate, duration of action and anti-suicidal activity of S-ketamine appear to be less pronounced. The σ1-protein interacts with μ-opioid and TrkB-receptors, whereas in preclinical experiments σ1-agonists reduce μ-receptor desensitization and improve TrkB signal transduction. TrkB activation occurs as a response to NMDA blockade. So, the σ1-activity of R-ketamine may not only enhance two pathways via which S-ketamine produces an antidepressant response, but it furthermore provides an antidepressant activity in its own right. These two factors could explain the apparently superior antidepressant effect observed with racemic ketamine compared to S-ketamine alone.
Collapse
Affiliation(s)
- Hans O Kalkman
- Retired Pharmacologist, Gänsbühlgartenweg 7, 4132 Muttenz, Switzerland
| |
Collapse
|
184
|
Fancy F, Haikazian S, Johnson DE, Chen-Li DCJ, Levinta A, Husain MI, Mansur RB, Rosenblat JD. Ketamine for bipolar depression: an updated systematic review. Ther Adv Psychopharmacol 2023; 13:20451253231202723. [PMID: 37771417 PMCID: PMC10524067 DOI: 10.1177/20451253231202723] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/31/2023] [Indexed: 09/30/2023] Open
Abstract
Background The therapeutic potential of subanesthetic doses of ketamine appears promising in unipolar depression; however, its effectiveness in treating bipolar depression (BD) remains uncertain. Objective This systematic review aimed to summarize findings on the use of ketamine for the treatment of BD by assessing its efficacy, safety, and tolerability. Design Systematic review. Methods We conducted a systematic review of studies that investigated the use of ketamine for adults with BD. We searched PubMed and Embase for relevant randomized-controlled trials, open-label trials, and retrospective chart analyses published from inception to 13 March 2023. Results Eight studies were identified [pooled n = 235; mean (SD) age: 45.55 (5.54)]. All participants who received intravenous (IV) ketamine were administered a dose of 0.5-0.75 mg/kg as an adjunctive treatment to a mood-stabilizing agent, whereas participants who received esketamine were administered a dosage ranging from 28 to 84 mg. Flexible dosing was used in real-world analyses. A total of 48% of participants receiving ketamine achieved a response (defined as ⩾50% reduction in baseline depression severity), whereas only 5% achieved a response with a placebo. Real-world studies demonstrated lower rates of response (30%) compared to the average across clinical trials (63%). Reductions in suicidal ideation were noted in some studies, although not all findings were statistically significant. Ketamine and esketamine were well tolerated in most participants; however, six participants (2% of the overall sample pool, 5 receiving ketamine) developed hypomanic/manic symptoms after infusions. Significant dissociative symptoms were observed at the 40-min mark in some trials. Conclusion Preliminary evidence suggests IV ketamine as being safe and effective for the treatment of BD. Future studies should focus on investigating the effects of repeated acute and maintenance infusions using a randomized study design.
Collapse
Affiliation(s)
- Farhan Fancy
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Sipan Haikazian
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Danica E. Johnson
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - David C. J. Chen-Li
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Anastasia Levinta
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Muhammad I. Husain
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Rodrigo B. Mansur
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Joshua D. Rosenblat
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, 399 Bathurst Street, Toronto, ON M5T 2S8, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, ON, Canada
| |
Collapse
|
185
|
Koncz S, Papp N, Pothorszki D, Bagdy G. (S)-Ketamine but Not (R)-Ketamine Shows Acute Effects on Depression-Like Behavior and Sleep-Wake Architecture in Rats. Int J Neuropsychopharmacol 2023; 26:618-626. [PMID: 37578355 PMCID: PMC10519815 DOI: 10.1093/ijnp/pyad050] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 08/09/2023] [Indexed: 08/15/2023] Open
Abstract
BACKGROUND Racemic ketamine consists of two enantiomers, namely (R)-ketamine and (S)-ketamine, with distinguishable pharmacological properties. Both enantiomers have been reported to show rapid antidepressant effects in rodents. Currently, the (S)-enantiomer has been approved for the treatment of major depression, whereas (R)-ketamine failed to show antidepressant effect in recent clinical studies. Major depressive disorder is frequently characterized by disinhibition of rapid eye movement (REM) sleep and disruption of non-REM (NREM) sleep. Racemic ketamine and most conventional antidepressants affect these parameters. However, it remains largely unknown which enantiomer is responsible for these effects. METHODS Here, we compared acute effects of the two ketamine enantiomers (15 mg/kg i.p.) on different sleep-wake stages in freely moving, EEG-equipped rats. We also evaluated the antidepressant-like activity of the enantiomers in a chronic restraint stress model of depression. RESULTS (S)-ketamine but not (R)-ketamine increased REM sleep latency and decreased REM sleep time at 2 and 3 hours, and increased electroencephalogram delta power during NREM sleep. In addition, only (S)-ketamine increased wakefulness and decreased NREM sleep in the first 2 hours. In the forced swimming test, only (S)-ketamine decreased the immobility time of chronically stressed rats. CONCLUSION Effects of the two ketamine enantiomers on rat sleep-wake architecture and behavior are markedly different when administered in the same dose. (S)-ketamine remarkably affects the sleep-wake cycle and very likely sleep-related neuroplasticity, which may be relevant for its antidepressant efficacy. Our results regarding (R)-ketamine's lack of effect on vigilance and behavior are in line with recent clinical studies.
Collapse
Affiliation(s)
- Szabolcs Koncz
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary
| | - Noémi Papp
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary
| | - Dóra Pothorszki
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary
| | - György Bagdy
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| |
Collapse
|
186
|
Salvatore SV, Lambert PM, Benz A, Rensing NR, Wong M, Zorumski CF, Mennerick S. Periodic and aperiodic changes to cortical EEG in response to pharmacological manipulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.21.558828. [PMID: 37790570 PMCID: PMC10542500 DOI: 10.1101/2023.09.21.558828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Cortical electroencephalograms (EEG) may help understanding of neuropsychiatric illness and new treatment mechanisms. The aperiodic component (1/ f ) of EEG power spectra is often treated as noise, but recent studies suggest that changes to the aperiodic exponent of power spectra may reflect changes in excitation/inhibition (E/I) balance, a concept linked to antidepressant effects, epilepsy, autism, and other clinical conditions. One confound of previous studies is behavioral state, because factors associated with behavioral state other than E/I ratio may alter EEG parameters. Thus, to test the robustness of the aperiodic exponent as a predictor of E/I ratio, we analyzed active exploration in mice using video EEG following various pharmacological manipulations with the Fitting Oscillations & One Over F (FOOOF) algorithm. We found that GABA A receptor (GABA A R) positive allosteric modulators increased the aperiodic exponent, consistent with the hypothesis that an increased exponent signals enhanced cortical inhibition, but other drugs (ketamine and GABA A R antagonists at sub-convulsive doses) did not follow the prediction. To tilt E/I ratio more selectively toward excitation, we suppressed the activity of parvalbumin (PV) interneurons with Designer Receptors Exclusively Activated by Designer Drugs (DREADDs). Contrary to our expectations and studies demonstrating increased cortical activity following PV suppression, circuit disinhibition with the DREADD increased the aperiodic exponent. We conclude that the aperiodic exponent of EEG power spectra does not yield a universally reliable marker of E/I ratio. Alternatively, the concept of E/I state may be sufficiently oversimplified that it cannot be mapped readily onto an EEG parameter. Significance StateBment Neuropsychiatric illness is widely prevalent and debilitating. Causes are not well understood, but some hypotheses point toward altered excitation/inhibition (E/I) balance. Here, we use cortical electroencephalograms (EEG) in mice, given applicability of cortical EEG across species, and evaluate the impact of validated drugs, including anxiolytics (pentobarbital and diazepam), along with novel rapid-acting antidepressants (ketamine and allopregnanolone). We focus on analyzing the aperiodic component of EEG power spectra, which may be associated with changes in E/I ratio. We show that aperiodic exponent of EEG power spectra is not a reliable marker of E/I ratio. Moreover, the concept of E/I ratio may be too broad and complex to be defined by an EEG parameter.
Collapse
|
187
|
Correia AS, Marques L, Cardoso A, Vale N. Exploring the Role of Drug Repurposing in Bridging the Hypoxia-Depression Connection. MEMBRANES 2023; 13:800. [PMID: 37755222 PMCID: PMC10537732 DOI: 10.3390/membranes13090800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 09/28/2023]
Abstract
High levels of oxidative stress are implicated in hypoxia, a physiological response to low levels of oxygen. Evidence supports a connection between this response and depression. Previous studies indicate that tryptophan hydroxylase can be negatively affected in hypoxia, impairing serotonin synthesis and downstream pathways. Some studies also hypothesize that increasing hypoxia-inducible factor-1 (HIF-1) levels may be a new therapeutic modality for depression. Hence, this study delved into the influence of hypoxia on the cellular response to drugs designed to act in depression. By the induction of hypoxia in SH-SY5Y cells through a hypoxia incubator chamber or Cobalt Chloride treatment, the effect of Mirtazapine, an antidepressant, and other drugs that interact with serotonin receptors (TCB-2, Dextromethorphan, Ketamine, Quetiapine, Scopolamine, Celecoxib, and Lamotrigine) on SH-SY5Y cellular viability and morphology was explored. The selection of drugs was initially conducted by literature search, focusing on compounds with established potential for employment in depression therapy. Subsequently, we employed in silico approaches to forecast their ability to traverse the blood-brain barrier (BBB). This step was particularly pertinent as we aimed to assess their viability for inducing potential antidepressant effects. The effect of these drugs in hypoxia under the inhibition of HIF-1 by Echinomycin was also tested. Our results revealed that all the potential repurposed drugs promoted cell viability, especially when hypoxia was chemically induced. When combined with Echinomycin, all drugs decreased cellular viability, possibly by the inability to interact with HIF-1.
Collapse
Affiliation(s)
- Ana Salomé Correia
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Lara Marques
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| | - Armando Cardoso
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
188
|
Dai B, Huo Y. A subanesthetic dose of esketamine combined with hip peripheral nerve block has good sedative and analgesic effects in elderly patients undergoing total hip arthroplasty: A randomized-controlled trial. Jt Dis Relat Surg 2023; 34:548-556. [PMID: 37750258 PMCID: PMC10546864 DOI: 10.52312/jdrs.2023.997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/16/2023] [Indexed: 09/27/2023] Open
Abstract
OBJECTIVES This study aims to observe the postoperative anesthetic effect of esketamine combined with hip capsule peripheral nerve block (PNB) in elderly patients undergoing total hip arthroplasty (THA). PATIENTS AND METHODS A total of 120 elderly patients (67 males, 53 females; mean age: 72.5±6.1 years; range, 60 to 89 years) who underwent THA between January 2020 and May 2021 were randomly divided into three groups including 40 patients in each group. The observation group (Group A) was treated with a subanesthetic dose of esketamine combined with hip capsule PNB; control group (Group B) was treated with a subanesthetic dose of esketamine combined with lumbar plexus block; and control group (Group C) was treated with a subanesthetic dose of esketamine for general anesthesia. The onset time of anesthesia, duration of block, postoperative recovery time, postoperative extubation time, mean arterial pressure (MAP), heart rate (HR) indexes, Visual Analog Scale (VAS) and Ramsay Sedation Scale (RSS) scores were recorded. RESULTS The onset time of anesthesia, duration of block, postoperative recovery time and postoperative extubation time in Group A were significantly lower than those in Groups B and C (p<0.001). Compared to Groups B and C, the MAP was higher at T1-T3 and the HR was significantly higher at T0-T1 (p<0.05) in Group A. The VAS scores after the operation in Group A were significantly lower than those in Groups B and C (p<0.001), and the RSS scores after the operation were significantly higher in this group than in Groups B and C (p<0.001). CONCLUSION A subanesthetic dose of esketamine combined with hip capsule PNB for elderly patients undergoing THA has better postoperative analgesic effects.
Collapse
Affiliation(s)
| | - Yanhua Huo
- Department of Anesthesiology, Changzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou 213000, Jiangsu, China.
| |
Collapse
|
189
|
Mitchell JS, Anijärv TE, Levenstein JL, Hermens DF, Lagopoulos J. Excitatory and inhibitory neurometabolites in anorexia nervosa: A systematic review of proton magnetic resonance spectroscopy studies. Neurosci Biobehav Rev 2023; 152:105279. [PMID: 37307945 DOI: 10.1016/j.neubiorev.2023.105279] [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: 01/25/2023] [Revised: 06/02/2023] [Accepted: 06/08/2023] [Indexed: 06/14/2023]
Abstract
The dysregulation of excitatory and inhibitory neurotransmission is considered a pathological marker of Anorexia Nervosa (AN), however, no systematic evaluation of the proton Magnetic Resonance Spectroscopy (1H-MRS) literature has been conducted to date. Accordingly, we conducted a systematic review of neurometabolite differences between individuals with AN and healthy controls (HC). A comprehensive database search (until June 2023) identified seven studies meeting inclusion criteria. Samples included adolescents and adults with similar mean age (AN: 22.20 HC: 22.60), and female percentages (AN: 98%; HC: 94%). The review found a considerable need for improving study design and the reporting of MRS sequence parameters and analysis. Reduced glutamate concentrations in the ACC and OCC, and reduced Glx concentrations in the ACC were reported by one and two studies, respectively. Lastly, only one study to date has quantified GABA concentrations, with no significant differences found. In conclusion, there is currently insufficient evidence of excitatory and inhibitory neurometabolites changes in AN. As the 1H-MRS literature in AN increases, the key questions herein proposed must be revisited.
Collapse
Affiliation(s)
- Jules S Mitchell
- Thompson Institute, University of Sunshine Coast, 12 Innovation Parkway, Birtinya, 4575 Sunshine Coast, Queensland, Australia.
| | - Toomas E Anijärv
- Thompson Institute, University of Sunshine Coast, 12 Innovation Parkway, Birtinya, 4575 Sunshine Coast, Queensland, Australia
| | - Jacob L Levenstein
- Thompson Institute, University of Sunshine Coast, 12 Innovation Parkway, Birtinya, 4575 Sunshine Coast, Queensland, Australia
| | - Daniel F Hermens
- Thompson Institute, University of Sunshine Coast, 12 Innovation Parkway, Birtinya, 4575 Sunshine Coast, Queensland, Australia
| | - Jim Lagopoulos
- Thompson Institute, University of Sunshine Coast, 12 Innovation Parkway, Birtinya, 4575 Sunshine Coast, Queensland, Australia
| |
Collapse
|
190
|
Serretti A. Anhedonia and Depressive Disorders. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2023; 21:401-409. [PMID: 37424409 PMCID: PMC10335915 DOI: 10.9758/cpn.23.1086] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/01/2023] [Indexed: 07/11/2023]
Abstract
Anhedonia is a core symptom of depression and of several psychiatric disorders. Anhedonia has however expanded from its original definition to encompass a spectrum of reward processing deficits that received much interest in the last decades. It is a relevant risk factor for possible suicidal behaviors, and that it may operate as an independent risk factor for suicidality apart from the episode severity. Anhedonia has also been linked to inflammation with a possible reciprocal deleterious effect on depression. Its neurophysiological bases mainly include alterations in striatal and prefrontal areas, with dopamine being the most involved neurotransmitter. Anhedonia is thought to have a significant genetic component and polygenic risk scores are a possible tool for predicting an individual's risk for developing anhedonia. Traditional antidepressants, such as selective serotonin reuptake inhibitors, showed a limited benefit on anhedonia, also considering their potential pro-anhedonic effect in some subjects. Other treatments may be more effective in treating anhedonia, such as agomelatine, vortioxetine, ketamine and transcranial magnetic stimulation. Psychotherapy is also widely supported, with cognitive-behavioral therapy and behavioral activation both showing benefit. In conclusion, a large body of evidence suggests that anhedonia is, at least partially, independent from depression, therefore it needs careful assessment and targeted treatment.
Collapse
Affiliation(s)
- Alessandro Serretti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
191
|
Kim J, Kim TE, Lee SH, Koo JW. The Role of Glutamate Underlying Treatment-resistant Depression. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2023; 21:429-446. [PMID: 37424412 PMCID: PMC10335903 DOI: 10.9758/cpn.22.1034] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 07/11/2023]
Abstract
The monoamine hypothesis has significantly improved our understanding of mood disorders and their treatment by linking monoaminergic abnormalities to the pathophysiology of mood disorders. Even 50 years after the monoamine hypothesis was established, some patients do not respond to treatments for depression, including selective serotonin reuptake drugs. Accumulating evidence shows that patients with treatment-resistant depression (TRD) have severe abnormalities in the neuroplasticity and neurotrophic factor pathways, indicating that different treatment approaches may be necessary. Therefore, the glutamate hypothesis is gaining attention as a novel hypothesis that can overcome monoamine restrictions. Glutamate has been linked to structural and maladaptive morphological alterations in several brain areas associated with mood disorders. Recently, ketamine, an N-methyl-D-aspartate receptor (NMDAR) antagonist, has shown efficacy in TRD treatment and has received the U.S. Food and Drug Administration approval, revitalizing psychiatry research. However, the mechanism by which ketamine improves TRD remains unclear. In this review, we re-examined the glutamate hypothesis, bringing the glutamate system onboard to join the modulation of the monoamine systems, emphasizing the most prominent ketamine antidepressant mechanisms, such as NMDAR inhibition and NMDAR disinhibition in GABAergic interneurons. Furthermore, we discuss the animal models used in preclinical studies and the sex differences in the effects of ketamine.
Collapse
Affiliation(s)
- Jeongseop Kim
- Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Korea
| | - Tae-Eun Kim
- Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Korea
| | - Seung-Hwan Lee
- Department of Psychiatry, Inje University Ilsan Paik Hospital, Goyang, Korea
| | - Ja Wook Koo
- Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Korea
| |
Collapse
|
192
|
van der Heijden AR, Houben T. Lipids in major depressive disorder: new kids on the block or old friends revisited? Front Psychiatry 2023; 14:1213011. [PMID: 37663599 PMCID: PMC10469871 DOI: 10.3389/fpsyt.2023.1213011] [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: 04/27/2023] [Accepted: 08/04/2023] [Indexed: 09/05/2023] Open
Abstract
Major depressive disorder (MDD) is a psychiatric mood disorder that results in substantial functional impairment and is characterized by symptoms such as depressed mood, diminished interest, impaired cognitive function, and vegetative symptoms such as disturbed sleep. Although the exact etiology of MDD is unclear, several underlying mechanisms (disturbances in immune response and/or stress response) have been associated with its development, with no single mechanism able to account for all aspects of the disorder. Currently, about 1 in 3 patients are resistant to current antidepressant therapies. Providing an alternative perspective on MDD could therefore pave the way for new, unexplored diagnostic and therapeutic solutions. The central nervous system harbors an enormous pool of lipids and lipid intermediates that have been linked to a plethora of its physiological functions. The aim of this review is therefore to provide an overview of the implications of lipids in MDD and highlight certain MDD-related underlying mechanisms that involve lipids and/or their intermediates. Furthermore, we will also focus on the bidirectional relationship between MDD and the lipid-related disorders obesity and type 2 diabetes.
Collapse
Affiliation(s)
| | - Tom Houben
- Department of Genetics and Cell Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, Netherlands
| |
Collapse
|
193
|
Joseph B, Nunez NA, Kung S, Vande Voort JL, Pazdernik VK, Schak KM, Boehm SM, Carpenter B, Johnson EK, Malyshev G, Smits N, Adewunmi DO, Brown SK, Singh B. Efficacy of Ketamine with and without Lamotrigine in Treatment-Resistant Depression: A Preliminary Report. Pharmaceuticals (Basel) 2023; 16:1164. [PMID: 37631079 PMCID: PMC10459873 DOI: 10.3390/ph16081164] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Intravenous (IV) ketamine and FDA-approved intranasal (IN) esketamine are increasingly used for treatment-resistant depression (TRD). Preliminary studies have suggested a synergistic effect of ketamine and lamotrigine, although the data are inconclusive. Herein, we report the response to serial ketamine/esketamine treatment among patients with TRD with or without lamotrigine therapy. In this historical cohort study, we included adult patients with TRD who received serial IV racemic ketamine (0.5 mg/kg over 40-100 min) or IN esketamine (56/84 mg) treatments. A change in depressive symptoms was assessed using the 16-item Quick Inventory of Depressive Symptomatology self-report (QIDS-SR) scale. There were no significant differences in response or remission rates among the patients on or not on lamotrigine during the ketamine/esketamine treatments. For a percent change in the QIDS-SR from baseline, no interaction was found between the lamotrigine groups and treatment number (p = 0.70), nor the overall effect of the group (p = 0.38). There was a trend towards lower dissociation (based on the CADSS score) among current lamotrigine users, especially in patients who received IV ketamine. A major limitation is the limited number of patients taking lamotrigine (n = 13). This preliminary study provides insufficient evidence that continuing lamotrigine therapy attenuates the antidepressant effect of repeated ketamine/esketamine; however, there seems to be a signal toward attenuating dissociation with lamotrigine in patients receiving serial ketamine treatments. Further observational studies or randomized controlled trials are needed to replicate these findings.
Collapse
Affiliation(s)
- Boney Joseph
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Nicolas A. Nunez
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Simon Kung
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Vanessa K. Pazdernik
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Kathryn M. Schak
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Stacey M. Boehm
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Brooke Carpenter
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Emily K. Johnson
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Grigoriy Malyshev
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Nathan Smits
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Daniel O. Adewunmi
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Sarah K. Brown
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Balwinder Singh
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
194
|
Zhang X, Asim M, Fang W, Md Monir H, Wang H, Kim K, Feng H, Wang S, Gao Q, Lai Y, He J. Cholecystokinin B receptor antagonists for the treatment of depression via blocking long-term potentiation in the basolateral amygdala. Mol Psychiatry 2023; 28:3459-3474. [PMID: 37365241 DOI: 10.1038/s41380-023-02127-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 06/01/2023] [Accepted: 06/13/2023] [Indexed: 06/28/2023]
Abstract
Depression is a common and severe mental disorder. Evidence suggested a substantial causal relationship between stressful life events and the onset of episodes of major depression. However, the stress-induced pathogenesis of depression and the related neural circuitry is poorly understood. Here, we investigated how cholecystokinin (CCK) and CCKBR in the basolateral amygdala (BLA) are implicated in stress-mediated depressive-like behavior. The BLA mediates emotional memories, and long-term potentiation (LTP) is widely considered a trace of memory. We identified that the cholecystokinin knockout (CCK-KO) mice impaired LTP in the BLA, while the application of CCK4 induced LTP after low-frequency stimulation (LFS). The entorhinal cortex (EC) CCK neurons project to the BLA and optogenetic activation of EC CCK afferents to BLA-promoted stress susceptibility through the release of CCK. We demonstrated that EC CCK neurons innervate CCKBR cells in the BLA and CCK-B receptor knockout (CCKBR-KO) mice impaired LTP in the BLA. Moreover, the CCKBR antagonists also blocked high-frequency stimulation (HFS) induced LTP formation in the BLA. Notably, CCKBR antagonists infusion into the BLA displayed an antidepressant-like effect in the chronic social defeat stress model. Together, these results indicate that CCKBR could be a potential target to treat depression.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
- Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
| | - Muhammad Asim
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
- Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
| | - Wei Fang
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
- Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
| | - Hossain Md Monir
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
- Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
| | - Huajie Wang
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
- Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
| | - Kyuhee Kim
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
- Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
| | - Hemin Feng
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
- Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Shujie Wang
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
- Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
| | - Qianqian Gao
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
- Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
| | - Yuanying Lai
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
- Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China
| | - Jufang He
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China.
- Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, 0000, Hong Kong SAR, PR China.
- City University of Hong Kong Shenzhen research institute, Shenzhen, 518507, PR China.
| |
Collapse
|
195
|
Burrows M, Kotoula V, Dipasquale O, Stringaris A, Mehta MA. Ketamine-induced changes in resting state connectivity, 2 h after the drug administration in patients with remitted depression. J Psychopharmacol 2023; 37:784-794. [PMID: 37491833 DOI: 10.1177/02698811231189432] [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] [Indexed: 07/27/2023]
Abstract
BACKGROUND Resting state connectivity studies link ketamine's antidepressant effects with normalisation of the brain connectivity changes that are observed in depression. These changes, however, usually co-occur with improvement in depressive symptoms, making it difficult to attribute these changes to ketamine's effects per se. AIMS Our aim is to examine the effects of ketamine in brain connectivity, 2 h after its administration in a cohort of volunteers with remitted depression. Any significant changes observed in this study could provide insight of ketamine's antidepressant mechanism as they are not accompanied by symptom changes. METHODS In total, 35 participants with remitted depression (21 females, mean age = 28.5 years) participated in a double-blind, placebo-controlled study of ketamine (0.5 mg/kg) or saline. Resting state scans were acquired approximately 2 h after the ketamine infusion. Brain connectivity was examined using a seed-based approach (ventral striatum, amygdala, hippocampus, posterior cingulate cortex and subgenual anterior cingulate cortex (sgACC)) and a brain network analysis (independent component analysis). RESULTS Decreased connectivity between the sgACC and the amygdala was observed approximately 2 h after the ketamine infusion, compared to placebo (pFWE < 0.05). The executive network presented with altered connectivity with different cortical and subcortical regions. Within the network, the left hippocampus and right amygdala had decreased connectivity (pFWE < 0.05). CONCLUSIONS Our findings support a model whereby ketamine would change the connectivity of brain areas and networks that are important for cognitive processing and emotional regulation. These changes could also be an indirect indicator of the plasticity changes induced by the drug.
Collapse
Affiliation(s)
- Matthew Burrows
- Centre for Neuroimaging Sciences, IoPPN, King's College London, London, UK
| | - Vasileia Kotoula
- Experimental Therapeutics and Pathophysiology Branch, NIMH, Bethesda, MA, USA
| | - Ottavia Dipasquale
- Centre for Neuroimaging Sciences, IoPPN, King's College London, London, UK
| | - Argyris Stringaris
- Division of Psychiatry and Department of Clinical, Educational & Health Psychology, UCL, London, UK
- First Department of Psychiatry, National and Kapodistrian University of Athens, Athens, Greece
| | - Mitul A Mehta
- Centre for Neuroimaging Sciences, IoPPN, King's College London, London, UK
| |
Collapse
|
196
|
Johnston JN, Allen J, Shkolnikov I, Sanchez-Lafuente CL, Reive BS, Scheil K, Liang S, Christie BR, Kalynchuk LE, Caruncho HJ. Reelin Rescues Behavioral, Electrophysiological, and Molecular Metrics of a Chronic Stress Phenotype in a Similar Manner to Ketamine. eNeuro 2023; 10:ENEURO.0106-23.2023. [PMID: 37550058 PMCID: PMC10431216 DOI: 10.1523/eneuro.0106-23.2023] [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: 03/27/2023] [Revised: 05/24/2023] [Accepted: 06/20/2023] [Indexed: 08/09/2023] Open
Abstract
Over the past decade, ketamine, an NMDA receptor antagonist, has demonstrated fast-acting antidepressant effects previously unseen with monoaminergic-based therapeutics. Concerns regarding psychotomimetic effects limit the use of ketamine for certain patient populations. Reelin, an extracellular matrix glycoprotein, has shown promise as a putative fast-acting antidepressant in a model of chronic stress. However, research has not yet demonstrated the changes that occur rapidly after peripheral reelin administration. To address this key gap in knowledge, male Long-Evans rats underwent a chronic corticosterone (CORT; or vehicle) paradigm (40 mg/kg, 21 d). On day 21, rats were then administered an acute dose of ketamine (10 mg/kg, i.p.), reelin (3 µg, i.v.), or vehicle. Twenty-four hours after administration, rats underwent behavioral or in vivo electrophysiological testing before killing. Immunohistochemistry was used to confirm changes in hippocampal reelin immunoreactivity. Lastly, the hippocampus was microdissected from fresh tissue to ascertain whole cell and synaptic-specific changes in protein expression through Western blotting. Chronic corticosterone induced a chronic stress phenotype in the forced swim test and sucrose preference test (SPT). Both reelin and ketamine rescued immobility and swimming, however reelin alone rescued latency to immobility. In vivo electrophysiology revealed decreases in hippocampal long-term potentiation (LTP) after chronic stress which was increased significantly by both ketamine and reelin. Reelin immunoreactivity in the dentate gyrus paralleled the behavioral and electrophysiological findings, but no significant changes were observed in synaptic-level protein expression. This exploratory research supports the putative rapid-acting antidepressant effects of an acute dose of reelin across behavioral, electrophysiological, and molecular measures.
Collapse
Affiliation(s)
- Jenessa N Johnston
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Josh Allen
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Irene Shkolnikov
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Carla L Sanchez-Lafuente
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Brady S Reive
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Kaylene Scheil
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Stanley Liang
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Brian R Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Lisa E Kalynchuk
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Hector J Caruncho
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| |
Collapse
|
197
|
Altê GA, Rodrigues ALS. Exploring the Molecular Targets for the Antidepressant and Antisuicidal Effects of Ketamine Enantiomers by Using Network Pharmacology and Molecular Docking. Pharmaceuticals (Basel) 2023; 16:1013. [PMID: 37513925 PMCID: PMC10383558 DOI: 10.3390/ph16071013] [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: 05/29/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Ketamine, a racemic mixture of esketamine (S-ketamine) and arketamine (R-ketamine), has received particular attention for its rapid antidepressant and antisuicidal effects. NMDA receptor inhibition has been indicated as one of the main mechanisms of action of the racemic mixture, but other pharmacological targets have also been proposed. This study aimed to explore the possible multiple targets of ketamine enantiomers related to their antidepressant and antisuicidal effects. To this end, targets were predicted using Swiss Target Prediction software for each ketamine enantiomer. Targets related to depression and suicide were collected by the Gene Cards database. The intersections of targets were analyzed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Network pharmacology analysis was performed using Gene Mania and Cytoscape software. Molecular docking was used to predict the main targets of the network. The results indicated that esketamine and arketamine share some biological targets, particularly NMDA receptor and phosphodiesterases 3A, 7A, and 5A but have specific molecular targets. While esketamine is predicted to interact with the GABAergic system, arketamine may interact with macrophage migration inhibitory factor (MIF). Both ketamine enantiomers activate neuroplasticity-related signaling pathways and show addiction potential. Our results identified novel, poorly explored molecular targets that may be related to the beneficial effects of esketamine and arketamine against depression and suicide.
Collapse
Affiliation(s)
- Glorister A Altê
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis 88037-000, SC, Brazil
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis 88037-000, SC, Brazil
| |
Collapse
|
198
|
Lin R, Kos A, Lopez JP, Dine J, Fiori LM, Yang J, Ben-Efraim Y, Aouabed Z, Ibrahim P, Mitsuhashi H, Wong TP, Ibrahim EC, Belzung C, Blier P, Farzan F, Frey BN, Lam RW, Milev R, Muller DJ, Parikh SV, Soares C, Uher R, Nagy C, Mechawar N, Foster JA, Kennedy SH, Chen A, Turecki G. SNORD90 induces glutamatergic signaling following treatment with monoaminergic antidepressants. eLife 2023; 12:e85316. [PMID: 37432876 PMCID: PMC10335830 DOI: 10.7554/elife.85316] [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: 12/02/2022] [Accepted: 06/26/2023] [Indexed: 07/13/2023] Open
Abstract
Pharmacotherapies for the treatment of major depressive disorder were serendipitously discovered almost seven decades ago. From this discovery, scientists pinpointed the monoaminergic system as the primary target associated with symptom alleviation. As a result, most antidepressants have been engineered to act on the monoaminergic system more selectively, primarily on serotonin, in an effort to increase treatment response and reduce unfavorable side effects. However, slow and inconsistent clinical responses continue to be observed with these available treatments. Recent findings point to the glutamatergic system as a target for rapid acting antidepressants. Investigating different cohorts of depressed individuals treated with serotonergic and other monoaminergic antidepressants, we found that the expression of a small nucleolar RNA, SNORD90, was elevated following treatment response. When we increased Snord90 levels in the mouse anterior cingulate cortex (ACC), a brain region regulating mood responses, we observed antidepressive-like behaviors. We identified neuregulin 3 (NRG3) as one of the targets of SNORD90, which we show is regulated through the accumulation of N6-methyladenosine modifications leading to YTHDF2-mediated RNA decay. We further demonstrate that a decrease in NRG3 expression resulted in increased glutamatergic release in the mouse ACC. These findings support a molecular link between monoaminergic antidepressant treatment and glutamatergic neurotransmission.
Collapse
Affiliation(s)
- Rixing Lin
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Department of Psychiatry, McGill UniversityMontrealCanada
- Integrated Program in Neuroscience, McGill UniversityMontrealCanada
| | - Aron Kos
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of PsychiatryMunichGermany
- Department of Brain Sciences, Weizmann Institute of ScienceRehovotIsrael
- Department of Molecular Neuroscience, Weizmann Institute of ScienceRehovotIsrael
| | - Juan Pablo Lopez
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of PsychiatryMunichGermany
- Department of Brain Sciences, Weizmann Institute of ScienceRehovotIsrael
- Department of Molecular Neuroscience, Weizmann Institute of ScienceRehovotIsrael
| | - Julien Dine
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of PsychiatryMunichGermany
- Department of Brain Sciences, Weizmann Institute of ScienceRehovotIsrael
- Department of Molecular Neuroscience, Weizmann Institute of ScienceRehovotIsrael
| | - Laura M Fiori
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Department of Psychiatry, McGill UniversityMontrealCanada
| | - Jennie Yang
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Department of Psychiatry, McGill UniversityMontrealCanada
| | - Yair Ben-Efraim
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of PsychiatryMunichGermany
- Department of Brain Sciences, Weizmann Institute of ScienceRehovotIsrael
- Department of Molecular Neuroscience, Weizmann Institute of ScienceRehovotIsrael
| | - Zahia Aouabed
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Department of Psychiatry, McGill UniversityMontrealCanada
| | - Pascal Ibrahim
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Department of Psychiatry, McGill UniversityMontrealCanada
- Integrated Program in Neuroscience, McGill UniversityMontrealCanada
| | - Haruka Mitsuhashi
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Department of Psychiatry, McGill UniversityMontrealCanada
- Integrated Program in Neuroscience, McGill UniversityMontrealCanada
| | - Tak Pan Wong
- Neuroscience Division, Douglas Research CentreMontrealCanada
- Department of Psychiatry, McGill UniversityMontrealCanada
| | - El Cherif Ibrahim
- Aix-Marseille Université, CNRS, INT, Institute Neuroscience TimoneMarseilleFrance
| | - Catherine Belzung
- UMR 1253, iBrain, UFR Sciences et Techniques; Parc GrandmontToursFrance
| | - Pierre Blier
- Mood Disorders Research Unit, University of Ottawa Institute of Mental Health ResearchOntarioCanada
| | | | - Benicio N Frey
- Department of Psychiatry and Behavioural Neurosciences, McMaster UniversityHamiltonCanada
- Mood Disorders Program, St. Joseph’s Healthcare HamiltonHamiltonCanada
| | - Raymond W Lam
- Department of Psychiatry, University of British ColumbiaColumbiaCanada
| | - Roumen Milev
- Departments of Psychiatry and Psychology, Queens UniversityOntarioCanada
| | - Daniel J Muller
- Department of Psychiatry, University Health Network, Krembil Research Institute, University of TorontoTorontoCanada
- Centre for Addiction and Mental HealthTorontoCanada
| | - Sagar V Parikh
- Department of Psychiatry, University of MichiganAnn ArborUnited States
| | - Claudio Soares
- Departments of Psychiatry and Psychology, Queens UniversityOntarioCanada
| | - Rudolf Uher
- Nova Scotia Health AuthorityHalifaxCanada
- Department of Psychiatry, Dalhousie UniversityHalifaxCanada
| | - Corina Nagy
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Department of Psychiatry, McGill UniversityMontrealCanada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Department of Psychiatry, McGill UniversityMontrealCanada
| | - Jane A Foster
- Department of Psychiatry and Behavioural Neurosciences, McMaster UniversityHamiltonCanada
- Mood Disorders Program, St. Joseph’s Healthcare HamiltonHamiltonCanada
- Department of Psychiatry, University Health Network, Krembil Research Institute, University of TorontoTorontoCanada
| | - Sidney H Kennedy
- Department of Psychiatry, University Health Network, Krembil Research Institute, University of TorontoTorontoCanada
- St Michael’s Hospital, Li Ka Shing Knowledge Institute, Centre for Depression and Suicide StudiesTorontoCanada
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of PsychiatryMunichGermany
- Department of Brain Sciences, Weizmann Institute of ScienceRehovotIsrael
- Department of Molecular Neuroscience, Weizmann Institute of ScienceRehovotIsrael
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Department of Psychiatry, McGill UniversityMontrealCanada
| |
Collapse
|
199
|
Popov MY, Lepik OV, Kozlovskii VL, Popov YV. Pharmacological strategies for appetite modulation in eating disorders: a narrative review. CONSORTIUM PSYCHIATRICUM 2023; 4:79-90. [PMID: 38250648 PMCID: PMC10795951 DOI: 10.17816/cp6150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/15/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND A substantial increase in the prevalence of eating disorders has been noticed over the past decades. Priority in the treatment of eating disorders is justifiably given to psychosocial interventions. However, it is also well known that centrally acting drugs can significantly affect appetite and food consumption. AIM To narratively review the available neurobiological data on the mechanisms of central regulation of eating behavior as a rationale to summarize pharmacological strategies for appetite modulation in eating disorders. METHODS The authors have carried out a narrative review of scientific papers published from January 2013 to March 2023 in the PubMed and Web of Science electronic databases. Studies were considered eligible if they included data on the neurobiological mechanisms of appetite regulation or the results of clinical trials of centrally acting drugs in eating disorders. Relevant studies were included regardless of their design. Descriptive analysis was used to summarize the obtained data. RESULTS The review included 51 studies. The available neurobiological and clinical data allowed us to identify the following pharmacological strategies for appetite modulation in eating disorders: serotonergic, catecholaminergic, amino acidergic and peptidergic. However, implementation of these data into clinical practice difficult due to an insufficient number of good-quality studies, which is particularly relevant for adolescents as there is a research gap in this population. CONCLUSION The progress in neurobiological understanding of the mechanisms of central regulation of appetite opens opportunities for new pharmacotherapeutic approaches aimed at changing the patterns of eating behavior. Obviously, treatment of eating disorders is a much broader problem and cannot be reduced to the correction of eating patterns. Nevertheless, at certain stages of treatment, drug-induced modulation of appetite can play an important role among multi-targeted biological and psychosocial interventions. Translation of neurobiological data into clinical practice requires a large number of clinical studies to confirm the long-term efficacy and safety of pharmacotherapeutic approaches and to develop personalized algorithms for the treatment of various forms of eating disorders in different age groups.
Collapse
Affiliation(s)
- Mikhail Y. Popov
- V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology
| | - Olga V. Lepik
- V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology
| | | | - Yuri V. Popov
- V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology
| |
Collapse
|
200
|
Begega A, Jove CI, López M, Moreno RD. Impact of environmental enrichment on the GABAergic neurons and glucocorticoid receptors in the hippocampus and nucleus accumbens of Wistar rats: pro-resilient effects. Brain Res Bull 2023; 200:110699. [PMID: 37406885 DOI: 10.1016/j.brainresbull.2023.110699] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/28/2023] [Accepted: 07/02/2023] [Indexed: 07/07/2023]
Abstract
The unpredictable chronic mild stress (UCMS) model has been used to induce depressive-like symptoms in animal models. Our work aims to evaluate the impact of environmental enrichment on male Wistar rats in an animal model for depression. For this purpose, we aim to assess changes in GR and GABAergic (PV+) density in cerebral regions related to cognitive-affective processes associated with depressive disorder, such as the dorsal- ventral hippocampus and accumbens nuclei. Three groups of rats were used: UCMs (unpredictable chronic mild stress), EE+ UCMs (enrichment + stress) and CONT (behavioral tests only). Hedonic responses elicited by sucrose solution were examined by licking behavior analysis; the anxiety level was evaluated using the elevated zero maze and the forced swimming (passive coping) tests. The environmental enrichment reduced the effects of chronic stress, promoting greater resilience. Thus, the UCMs group showed an anhedonia response, more anxiety and immobility behavior than either the control or the EE+ UCMs groups. Regarding immunochemistry results, there was a reduction in GABAergic activity coupled with increased activation of GR in UCMs in the dorsal hippocampus, but there were no differences between groups in the ventral hippocampus. These results suggest environmental enrichment could enhance greater resilience, reducing the vulnerability of the subjects to develop disorders such as depression and anxiety.
Collapse
Affiliation(s)
- Azucena Begega
- Laboratory of Neuroscience. Faculty of Psychology. Plaza Feijoo s/n Oviedo, 33003. Principado de Asturias, Spain; Institute of Neuroscience of Principado Asturias, INEUROPA. Plaza Feijoo s/n Oviedo, 33003. Principado de Asturias, Spain.
| | - Claudia I Jove
- Laboratory of Neuroscience. Faculty of Psychology. Plaza Feijoo s/n Oviedo, 33003. Principado de Asturias, Spain
| | - Matías López
- Institute of Neuroscience of Principado Asturias, INEUROPA. Plaza Feijoo s/n Oviedo, 33003. Principado de Asturias, Spain; Basic Psychology Area. Faculty of Psychology. Plaza Feijoo s/n Oviedo, 33003. Principado de Asturias, Spain
| | - Román-Darío Moreno
- Faculty of Education and Psychology. University Francisco de Vitoria, Pozuelo de Alarcón, 28223. Madrid, Spain
| |
Collapse
|