1
|
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
|
2
|
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
|
3
|
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
|
4
|
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
|
5
|
Chen T, Cheng L, Ma J, Yuan J, Pi C, Xiong L, Chen J, Liu H, Tang J, Zhong Y, Zhang X, Liu Z, Zuo Y, Shen H, Wei Y, Zhao L. Molecular mechanisms of rapid-acting antidepressants: New perspectives for developing antidepressants. Pharmacol Res 2023; 194:106837. [PMID: 37379962 DOI: 10.1016/j.phrs.2023.106837] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 06/11/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023]
Abstract
Major depressive disorder (MDD) is a chronic relapsing psychiatric disorder. Conventional antidepressants usually require several weeks of continuous administration to exert clinically significant therapeutic effects, while about two-thirds of the patients are prone to relapse of symptoms or are completely ineffective in antidepressant treatment. The recent success of the N-methyl-D-aspartic acid (NMDA) receptor antagonist ketamine as a rapid-acting antidepressant has propelled extensive research on the action mechanism of antidepressants, especially in relation to its role in synaptic targets. Studies have revealed that the mechanism of antidepressant action of ketamine is not limited to antagonism of postsynaptic NMDA receptors or GABA interneurons. Ketamine produces powerful and rapid antidepressant effects by affecting α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptors, adenosine A1 receptors, and the L-type calcium channels, among others in the synapse. More interestingly, the 5-HT2A receptor agonist psilocybin has demonstrated potential for rapid antidepressant effects in depressed mouse models and clinical studies. This article focuses on a review of new pharmacological target studies of emerging rapid-acting antidepressant drugs such as ketamine and hallucinogens (e.g., psilocybin) and briefly discusses the possible strategies for new targets of antidepressants, with a view to shed light on the direction of future antidepressant research.
Collapse
Affiliation(s)
- Tao Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou 646000 China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000 China; Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan 646000, China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ling Cheng
- Hospital-Acquired Infection Control Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jingwen Ma
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou 646000 China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000 China; Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan 646000, China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jiyuan Yuan
- Clinical trial center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chao Pi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou 646000 China
| | - Linjin Xiong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou 646000 China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000 China; Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan 646000, China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jinglin Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou 646000 China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000 China; Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan 646000, China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Huiyang Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou 646000 China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000 China; Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan 646000, China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jia Tang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou 646000 China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000 China; Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan 646000, China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yueting Zhong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou 646000 China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000 China; Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan 646000, China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaomei Zhang
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, Institute of medicinal chemistry of Chinese Medicine, Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China
| | - Zerong Liu
- Central Nervous System Drug Key Laboratory of Sichuan Province, Sichuan Credit Pharmaceutical CO., Ltd., Luzhou, Sichuan 646000, China; Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Ying Zuo
- Department of Comprehensive Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University; Luzhou, Sichuan 646000, China
| | - Hongping Shen
- Clinical trial center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Yumeng Wei
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou 646000 China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Ling Zhao
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000 China; Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan 646000, China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
6
|
Hashimoto K, Chaki S. Ketamine and its metabolites: Potential as novel treatments for depression. Neuropharmacology 2023; 230:109492. [PMID: 36889434 DOI: 10.1016/j.neuropharm.2023.109492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Affiliation(s)
- Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan.
| | - Shigeyuki Chaki
- Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama, Saitama, 331-9530, Japan.
| |
Collapse
|
7
|
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
|
8
|
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: 39] [Impact Index Per Article: 19.5] [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
|
9
|
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
|
10
|
Georgiou P, Zanos P, Mou TCM, An X, Gerhard DM, Dryanovski DI, Potter LE, Highland JN, Jenne CE, Stewart BW, Pultorak KJ, Yuan P, Powels CF, Lovett J, Pereira EFR, Clark SM, Tonelli LH, Moaddel R, Zarate CA, Duman RS, Thompson SM, Gould TD. Experimenters' sex modulates mouse behaviors and neural responses to ketamine via corticotropin releasing factor. Nat Neurosci 2022; 25:1191-1200. [PMID: 36042309 PMCID: PMC10186684 DOI: 10.1038/s41593-022-01146-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/14/2022] [Indexed: 11/09/2022]
Abstract
We show that the sex of human experimenters affects mouse behaviors and responses following administration of the rapid-acting antidepressant ketamine and its bioactive metabolite (2R,6R)-hydroxynorketamine. Mice showed aversion to the scent of male experimenters, preference for the scent of female experimenters and increased stress susceptibility when handled by male experimenters. This human-male-scent-induced aversion and stress susceptibility was mediated by the activation of corticotropin-releasing factor (CRF) neurons in the entorhinal cortex that project to hippocampal area CA1. Exposure to the scent of male experimenters before ketamine administration activated CA1-projecting entorhinal cortex CRF neurons, and activation of this CRF pathway modulated in vivo and in vitro antidepressant-like effects of ketamine. A better understanding of the specific and quantitative contributions of the sex of human experimenters to study outcomes in rodents may improve replicability between studies and, as we have shown, reveal biological and pharmacological mechanisms.
Collapse
Affiliation(s)
- Polymnia Georgiou
- Veterans Affairs Maryland Health Care System, Baltimore, MD, USA.,Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA.,Department of Biology, University of Cyprus, Nicosia, Cyprus
| | - Panos Zanos
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA.,Department of Psychology, University of Cyprus, Nicosia, Cyprus
| | - Ta-Chung M Mou
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Xiaoxian An
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Danielle M Gerhard
- Department of Psychiatry, Yale University, New Haven, CT, USA.,Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Dilyan I Dryanovski
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Liam E Potter
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA.,Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jaclyn N Highland
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA.,The Graduate Program in Toxicology, University of Maryland, Baltimore, MD, USA
| | - Carleigh E Jenne
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Brent W Stewart
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA.,The Graduate Program in Neuroscience, University of Maryland, Baltimore, MD, USA
| | - Katherine J Pultorak
- The Graduate Program in Neuroscience, University of Maryland, Baltimore, MD, USA
| | - Peixiong Yuan
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Chris F Powels
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Jacqueline Lovett
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Edna F R Pereira
- Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Sarah M Clark
- Veterans Affairs Maryland Health Care System, Baltimore, MD, USA.,Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Leonardo H Tonelli
- Veterans Affairs Maryland Health Care System, Baltimore, MD, USA.,Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Ruin Moaddel
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Ronald S Duman
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Scott M Thompson
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA.,Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Todd D Gould
- Veterans Affairs Maryland Health Care System, Baltimore, MD, USA. .,Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA. .,Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD, USA. .,Department of Anatomy and Neurobiology, School of Medicine, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|