1
|
Riggs LM, Aronson S, Mou TCM, Pereira EFR, Thompson SM, Gould TD. Bioactive ketamine metabolite exerts in vivo neuroplastogenic effects to improve hippocampal function in a treatment-resistant depression model. Cell Rep 2025; 44:115743. [PMID: 40408248 DOI: 10.1016/j.celrep.2025.115743] [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: 02/04/2025] [Revised: 04/04/2025] [Accepted: 05/05/2025] [Indexed: 05/25/2025] Open
Abstract
An acute increase in excitatory synaptic transmission contributes to the rapid antidepressant actions of neuroplastogens, including ketamine and its bioactive metabolite, (2R,6R)-hydroxynorketamine (HNK). It is hypothesized that drug-induced metaplastic changes in synaptic strength account for therapeutically relevant behavioral adaptations in vivo. Using the plasticity-deficient Wistar Kyoto model of treatment-resistant depression, we demonstrate that (2R,6R)-HNK potentiates glutamatergic transmission, promotes synaptic strength, restores long-term potentiation (LTP), and reverses deficits in hippocampal-dependent synaptic activity and behavior. (2R,6R)-HNK selectively potentiated CA1 pyramidal neuron activity during novelty exploration and restored Schaffer collateral-dependent spatial recognition memory. Prior experience with spatial learning partially occluded LTP in control rats, an effect mimicked in LTP-impaired rats in which spatial learning deficits were reversed by (2R,6R)-HNK. These findings demonstrate that (2R,6R)-HNK exerts rapid neuroplastogenic effects in vivo, which improve cognitive function and promote adaptive changes in synaptic strength at functionally impaired synapses.
Collapse
Affiliation(s)
- Lace M Riggs
- Program in Neuroscience and Training Program in Integrative Membrane Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | - Ta-Chung M Mou
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Edna F R Pereira
- Department of Pharmacology and Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Scott M Thompson
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Pharmacology and Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Pharmacology and Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Veterans Affairs Maryland Health Care System, Baltimore, MD 21201, USA.
| |
Collapse
|
2
|
Brown KA, Ajibola MI, Gould TD. Rapid hippocampal synaptic potentiation induced by ketamine metabolite (2R,6R)-hydroxynorketamine persistently primes synaptic plasticity. Neuropsychopharmacology 2025; 50:928-940. [PMID: 40097740 PMCID: PMC12032166 DOI: 10.1038/s41386-025-02085-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/20/2025] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
The pharmacologically active (R,S)-ketamine (ketamine) metabolite (2 R,6 R)-hydroxynorketamine (HNK) maintains ketamine's preclinical antidepressant profile without adverse effects. While hypotheses have been proposed to explain how ketamine and its metabolites initiate their antidepressant-relevant effects, it remains unclear how sustained therapeutic actions arise following drug elimination. To distinguish the physiological mechanisms involved in the rapid from sustained actions of HNK, we utilized extracellular electrophysiology combined with pharmacology to develop an in vitro hippocampal slice incubation model that exhibited pharmacological fidelity to the 1) rapid synaptic potentiation induced by HNK at the Schaffer collateral-CA1 (SC-CA1) synapse during bath-application to slices collected from mice, and 2) maintenance of metaplastic (priming) activity that enhanced N-methyl-D-aspartate receptor (NMDAR) activation-dependent long-term potentiation (LTP) hours after in vivo dosing. We used this model to reveal novel mechanisms engaged in HNK's temporally-sensitive antidepressant-relevant synaptic actions, finding that the induction of synaptic potentiation by HNK did not require NMDAR activity, but NMDAR activity was necessary to maintain synaptic priming. HNK required protein kinase A (PKA) activity to rapidly potentiate SC-CA1 neurotransmission to facilitate synaptic priming that persistently promoted LTP formation. HNK's rapid actions were blocked by inhibitors of adenylyl cyclase 1 (AC1), but not an AC5 inhibitor. We conclude that HNK rapidly potentiates SC-CA1 synaptic efficacy, which then stimulates priming mechanisms that persistently favor plasticity. Targeting such priming mechanisms may be an effective antidepressant strategy, and our incubation model may aid in revealing novel pharmacological targets.
Collapse
Affiliation(s)
- Kyle A Brown
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA
| | - Musa I Ajibola
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA
| | - Todd D Gould
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA.
- Department of Pharmacology and Physiology, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA.
- Department of Neurobiology, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA.
- Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA.
| |
Collapse
|
3
|
Koutrouli I, Mazochová K, Horsley RR. The antidepressant potential of (2R,6R)-hydroxynorketamine: A detailed review of pre-clinical findings. Eur J Pharmacol 2025; 999:177604. [PMID: 40209847 DOI: 10.1016/j.ejphar.2025.177604] [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: 10/04/2024] [Revised: 04/05/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
Depression affects hundreds of millions globally, and in 2019, esketamine, an S-enantiomer of ketamine, was approved for treatment-resistant depression (TRD). While effective, esketamine carries risks, including abuse potential and adverse effects even at low doses. As a result, ketamine's metabolite, (2R,6R)-hydroxynorketamine ((2R,6R)-HNK), has garnered attention for its potential antidepressant effects without these drawbacks. This selective review evaluates preclinical behavioral evidence for (2R,6R)-HNK's antidepressant properties, focusing on rodent studies that used established depression models. Results showed that (2R,6R)-HNK reduced behavioral despair, anhedonia, anxiety, and social avoidance in both stressed and non-stressed rodents. Antidepressant effects were observed at doses between 5 and 125 mg/kg, with rapid onset (30 min) and long-lasting effects (up to 21 days). However, some studies failed to demonstrate significant antidepressant effects at doses below 40 mg/kg, often in models with pre-induced depression. No significant adverse effects were reported, but data on side effects were limited. In conclusion, (2R,6R)-HNK shows promise as a next-generation antidepressant. However, further research is needed to fully understand its long-term safety and mechanisms, and to determine its advantages over existing treatments like esketamine, particularly for TRD patients.
Collapse
Affiliation(s)
- Isis Koutrouli
- Psychedelic Research Center, National Institute of Mental Health, Topolová 748, Klecany, Czech Republic; 3rd Faculty of Medicine (3.LF) Charles University, Czech Republic.
| | - Kristýna Mazochová
- Psychedelic Research Center, National Institute of Mental Health, Topolová 748, Klecany, Czech Republic
| | - Rachel R Horsley
- Psychedelic Research Center, National Institute of Mental Health, Topolová 748, Klecany, Czech Republic
| |
Collapse
|
4
|
Michael A, Onisiforou A, Georgiou P, Koumas M, Powels C, Mammadov E, Georgiou AN, Zanos P. (2R,6R)-hydroxynorketamine prevents opioid abstinence-related negative affect and stress-induced reinstatement in mice. Br J Pharmacol 2025. [PMID: 40155780 DOI: 10.1111/bph.70018] [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: 11/22/2024] [Revised: 01/09/2025] [Accepted: 02/05/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND AND PURPOSE Opioid use disorder (OUD) is a pressing public health concern marked by frequent relapse during periods of abstinence, perpetuated by negative affective states. Classical antidepressants or the currently prescribed opioid pharmacotherapies have limited efficacy to reverse the negative affect or prevent relapse. EXPERIMENTAL APPROACH Using mouse models, we investigated the effects of ketamine's metabolite (2R,6R)-hydroxynorketamine (HNK) on reversing conditioning to sub-effective doses of morphine in stress-susceptible mice, preventing conditioned-place aversion and alleviating acute somatic abstinence symptoms in opioid-dependent mice. Additionally, we evaluated its effects on anhedonia, anxiety-like behaviours and cognitive impairment during protracted opioid abstinence, while mechanistic studies examined cortical EEG oscillations and synaptic plasticity markers. KEY RESULTS (2R,6R)-HNK reversed conditioning to sub-effective doses of morphine in stress-susceptible mice and prevented conditioned-place aversion and acute somatic abstinence symptoms in opioid-dependent mice. In addition, (2R,6R)-HNK reversed anhedonia, anxiety-like behaviours and cognitive impairment emerging during protracted opioid abstinence plausibly via a restoration of impaired cortical high-frequency EEG oscillations, through a GluN2A-NMDA receptor-dependent mechanism. Notably, (2R,6R)-HNK facilitated the extinction of opioid conditioning, prevented stress-induced reinstatement of opioid-seeking behaviours and reduced the propensity for enhanced morphine self-consumption in mice previously exposed to opioids. CONCLUSIONS AND IMPLICATIONS These findings emphasize the therapeutic potential of (2R,6R)-HNK, which is currently in Phase II clinical trials, in addressing stress-related opioid responses. Reducing the time and cost required for development of new medications for the treatment of OUDs via drug repurposing is critical due to the opioid crisis we currently face.
Collapse
Affiliation(s)
- Andria Michael
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Center for Applied Neuroscience (CAN), University of Cyprus, Nicosia, Cyprus
| | - Anna Onisiforou
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Center for Applied Neuroscience (CAN), University of Cyprus, Nicosia, Cyprus
| | - Polymnia Georgiou
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, USA
| | - Morfeas Koumas
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | - Chris Powels
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Elmar Mammadov
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Andrea N Georgiou
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Center for Applied Neuroscience (CAN), University of Cyprus, Nicosia, Cyprus
| | - Panos Zanos
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Center for Applied Neuroscience (CAN), University of Cyprus, Nicosia, Cyprus
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| |
Collapse
|
5
|
Fogaça MV, Daher F, Picciotto MR. Effects of ketamine on GABAergic and glutamatergic activity in the mPFC: biphasic recruitment of GABA function in antidepressant-like responses. Neuropsychopharmacology 2025; 50:673-684. [PMID: 39390105 PMCID: PMC11845475 DOI: 10.1038/s41386-024-02002-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 10/12/2024]
Abstract
Major depressive disorder (MDD) is associated with disruptions in glutamatergic and GABAergic activity in the medial prefrontal cortex (mPFC), leading to altered synaptic formation and function. Low doses of ketamine rapidly rescue these deficits, inducing fast and sustained antidepressant effects. While it is suggested that ketamine produces a rapid glutamatergic enhancement in the mPFC, the temporal dynamics and the involvement of GABA interneurons in its sustained effects remain unclear. Using simultaneous photometry recordings of calcium activity in mPFC pyramidal and GABA neurons, as well as chemogenetic approaches in Gad1-Cre mice, we explored the hypothesis that initial effects of ketamine on glutamate signaling trigger subsequent enhancement of GABAergic responses, contributing to its sustained antidepressant responses. Calcium recordings revealed a biphasic effect of ketamine on activity of mPFC GABA neurons, characterized by an initial transient decrease (phase 1, <30 min) followed by an increase (phase 2, >60 min), in parallel with a transient increase in excitation/inhibition levels (10 min) and lasting enhancement of glutamatergic activity (30-120 min). Previous administration of ketamine enhanced GABA neuron activity during the sucrose splash test (SUST) and novelty suppressed feeding test (NSFT), 24 h and 72 h post-treatment, respectively. Chemogenetic inhibition of GABA interneurons during the surge of GABAergic activity (phase 2), or immediately before the SUST or NSFT, occluded ketamine's behavioral actions. These results indicate that time-dependent modulation of GABAergic activity is required for the sustained antidepressant-like responses induced by ketamine, suggesting that approaches to enhance GABAergic plasticity and function are promising therapeutic targets for antidepressant development.
Collapse
Affiliation(s)
- Manoela V Fogaça
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA.
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT, 06519, USA.
| | - Fernanda Daher
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Marina R Picciotto
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT, 06519, USA
| |
Collapse
|
6
|
Lima da Cruz RV, Leão RN, Moulin TC. Effects of psychedelics on neurogenesis and broader neuroplasticity: a systematic review. Mol Med 2024; 30:244. [PMID: 39701927 DOI: 10.1186/s10020-024-01013-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024] Open
Abstract
In the mammalian brain, new neurons continue to be generated throughout life in a process known as adult neurogenesis. The role of adult-generated neurons has been broadly studied across laboratories, and mounting evidence suggests a strong link to the HPA axis and concomitant dysregulations in patients diagnosed with mood disorders. Psychedelic compounds, such as phenethylamines, tryptamines, cannabinoids, and a variety of ever-growing chemical categories, have emerged as therapeutic options for neuropsychiatric disorders, while numerous reports link their effects to increased adult neurogenesis. In this systematic review, we examine studies assessing neurogenesis or other neurogenesis-associated brain plasticity after psychedelic interventions and aim to provide a comprehensive picture of how this vast category of compounds regulates the generation of new neurons. We conducted a literature search on PubMed and Science Direct databases, considering all articles published until January 31, 2023, and selected articles containing both the words "neurogenesis" and "psychedelics". We analyzed experimental studies using either in vivo or in vitro models, employing classical or atypical psychedelics at all ontogenetic windows, as well as human studies referring to neurogenesis-associated plasticity. Our findings were divided into five main categories of psychedelics: CB1 agonists, NMDA antagonists, harmala alkaloids, tryptamines, and entactogens. We described the outcomes of neurogenesis assessments and investigated related results on the effects of psychedelics on brain plasticity and behavior within our sample. In summary, this review presents an extensive study into how different psychedelics may affect the birth of new neurons and other brain-related processes. Such knowledge may be valuable for future research on novel therapeutic strategies for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Rafael V Lima da Cruz
- Neurodynamics Lab, Brain Institute (ICe), Universidade Federal do Rio Grande do Norte, Natal, Brazil.
| | - Richardson N Leão
- Neurodynamics Lab, Brain Institute (ICe), Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Thiago C Moulin
- Department of Experimental Medical Science, Lund University, Lund, Sweden.
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
7
|
Liu L, Li R, Wu L, Guan Y, Miao M, Wang Y, Li C, Wu C, Lu G, Hu X, Sun L. (2R,6R)-hydroxynorketamine alleviates PTSD-like endophenotypes by regulating the PI3K/AKT signaling pathway in rats. Pharmacol Biochem Behav 2024; 245:173891. [PMID: 39369910 DOI: 10.1016/j.pbb.2024.173891] [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: 07/30/2024] [Revised: 09/08/2024] [Accepted: 10/01/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Patients diagnosed with post-traumatic stress disorder (PTSD) mainly exhibit enduring adverse emotions, heightening susceptibility to suicidal thoughts and behaviors. Notably, metabolites of ketamine, particularly (2R,6R)-hydroxyketamine (HNK), have demonstrated favorable antidepressant properties. However, the precise mechanism through which HNK exerts its therapeutic effects on negative emotional symptoms in PTSD patients should be fully elucidated. METHODS In this investigation, a model involving a single prolonged stress and plantar shock (SPS&S) was utilized, followed by the administration of (2R, 6R)-HNK into the lateral ventricle subsequent to the recovery phase. The evaluation of PTSD-related behaviors was conducted through the open field test (OFT), elevated plus maze test (EMPT), and forced swim test (FST). The expression of phosphatidylinositol 3-kinase (PI3K)/phosphokinase B (AKT) signaling pathway in rat brain regions was analyzed using molecular biology experiments. RESULTS SPS&S rats displayed adverse emotional behaviors characterized by depression and anxiety. Treatment with (2R, 6R)-HNK enhanced exploratory behavior and reversed negative emotional behaviors. This intervention mitigated disruptions in the expression levels of PI3K/AKT signaling pathway-associated proteins in the HIP and PFC, without influencing PI3K/AKT signaling in the AMY of SPS&S rats. CONCLUSION Traumatic stress can trigger negative emotional reactions in rats, potentially involving the PI3K/AKT signaling pathway in the HIP, PFC, and AMY. The (2R, 6R)-HNK compounds have demonstrated the potential to mitigate adverse emotions in rats subjected to the SPS&S paradigm. This effect may be attributed to the modulation of the PI3K/AKT signaling pathway in the HIP, and PFC, with a particularly notable impact observed in the HIP region.
Collapse
Affiliation(s)
- Lifen Liu
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Rui Li
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Lanxia Wu
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Yubo Guan
- School of Clinical Medicine, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Miao Miao
- School of Clinical Medicine, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Yuxuan Wang
- School of Clinical Medicine, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Changjiang Li
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Chunyan Wu
- Department of Neurology, Affiliated Hospital of Shandong Second Medical University, Weifang, PR China
| | - Guohua Lu
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Xinyu Hu
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China; CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, PR China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, PR China.
| | - Lin Sun
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China; Department of Neurosurgery, Shanting District People's Hospital, Beijing Road, New Town, Zaozhuang, Shandong 277200, PR China; Management Committee of Shanting Economic Development Zone, No.37, Fuqian Road, Zaozhuang, Shandong 277200, PR China.
| |
Collapse
|
8
|
Raja SM, Guptill JT, Mack M, Peterson M, Byard S, Twieg R, Jordan L, Rich N, Castledine R, Bourne S, Wilmshurst M, Oxendine S, Avula SG, Zuleta H, Quigley P, Lawson S, McQuaker SJ, Ahmadkhaniha R, Appelbaum LG, Kowalski K, Barksdale CT, Gufford BT, Awan A, Sancho AR, Moore MC, Berrada K, Cogan GB, DeLaRosa J, Radcliffe J, Pao M, Kennedy M, Lawrence Q, Goldfeder L, Amanfo L, Zanos P, Gilbert JR, Morris PJ, Moaddel R, Gould TD, Zarate CA, Thomas CJ. A Phase 1 Assessment of the Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of (2R,6R)-Hydroxynorketamine in Healthy Volunteers. Clin Pharmacol Ther 2024; 116:1314-1324. [PMID: 39054770 PMCID: PMC11479831 DOI: 10.1002/cpt.3391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
(R,S)-Ketamine (ketamine) is a dissociative anesthetic that also possesses analgesic and antidepressant activity. Undesirable dissociative side effects and misuse potential limit expanded use of ketamine in several mental health disorders despite promising clinical activity and intensifying medical need. (2R,6R)-Hydroxynorketamine (RR-HNK) is a metabolite of ketamine that lacks anesthetic and dissociative activity but maintains antidepressant and analgesic activity in multiple preclinical models. To enable future assessments in selected human indications, we report the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of RR-HNK in a Phase 1 study in healthy volunteers (NCT04711005). A six-level single-ascending dose (SAD) (0.1-4 mg/kg) and a two-level multiple ascending dose (MAD) (1 and 2 mg/kg) study was performed using a 40-minute IV administration emulating the common practice for ketamine administration for depression. Safety assessments showed RR-HNK possessed a minimal adverse event profile and no serious adverse events at all doses examined. Evaluations of dissociation and sedation demonstrated that RR-HNK did not possess anesthetic or dissociative characteristics in the doses examined. RR-HNK PK parameters were measured in both the SAD and MAD studies and exhibited dose-proportional increases in exposure. Quantitative electroencephalography (EEG) measurements collected as a PD parameter based on preclinical findings and ketamine's established effect on gamma-power oscillations demonstrated increases of gamma power in some participants at the lower/mid-range doses examined. Cerebrospinal fluid examination confirmed RR-HNK exposure within the central nervous system (CNS). Collectively, these data demonstrate RR-HNK is well tolerated with an acceptable PK profile and promising PD outcomes to support the progression into Phase 2.
Collapse
Affiliation(s)
- Shruti M. Raja
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jeffrey T. Guptill
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
- Argenx BV, 9052 Gent, Belgium
| | - Michelle Mack
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | | | - Stephen Byard
- Quotient Sciences, Alnwick, Northumberland, NE66 2DH, England
| | - Robert Twieg
- Labcorp Bioanalytical Services, Indianapolis, IN, 46214, USA
| | - Lynn Jordan
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | | | | | - Samuel Bourne
- Quotient Sciences, Alnwick, Northumberland, NE66 2DH, England
| | | | - Sarah Oxendine
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | | | - Helen Zuleta
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Paul Quigley
- Quotient Sciences, Alnwick, Northumberland, NE66 2DH, England
| | - Sheila Lawson
- Quotient Sciences, Alnwick, Northumberland, NE66 2DH, England
| | | | - Reza Ahmadkhaniha
- National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, 22124, USA
| | - Lawrence G. Appelbaum
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kevin Kowalski
- Labcorp Bioanalytical Services, Indianapolis, IN, 46214, USA
| | | | - Brandon T. Gufford
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Asaad Awan
- Office of the Director, Intramural Research Program, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Alfredo R. Sancho
- Office of the Director, Intramural Research Program, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Max C. Moore
- Drug Discovery and Development Program, Frederick National Laboratory, Fredrick, MD, 21701, USA
| | - Karim Berrada
- Drug Discovery and Development Program, Frederick National Laboratory, Fredrick, MD, 21701, USA
| | - Gregory B. Cogan
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jesse DeLaRosa
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jeanne Radcliffe
- Office of the Clinical Director, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Maryland Pao
- Office of the Clinical Director, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | | | - Lisa Goldfeder
- Office of the Director, Intramural Research Program, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Leslie Amanfo
- Office of the Director, Intramural Research Program, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Panos Zanos
- Department of Psychology, University of Cyprus, Nicosia, 2109, Cyprus
| | - Jessica R Gilbert
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Patrick J. Morris
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, 20850, USA
| | - Ruin Moaddel
- National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, 22124, USA
| | - Todd D. Gould
- Departments of Psychiatry, Pharmacology, and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Baltimore Veterans Affairs Medical Center, Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA
| | - Carlos A. Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Craig J. Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, 20850, USA
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
9
|
Loan Nguyen TM, Guilloux JP, Defaix C, Mendez-David I, Etting I, Alvarez JC, McGowan JC, Highland JN, Zanos P, Lovett J, Moaddel R, Corruble E, David DJ, Gould TD, Denny CA, Gardier AM. Ketamine metabolism via hepatic CYP450 isoforms contributes to its sustained antidepressant actions. Neuropharmacology 2024; 258:110065. [PMID: 39004413 PMCID: PMC11492263 DOI: 10.1016/j.neuropharm.2024.110065] [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: 04/02/2024] [Revised: 07/03/2024] [Accepted: 07/06/2024] [Indexed: 07/16/2024]
Abstract
(R,S)-ketamine (ketamine) has rapid and sustained antidepressant (AD) efficacy at sub-anesthetic doses in depressed patients. A metabolite of ketamine, including (2R,6R)-hydroxynorketamine ((6)-HNKs) has been reported to exert antidepressant actions in rodent model of anxiety/depression. To further understand the specific role of ketamine's metabolism in the AD actions of the drug, we evaluated the effects of inhibiting hepatic cytochrome P450 enzymes on AD responses. We assessed whether pre-treatment with fluconazole (10 and 20 mg/kg, i. p.) 1 h prior to ketamine or HNKs (10 mg/kg, i. p.) administration would alter behavioral and neurochemical actions of the drugs in male BALB/cJ mice with a highly anxious phenotype. Extracellular microdialysate levels of glutamate and GABA (Gluext, GABAext) were also measured in the medial prefrontal cortex (mPFC). Pre-treatment with fluconazole altered the pharmacokinetic profile of ketamine, by increasing both plasma and brain levels of ketamine and (R,S)-norketamine, while robustly reducing those of (6)-HNKs. At 24 h post-injection (t24 h), fluconazole prevented the sustained AD-like response of ketamine responses in the forced swim test and splash test, as well as the enhanced cortical GABA levels produced by ketamine. A single (2R,6R)-HNK administration resulted in prevention of the effects of fluconazole on the antidepressant-like activity of ketamine in mice. Overall, these findings are consistent with an essential contribution of (6)-HNK to the sustained antidepressant-like effects of ketamine and suggest potential interactions between pharmacological CYPIs and ketamine during antidepressant treatment in patients.
Collapse
Affiliation(s)
- Thi Mai Loan Nguyen
- Université Paris-Saclay, Faculté de Pharmacie, Inserm UMR 1018, CESP, MOODS Team, 91400 Orsay, France
| | - Jean-Philippe Guilloux
- Université Paris-Saclay, Faculté de Pharmacie, Inserm UMR 1018, CESP, MOODS Team, 91400 Orsay, France
| | - Céline Defaix
- Université Paris-Saclay, Faculté de Pharmacie, Inserm UMR 1018, CESP, MOODS Team, 91400 Orsay, France
| | - Indira Mendez-David
- Université Paris-Saclay, Faculté de Pharmacie, Inserm UMR 1018, CESP, MOODS Team, 91400 Orsay, France
| | - Isabelle Etting
- Service de Pharmacologie-Toxicologie, Hôpital Raymond Poincaré, Groupe Hospitalier Universitaires AP-HP, Université Paris-Saclay, Inserm U-1018, CESP, MOODS Team, 92380 Garches, France
| | - Jean-Claude Alvarez
- Service de Pharmacologie-Toxicologie, Hôpital Raymond Poincaré, Groupe Hospitalier Universitaires AP-HP, Université Paris-Saclay, Inserm U-1018, CESP, MOODS Team, 92380 Garches, France
| | - Josephine C McGowan
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA; Division of Systems Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH) /New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA; Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Jaclyn N Highland
- Departments of Psychiatry, Baltimore, MD, USA; Departments of Program in Toxicology, University of Maryland School of Medicine, Baltimore, MD, USA; Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA
| | - Panos Zanos
- Departments of Psychiatry, Baltimore, MD, USA; Departments of Pharmacology, Baltimore, MD, USA; Departments of Physiology, Baltimore, MD, USA; Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA; Department of Psychology, University of Cyprus, Nicosia, 2109, Cyprus
| | - Jacqueline Lovett
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Ruin Moaddel
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Emmanuelle Corruble
- Université Paris-Saclay, Faculté de Médecine, Inserm UMR 1018, CESP, MOODS Team, 94270 Bicêtre Hospital, 94270 Le Kremin-Bicêtre, France; Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Saclay, Hôpital de Bicêtre, 94270 Le Kremlin Bicêtre, France
| | - Denis J David
- Université Paris-Saclay, Faculté de Pharmacie, Inserm UMR 1018, CESP, MOODS Team, 91400 Orsay, France
| | - Todd D Gould
- Departments of Psychiatry, Baltimore, MD, USA; Departments of Pharmacology, Baltimore, MD, USA; Departments of Physiology, Baltimore, MD, USA; Departments of Neurobiology, Baltimore, MD, USA; Departments of Program in Toxicology, University of Maryland School of Medicine, Baltimore, MD, USA; Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA; Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Christine A Denny
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA; Division of Systems Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH) /New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA
| | - Alain M Gardier
- Université Paris-Saclay, Faculté de Pharmacie, Inserm UMR 1018, CESP, MOODS Team, 91400 Orsay, France.
| |
Collapse
|
10
|
Brown KA, Ajibola MI, Gould TD. Rapid Hippocampal Synaptic Potentiation Induced by Ketamine Metabolite ( 2R , 6R )-Hydroxynorketamine Persistently Primes Synaptic Plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.619152. [PMID: 39484512 PMCID: PMC11526997 DOI: 10.1101/2024.10.18.619152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
The pharmacologically active ( R , S )-ketamine (ketamine) metabolite ( 2R , 6R )-hydroxynorketamine (HNK) maintains ketamine's preclinical antidepressant profile without adverse effects. While hypotheses have been proposed to explain how ketamine and its metabolites initiate their antidepressant-relevant effects, it remains unclear how sustained therapeutic actions arise following drug elimination. To distinguish the physiological mechanisms involved in the rapid from sustained actions of HNK, we utilized extracellular electrophysiology combined with pharmacology to develop an in vitro hippocampal slice incubation model that exhibited pharmacological fidelity to the 1) rapid synaptic potentiation induced by HNK at the Schaffer collateral-CA1 (SC-CA1) synapse during bath-application to slices collected from mice, and 2) maintenance of metaplastic (priming) activity that lowered the threshold for N- methyl-D-aspartate receptor (NMDAR) activation-dependent long-term potentiation (LTP) hours after in vivo dosing. We then used this model to reveal novel druggable mechanisms engaged in HNK's temporally-sensitive antidepressant synaptic actions, finding that the induction of synaptic potentiation by HNK did not require NMDAR activity, but NMDAR activity was necessary to maintain synaptic priming. HNK required protein kinase A (PKA) activity to rapidly potentiate SC-CA1 neurotransmission to facilitate synaptic priming that persistently promoted LTP formation. HNK's rapid actions were blocked by inhibitors of adenylyl cyclase 1 (AC1), but not an AC5 inhibitor. We conclude that HNK rapidly potentiates SC-CA1 synaptic efficacy, which then stimulates priming mechanisms that persistently favor antidepressant-relevant plasticity. Targeting such priming mechanisms may be an effective antidepressant strategy, and using approaches such as our incubation model may aid in revealing novel pharmacological targets.
Collapse
|
11
|
Freudenberg F, Reif-Leonhard C, Reif A. Advancing past ketamine: emerging glutamatergic compounds for the treatment of depression. Eur Arch Psychiatry Clin Neurosci 2024:10.1007/s00406-024-01875-z. [PMID: 39207462 DOI: 10.1007/s00406-024-01875-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
Changes in glutamatergic neuroplasticity has been proposed as one of the core mechanisms underlying the pathophysiology of depression. In consequence components of the glutamatergic synapse have been explored as potential targets for antidepressant treatment. The rapid antidepressant effect of the NMDA receptor antagonist ketamine and subsequent approval of its S-enantiomer (i.e. esketamine), have set the precedent for investigation into other glutamatergic rapid acting antidepressants (RAADs). In this review, we discuss the potential of the different glutamatergic targets for antidepressant treatment. We describe important clinical outcomes of several key molecules targeting components of the glutamatergic synapse and their applicability as RAADs. Specifically, here we focus on substances beyond (es)ketamine, for which meaningful data from clinical trials are available, including arketamine, esmethadone, nitrous oxide and other glutamate receptor modulators. Molecules only successful in preclinical settings and case reports/series are only marginally discussed. With this review, we aim underscore the critical role of glutamatergic modulation in advancing antidepressant therapy, thereby possibly enhancing clinical outcomes but also to reducing the burden of depression through faster therapeutic effects.
Collapse
Affiliation(s)
- Florian Freudenberg
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt am Main, Germany.
| | - Christine Reif-Leonhard
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt am Main, Germany
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596, Frankfurt Am Main, Germany
| |
Collapse
|
12
|
Ribeiro FC, Cozachenco D, Argyrousi EK, Staniszewski A, Wiebe S, Calixtro JD, Soares‐Neto R, Al‐Chami A, Sayegh FE, Bermudez S, Arsenault E, Cossenza M, Lacaille J, Nader K, Sun H, De Felice FG, Lourenco MV, Arancio O, Aguilar‐Valles A, Sonenberg N, Ferreira ST. The ketamine metabolite (2R,6R)-hydroxynorketamine rescues hippocampal mRNA translation, synaptic plasticity and memory in mouse models of Alzheimer's disease. Alzheimers Dement 2024; 20:5398-5410. [PMID: 38934107 PMCID: PMC11350050 DOI: 10.1002/alz.14034] [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: 12/02/2023] [Revised: 04/16/2024] [Accepted: 05/06/2024] [Indexed: 06/28/2024]
Abstract
INTRODUCTION Impaired brain protein synthesis, synaptic plasticity, and memory are major hallmarks of Alzheimer's disease (AD). The ketamine metabolite (2R,6R)-hydroxynorketamine (HNK) has been shown to modulate protein synthesis, but its effects on memory in AD models remain elusive. METHODS We investigated the effects of HNK on hippocampal protein synthesis, long-term potentiation (LTP), and memory in AD mouse models. RESULTS HNK activated extracellular signal-regulated kinase 1/2 (ERK1/2), mechanistic target of rapamycin (mTOR), and p70S6 kinase 1 (S6K1)/ribosomal protein S6 signaling pathways. Treatment with HNK rescued hippocampal LTP and memory deficits in amyloid-β oligomers (AβO)-infused mice in an ERK1/2-dependent manner. Treatment with HNK further corrected aberrant transcription, LTP and memory in aged APP/PS1 mice. DISCUSSION Our findings demonstrate that HNK induces signaling and transcriptional responses that correct synaptic and memory deficits in AD mice. These results raise the prospect that HNK could serve as a therapeutic approach in AD. HIGHLIGHTS The ketamine metabolite HNK activates hippocampal ERK/mTOR/S6 signaling pathways. HNK corrects hippocampal synaptic and memory defects in two mouse models of AD. Rescue of synaptic and memory impairments by HNK depends on ERK signaling. HNK corrects aberrant transcriptional signatures in APP/PS1 mice.
Collapse
Affiliation(s)
- Felipe C. Ribeiro
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
| | - Danielle Cozachenco
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
| | - Elentina K. Argyrousi
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
| | - Agnieszka Staniszewski
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
| | - Shane Wiebe
- Department of BiochemistryMcGill UniversityMontrealQuebecCanada
| | - Joao D. Calixtro
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
| | - Rubens Soares‐Neto
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
| | - Aycheh Al‐Chami
- Department of NeuroscienceCarleton UniversityOttawaOntarioCanada
| | - Fatema El Sayegh
- Department of NeuroscienceCarleton UniversityOttawaOntarioCanada
| | - Sara Bermudez
- Department of BiochemistryMcGill UniversityMontrealQuebecCanada
| | - Emily Arsenault
- Department of NeuroscienceCarleton UniversityOttawaOntarioCanada
| | - Marcelo Cossenza
- Department of Physiology and Pharmacology, Fluminense Federal UniversityBiomedical InstituteNiteróiRio de JaneiroBrazil
| | - Jean‐Claude Lacaille
- Department of Neurosciences, Université de MontréalCentre for Interdisciplinary Research on Brain and Learning and Research Group on Neural Signaling and CircuitsMontrealQuebecCanada
| | - Karim Nader
- Department of PsychologyMcGill UniversityMontrealQuebecCanada
| | - Hongyu Sun
- Department of NeuroscienceCarleton UniversityOttawaOntarioCanada
| | - Fernanda G. De Felice
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
- Department of Biomedical and Molecular Sciences, Centre for Neuroscience StudiesQueen's UniversityKingstonOntarioCanada
- Department of PsychiatryQueen's UniversityKingstonOntarioCanada
- D'Or Institute for Research and EducationRio de JaneiroRio de JaneiroBrazil
| | - Mychael V. Lourenco
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
| | | | - Nahum Sonenberg
- Department of BiochemistryMcGill UniversityMontrealQuebecCanada
| | - Sergio T. Ferreira
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
- D'Or Institute for Research and EducationRio de JaneiroRio de JaneiroBrazil
- Institute of Biophysics Carlos Chagas FilhoFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
| |
Collapse
|
13
|
Fogaça MV, Daher F, Picciotto MR. Effects of ketamine on GABAergic and glutamatergic activity in the mPFC: biphasic recruitment of GABA function in antidepressant-like responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.29.605610. [PMID: 39131322 PMCID: PMC11312475 DOI: 10.1101/2024.07.29.605610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Major depressive disorder (MDD) is associated with disruptions in glutamatergic and GABAergic activity in the medial prefrontal cortex (mPFC), leading to altered synaptic formation and function. Low doses of ketamine rapidly rescue these deficits, inducing fast and sustained antidepressant effects. While it is suggested that ketamine produces a rapid glutamatergic enhancement in the mPFC, the temporal dynamics and the involvement of GABA interneurons in its sustained effects remain unclear. Using simultaneous photometry recordings of calcium activity in mPFC pyramidal and GABA neurons, as well as chemogenetic approaches in Gad1-Cre mice, we explored the hypothesis that initial effects of ketamine on glutamate signaling trigger subsequent enhancement of GABAergic responses, contributing to its sustained antidepressant responses. Calcium recordings revealed a biphasic effect of ketamine on activity of mPFC GABA neurons, characterized by an initial transient decrease (phase 1, <30 min) followed by an increase (phase 2, >60 min), in parallel with a transient increase in excitation/inhibition levels (10 min) and lasting enhancement of glutamatergic activity (30-120 min). Previous administration of ketamine enhanced GABA neuron activity during the sucrose splash test (SUST) and novelty suppressed feeding test (NSFT), 24 h and 72 h post-treatment, respectively. Chemogenetic inhibition of GABA interneurons during the surge of GABAergic activity (phase 2), or immediately before the SUST or NSFT, occluded ketamine's behavioral actions. These results indicate that time-dependent modulation of GABAergic activity is required for the sustained antidepressant-like responses induced by ketamine, suggesting that approaches to enhance GABAergic plasticity and function are promising therapeutic targets for antidepressant development.
Collapse
Affiliation(s)
- Manoela V. Fogaça
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06519, USA
| | - Fernanda Daher
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Marina R. Picciotto
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06519, USA
| |
Collapse
|
14
|
Jiang Y, Dong Y, Hu H. The N-methyl-d-aspartate receptor hypothesis of ketamine's antidepressant action: evidence and controversies. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230225. [PMID: 38853549 PMCID: PMC11343275 DOI: 10.1098/rstb.2023.0225] [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: 10/10/2023] [Revised: 12/26/2023] [Accepted: 01/02/2024] [Indexed: 06/11/2024] Open
Abstract
Substantial clinical evidence has unravelled the superior antidepressant efficacy of ketamine: in comparison to traditional antidepressants targeting the monoamine systems, ketamine, as an N-methyl-d-aspartate receptor (NMDAR) antagonist, acts much faster and more potently. Surrounding the antidepressant mechanisms of ketamine, there is ample evidence supporting an NMDAR-antagonism-based hypothesis. However, alternative arguments also exist, mostly derived from the controversial clinical results of other NMDAR inhibitors. In this article, we first summarize the historical development of the NMDAR-centred hypothesis of rapid antidepressants. We then classify different NMDAR inhibitors based on their mechanisms of inhibition and evaluate preclinical as well as clinical evidence of their antidepressant effects. Finally, we critically analyse controversies and arguments surrounding ketamine's NMDAR-dependent and NMDAR-independent antidepressant action. A better understanding of ketamine's molecular targets and antidepressant mechanisms should shed light on the future development of better treatment for depression. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.
Collapse
Affiliation(s)
- Yihao Jiang
- Department of Affiliated Mental Health Center & Hangzhou Seventh People’s Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou310058, People's Republic of China
- Nanhu Brain-Computer Interface Institute, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou311100, People's Republic of China
| | - Yiyan Dong
- Department of Affiliated Mental Health Center & Hangzhou Seventh People’s Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou310058, People's Republic of China
| | - Hailan Hu
- Department of Affiliated Mental Health Center & Hangzhou Seventh People’s Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou310058, People's Republic of China
- Nanhu Brain-Computer Interface Institute, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou311100, People's Republic of China
| |
Collapse
|
15
|
Ahuja J, Leontieva L. Ketamine Therapy in Complex Cases: A Cautionary Tale of Exacerbated Personality Traits and the Crucial Role of Comprehensive Follow-Up and Psychosocial Interventions. Case Rep Psychiatry 2024; 2024:2143372. [PMID: 38939043 PMCID: PMC11208781 DOI: 10.1155/2024/2143372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/29/2024] Open
Abstract
This case report examines the unexpected increase in suicidal ideation following ketamine infusion therapy in a 75-year-old female with a history of treatment-resistant depression. Despite ketamine's established efficacy in treating depression and acute suicidality, this patient's condition deteriorated posttreatment. The report delves into the patient's complex background, including psychosocial stressors, genetic predisposition to depression, and a history of personality traits that may have influenced her response to ketamine. This case underscores the importance of cautious administration of ketamine, especially in patients with personality disorders, and calls for deeper understanding and individualized treatment plans in mental health care. It is a reminder of the complexities involved in treating mental health conditions and the varying effects of treatments like ketamine on different individuals.
Collapse
Affiliation(s)
- Jai Ahuja
- Department of PsychiatrySUNY Upstate Medical University, Syracuse, NY, USA
| | - Luba Leontieva
- Department of PsychiatrySUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
16
|
Li Z, Lee CS, Peng HY, Lin TB, Hsieh MC, Lai CY, Chou D. Lights at night mediate depression-like behavioral and molecular phenotypes in a glucocorticoid-dependent manner in male rats. Neuropharmacology 2024; 248:109888. [PMID: 38403262 DOI: 10.1016/j.neuropharm.2024.109888] [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: 11/28/2023] [Revised: 02/12/2024] [Accepted: 02/17/2024] [Indexed: 02/27/2024]
Abstract
Nocturnal light pollution, an underappreciated mood manipulator, disturbs the circadian rhythms of individuals in modern society. Preclinical and clinical studies have suggested that exposure to lights at night (LANs) results in depression-like phenotypes. However, the mechanism underlying the action of LANs remains unclear. Therefore, this study explored the potential influence of LANs on depression-related brain regions by testing brain-derived neurotrophic factor (BDNF), synaptic transmission, and plasticity in male Sprague-Dawley rats. Depression-related behavioral tests, enzyme-linked immunosorbent assays, and intracellular and extracellular electrophysiological recordings were performed. Resultantly, rats exposed to either white or blue LAN for 5 or 21 days exhibited depression-like behaviors. Both white and blue LANs reduced BDNF expression in the medial prefrontal cortex (mPFC) and ventrolateral periaqueductal gray (vlPAG). Moreover, both lights at night (LANs) elevated the plasma corticosterone levels. Pharmacologically, the activation of glucocorticoid receptors mimics the LAN-mediated effects on depression-like behaviors and reduces BDNF levels, whereas the inhibition of glucocorticoid receptors blocks LAN-mediated behavioral and molecular actions. Electrophysiologically, both LANs attenuated the stimulation-response curve, increased the paired-pulse ratio, and decreased the frequency and amplitude of miniature excitatory postsynaptic currents in the vlPAG. In the mPFC, LANs attenuate long-term potentiation and long-term depression. Collectively, these results suggested that white and blue LANs disturbed BDNF expression, synaptic transmission, and plasticity in the vlPAG and mPFC in a glucocorticoid-dependent manner. The results of the present study provide a theoretical basis for understanding the effects of nocturnal light exposure on depression-like phenotypes.
Collapse
Affiliation(s)
- Zhenlong Li
- School of Basic Medical Sciences, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China.
| | - Chau-Shoun Lee
- Department of Medicine, MacKay Medical College, New Taipei, Taiwan; Department of Psychiatry, MacKay Memorial Hospital, Taipei, Taiwan.
| | - Hsien-Yu Peng
- Department of Medicine, MacKay Medical College, New Taipei, Taiwan; Institute of Biomedical Sciences, MacKay Medical College, New Taipei, Taiwan.
| | - Tzer-Bin Lin
- Institute of New Drug Development, College of Medicine, China Medical University, Taichung, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Ming-Chun Hsieh
- Department of Medicine, MacKay Medical College, New Taipei, Taiwan.
| | - Cheng-Yuan Lai
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei, Taiwan.
| | - Dylan Chou
- Department of Medicine, MacKay Medical College, New Taipei, Taiwan.
| |
Collapse
|
17
|
Ruan Y, Yuan R, He J, Jiang Y, Chu S, Chen N. New perspective on sustained antidepressant effect: focus on neurexins regulating synaptic plasticity. Cell Death Discov 2024; 10:205. [PMID: 38693106 PMCID: PMC11063156 DOI: 10.1038/s41420-024-01974-9] [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: 02/17/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 05/03/2024] Open
Abstract
Depression is highly prevalent globally, however, currently available medications face challenges such as low response rates and short duration of efficacy. Additionally, depression mostly accompany other psychiatric disorders, further progressing to major depressive disorder without long-term effective management. Thus, sustained antidepressant strategies are urgently needed. Recently, ketamine and psilocybin gained attention as potential sustained antidepressants. Review of recent studies highlights that synaptic plasticity changes as key events of downstream long-lasting changes in sustained antidepressant effect. This underscores the significance of synaptic plasticity in sustained antidepressant effect. Moreover, neurexins, key molecules involved in the regulation of synaptic plasticity, act as critical links between synaptic plasticity and sustained antidepressant effects, involving mechanisms including protein level, selective splicing, epigenetics, astrocytes, positional redistribution and protein structure. Based on the regulation of synaptic plasticity by neurexins, several drugs with potential for sustained antidepressant effect are also discussed. Focusing on neurexins in regulating synaptic plasticity promises much for further understanding underlying mechanisms of sustained antidepressant and the next step in new drug development. This research represents a highly promising future research direction.
Collapse
Affiliation(s)
- Yuan Ruan
- Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Ruolan Yuan
- Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Jiaqi He
- Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Yutong Jiang
- Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Shifeng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China.
| | - Naihong Chen
- Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China.
| |
Collapse
|
18
|
Zhang S, Pu Y, Liu J, Li L, An C, Wu Y, Zhang W, Zhang W, Qu S, Yan W. Exploring the multifaceted potential of (R)-ketamine beyond antidepressant applications. Front Pharmacol 2024; 15:1337749. [PMID: 38666026 PMCID: PMC11043571 DOI: 10.3389/fphar.2024.1337749] [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: 11/13/2023] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
(R, S)- and (S)-ketamine have made significant progress in the treatment of treatment-resistant depression (TRD) and have become a research focus in recent years. However, they both have risks of psychomimetic effects, dissociative effects, and abuse liability, which limit their clinical use. Recent preclinical and clinical studies have shown that (R)-ketamine has a more efficient and lasting antidepressant effect with fewer side effects compared to (R, S)- and (S)-ketamine. However, a recent small-sample randomized controlled trial found that although (R)-ketamine has a lower incidence of adverse reactions in adult TRD treatment, its antidepressant efficacy is not superior to the placebo group, indicating its antidepressant advantage still needs further verification and clarification. Moreover, an increasing body of research suggests that (R)-ketamine might also have significant applications in the prevention and treatment of medical fields or diseases such as cognitive disorders, perioperative anesthesia, ischemic stroke, Parkinson's disease, multiple sclerosis, osteoporosis, substance use disorders, inflammatory diseases, COVID-19, and organophosphate poisoning. This article briefly reviews the mechanism of action and research on antidepressants related to (R)-ketamine, fully revealing its application potential and development prospects, and providing some references and assistance for subsequent expanded research.
Collapse
Affiliation(s)
- Senbing Zhang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
- Department of Anesthesiology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei, China
| | - Yanzhu Pu
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Jianning Liu
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Lewen Li
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Chibing An
- Department of Anesthesiology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei, China
| | - Yumin Wu
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Wenjie Zhang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Wenxia Zhang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Song Qu
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Wenjun Yan
- Department of Anesthesiology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| |
Collapse
|
19
|
Brown KA, Gould TD. Targeting metaplasticity mechanisms to promote sustained antidepressant actions. Mol Psychiatry 2024; 29:1114-1127. [PMID: 38177353 PMCID: PMC11176041 DOI: 10.1038/s41380-023-02397-1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
The discovery that subanesthetic doses of (R, S)-ketamine (ketamine) and (S)-ketamine (esketamine) rapidly induce antidepressant effects and promote sustained actions following drug clearance in depressed patients who are treatment-resistant to other therapies has resulted in a paradigm shift in the conceptualization of how rapidly and effectively depression can be treated. Consequently, the mechanism(s) that next generation antidepressants may engage to improve pathophysiology and resultant symptomology are being reconceptualized. Impaired excitatory glutamatergic synapses in mood-regulating circuits are likely a substantial contributor to the pathophysiology of depression. Metaplasticity is the process of regulating future capacity for plasticity by priming neurons with a stimulation that alters later neuronal plasticity responses. Accordingly, the development of treatment modalities that specifically modulate the duration, direction, or magnitude of glutamatergic synaptic plasticity events such as long-term potentiation (LTP), defined here as metaplastogens, may be an effective approach to reverse the pathophysiology underlying depression and improve depression symptoms. We review evidence that the initiating mechanisms of pharmacologically diverse rapid-acting antidepressants (i.e., ketamine mimetics) converge on consistent downstream molecular mediators that facilitate the expression/maintenance of increased synaptic strength and resultant persisting antidepressant effects. Specifically, while the initiating mechanisms of these therapies may differ (e.g., cell type-specificity, N-methyl-D-aspartate receptor (NMDAR) subtype-selective inhibition vs activation, metabotropic glutamate receptor 2/3 antagonism, AMPA receptor potentiation, 5-HT receptor-activating psychedelics, etc.), the sustained therapeutic mechanisms of putative rapid-acting antidepressants will be mediated, in part, by metaplastic effects that converge on consistent molecular mediators to enhance excitatory neurotransmission and altered capacity for synaptic plasticity. We conclude that the convergence of these therapeutic mechanisms provides the opportunity for metaplasticity processes to be harnessed as a druggable plasticity mechanism by next-generation therapeutics. Further, targeting metaplastic mechanisms presents therapeutic advantages including decreased dosing frequency and associated diminished adverse responses by eliminating the requirement for the drug to be continuously present.
Collapse
Affiliation(s)
- Kyle A Brown
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA.
| |
Collapse
|
20
|
Rodrigues NB, Chen-Li D, Di Vincenzo JD, Juneja A, Pinder BD, McIntyre RS, Rosenblat JD. Brain-derived neurotrophic factor Val66Met and CYP2B6 polymorphisms as predictors for ketamine effectiveness in patients with treatment-resistant depression. J Psychopharmacol 2024; 38:375-381. [PMID: 38477185 PMCID: PMC11010549 DOI: 10.1177/02698811241238284] [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: 03/14/2024]
Abstract
BACKGROUND Converging lines of evidence indicate that ketamine is a rapid antidepressant for individuals with treatment-resistant depression. Hitherto, no reliable a priori predictors of ketamine response have been reported. Pharmacogenetic biomarkers have yielded mixed results regarding potential candidate genes associated with ketamine's biochemistry as reliable predictors of response. AIMS No studies have examined the effects of Val66Met and CYP2B6 genotypes on patients receiving repeated infusions of intravenous ketamine. METHODS In all, 85 participants with major depressive disorder who had previously received four infusions of intravenous ketamine were recruited to the foregoing study. Buccal swabs were collected and genotype variants across the Val66Met and CYP2B6 genes were analyzed. A repeated measures mixed linear model was used to assess change in depressive symptoms, suicidality, and anxiety, correcting for sex and age. Multiple regression was run to determine whether these genetic markers were associated with treatment efficacy for depressive severity, suicidal ideation, anxiolytic response, and degree of dissociation to intravenous ketamine. RESULTS Participants experienced significant overall reductions in depression, suicide, and anxiety. Overall, 25% met the response criteria and 15% met the remission criteria. However, Val66Met and CYP2B6 did not significantly predict changes in symptoms of depression, suicide, anxiety, or average dissociation. CONCLUSIONS This study contributes to the growing literature that ketamine efficacy is unlikely to be predicted by single genes, and a pleiotropic approach may likely be necessary for developing reliable predictors of clinical benefits.
Collapse
Affiliation(s)
- Nelson B Rodrigues
- Braxia Health, Mississauga, ON, Canada
- Department of Psychology, Neuropsychology Track, University of Windsor, Windsor, ON, Canada
| | | | | | | | | | - Roger S McIntyre
- Braxia Health, Mississauga, ON, Canada
- Brain and Cognition Discovery Foundation, Canada, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Joshua D Rosenblat
- Braxia Health, Mississauga, ON, Canada
- Brain and Cognition Discovery Foundation, Canada, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
21
|
Zhang L, Zheng J, Liu SY, Hou LL, Zhang B, Tian SW. Acute Administration of Lactate Exerts Antidepressant-like Effect Through cAMP-dependent Protein Synthesis. Neuroscience 2024; 542:11-20. [PMID: 38336096 DOI: 10.1016/j.neuroscience.2024.02.003] [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: 09/14/2023] [Revised: 01/16/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
Lactate acts as an important metabolic substrate and signalling molecule modulating neural activities in the brain, and recent preclinical and clinical studies have revealed its antidepressant effect after acute or chronic peripheral administration. However, the neural mechanism underlying the antidepressant effect of lactate, in particular when lactate is acutely administered remains largely unknown. In the current study, we focused on forced swimming test (FST) to elucidate the neural mechanisms through which acute intracerebroventricular (ICV) infusion of lactate exerts antidepressant-like effect. A total of 238 male Sprague Dawley rats were used as experimental subjects. Results showed lactate produced antidepressant-like effect, as indicated by reduced immobility, in a dose- and time-dependent manner. Moreover, the antidepressant-like effect of lactate was dependent of new protein synthesis but not new gene expression, lactate's metabolic effect or hydroxy-carboxylic acid receptor 1 (HCAR1) activation. Furthermore, lactate rapidly promoted dephosphorylation of eukaryotic elongation factor 2 (eEF2) and increased brain-derived neurotrophic factor (BDNF) protein synthesis in the hippocampus in a cyclic adenosine monophosphate (cAMP)-dependent manner. Finally, inhibition of cAMP production blocked the antidepressant-like effect of lactate. These findings suggest that acute administration of lactate exerts antidepressant-like effect through cAMP-dependent protein synthesis.
Collapse
Affiliation(s)
- Liang Zhang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin, Guangxi 541199, China; Department of Anesthesiology, National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong 518112, China; Department of Anesthesiology, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan 421001, China
| | - Jing Zheng
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Shi-Yan Liu
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Li-Li Hou
- Department of Anesthesiology, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan 421001, China
| | - Bo Zhang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Shao-Wen Tian
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin, Guangxi 541199, China.
| |
Collapse
|
22
|
Zhang Q, Xue Y, Wei K, Wang H, Ma Y, Wei Y, Fan Y, Gao L, Yao H, Wu F, Ding X, Zhang Q, Ding J, Fan Y, Lu M, Hu G. Locus Coeruleus-Dorsolateral Septum Projections Modulate Depression-Like Behaviors via BDNF But Not Norepinephrine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303503. [PMID: 38155473 PMCID: PMC10933643 DOI: 10.1002/advs.202303503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 12/14/2023] [Indexed: 12/30/2023]
Abstract
Locus coeruleus (LC) dysfunction is involved in the pathophysiology of depression; however, the neural circuits and specific molecular mechanisms responsible for this dysfunction remain unclear. Here, it is shown that activation of tyrosine hydroxylase (TH) neurons in the LC alleviates depression-like behaviors in susceptible mice. The dorsolateral septum (dLS) is the most physiologically relevant output from the LC under stress. Stimulation of the LCTH -dLSSST innervation with optogenetic and chemogenetic tools bidirectionally can regulate depression-like behaviors in both male and female mice. Mechanistically, it is found that brain-derived neurotrophic factor (BDNF), but not norepinephrine, is required for the circuit to produce antidepressant-like effects. Genetic overexpression of BDNF in the circuit or supplementation with BDNF protein in the dLS is sufficient to produce antidepressant-like effects. Furthermore, viral knockdown of BDNF in this circuit abolishes the antidepressant-like effect of ketamine, but not fluoxetine. Collectively, these findings underscore the notable antidepressant-like role of the LCTH -dLSSST pathway in depression via BDNF-TrkB signaling.
Collapse
Affiliation(s)
- Qian Zhang
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - You Xue
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Ke Wei
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Hao Wang
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Yuan Ma
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Yao Wei
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Yi Fan
- Department of NeurologyAffiliated Nanjing Brain HospitalNanjing Medical UniversityNanjing210024China
| | - Lei Gao
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Hang Yao
- Jiangsu Key Laboratory of NeurodegenerationDepartment of PharmacologyNanjing Medical UniversityNanjing211166China
| | - Fangfang Wu
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Xin Ding
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Qingyu Zhang
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Jianhua Ding
- Jiangsu Key Laboratory of NeurodegenerationDepartment of PharmacologyNanjing Medical UniversityNanjing211166China
| | - Yi Fan
- Jiangsu Key Laboratory of NeurodegenerationDepartment of PharmacologyNanjing Medical UniversityNanjing211166China
| | - Ming Lu
- Jiangsu Key Laboratory of NeurodegenerationDepartment of PharmacologyNanjing Medical UniversityNanjing211166China
| | - Gang Hu
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
- Jiangsu Key Laboratory of NeurodegenerationDepartment of PharmacologyNanjing Medical UniversityNanjing211166China
| |
Collapse
|
23
|
Famuła A, Radoszewski J, Czerwiec T, Sobiś J, Więckiewicz G. Ketamine in Substance Use Disorder Treatment: A Narrative Review. ALPHA PSYCHIATRY 2024; 25:206-211. [PMID: 38798813 PMCID: PMC11117434 DOI: 10.5152/alphapsychiatry.2024.241522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/06/2024] [Indexed: 05/29/2024]
Abstract
Substance use disorder (SUD) continues to pose a significant global health challenge, necessitating innovative and effective therapeutic interventions. Ketamine, traditionally recognized for its anesthetic properties, has emerged as a novel and promising avenue for the treatment of SUD. This narrative review critically examines the current body of literature surrounding the use of ketamine in various forms and settings for individuals grappling with substance abuse. The review explores the neurobiological underpinnings of ketamine's potential therapeutic effects in SUD, shedding light on its impact on glutamatergic neurotransmission, neuroplasticity, and reward pathways. Special attention is given to the psychotropic and dissociative properties of ketamine, exploring their implications for both therapeutic outcomes and patient experience. Ultimately, this review aims to provide a comprehensive overview of the current state of knowledge regarding ketamine's role in the treatment of SUD, emphasizing the need for further research and clinical exploration. As we navigate the complex terrain of addiction medicine, understanding the nuances of ketamine's potential in SUD holds promise for the development of more effective and personalized therapeutic strategies.
Collapse
Affiliation(s)
- Anna Famuła
- Department of Psychiatry, Medical University of Silesia, Faculty of Medical Sciences, Zabrze, Poland
- Intravenous Ketamine Treatment Laboratory, Multispecialty County Hospital, Tarnowskie Góry, Poland
| | - Jakub Radoszewski
- Department and Clinical Division of Psychiatry and Psychotherapy of Developmental Age, Medical University of Silesia, Faculty of Medical Sciences, Katowice, Poland
| | - Tomasz Czerwiec
- Department and Clinical Division of Psychiatry and Psychotherapy of Developmental Age, Medical University of Silesia, Faculty of Medical Sciences, Katowice, Poland
| | - Jarosław Sobiś
- Department of Psychiatry, Medical University of Silesia, Faculty of Medical Sciences, Zabrze, Poland
| | - Gniewko Więckiewicz
- Department of Psychiatry, Medical University of Silesia, Faculty of Medical Sciences, Zabrze, Poland
- Intravenous Ketamine Treatment Laboratory, Multispecialty County Hospital, Tarnowskie Góry, Poland
| |
Collapse
|
24
|
Kuntz S, Kunz C, Borsch C, Hill D, Morrin S, Buck R, Rudloff S. Influence of microbially fermented 2´-fucosyllactose on neuronal-like cell activity in an in vitro co-culture system. Front Nutr 2024; 11:1351433. [PMID: 38389793 PMCID: PMC10881714 DOI: 10.3389/fnut.2024.1351433] [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: 12/06/2023] [Accepted: 01/18/2024] [Indexed: 02/24/2024] Open
Abstract
Scope 2´-Fucosyllactose (2´-FL), the most abundant oligosaccharide in human milk, plays an important role in numerous biological functions, including improved learning. It is not clear, however, whether 2´-FL or a cleavage product could influence neuronal cell activity. Thus, we investigated the effects of 2´-FL, its monosaccharide fucose (Fuc), and microbial fermented 2´-FL and Fuc on the parameters of neuronal cell activity in an intestinal-neuronal transwell co-culture system in vitro. Methods Native 13C-labeled 2´-FL and 13C-Fuc or their metabolites, fermented with Bifidobacterium (B.) longum ssp. infantis and B. breve, which were taken from the lag-, log- and stationary (stat-) growth phases of batch cultures, were applied to the apical compartment of the co-culture system with Caco-2 cells representing the intestinal layer and all-trans-retinoic acid-differentiated SH-SY5Y (SH-SY5YATRA) cells mimicking neuronal-like cells. After 3 h of incubation, the culture medium in the basal compartment was monitored for 13C enrichment by using elemental analysis isotope-ratio mass spectrometry (EA-IRMS) and effects on cell viability, plasma, and mitochondrial membrane potential. The neurotransmitter activation (BDNF, GABA, choline, and glutamate) of SH-SY5YATRA cells was also determined. Furthermore, these effects were also measured by the direct application of 13C-2´-FL and 13C-Fuc to SH-SY5YATRA cells. Results While no effects on neuronal-like cell activities were observed after intact 2´-FL or Fuc was incubated with SH-SY5YATRA cells, supernatants from the stat-growth phase of 2´-FL, fermented by B. longum ssp. infantis alone and together with B. breve, significantly induced BDNF release from SH-SY5YATRA cells. No such effects were found for 2´-FL, Fuc, or their fermentation products from B. breve. The BDNF release occurred from an enhanced vesicular release, which was confirmed by the use of the Ca2+-channel blocker verapamil. Concomitant with this event, 13C enrichment was also observed in the basal compartment when supernatants from the stat-growth phase of fermentation by B. longum ssp. infantis alone or together with B. breve were used. Conclusion The results obtained in this study suggest that microbial products of 2´-FL rather than the oligosaccharide itself may influence neuronal cell activities.
Collapse
Affiliation(s)
- Sabine Kuntz
- Department of Nutritional Science, Justus Liebig University Giessen, Giessen, Germany
| | - Clemens Kunz
- Department of Nutritional Science, Justus Liebig University Giessen, Giessen, Germany
| | - Christian Borsch
- Department of Nutritional Science, Justus Liebig University Giessen, Giessen, Germany
| | - David Hill
- Abbott, Nutrition Division, Columbus, OH, United States
| | - Sinéad Morrin
- Abbott, Nutrition Division, Columbus, OH, United States
| | - Rachael Buck
- Abbott, Nutrition Division, Columbus, OH, United States
| | - Silvia Rudloff
- Department of Nutritional Science, Justus Liebig University Giessen, Giessen, Germany
- Department of Pediatrics, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
25
|
Elmeseiny OSA, Müller HK. A molecular perspective on mGluR5 regulation in the antidepressant effect of ketamine. Pharmacol Res 2024; 200:107081. [PMID: 38278430 DOI: 10.1016/j.phrs.2024.107081] [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: 10/30/2023] [Revised: 01/16/2024] [Accepted: 01/23/2024] [Indexed: 01/28/2024]
Abstract
Ketamine, a non-competitive N-methyl-D-aspartate receptor (NMDAR) antagonist, has received much attention for its rapid antidepressant effects. A single administration of ketamine elicits rapid and sustained antidepressant effects in both humans and animals. Current efforts are focused on uncovering molecular mechanisms responsible for ketamine's antidepressant activity. Ketamine primarily acts via the glutamatergic pathway, and increasing evidence suggests that ketamine induces synaptic and structural plasticity through increased translation and release of neurotrophic factors, activation of mammalian target of rapamycin (mTOR), and α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR)-mediated synaptic potentiation. However, the initial events triggering activation of intracellular signaling cascades and the mechanisms responsible for the sustained antidepressant effects of ketamine remain poorly understood. Over the last few years, it has become apparent that in addition to the fast actions of the ligand-gated AMPARs and NMDARs, metabotropic glutamate receptors (mGluRs), and particularly mGluR5, may also play a role in the antidepressant action of ketamine. Although research on mGluR5 in relation to the beneficial actions of ketamine is still in its infancy, a careful evaluation of the existing literature can identify converging trends and provide new interpretations. Here, we review the current literature on mGluR5 regulation in response to ketamine from a molecular perspective and propose a possible mechanism linking NMDAR inhibition to mGluR5 modulation.
Collapse
Affiliation(s)
- Ola Sobhy A Elmeseiny
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Heidi Kaastrup Müller
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
26
|
Kim J, Seol S, Kim TE, Lee J, Koo JW, Kang HJ. Synaptotagmin-4 induces anhedonic responses to chronic stress via BDNF signaling in the medial prefrontal cortex. Exp Mol Med 2024; 56:329-343. [PMID: 38297157 PMCID: PMC10907712 DOI: 10.1038/s12276-024-01156-8] [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: 04/04/2023] [Revised: 11/22/2023] [Accepted: 12/08/2023] [Indexed: 02/02/2024] Open
Abstract
Stressful circumstances are significant contributors to mental illnesses such as major depressive disorder. Anhedonia, defined as loss of the ability to enjoy pleasure in pleasurable situations, including rewarding activities or social contexts, is considered a key symptom of depression. Although stress-induced depression is associated with anhedonia in humans and animals, the underlying molecular mechanisms of anhedonic responses remain poorly understood. In this study, we demonstrated that synaptotagmin-4 (SYT4), which is involved in the release of neurotransmitters and neurotrophic factors, is implicated in chronic stress-induced anhedonia. Employing chronic unpredictable stress (CUS), we evaluated two subpopulations of mice, susceptible (SUS, anhedonic) and resilient (RES, nonanhedonic), based on sucrose preference, which was strongly correlated with social reward. The FosTRAP (targeted recombination in active populations) system and optogenetic approach revealed that neural activity in the medial prefrontal cortex (mPFC) was significantly associated with CUS-induced anhedonic behavioral phenotypes. By conducting weighted gene coexpression network analysis of RNA sequencing data from the mPFC of SUS and RES mice, we identified Syt4 as a hub gene in a gene network that was unique to anhedonia. We also confirmed that Syt4 overexpression in the mPFC was pro-susceptible, while Syt4 knockdown was pro-resilient; the pro-susceptible effects of SYT4 were mediated through a reduction in brain-derived neurotrophic factor (BDNF)-tropomyosin receptor kinase B (TrkB) signaling in the mPFC. These findings suggest that SYT4-BDNF interactions in the mPFC represent a crucial regulatory mechanism of anhedonic susceptibility to chronic stress.
Collapse
Affiliation(s)
- Jeongseop Kim
- Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute (KBRI), Dong-gu, Daegu, 41062, Republic of Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Dalseong-gun, Daegu, 42988, Republic of Korea
| | - Sihwan Seol
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Tae-Eun Kim
- Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute (KBRI), Dong-gu, Daegu, 41062, Republic of Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Dalseong-gun, Daegu, 42988, Republic of Korea
| | - Joonhee Lee
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Ja Wook Koo
- Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute (KBRI), Dong-gu, Daegu, 41062, Republic of Korea.
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Dalseong-gun, Daegu, 42988, Republic of Korea.
| | - Hyo Jung Kang
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
27
|
Guhathakurta D, Petrušková A, Akdaş EY, Perelló-Amorós B, Frischknecht R, Anni D, Weiss EM, Walter M, Fejtová A. Hydroxynorketamine, but not ketamine, acts via α7 nicotinic acetylcholine receptor to control presynaptic function and gene expression. Transl Psychiatry 2024; 14:47. [PMID: 38253622 PMCID: PMC10803733 DOI: 10.1038/s41398-024-02744-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Ketamine is clinically used fast-acting antidepressant. Its metabolite hydroxynorketamine (HNK) shows a robust antidepressant effect in animal studies. It is unclear, how these chemically distinct compounds converge on similar neuronal effects. While KET acts mostly as N-methyl-d-aspartate receptor (NMDAR) antagonist, the molecular target of HNK remains enigmatic. Here, we show that KET and HNK converge on rapid inhibition of glutamate release by reducing the release competence of synaptic vesicles and induce nuclear translocation of pCREB that controls expression of neuroplasticity genes connected to KET- and HNK-mediated antidepressant action. Ro25-6981, a selective antagonist of GluN2B, mimics effect of KET indicating that GluN2B-containing NMDAR might mediate the presynaptic effect of KET. Selective antagonist of α7 nicotinic acetylcholine receptors (α7nAChRs) or genetic deletion of Chrna7, its pore-forming subunit, fully abolishes HNK-induced synaptic and nuclear regulations, but leaves KET-dependent cellular effects unaffected. Thus, KET or HNK-induced modulation of synaptic transmission and nuclear translocation of pCREB can be mediated by selective signaling via NMDAR or α7nAChRs, respectively. Due to the rapid metabolism of KET to HNK, it is conceivable that subsequent modulation of glutamatergic and cholinergic neurotransmission affects circuits in a cell-type-specific manner and contributes to the therapeutic potency of KET. This finding promotes further exploration of new combined medications for mood disorders.
Collapse
Affiliation(s)
- Debarpan Guhathakurta
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Aneta Petrušková
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- National Institute of Mental Health, Klecany, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Enes Yağız Akdaş
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Bartomeu Perelló-Amorós
- Department of Biology, Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Renato Frischknecht
- Department of Biology, Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Daniela Anni
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Eva-Maria Weiss
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Walter
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Jena, Jena, Germany
| | - Anna Fejtová
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
28
|
Riggs LM, Pereira EFR, Thompson SM, Gould TD. cAMP-dependent protein kinase signaling is required for ( 2R,6R)-hydroxynorketamine to potentiate hippocampal glutamatergic transmission. J Neurophysiol 2024; 131:64-74. [PMID: 38050689 PMCID: PMC11286304 DOI: 10.1152/jn.00326.2023] [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: 08/30/2023] [Revised: 11/10/2023] [Accepted: 11/23/2023] [Indexed: 12/06/2023] Open
Abstract
(2R,6R)-Hydroxynorketamine (HNK) is a ketamine metabolite that shows rapid antidepressant-like effects in preclinical studies and lacks the adverse N-methyl-d-aspartate receptor (NMDAR) inhibition-related properties of ketamine. Investigating how (2R,6R)-HNK exerts its antidepressant actions may be informative in the design of novel pharmacotherapies with improved safety and efficacy. We sought to identify the molecular substrates through which (2R,6R)-HNK induces functional changes at excitatory synapses, a prevailing hypothesis for how rapid antidepressant effects are initiated. We recorded excitatory postsynaptic potentials in hippocampal slices from male Wistar Kyoto rats, which have impaired hippocampal plasticity and are resistant to traditional antidepressants. (2R,6R)-HNK (10 µM) led to a rapid potentiation of electrically evoked excitatory postsynaptic potentials at Schaffer collateral CA1 stratum radiatum synapses. This potentiation was associated with a decrease in paired pulse facilitation, suggesting an increase in the probability of glutamate release. The (2R,6R)-HNK-induced potentiation was blocked by inhibiting either cyclic adenosine monophosphate (cAMP) or its downstream target, cAMP-dependent protein kinase (PKA). As cAMP is a potent regulator of brain-derived neurotrophic factor (BDNF) release, we assessed whether (2R,6R)-HNK exerts this acute potentiation through a rapid increase in cAMP-dependent BDNF-TrkB signaling. We found that the cAMP-PKA-dependent potentiation was not dependent on TrkB activation by BDNF, which functionally delimits the acute synaptic effects of (2R,6R)-HNK from its sustained BDNF-dependent actions in vivo. These results suggest that, by potentiating glutamate release via cAMP-PKA signaling, (2R,6R)-HNK initiates acute adaptations in fast excitatory synaptic transmission that promote structural plasticity leading to maintained antidepressant action.NEW & NOTEWORTHY Ketamine is a rapid-acting antidepressant and its preclinical effects are mimicked by its (2R,6R)-(HNK) metabolite. We found that (2R,6R)-HNK initiates acute adaptations in fast excitatory synaptic transmission by potentiating glutamate release via cAMP-PKA signaling at hippocampal Schaffer collateral synapses. This cAMP-PKA-dependent potentiation was not dependent on TrkB activation by BDNF, which functionally delimits the rapid synaptic effects of (2R,6R)-HNK from its sustained BDNF-dependent actions that are thought to maintain antidepressant action in vivo.
Collapse
Affiliation(s)
- Lace M Riggs
- Program in Neuroscience and Training Program in Integrative Membrane Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Edna F R Pereira
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Scott M Thompson
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Veterans Affairs Maryland Health Care System, Baltimore, Maryland, United States
| |
Collapse
|
29
|
Kim J, He MJ, Widmann AK, Lee FS. The role of neurotrophic factors in novel, rapid psychiatric treatments. Neuropsychopharmacology 2024; 49:227-245. [PMID: 37673965 PMCID: PMC10700398 DOI: 10.1038/s41386-023-01717-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 07/11/2023] [Accepted: 07/26/2023] [Indexed: 09/08/2023]
Abstract
Neurotrophic factors are a family of growth factors that modulate cellular growth, survival, and differentiation. For many decades, it has been generally believed that a lack of neurotrophic support led to the decreased neuronal synaptic plasticity, death, and loss of non-neuronal supportive cells seen in neuropsychiatric disorders. Traditional psychiatric medications that lead to immediate increases in neurotransmitter levels at the synapse have been shown also to elevate synaptic neurotrophic levels over weeks, correlating with the time course of the therapeutic effects of these drugs. Recent advances in psychiatric treatments, such as ketamine and psychedelics, have shown a much faster onset of therapeutic effects (within minutes to hours). They have also been shown to lead to a rapid release of neurotrophins into the synapse. This has spurred a significant shift in understanding the role of neurotrophins and how the receptor tyrosine kinases that bind neurotrophins may work in concert with other signaling systems. In this review, this renewed understanding of synaptic receptor signaling interactions and the clinical implications of this mechanistic insight will be discussed within the larger context of the well-established roles of neurotrophic factors in psychiatric disorders and treatments.
Collapse
Affiliation(s)
- Jihye Kim
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Michelle J He
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Alina K Widmann
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Francis S Lee
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, 10065, USA.
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
30
|
Drinkuth CR, Lehane MJ, Sartor GC. The effects of (2R,6R)-hydroxynorketamine on oxycodone withdrawal and reinstatement. Drug Alcohol Depend 2023; 253:110987. [PMID: 37864957 PMCID: PMC10842506 DOI: 10.1016/j.drugalcdep.2023.110987] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/13/2023] [Accepted: 09/30/2023] [Indexed: 10/23/2023]
Abstract
Despite the thousands of lives lost during the ongoing opioid crisis, a scarcity of new and effective clinical treatments for opioid use disorder (OUD) remains. To address this unmet need, some researchers have turned to dissociative and psychedelic drugs to treat multiple psychiatric conditions. In particular, low doses of ketamine have been shown to attenuate opioid withdrawal and drug use in clinical and preclinical studies. However, ketamine has misuse liability and dissociative side effects that may limit its widespread application as a treatment for OUD. More recently, (2R,6R)-hydroxynorketamine (HNK), a ketamine metabolite that lacks misuse potential, has gained attention for its effectiveness in depression and stress models. To uncover its role in OUD, we tested the time-dependent effects of (2R,6R)-HNK on oxycodone withdrawal and reinstatement of oxycodone conditioned place preference (CPP). In male and female oxycodone-dependent mice, we found that 24h pretreatment with (2R,6R)-HNK (10 or 30mg/kg, s.c.) reduced the frequency of withdrawal-like behaviors and global withdrawal scores during naloxone-precipitated withdrawal, whereas 1h pretreatment with (2R,6R)-HNK only reduced paw tremors and the sum of global withdrawal scores but not GWS Z-scores. In other experiments, both 1h and 24h pretreatment with (2R,6R)-HNK (30mg/kg, s.c.) blocked drug-induced reinstatement of oxycodone CPP. Finally, we found (2R,6R)-HNK (30mg/kg, sc) had no effect on locomotor activity and thigmotaxis. Together, these results indicate that acute (2R,6R)-HNK has efficacy in some preclinical models of OUD without producing locomotor or anxiety-like side effects.
Collapse
Affiliation(s)
- Caryssa R Drinkuth
- Department of Pharmaceutical Sciences, Connecticut Institute for the Brain and Cognitive Sciences (IBACS), University of Connecticut, Storrs, CT 06269, United States
| | - Michael J Lehane
- Department of Pharmaceutical Sciences, Connecticut Institute for the Brain and Cognitive Sciences (IBACS), University of Connecticut, Storrs, CT 06269, United States
| | - Gregory C Sartor
- Department of Pharmaceutical Sciences, Connecticut Institute for the Brain and Cognitive Sciences (IBACS), University of Connecticut, Storrs, CT 06269, United States.
| |
Collapse
|
31
|
Kuo J, Block T, Nicklay M, Lau B, Green M. Interventional Mental Health: A Transdisciplinary Approach to Novel Psychiatric Care Delivery. Cureus 2023; 15:e43533. [PMID: 37719598 PMCID: PMC10501497 DOI: 10.7759/cureus.43533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2023] [Indexed: 09/19/2023] Open
Abstract
Mental health disorders are among the most common health conditions in the United States. Traditional clinical treatments rely on psychiatric counseling and, in many cases, prescription medications. We propose an innovative model, Interventional Mental Health, which employs a combination of modalities through a multifaceted approach to treat conditions that have exhibited limited responsiveness to traditional methods and individuals afflicted with multiple comorbidities simultaneously. We hypothesize that creating a unique treatment algorithm combining current therapeutic modalities such as Stellate Ganglion Blocks (SGB), Transcranial Magnetic Stimulation (TMS) therapy, and ketamine therapy, within a consolidated timeframe, will yield synergistic outcomes among patients presenting with comorbid post-traumatic stress disorder (PTSD), depression, and/or anxiety.
Collapse
Affiliation(s)
- Jonathann Kuo
- Regenerative and Anti-Aging Medicine, Hudson Health, New York, USA
| | | | | | | | | |
Collapse
|
32
|
Fogaça MV, Wu M, Li C, Li XY, Duman RS, Picciotto MR. M1 acetylcholine receptors in somatostatin interneurons contribute to GABAergic and glutamatergic plasticity in the mPFC and antidepressant-like responses. Neuropsychopharmacology 2023; 48:1277-1287. [PMID: 37142667 PMCID: PMC10354201 DOI: 10.1038/s41386-023-01583-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/28/2023] [Accepted: 04/01/2023] [Indexed: 05/06/2023]
Abstract
Alterations in glutamatergic and GABAergic function in the medial prefrontal cortex (mPFC) are prevalent in individuals with major depressive disorder, resulting in impaired synaptic plasticity that compromises the integrity of signal transfer to limbic regions. Scopolamine, a non-selective muscarinic receptor antagonist, produces rapid antidepressant-like effects by targeting M1-type acetylcholine receptors (M1R) on somatostatin (SST) interneurons. So far, these effects have been investigated with relatively short-term manipulations, and long-lasting synaptic mechanisms involved in these responses are still unknown. Here, we generated mice with conditional deletion of M1R (M1f/fSstCre+) only in SST interneurons to determine the role of M1R in modulating long-term GABAergic and glutamatergic plasticity in the mPFC that leads to attenuation of stress-relevant behaviors. We have also investigated whether the molecular and antidepressant-like effects of scopolamine could be mimicked or occluded in male M1f/fSstCre+ mice. M1R deletion in SST-expressing neurons occluded the rapid and sustained antidepressant-like effects of scopolamine, as well as scopolamine-induced increases in c-Fos+/CaMKIIα cells and proteins necessary for glutamatergic and GABAergic function in the mPFC. Importantly, M1R SST deletion resulted in resilience to chronic unpredictable stress in behaviors relevant to coping strategies and motivation, and to a lesser extent, in behaviors relevant to avoidance. Finally, M1R SST deletion also prevented stress-induced impairments in the expression of GABAergic and glutamatergic markers in the mPFC. These findings suggest that the antidepressant-like effects of scopolamine result from modulation of excitatory and inhibitory plasticity via M1R blockade in SST interneurons. This mechanism could represent a promising strategy for antidepressant development.
Collapse
Affiliation(s)
- Manoela V Fogaça
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT, 06519, USA.
| | - Min Wu
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT, 06519, USA
| | - Chan Li
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT, 06519, USA
| | - Xiao-Yuan Li
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT, 06519, USA
| | - Ronald S Duman
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT, 06519, USA
| | - Marina R Picciotto
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT, 06519, USA
| |
Collapse
|
33
|
Li J, Zhang M, Pei Y, Yang Q, Zheng L, Wang G, Sun Y, Yang W, Liu L. The total alkaloids of Sophora alopecuroides L. improve depression-like behavior in mice via BDNF-mediated AKT/mTOR signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023:116723. [PMID: 37271329 DOI: 10.1016/j.jep.2023.116723] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/02/2023] [Accepted: 06/01/2023] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Depression has become a global public health problem and the development of new highly effective, low-toxicity antidepressants is imminent. Sophora alopecuroides L. is a common medicinal plant, which has therapeutic effect on central nervous system diseases. AIM OF THE STUDY In this study, the antidepressant effect of total alkaloids (ALK) isolated from Sophora alopecuroides L. was explored and the mechanism was further elucidated. MATERIALS AND METHODS A primary neuronal injury model was established in vitro by corticosterone. ICR mice were then selected to construct an in vivo model of chronic unpredictable mild stress (CUMS)-induced depression, and the ameliorative effects of ALK on depression were examined by various behavioral tests. The antidepressant molecular mechanism of ALK was subsequently revealed by ELISA, Western blot, immunohistochemistry and Golgi staining. RESULTS BDNF secretion as well as TrkB and ERK phosphorylated protein levels were found to be improved in primary cortical neurons, along with improved dendritic complexity of neurons. The results of in vivo showed that the depression-like behavior of CUMS-induced mice was reversed after 2 weeks of continuous gavage administration of ALK, and the neurotransmitter levels in the plasma of mice were increased. Moreover, the expression levels of key proteins of BDNF-AKT-mTOR pathway and the complexity of neuronal dendrites were improved in the prefrontal cortex of mice. CONCLUSIONS These findings indicate that ALK of Sophora alopecuroides L. can effectively improve the depressive phenotype of mice, possibly by promoting the expression of BDNF in prefrontal cortex, activating the downstream AKT/mTOR signal pathway, and ultimately enhancing neuronal dendritic complexity.
Collapse
Affiliation(s)
- Jingyi Li
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| | - Ming Zhang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Yiying Pei
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| | - Qifang Yang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| | - Lihua Zheng
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| | - Guannan Wang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| | - Ying Sun
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China.
| | - Lei Liu
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China.
| |
Collapse
|
34
|
Wang YB, Song NN, Ding YQ, Zhang L. Neural plasticity and depression treatment. IBRO Neurosci Rep 2023; 14:160-184. [PMID: 37388497 PMCID: PMC10300479 DOI: 10.1016/j.ibneur.2022.09.001] [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: 05/31/2022] [Revised: 07/29/2022] [Accepted: 09/01/2022] [Indexed: 12/08/2022] Open
Abstract
Depression is one of the most common mental disorders, which can lead to a variety of emotional problems and even suicide at its worst. As this neuropsychiatric disorder causes the patients to suffer a lot and function poorly in everyday life, it is imposing a heavy burden on the affected families and the whole society. Several hypotheses have been proposed to elucidate the pathogenesis of depression, such as the genetic mutations, the monoamine hypothesis, the hypothalamic-pituitary-adrenal (HPA) axis hyperactivation, the inflammation and the neural plasticity changes. Among these models, neural plasticity can occur at multiple levels from brain regions, cells to synapses structurally and functionally during development and in adulthood. In this review, we summarize the recent progresses (especially in the last five years) on the neural plasticity changes in depression under different organizational levels and elaborate different treatments for depression by changing the neural plasticity. We hope that this review would shed light on the etiological studies for depression and on the development of novel treatments.
Collapse
Affiliation(s)
- Yu-Bing Wang
- Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center) and Department of Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai 200092, China
| | - Ning-Ning Song
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudfan University, Shanghai 200032, China
| | - Yu-Qiang Ding
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudfan University, Shanghai 200032, China
| | - Lei Zhang
- Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center) and Department of Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai 200092, China
| |
Collapse
|
35
|
Borsellino P, Krider RI, Chea D, Grinnell R, Vida TA. Ketamine and the Disinhibition Hypothesis: Neurotrophic Factor-Mediated Treatment of Depression. Pharmaceuticals (Basel) 2023; 16:ph16050742. [PMID: 37242525 DOI: 10.3390/ph16050742] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Ketamine is a promising alternative to traditional pharmacotherapies for major depressive disorder, treatment-resistant depression, and other psychiatric conditions that heavily contribute to the global disease burden. In contrast to the current standard of care medications for these disorders, ketamine offers rapid onset, enduring clinical efficacy, and unique therapeutic potential for use in acute, psychiatric emergencies. This narrative presents an alternative framework for understanding depression, as mounting evidence supports a neuronal atrophy and synaptic disconnection theory, rather than the prevailing monoamine depletion hypothesis. In this context, we describe ketamine, its enantiomers, and various metabolites in a range of mechanistic actions through multiple converging pathways, including N-methyl-D-aspartate receptor (NMDAR) inhibition and the enhancement of glutamatergic signaling. We describe the disinhibition hypothesis, which posits that ketamine's pharmacological action ultimately results in excitatory cortical disinhibition, causing the release of neurotrophic factors, the most important of which is brain-derived neurotrophic factor (BDNF). BDNF-mediated signaling along with vascular endothelial growth factor (VEGF) and insulin-like growth factor 1 (IGF-1) subsequently give rise to the repair of neuro-structural abnormalities in patients with depressive disorders. Ketamine's efficacious amelioration of treatment-resistant depression is revolutionizing psychiatric treatment and opening up fresh vistas for understanding the underlying causes of mental illness.
Collapse
Affiliation(s)
- Philip Borsellino
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA
| | - Reese I Krider
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA
| | - Deanna Chea
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA
| | - Ryan Grinnell
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA
| | - Thomas A Vida
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA
| |
Collapse
|
36
|
Gou L, Li Y, Liu S, Sang H, Lan J, Chen J, Wang L, Li C, Lian B, Zhang X, Sun H, Sun L. (2R,6R)-hydroxynorketamine improves PTSD-associated behaviors and structural plasticity via modulating BDNF-mTOR signaling in the nucleus accumbens. J Affect Disord 2023; 335:129-140. [PMID: 37137411 DOI: 10.1016/j.jad.2023.04.101] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 04/16/2023] [Accepted: 04/28/2023] [Indexed: 05/05/2023]
Abstract
BACKGROUND Post-traumatic stress disorder (PTSD) is a mental illness caused by either experiencing or observing a traumatic event that is perceived to pose a serious risk to one's life. (2R,6R)-HNK has an alleviating effect on negative emotions, nevertheless, the mechanism of (2R,6R)-HNK action is unclear. METHODS In this study, the single prolonged stress and electric foot shock (SPS&S) method was used to establish a rat model of PTSD. After determining the validity of the model, (2R,6R)-HNK was administered to the NAc by microinjection using a concentration gradient of 10, 50, and 100 μM, and the effects of the drug in the SPS&S rat model were evaluated. Moreover, our study measured changes in related proteins in the NAc (BDNF, p-mTOR/mTOR, and PSD95) and synaptic ultrastructure. RESULTS In the SPS&S group, the protein expression of brain-derived neurotrophic factor (BDNF), mammalian target of rapamycin (mTOR), and PSD95 was reduced and synaptic morphology was damaged in the NAc. In contrast, after the administration of 50 μM (2R,6R)-HNK, SPS&S-treated rats improved their exploration and depression-linked behavior, while protein levels and synaptic ultrastructure were also restored in the NAc. With the administration of 100 μM (2R,6R)-HNK, locomotor behavior, and social interaction improved in the PTSD model. LIMITATIONS The mechanism of BDNF-mTOR signaling after (2R,6R)-HNK administration was not explored. CONCLUSION (2R,6R)-HNK may ameliorate negative mood and social avoidance symptoms in PTSD rats by regulating BDNF/mTOR-mediated synaptic structural plasticity in the NAc, providing new targets for the development of anti-PTSD drugs.
Collapse
Affiliation(s)
- Luping Gou
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, China
| | - Yu Li
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, China
| | - Shiqi Liu
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, China
| | - Haohan Sang
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, China
| | - Jiajun Lan
- School of Clinical Medical, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, China
| | - Jinhong Chen
- College of Extended Education, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, China
| | - Ling Wang
- Clinical Competency Training Center, Medical Experiment and Training Center, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong, 261053, China
| | - Changjiang Li
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, China
| | - Bo Lian
- Department of Bioscience and Technology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, China
| | - Xianqiang Zhang
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, China; National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital/Institute of Mental Health and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| | - Hongwei Sun
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, China.
| | - Lin Sun
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, China.
| |
Collapse
|
37
|
Wang H, Yang Y, Pei G, Wang Z, Chen N. Neurotrophic basis to the pathogenesis of depression and phytotherapy. Front Pharmacol 2023; 14:1182666. [PMID: 37089920 PMCID: PMC10115971 DOI: 10.3389/fphar.2023.1182666] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 03/28/2023] [Indexed: 04/09/2023] Open
Abstract
Depression is a major neuropsychiatric disease that considerably impacts individuals’ psychosocial function and life quality. Neurotrophic factors are now connected to the pathogenesis of depression, while the definitive neurotrophic basis remains elusive. Besides, phytotherapy is alternative to conventional antidepressants that may minimize undesirable adverse reactions. Thus, further research into the interaction between neurotrophic factors and depression and phytochemicals that repair neurotrophic factors deficit is highly required. This review highlighted the implication of neurotrophic factors in depression, with a focus on the brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF), vascular endothelial growth factor (VEGF), and nerve growth factor (NGF), and detailed the antidepressant activities of various phytochemicals targeting neurotrophic factors. Additionally, we presented future opportunities for novel diagnostic and therapeutic strategies for depression and provided solutions to challenges in this area to accelerate the clinical translation of neurotrophic factors for the treatment of depression.
Collapse
Affiliation(s)
- Huiqin Wang
- Hunan University of Chinese Medicine and Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, Hunan, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yantao Yang
- Hunan University of Chinese Medicine and Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, Hunan, China
| | - Gang Pei
- Hunan University of Chinese Medicine and Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, Hunan, China
| | - Zhenzhen Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Naihong Chen
- Hunan University of Chinese Medicine and Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, Hunan, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Naihong Chen,
| |
Collapse
|
38
|
Barrutieta-Arberas I, Ortuzar N, Vaquero-Rodríguez A, Picó-Gallardo M, Bengoetxea H, Guevara MA, Gargiulo PA, Lafuente JV. The role of ketamine in major depressive disorders: Effects on parvalbumin-positive interneurons in hippocampus. Exp Biol Med (Maywood) 2023; 248:588-595. [PMID: 37158084 PMCID: PMC10350797 DOI: 10.1177/15353702231170007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023] Open
Abstract
Major depressive disorder (MDD) is a complex illness that is arising as a growing public health concern. Although several brain areas are related to this type of disorders, at the cellular level, the parvalbumin-positive cells of the hippocampus interplay a very relevant role. They control pyramidal cell bursts, neuronal networks, basic microcircuit functions, and other complex neuronal tasks involved in mood disorders. In resistant depressions, the efficacy of current antidepressant treatments drops dramatically, so the new rapid-acting antidepressants (RAADs) are being postulated as novel treatments. Ketamine at subanesthetic doses and its derivative metabolites have been proposed as RAADs due to their rapid and sustained action by blocking N-methyl-d-aspartate (NMDA) receptors, which in turn lead to the release of brain-derived neurotrophic factor (BDNF). This mechanism produces a rapid plasticity activation mediated by neurotransmitter homeostasis, synapse recovery, and increased dendritic spines and therefore, it is a promising therapeutic approach to improve cognitive symptoms in MDD.
Collapse
Affiliation(s)
- I Barrutieta-Arberas
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - N Ortuzar
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Neurodegenerative Diseases Group, BioCruces Health Research Institute, 48903 Barakaldo, Spain
| | - A Vaquero-Rodríguez
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Neurodegenerative Diseases Group, BioCruces Health Research Institute, 48903 Barakaldo, Spain
| | - M Picó-Gallardo
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - H Bengoetxea
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Neurodegenerative Diseases Group, BioCruces Health Research Institute, 48903 Barakaldo, Spain
| | - MA Guevara
- Laboratory of Neurosciences and Experimental Psychology, Area of Pharmacology, Department of Pathology, Faculty of Medical Sciences, National Council of Scientific and Technical Research, National University of Cuyo, 5502 Mendoza, Argentina
| | - PA Gargiulo
- Laboratory of Neurosciences and Experimental Psychology, Area of Pharmacology, Department of Pathology, Faculty of Medical Sciences, National Council of Scientific and Technical Research, National University of Cuyo, 5502 Mendoza, Argentina
| | - JV Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Neurodegenerative Diseases Group, BioCruces Health Research Institute, 48903 Barakaldo, Spain
| |
Collapse
|
39
|
Johnston JN, Greenwald MS, Henter ID, Kraus C, Mkrtchian A, Clark NG, Park LT, Gold P, Zarate CA, Kadriu B. Inflammation, stress and depression: An exploration of ketamine's therapeutic profile. Drug Discov Today 2023; 28:103518. [PMID: 36758932 PMCID: PMC10050119 DOI: 10.1016/j.drudis.2023.103518] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/13/2022] [Accepted: 01/31/2023] [Indexed: 02/09/2023]
Abstract
Well-established animal models of depression have described a proximal relationship between stress and central nervous system (CNS) inflammation - a relationship mirrored in the peripheral inflammatory biomarkers of individuals with depression. Evidence also suggests that stress-induced proinflammatory states can contribute to the neurobiology of treatment-resistant depression. Interestingly, ketamine, a rapid-acting antidepressant, can partially exert its therapeutic effects via anti-inflammatory actions on the hypothalamic-pituitary adrenal (HPA) axis, the kynurenine pathway or by cytokine suppression. Further investigations into the relationship between ketamine, inflammation and stress could provide insight into ketamine's unique therapeutic mechanisms and stimulate efforts to develop rapid-acting, anti-inflammatory-based antidepressants.
Collapse
Affiliation(s)
- Jenessa N Johnston
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Maximillian S Greenwald
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Ioline D Henter
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Christoph Kraus
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Anahit Mkrtchian
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Neil G Clark
- US School of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Lawrence T Park
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Philip Gold
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Carlos A Zarate
- 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
| |
Collapse
|
40
|
Garcia GP, Perez GM, Gasperi RD, Sosa MAG, Otero-Pagan A, Abutarboush R, Kawoos U, Statz JK, Patterson J, Zhu CW, Hof PR, Cook DG, Ahlers ST, Elder GA. (2R,6R)-Hydroxynorketamine Treatment of Rats Exposed to Repetitive Low-Level Blast Injury. Neurotrauma Rep 2023; 4:197-217. [PMID: 37020715 PMCID: PMC10068674 DOI: 10.1089/neur.2022.0088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023] Open
Abstract
Many military veterans who experienced blast-related traumatic brain injuries (TBIs) in the conflicts in Iraq and Afghanistan suffer from chronic cognitive and mental health problems, including post-traumatic stress disorder (PTSD). Male rats subjected to repetitive low-level blast exposure develop chronic cognitive and PTSD-related traits that develop in a delayed manner. Ketamine has received attention as a treatment for refractory depression and PTSD. (2R,6R)-hydroxynorketamine [(2R,6R)-HNK] is a ketamine metabolite that exerts rapid antidepressant actions. (2R,6R)-HNK has become of clinical interest because of its favorable side-effect profile, low abuse potential, and oral route of administration. We treated three cohorts of blast-exposed rats with (2R,6R)-HNK, beginning 7-11 months after blast exposure, a time when the behavioral phenotype is established. Each cohort consisted of groups (n = 10-13/group) as follows: 1) Sham-exposed treated with saline, 2) blast-exposed treated with saline, and 3) blast-exposed treated with a single dose of 20 mg/kg of (2R,6R)-HNK. (2R,6R)-HNK rescued blast-induced deficits in novel object recognition (NOR) and anxiety-related features in the elevated zero maze (EZM) in all three cohorts. Exaggerated acoustic startle was reversed in cohort 1, but not in cohort 3. (2R,6R)-HNK effects were still present in the EZM 12 days after administration in cohort 1 and 27 days after administration in NOR testing of cohorts 2 and 3. (2R,6R)-HNK may be beneficial for the neurobehavioral syndromes that follow blast exposure in military veterans. Additional studies will be needed to determine whether higher doses or more extended treatment regimens may be more effective.
Collapse
Affiliation(s)
- Georgina Perez Garcia
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Gissel M. Perez
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Rita De Gasperi
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Miguel A. Gama Sosa
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- General Medical Research Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Alena Otero-Pagan
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Rania Abutarboush
- Department of Neurotrauma, Naval Medical Research Center, Silver Spring, Maryland, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Usmah Kawoos
- Department of Neurotrauma, Naval Medical Research Center, Silver Spring, Maryland, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Jonathan K. Statz
- Department of Neurotrauma, Naval Medical Research Center, Silver Spring, Maryland, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Jacob Patterson
- Department of Neurotrauma, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Carolyn W. Zhu
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mount Sinai Alzheimer's Disease Research Center and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Patrick R. Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mount Sinai Alzheimer's Disease Research Center and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - David G. Cook
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Stephen T. Ahlers
- Department of Neurotrauma, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Gregory A. Elder
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mount Sinai Alzheimer's Disease Research Center and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| |
Collapse
|
41
|
Xu Y, Yu Z, Chen S, Li Z, Long X, Chen M, Lee CS, Peng HY, Lin TB, Hsieh MC, Lai CY, Chou D. (2R,6R)-hydroxynorketamine targeting the basolateral amygdala regulates fear memory. Neuropharmacology 2023; 225:109402. [PMID: 36565854 DOI: 10.1016/j.neuropharm.2022.109402] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/28/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
(2R,6R)-Hydroxynorketamine (HNK), a ketamine metabolite, has been proposed as an ideal next-generation antidepressant due to its rapid-acting and long-lasting antidepression-relevant actions. Interestingly, recent studies have shown that (2R,6R)-HNK may have diverse impacts on memory formation. However, its effect on fear memory extinction is still unknown. In the present study, we assessed the effects of (2R,6R)-HNK on synaptic transmission and plasticity in the basolateral amygdala (BLA) and explored its actions on auditory fear memory extinction. Adult male C57BL/6J mice were used in this study. The extracellular electrophysiological recording was conducted to assay synaptic transmission and plasticity. The auditory fear conditioning paradigm was performed to test fear extinction. The results showed that (2R,6R)-HNK at 30 mg/kg increased the number of c-fos-positive cells in the BLA. Moreover, (2R,6R)-HNK enhanced the induction and maintenance of long-term potentiation (LTP) in the BLA in a dose-dependent manner (at 1, 10, and 30 mg/kg). In addition, (2R,6R)-HNK at 30 mg/kg and directly slice perfusion of (2R,6R)-HNK enhanced BLA synaptic transmission. Furthermore, intra-BLA application and systemic administration of (2R,6R)-HNK reduced the retrieval of recent fear memory and decreased the retrieval of remote fear memory. Both local and systemic (2R,6R)-HNK also inhibited the spontaneous recovery of remote fear memory. Taken together, these results indicated that (2R,6R)-HNK could regulate BLA synaptic transmission and plasticity and act through the BLA to modulate fear memory. The results revealed that (2R,6R)-HNK may be a potential drug to treat posttraumatic stress disorder (PTSD) patients.
Collapse
Affiliation(s)
- Yuanyuan Xu
- Department of Pharmacology, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China.
| | - Zhenfei Yu
- Department of Pharmacology, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China.
| | - Si Chen
- Department of Human Anatomy and Histology & Embryology, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China.
| | - Zhenlong Li
- School of Basic Medical Sciences, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China.
| | - Xiting Long
- Department of Pharmacology, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China.
| | - Mengxu Chen
- Department of Pharmacology, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China.
| | - Chau-Shoun Lee
- Department of Medicine, MacKay Medical College, New Taipei, Taiwan; Department of Psychiatry, MacKay Memorial Hospital, Taipei, Taiwan.
| | - Hsien-Yu Peng
- Department of Medicine, MacKay Medical College, New Taipei, Taiwan.
| | - Tzer-Bin Lin
- Institute of New Drug Development, College of Medicine, China Medical University, Taichung, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Ming-Chun Hsieh
- Department of Medicine, MacKay Medical College, New Taipei, Taiwan.
| | - Cheng-Yuan Lai
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei, Taiwan.
| | - Dylan Chou
- Department of Medicine, MacKay Medical College, New Taipei, Taiwan; Department of Physiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China.
| |
Collapse
|
42
|
Zanos P, Brown KA, Georgiou P, Yuan P, Zarate CA, Thompson SM, Gould TD. NMDA Receptor Activation-Dependent Antidepressant-Relevant Behavioral and Synaptic Actions of Ketamine. J Neurosci 2023; 43:1038-1050. [PMID: 36596696 PMCID: PMC9908316 DOI: 10.1523/jneurosci.1316-22.2022] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 11/30/2022] [Accepted: 12/18/2022] [Indexed: 01/05/2023] Open
Abstract
Ketamine is a well-characterized NMDA receptor (NMDAR) antagonist, although the relevance of this pharmacology to its rapid (within hours of administration) antidepressant actions, which depend on mechanisms convergent with strengthening of excitatory synapses, is unclear. Activation of synaptic NMDARs is necessary for the induction of canonical long-term potentiation (LTP) leading to a sustained expression of increased synaptic strength. We tested the hypothesis that induction of rapid antidepressant effects requires NMDAR activation, by using behavioral pharmacology, western blot quantification of hippocampal synaptoneurosomal protein levels, and ex vivo hippocampal slice electrophysiology in male mice. We found that ketamine exerts an inverted U-shaped dose-response in antidepressant-sensitive behavioral tests, suggesting that an excessive NMDAR inhibition can prevent ketamine's antidepressant effects. Ketamine's actions to induce antidepressant-like behavioral effects, up-regulation of hippocampal AMPAR subunits GluA1 and GluA2, as well as metaplasticity measured ex vivo using electrically-stimulated LTP, were abolished by pretreatment with other non-antidepressant NMDAR antagonists, including MK-801 and CPP. Similarly, the antidepressant-like actions of other putative rapid-acting antidepressant drugs (2R,6R)-hydroxynorketamine (ketamine metabolite), MRK-016 (GABAAα5 negative allosteric modulator), and LY341495 (mGlu2/3 receptor antagonist) were blocked by NMDAR inhibition. Ketamine acted synergistically with an NMDAR positive allosteric modulator to exert antidepressant-like behavioral effects and activation of the NMDAR subunit GluN2A was necessary and sufficient for such relevant effects. We conclude rapid-acting antidepressant compounds share a common downstream NMDAR-activation dependent effector mechanism, despite variation in initial pharmacological targets. Promoting NMDAR signaling or other approaches that enhance NMDAR-dependent LTP-like synaptic potentiation may be an effective antidepressant strategy.SIGNIFICANCE STATEMENT The anesthetic and antidepressant drug ketamine is well-characterized as an NMDA receptor (NMDAR) antagonist; though, the relevance and full impact of this pharmacology to its antidepressant actions is unclear. We found that NMDAR activation, which occurs downstream of their initial actions, is necessary for the beneficial effects of ketamine and several other putative antidepressant compounds. As such, promoting NMDAR signaling, or other approaches that enhance NMDAR-dependent long-term potentiation (LTP)-like synaptic potentiation in vivo may be an effective antidepressant strategy directly, or acting synergistically with other drug or interventional treatments.
Collapse
Affiliation(s)
- Panos Zanos
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Maryland 21201
- Department of Psychology, University of Cyprus, Nicosia 2109, Cyprus
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| | - Kyle A Brown
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| | - Polymnia Georgiou
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Maryland 21201
- Department of Biology, University of Cyprus, Nicosia 2109, Cyprus
| | - Peixiong Yuan
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Scott M Thompson
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Maryland 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| | - Todd D Gould
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Maryland 21201
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
- Department of Anatomy & Neurobiology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
- Veterans Affairs Maryland Health Care System, Baltimore, Maryland 21201
| |
Collapse
|
43
|
Toki H, Yamaguchi JI, Mizuno-Yasuhira A, Endo H. Chiral LC-MS/MS method for the simultaneous determination of (R,S)-ketamine, (R,S)-norketamine, and (2R,6R;2S,6S)-hydroxynorketamine in mouse plasma and brain. J Pharm Biomed Anal 2023; 224:115168. [PMID: 36473323 DOI: 10.1016/j.jpba.2022.115168] [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: 08/08/2022] [Revised: 11/13/2022] [Accepted: 11/14/2022] [Indexed: 11/25/2022]
Abstract
A convenient LC-MS/MS assay method to simultaneously and sensitively determine (R,S)-ketamine (Ket), (R,S)-norketamine (NK), and (2R,6R;2S,6S)-hydroxynorketamine (HNK) enantiomers in plasma and brain from mice was developed. This method enables the chiral separations of these six enantiomers in one analysis by constructing a column-switching system composed of one achiral column and two chiral columns with a relatively short analysis time (17 min). The chromatography involves the separation of (2R,6R;2S,6S)-HNK from (R,S)-Ket and (R,S)-NK on an octadecyl-silica column, followed by chiral separations on a CHIRALPAK AY-RH column for (2R,6R;2S,6S)-HNK or on a CHIRALPAK AS-RH column for the other analytes. The calibration curves for plasma and brain showed a good linearity in the range of 3-1000 ng/mL and 1.5-500 ng/g, respectively. The accuracy ranged from 90.0% to 104.0% in within-run and between-run. This validated method was applicable to determine the stereoselective pharmacokinetic profiles of (R,S)-Ket, (R,S)-NK, and (2R,6R;2S,6S)-HNK in plasma and brain collected from individual mice after a single intraperitoneal dosing of racemic Ket at an antidepressant dose. It is hoped that this assay will greatly help for understanding the relationship between the antidepressant actions of (R,S)-Ket enantiomers or their metabolites and their pharmacokinetics.
Collapse
Affiliation(s)
- Hidetoh Toki
- Drug Metabolism and Pharmacokinetics, Drug Safety and Pharmacokinetics Laboratories Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-cho, Kita-ku, Saitama-shi, Saitama 331-9530, Japan.
| | - Jun-Ichi Yamaguchi
- Drug Metabolism and Pharmacokinetics, Drug Safety and Pharmacokinetics Laboratories Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-cho, Kita-ku, Saitama-shi, Saitama 331-9530, Japan
| | - Akiko Mizuno-Yasuhira
- Drug Metabolism and Pharmacokinetics, Drug Safety and Pharmacokinetics Laboratories Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-cho, Kita-ku, Saitama-shi, Saitama 331-9530, Japan
| | - Hiromi Endo
- Drug Metabolism and Pharmacokinetics, Drug Safety and Pharmacokinetics Laboratories Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-cho, Kita-ku, Saitama-shi, Saitama 331-9530, Japan
| |
Collapse
|
44
|
Onisiforou A, Georgiou P, Zanos P. Role of group II metabotropic glutamate receptors in ketamine's antidepressant actions. Pharmacol Biochem Behav 2023; 223:173531. [PMID: 36841543 DOI: 10.1016/j.pbb.2023.173531] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/08/2023] [Accepted: 02/15/2023] [Indexed: 02/26/2023]
Abstract
Major Depressive Disorder (MDD) is a serious neuropsychiatric disorder afflicting around 16-17 % of the global population and is accompanied by recurrent episodes of low mood, hopelessness and suicidal thoughts. Current pharmacological interventions take several weeks to even months for an improvement in depressive symptoms to emerge, with a significant percentage of individuals not responding to these medications at all, thus highlighting the need for rapid and effective next-generation treatments for MDD. Pre-clinical studies in animals have demonstrated that antagonists of the metabotropic glutamate receptor subtype 2/3 (mGlu2/3 receptor) exert rapid antidepressant-like effects, comparable to the actions of ketamine. Therefore, it is possible that mGlu2 or mGlu3 receptors to have a regulatory role on the unique antidepressant properties of ketamine, or that convergent intracellular mechanisms exist between mGlu2/3 receptor signaling and ketamine's effects. Here, we provide a comprehensive and critical evaluation of the literature on these convergent processes underlying the antidepressant action of mGlu2/3 receptor inhibitors and ketamine. Importantly, combining sub-threshold doses of mGlu2/3 receptor inhibitors with sub-antidepressant ketamine doses induce synergistic antidepressant-relevant behavioral effects. We review the evidence supporting these combinatorial effects since sub-effective dosages of mGlu2/3 receptor antagonists and ketamine could reduce the risk for the emergence of significant adverse events compared with taking normal dosages. Overall, deconvolution of ketamine's pharmacological targets will give critical insights to influence the development of next-generation antidepressant treatments with rapid actions.
Collapse
Affiliation(s)
- Anna Onisiforou
- Department of Psychology, University of Cyprus, Nicosia 2109, Cyprus
| | - Polymnia Georgiou
- Department of Biological Sciences, University of Cyprus, Nicosia 2109, Cyprus; Department of Psychology, University of Wisconsin Milwaukee, WI 53211, USA
| | - Panos Zanos
- Department of Psychology, University of Cyprus, Nicosia 2109, Cyprus.
| |
Collapse
|
45
|
Ago Y, Yokoyama R, Asano S, Hashimoto H. Roles of the monoaminergic system in the antidepressant effects of ketamine and its metabolites. Neuropharmacology 2023; 223:109313. [PMID: 36328065 DOI: 10.1016/j.neuropharm.2022.109313] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
Abstract
While the molecular target of (R,S)-ketamine (ketamine) is thought to be the NMDA receptor, subanesthetic doses of ketamine have been known to modulate monoaminergic neurotransmission in the central nervous system. Although the involvement of the serotonergic system in the antidepressant effects of ketamine has been reported in most studies of this topic, some recent studies have reported that the dopaminergic system plays a key role in the effects of ketamine. Additionally, several lines of evidence suggest that the antidepressant-like effects of (R)-ketamine might be independent of the monoaminergic system. Ketamine metabolites also differ considerably in their ability to regulate monoamine neurotransmitters relative to (S)-ketamine and (R)-ketamine, while (2R,6R)-hydroxynorketamine might share common serotonergic signaling mechanisms with ketamine. In the current review, we summarize the effects of ketamine and its metabolites on monoamine neurotransmission in the brain and discuss the potential roles of the monoaminergic system in the mechanism of action of ketamine.
Collapse
Affiliation(s)
- Yukio Ago
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, 734-8553, Japan.
| | - Rei Yokoyama
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Satoshi Asano
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, 734-8553, Japan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, 565-0871, Japan; Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka, 565-0871, Japan; Division of Bioscience, Institute for Datability Science, Osaka University, Suita, Osaka, 565-0871, Japan; Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, 565-0871, Japan; Department of Molecular Pharmaceutical Science, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
46
|
Goswami N, Aleem M, Manda K. Intranasal (2R, 6R)-hydroxynorketamine for acute pain: Behavioural and neurophysiological safety analysis in mice. Clin Exp Pharmacol Physiol 2023; 50:169-177. [PMID: 36371631 DOI: 10.1111/1440-1681.13737] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 10/12/2022] [Accepted: 11/09/2022] [Indexed: 11/14/2022]
Abstract
Ketamine is known for its antinociceptive effect and is also used for treatment-resistant depression. However, the efficacy and safety of (2R, 6R)-hydroxynorketamine (HNK), a ketamine metabolite has been sparingly investigated for acute pain management. The current study aims at investigating the antinociceptive effect of intranasal (2R, 6R)-HNK using pre-clinical models of acute pain. Additionally, the behavioural and neurophysiological safety analyses were carried out for the effective time window. Antinociceptive efficacy of (2R, 6R)-HNK was evaluated using the hot plate test and Hargreaves' plantar test. The formalin test was carried out in both the acute and tonic phases. The neurophysiological and behavioural safety analyses were carried out separately for the haemodynamic function, cortical electroencephalography (EEG), and spontaneous behavioural functions. Analgesic effect of (2R, 6R)-HNK was evident by a significant increase in paw-withdrawal latency in both Hargreaves' and hot plate tests. Additionally, the (2R, 6R)-HNK showed a significant ameliorative effect on pain-related behaviour in the second phase of the formalin test. (2R, 6R)-HNK exhibited an anxiolytic effect without causing any significant changes in locomotor activity and haemodynamic parameters. Power spectral density (PSD) analysis of electroencephalogram revealed no significant changes except a comparative increase in the gamma band range. Both the locomotor functions in the open field test and the PSD value of delta wave indicated no sedative effect at the given dose of (2R, 6R)-HNK. The results demonstrated the pain-alleviating effect of (2R, 6R)-HNK without compromising the neurophysiological and behavioural function. Therefore, intranasal (2R, 6R)-HNK is suggested as a safe candidate for further clinical study in the management of acute pain.
Collapse
Affiliation(s)
- Nidhi Goswami
- Division of Behavioral Neuroscience, Institute of Nuclear Medicine & Allied Sciences, Delhi, India
| | - Mohd Aleem
- Division of Behavioral Neuroscience, Institute of Nuclear Medicine & Allied Sciences, Delhi, India
| | - Kailash Manda
- Division of Behavioral Neuroscience, Institute of Nuclear Medicine & Allied Sciences, Delhi, India
| |
Collapse
|
47
|
Role of mTOR1 signaling in the antidepressant effects of ketamine and the potential of mTORC1 activators as novel antidepressants. Neuropharmacology 2023; 223:109325. [PMID: 36334763 DOI: 10.1016/j.neuropharm.2022.109325] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/29/2022] [Accepted: 10/31/2022] [Indexed: 11/11/2022]
Abstract
Conventional antidepressant medications act on monoaminergic systems and have important limitations, including a therapeutic delay of weeks to months and low rates of efficacy. Recently, clinical findings have indicated that ketamine, a dissociative anesthetic that blocks N-methyl-d-aspartate receptor channel activity, causes rapid and long-lasting antidepressant effects. Although the exact mechanisms underlying the antidepressant effects of ketamine are not fully known, preclinical studies have demonstrated a key role for mechanistic target of rapamycin complex 1 (mTORC1) signaling and a subsequent increase in synapse formation in the medial prefrontal cortex. In this review, we discuss the role of mTORC1 and its subsequent signaling cascade in the antidepressant effects of ketamine and other compounds with glutamatergic mechanisms of action. We also present the possibility that mTORC1 signaling itself is a drug discovery target.
Collapse
|
48
|
Abdulghani A, Poghosyan M, Mehren A, Philipsen A, Anderzhanova E. Neuroplasticity to autophagy cross-talk in a therapeutic effect of physical exercises and irisin in ADHD. Front Mol Neurosci 2023; 15:997054. [PMID: 36776770 PMCID: PMC9909442 DOI: 10.3389/fnmol.2022.997054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 12/30/2022] [Indexed: 01/28/2023] Open
Abstract
Adaptive neuroplasticity is a pivotal mechanism for healthy brain development and maintenance, as well as its restoration in disease- and age-associated decline. Management of mental disorders such as attention deficit hyperactivity disorder (ADHD) needs interventions stimulating adaptive neuroplasticity, beyond conventional psychopharmacological treatments. Physical exercises are proposed for the management of ADHD, and also depression and aging because of evoked brain neuroplasticity. Recent progress in understanding the mechanisms of muscle-brain cross-talk pinpoints the role of the myokine irisin in the mediation of pro-cognitive and antidepressant activity of physical exercises. In this review, we discuss how irisin, which is released in the periphery as well as derived from brain cells, may interact with the mechanisms of cellular autophagy to provide protein recycling and regulation of brain-derived neurotrophic factor (BDNF) signaling via glia-mediated control of BDNF maturation, and, therefore, support neuroplasticity. We propose that the neuroplasticity associated with physical exercises is mediated in part by irisin-triggered autophagy. Since the recent findings give objectives to consider autophagy-stimulating intervention as a prerequisite for successful therapy of psychiatric disorders, irisin appears as a prototypic molecule that can activate autophagy with therapeutic goals.
Collapse
Affiliation(s)
- Alhasan Abdulghani
- C. and O. Vogt Institute for Brain Research, Medical Faculty and University Hospital Düsseldorf, Henrich Heine University, Düsseldorf, Düsseldorf, Germany,*Correspondence: Alhasan Abdulghani,
| | - Mikayel Poghosyan
- Institute for Biology-Neurobiology, Freie University of Berlin, Berlin, Germany
| | - Aylin Mehren
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Alexandra Philipsen
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Elmira Anderzhanova
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
49
|
Ji N, Lei M, Chen Y, Tian S, Li C, Zhang B. How Oxidative Stress Induces Depression? ASN Neuro 2023; 15:17590914231181037. [PMID: 37331994 DOI: 10.1177/17590914231181037] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023] Open
Abstract
Depression increasingly affects a wide range and a large number of people worldwide, both physically and psychologically, which makes it a social problem requiring prompt attention and management. Accumulating clinical and animal studies have provided us with substantial insights of disease pathogenesis, especially central monoamine deficiency, which considerably promotes antidepressant research and clinical treatment. The first-line antidepressants mainly target the monoamine system, whose drawbacks mainly include slow action and treatment resistant. The novel antidepressant esketamine, targeting on central glutamatergic system, rapidly and robustly alleviates depression (including treatment-resistant depression), whose efficiency is shadowed by potential addictive and psychotomimetic side effects. Thus, exploring novel depression pathogenesis is necessary, for seeking more safe and effective therapeutic methods. Emerging evidence has revealed vital involvement of oxidative stress (OS) in depression, which inspires us to pursue antioxidant pathway for depression prevention and treatment. Fully uncovering the underlying mechanisms of OS-induced depression is the first step towards the avenue, thus we summarize and expound possible downstream pathways of OS, including mitochondrial impairment and related ATP deficiency, neuroinflammation, central glutamate excitotoxicity, brain-derived neurotrophic factor/tyrosine receptor kinase B dysfunction and serotonin deficiency, the microbiota-gut-brain axis disturbance and hypothalamic-pituitary-adrenocortical axis dysregulation. We also elaborate on the intricate interactions between the multiple aspects, and molecular mechanisms mediating the interplay. Through reviewing the related research progress in the field, we hope to depict an integral overview of how OS induces depression, in order to provide fresh ideas and novel targets for the final goal of efficient treatment of the disease.
Collapse
Affiliation(s)
- Na Ji
- The School of Public Health, Faculty of Basic Medical Sciences, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin Guangxi, China
| | - Mengzhu Lei
- The School of Public Health, Faculty of Basic Medical Sciences, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin Guangxi, China
| | - Yating Chen
- The School of Public Health, Faculty of Basic Medical Sciences, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin Guangxi, China
| | - Shaowen Tian
- The School of Public Health, Faculty of Basic Medical Sciences, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin Guangxi, China
| | - Chuanyu Li
- The School of Public Health, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin Guangxi, China
| | - Bo Zhang
- The School of Public Health, Faculty of Basic Medical Sciences, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin Guangxi, China
| |
Collapse
|
50
|
Kan HW, Peng WH, Wu CC, Wang DW, Lee MT, Lee YK, Chu TH, Ho YC. Rapid antidepressant-like effects of muscarinic receptor antagonists require BDNF-dependent signaling in the ventrolateral periaqueductal gray. Psychopharmacology (Berl) 2022; 239:3805-3818. [PMID: 36221037 DOI: 10.1007/s00213-022-06250-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/26/2022] [Indexed: 10/17/2022]
Abstract
RATIONALE Clinical reports reveal that scopolamine, an acetylcholine muscarinic receptor antagonist, exerts rapid antidepressant effects in depressed patients, but the mechanisms underlying the therapeutic effects have not been fully identified. OBJECTIVES The present study examines the cellular mechanisms by which scopolamine produces antidepressant-like effects through its action in the ventrolateral midbrain periaqueductal gray (vlPAG). METHODS We used a well-established mouse model of depression induced by chronic restraint stress (CRS) exposure for 14 days. Behaviors were tested using the forced swim test (FST), tail suspension test (TST), female urine sniffing test (FUST), novelty-suppressed feeding test (NSFT), and locomotor activity (LMA). Synaptic transmission in the vlPAG was measured by whole-cell patch-clamp recordings. IntravlPAG microinjection was used to pharmacologically verify the signaling cascades of scopolamine in the vlPAG. RESULTS The results demonstrated that intraperitoneal injection of scopolamine produced antidepressant-like effects in a dose-dependent manner without affecting locomotor activity. CRS elicited depression-like behaviors, whereas intraperitoneal injection of scopolamine alleviated CRS-induced depression-like behaviors. CRS diminished glutamatergic transmission in the vlPAG, while scopolamine reversed the above effects. Moreover, intravlPAG microinjection of the L-type voltage-dependent calcium channel (VDCC) blocker verapamil, tropomyosin-related kinase B (TrkB) receptor antagonist ANA-12, mammalian target of rapamycin complex 1 (mTORC1) inhibitor rapamycin, and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPA) antagonist CNQX prevented scopolamine-induced antidepressant-like effects. CONCLUSIONS Scopolamine ameliorated CRS-elicited depression-like behavior required activation of VDCC, resulting in activity-dependent release of brain-derived neurotrophic factor (BDNF), engaging the TrkB receptor and downstream mTORC1 signaling in the vlPAG.
Collapse
Affiliation(s)
- Hung-Wei Kan
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung City, 82445, Taiwan, Republic of China
| | - Wei-Hao Peng
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung City, 82445, Taiwan, Republic of China.,School of Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan, Republic of China
| | - Cheng-Chun Wu
- School of Medicine, College of Medicine, I-Shou University, No.8, Yida Rd., Yanchao District, Kaohsiung City, 82445, Taiwan, Republic of China
| | - Deng-Wu Wang
- School of Medicine, College of Medicine, I-Shou University, No.8, Yida Rd., Yanchao District, Kaohsiung City, 82445, Taiwan, Republic of China.,Department of Psychiatry, E-Da Hospital, Kaohsiung City, 82445, Taiwan, Republic of China
| | - Ming Tatt Lee
- Faculty of Pharmaceutical Sciences, UCSI University, 56000, Cheras, Kuala Lumpur, Malaysia
| | - Yung-Kuo Lee
- Medical Laboratory, Medical Education and Research Center, Kaohsiung Armed Forces General Hospital, Kaohsiung City, 80284, Taiwan, Republic of China
| | - Tian-Huei Chu
- Medical Laboratory, Medical Education and Research Center, Kaohsiung Armed Forces General Hospital, Kaohsiung City, 80284, Taiwan, Republic of China
| | - Yu-Cheng Ho
- School of Medicine, College of Medicine, I-Shou University, No.8, Yida Rd., Yanchao District, Kaohsiung City, 82445, Taiwan, Republic of China.
| |
Collapse
|