1
|
Wang H, He Y, Tang J, Liu Y, Wu C, Li C, Sun H, Sun L. (2R, 6R)-hydroxynorketamine ameliorates PTSD-like behaviors during the reconsolidation phase of fear memory in rats by modulating the VGF/BDNF/GluA1 signaling pathway in the hippocampus. Behav Brain Res 2025; 476:115273. [PMID: 39326635 DOI: 10.1016/j.bbr.2024.115273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 09/28/2024]
Abstract
RATIONALE Fear memory, a fundamental symptom of post-traumatic stress disorder (PTSD), is improved by (2R, 6R)-hydroxynorketamine ((2R, 6R)-HNK) administration. However, the phase of fear memory in which the injected drug is the most effective at mitigating PTSD-like effects remains unknown. OBJECTIVE This study aimed to explore the effects of (2 R, 6 R)-HNK administration during three phases [acquisition (AP), reconsolidation (RP), and extinction (EP)] on PTSD-like behaviors in single prolonged stress (SPS) and contextual fear conditioning (CFC) rat models. The effects of VGF-inducible type of nerve growth factor (VGF), brain-derived neurotrophic factor (BDNF), and GluA1 on hippocampus (HIP) expression were also explored. METHODS SPS and CFC (SPSC) were used to establish a PTSD rat model. After lateral ventricle injection of 5 μL (2 R, 6 R)-HNK (0.5 nmol). Anxiety-depression-like behaviors were assessed in rats by the open field test (OFT) and elevated plus maze test (EPMT). Situational fear responses were evaluated in rodents by freezing behavior test (FBT) test. In addition, GluA1, VGF, and BDNF were assessed in the hippocampus by Western blot assay (WB) and Immunohistochemistry assay (IF). RESULTS SPSC procedure induced PTSD-like behaviors. The SPSC group had decreased spontaneous exploratory behavior and increased fear response. The (2R, 6R)-HNK group showed improved SPSC-induced reduction in GluA1, VGF, and BDNF levels in the HIP. During RP, anxiety and fear avoidance behaviors were alleviated, and the protein levels of GluA1, VGF, and BDNF in the HIP were restored. In contrast, no significant improvement was noted during AP and EP. CONCLUSIONS (2R,6R)-HNK modulates the VGF/BDNF/GluA1 signaling pathway in the hippocampus and improves PTSD-like behaviors during the reconsolidation phase of fear memory in rats, which may provide a new target for the clinical treatment and prevention of fear-related disorders such as PTSD.
Collapse
Affiliation(s)
- Han Wang
- School of Mental Health, Jining Medical University, Jining, Shandong 272067, China; School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Yuxuan He
- Department of Clinical Medicine, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Jiahao Tang
- Department of Clinical Medicine, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Yang Liu
- 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 261031, China
| | - Changjiang Li
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Hongwei Sun
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Lin Sun
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China.
| |
Collapse
|
2
|
Lakhawat SS, Mech P, Kumar A, Malik N, Kumar V, Sharma V, Bhatti JS, Jaswal S, Kumar S, Sharma PK. Intricate mechanism of anxiety disorder, recognizing the potential role of gut microbiota and therapeutic interventions. Metab Brain Dis 2024; 40:64. [PMID: 39671133 DOI: 10.1007/s11011-024-01453-1] [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: 02/21/2024] [Accepted: 09/29/2024] [Indexed: 12/14/2024]
Abstract
Anxiety is a widespread psychological disorder affecting both humans and animals. It is a typical stress reaction; however, its longer persistence can cause severe health disorders affecting the day-to-day life activities of individuals. An intriguing facet of the anxiety-related disorder can be addressed better by investigating the role of neurotransmitters in regulating emotions, provoking anxiety, analyzing the cross-talks between neurotransmitters, and, most importantly, identifying the biomarkers of the anxiety. Recent years have witnessed the potential role of the gut microbiota in human health and disorders, including anxiety. Animal models are commonly used to study anxiety disorder as they offer a simpler and more controlled environment than humans. Ultimately, developing new strategies for diagnosing and treating anxiety is of paramount interest to medical scientists. Altogether, this review article shall highlight the intricate mechanisms of anxiety while emphasizing the emerging role of gut microbiota in regulating metabolic pathways through various interaction networks in the host. In addition, the review will foster information about the therapeutic interventions of the anxiety and related disorder.
Collapse
Affiliation(s)
- Sudarshan Singh Lakhawat
- Amity Institute of Biotechnology, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur, Rajasthan, 303002, India
| | - Priyanka Mech
- Amity Institute of Biotechnology, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur, Rajasthan, 303002, India
| | - Akhilesh Kumar
- Amity Institute of Biotechnology, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur, Rajasthan, 303002, India
| | - Naveen Malik
- Amity Institute of Biotechnology, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur, Rajasthan, 303002, India
| | - Vikram Kumar
- Amity Institute of Pharmacy, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur, Rajasthan, India
| | - Vinay Sharma
- Amity Institute of Biotechnology, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur, Rajasthan, 303002, India
| | - Jasvinder Singh Bhatti
- Department of Environmental Sciences, Himachal Pradesh University, Summer Hill, Shimla, 171005, India
| | - Sunil Jaswal
- Department of Human Genetics and Molecular Medicine Central University Punjab, Bathinda, 151401, India
| | - Sunil Kumar
- Amity Institute of Biotechnology, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur, Rajasthan, 303002, India
| | - Pushpender Kumar Sharma
- Amity Institute of Biotechnology, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur, Rajasthan, 303002, India.
- Amity Centre for Nanobiotechnology and Nanomedicine, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur, Rajasthan, 303002, India.
| |
Collapse
|
3
|
Pretzel CW, Borba JV, Resmim CM, De Abreu MS, Kalueff AV, Fontana BD, Canzian J, Rosemberg DB. Ketamine modulates the exploratory dynamics and homebase-related behaviors of adult zebrafish. Pharmacol Biochem Behav 2024; 245:173892. [PMID: 39378930 DOI: 10.1016/j.pbb.2024.173892] [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: 08/15/2024] [Revised: 09/26/2024] [Accepted: 10/02/2024] [Indexed: 10/10/2024]
Abstract
Anxiety can be a protective emotion when animals face aversive conditions, but is commonly associated with various neuropsychiatric disorders when pathologically exacerbated. Drug repurposing has emerged as a valuable strategy based on utilizing the existing pharmaceuticals for new therapeutic purposes. Ketamine, traditionally used as an anesthetic, acts as a non-competitive antagonist of the glutamate N-methyl-d-aspartate (NMDA) receptor, and shows potential anxiolytic and antidepressant effects at subanesthetic doses. However, the influence of ketamine on multiple behavioral domains in vertebrates is not completely understood. Here, we evaluated the potential modulatory effect of ketamine on the spatio-temporal exploratory dynamics and homebase-related behaviors in adult zebrafish using the open field test (OFT). Animals were exposed to subanesthetic concentrations of ketamine (0, 2, 20, and 40 mg/L) for 20 min and their locomotion-, exploration- and homebase-related behaviors were assessed in a single 30-min trial. Our data revealed that acute ketamine (20 and 40 mg/L) induced hyperlocomotion, as verified by the increased total distance traveled. All concentrations tested elicited circling behavior, a stereotyped-like response which gradually reduced across the periods of test. We also observed modulatory effects of ketamine on the spatio-temporal exploratory pattern, in which the reduced thigmotaxis and homebase activity, associated with the increased average length of trips, suggest anxiolytic-like effects. Collectively, our findings support the modulatory effects of ketamine on the spatio-temporal exploratory activity, and corroborate the utility of homebase-related measurements to evaluate the behavioral dynamics in zebrafish models.
Collapse
Affiliation(s)
- Camilla W Pretzel
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - João V Borba
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Cássio M Resmim
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Murilo S De Abreu
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS, Brazil; Western Caspian University, Baku, Azerbaijan
| | - Allan V Kalueff
- Institute of Translational Biomedicine (ITBM), St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China; Suzhou Key Laboratory on Neurobiology and Cell Signaling, Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Barbara D Fontana
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Julia Canzian
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Denis B Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), USA.
| |
Collapse
|
4
|
Fan Y, Luan X, Wang X, Li H, Zhao H, Li S, Li X, Qiu Z. Exploring the Association between BDNF related Signaling Pathways and Depression: A Literature Review. Brain Res Bull 2024; 220:111143. [PMID: 39608613 DOI: 10.1016/j.brainresbull.2024.111143] [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: 09/13/2024] [Revised: 11/13/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024]
Abstract
Depression is a debilitating mental disease that inflicts significant harm upon individuals and society, yet effective treatment options remain elusive. At present, the pathogenesis of multiple depression is not fully clear, but its occurrence can be related to biological or environmental pathways, among which Brain-derived neurotrophic factor (BDNF) can unequivocally act on two downstream receptors, tyrosine kinase receptor (TrkB) and the p75 neurotrophin receptor (p75NTR), then affect the related signal pathways, affecting the occurrence and development of depression. Accumulating studies have revealed that BDNF-related pathways are critical in the pathophysiology of depression, and their interaction can further influence the efficacy of depression treatment. In this review, we mainly summarized the signaling pathways associated with BDNF and classified them according to different receptors and related molecules, providing promising insights and future directions in the treatment of depression.
Collapse
Affiliation(s)
- Yuchen Fan
- Interventional Medical Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China; Qingdao medical college, Qingdao University, Qingdao, Shandong, China.
| | - Xinchi Luan
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| | - Xuezhe Wang
- Qingdao medical college, Qingdao University, Qingdao, Shandong, China.
| | - Hongchi Li
- Qingdao medical college, Qingdao University, Qingdao, Shandong, China.
| | - Hongjiao Zhao
- Qingdao medical college, Qingdao University, Qingdao, Shandong, China.
| | - Sheng Li
- Qingdao medical college, Qingdao University, Qingdao, Shandong, China.
| | - Xiaoxuan Li
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| | - Zhenkang Qiu
- Interventional Medical Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
5
|
Bharmauria V, Ramezanpour H, Ouelhazi A, Yahia Belkacemi Y, Flouty O, Molotchnikoff S. KETAMINE: Neural- and network-level changes. Neuroscience 2024; 559:188-198. [PMID: 39245312 DOI: 10.1016/j.neuroscience.2024.09.010] [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: 07/08/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Ketamine is a widely used clinical drug that has several functional and clinical applications, including its use as an anaesthetic, analgesic, anti-depressive, anti-suicidal agent, among others. Among its diverse behavioral effects, it influences short-term memory and induces psychedelic effects. At the neural level across different brain areas, it modulates neural firing rates, neural tuning, brain oscillations, and modularity, while promoting hypersynchrony and random connectivity between neurons. In our recent studies we demonstrated that topical application of ketamine on the visual cortex alters neural tuning and promotes vigorous connectivity between neurons by decreasing their firing variability. Here, we begin with a brief review of the literature, followed by results from our lab, where we synthesize a dendritic model of neural tuning and network changes following ketamine application. This model has potential implications for focused modulation of cortical networks in clinical settings. Finally, we identify current gaps in research and suggest directions for future studies, particularly emphasizing the need for more animal experiments to establish a platform for effective translation and synergistic therapies combining ketamine with other protocols such as training and adaptation. In summary, investigating ketamine's broader systemic effects, not only provides deeper insight into cognitive functions and consciousness but also paves the way to advance therapies for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Vishal Bharmauria
- The Tampa Human Neurophysiology Lab & Department of Neurosurgery and Brain Repair, Morsani College of Medicine, 2 Tampa General Circle, University of South Florida, Tampa, FL 33606, USA; Centre for Vision Research and Centre for Integrative and Applied Neuroscience, York University, 4700 Keele Street, Toronto, Ontario M3J 1P3, Canada.
| | - Hamidreza Ramezanpour
- Department of Biology, York University, 4700 Keele Street, Toronto, Ontario M3J 1P3, Canada
| | - Afef Ouelhazi
- Neurophysiology of the Visual system, Département de Sciences Biologiques, 1375 Av. Thérèse-Lavoie-Roux, Université de Montréal, Montréal, Québec H2V 0B3, Canada
| | - Yassine Yahia Belkacemi
- Neurophysiology of the Visual system, Département de Sciences Biologiques, 1375 Av. Thérèse-Lavoie-Roux, Université de Montréal, Montréal, Québec H2V 0B3, Canada
| | - Oliver Flouty
- The Tampa Human Neurophysiology Lab & Department of Neurosurgery and Brain Repair, Morsani College of Medicine, 2 Tampa General Circle, University of South Florida, Tampa, FL 33606, USA
| | - Stéphane Molotchnikoff
- Neurophysiology of the Visual system, Département de Sciences Biologiques, 1375 Av. Thérèse-Lavoie-Roux, Université de Montréal, Montréal, Québec H2V 0B3, Canada
| |
Collapse
|
6
|
Acero-Castillo MC, Correia MBM, Caixeta FV, Motta V, Barros M, Maior RS. Is the antidepressant effect of ketamine separate from its psychotomimetic effect? A review of rodent models. Neuropharmacology 2024; 258:110088. [PMID: 39032814 DOI: 10.1016/j.neuropharm.2024.110088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/09/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Ketamine is an NMDA (N-methyl-d-aspartate) glutamate receptor antagonist, which has a myriad of dose-dependent pharmacological and behavioral effects, including anesthetic, sedative, amnestic, analgesic, and anti-inflammatory properties. Intriguingly, ketamine at subanesthetic doses displays a relevant profile both in mimicking symptoms of schizophrenia and also as the first fast-acting treatment for depression. Here, we present an overview of the state-of-the-art knowledge about ketamine as an antidepressant as well as a pharmacological model of schizophrenia in animal models and human participants. Ketamine's dual effect appears to arise from its mechanism of action involving NMDA receptors, with both immediate and downstream consequences being triggered as a result. Finally, we discuss the feasibility of a unified approach linking the glutamatergic hypothesis of schizophrenia to the promising preclinical and clinical success of ketamine in the treatment of refractory depression.
Collapse
Affiliation(s)
- M C Acero-Castillo
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - M B M Correia
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil; Department of Anthropology, Emory University, Atlanta GA, ZIP 30322, USA
| | - F V Caixeta
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - V Motta
- Department of Basic Psychological Processes, Institute of Psychology, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - M Barros
- Department of Pharmacy, School of Health Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - R S Maior
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil.
| |
Collapse
|
7
|
Gomes I, Gupta A, Margolis EB, Fricker LD, Devi LA. Ketamine and Major Ketamine Metabolites Function as Allosteric Modulators of Opioid Receptors. Mol Pharmacol 2024; 106:240-252. [PMID: 39187388 PMCID: PMC11493337 DOI: 10.1124/molpharm.124.000947] [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: 05/08/2024] [Revised: 08/18/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024] Open
Abstract
Ketamine is a glutamate receptor antagonist that was developed over 50 years ago as an anesthetic agent. At subanesthetic doses, ketamine and some metabolites are analgesics and fast-acting antidepressants, presumably through targets other than glutamate receptors. We tested ketamine and its metabolites for activity as allosteric modulators of opioid receptors expressed as recombinant receptors in heterologous systems and with native receptors in rodent brain; signaling was examined by measuring GTP binding, β-arrestin recruitment, MAPK activation, and neurotransmitter release. Although micromolar concentrations of ketamine alone had weak agonist activity at μ opioid receptors, the combination of submicromolar concentrations of ketamine with endogenous opioid peptides produced robust synergistic responses with statistically significant increases in efficacies. All three opioid receptors (μ, δ, and κ) showed synergism with submicromolar concentrations of ketamine and either methionine-enkephalin (Met-enk), leucine-enkephalin (Leu-enk), and/or dynorphin A17 (Dyn A17), albeit the extent of synergy was variable between receptors and peptides. S-ketamine exhibited higher modulatory effects compared with R-ketamine or racemic ketamine, with ∼100% increase in efficacy. Importantly, the ketamine metabolite 6-hydroxynorketamine showed robust allosteric modulatory activity at μ opioid receptors; this metabolite is known to have analgesic and antidepressant activity but does not bind to glutamate receptors. Ketamine enhanced potency and efficacy of Met-enkephalin signaling both in mouse midbrain membranes and in rat ventral tegmental area neurons as determined by electrophysiology recordings in brain slices. Taken together, these findings support the hypothesis that some of the therapeutic effects of ketamine and its metabolites are mediated by directly engaging the endogenous opioid system. SIGNIFICANCE STATEMENT: This study found that ketamine and its major biologically active metabolites function as potent allosteric modulators of μ, δ, and κ opioid receptors, with submicromolar concentrations of these compounds synergizing with endogenous opioid peptides, such as enkephalin and dynorphin. This allosteric activity may contribute to ketamine's therapeutic effectiveness for treating acute and chronic pain and as a fast-acting antidepressant drug.
Collapse
Affiliation(s)
- Ivone Gomes
- Departments of Pharmacological Sciences (I.G., A.G., L.A.D.) and Psychiatry (L.A.D.), and Nash Family Department of Neuroscience (L.A.D.), Icahn School of Medicine at Mount Sinai, New York, New York; UCSF Weill Institute for Neurosciences, Department of Neurology, Neuroscience Graduate Program, University of California, San Francisco, California (E.B.M.); and Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (L.D.F.)
| | - Achla Gupta
- Departments of Pharmacological Sciences (I.G., A.G., L.A.D.) and Psychiatry (L.A.D.), and Nash Family Department of Neuroscience (L.A.D.), Icahn School of Medicine at Mount Sinai, New York, New York; UCSF Weill Institute for Neurosciences, Department of Neurology, Neuroscience Graduate Program, University of California, San Francisco, California (E.B.M.); and Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (L.D.F.)
| | - Elyssa B Margolis
- Departments of Pharmacological Sciences (I.G., A.G., L.A.D.) and Psychiatry (L.A.D.), and Nash Family Department of Neuroscience (L.A.D.), Icahn School of Medicine at Mount Sinai, New York, New York; UCSF Weill Institute for Neurosciences, Department of Neurology, Neuroscience Graduate Program, University of California, San Francisco, California (E.B.M.); and Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (L.D.F.)
| | - Lloyd D Fricker
- Departments of Pharmacological Sciences (I.G., A.G., L.A.D.) and Psychiatry (L.A.D.), and Nash Family Department of Neuroscience (L.A.D.), Icahn School of Medicine at Mount Sinai, New York, New York; UCSF Weill Institute for Neurosciences, Department of Neurology, Neuroscience Graduate Program, University of California, San Francisco, California (E.B.M.); and Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (L.D.F.)
| | - Lakshmi A Devi
- Departments of Pharmacological Sciences (I.G., A.G., L.A.D.) and Psychiatry (L.A.D.), and Nash Family Department of Neuroscience (L.A.D.), Icahn School of Medicine at Mount Sinai, New York, New York; UCSF Weill Institute for Neurosciences, Department of Neurology, Neuroscience Graduate Program, University of California, San Francisco, California (E.B.M.); and Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (L.D.F.)
| |
Collapse
|
8
|
Mastrodonato A, Jin M, Kee N, Lanio M, Tapia J, Quintana L, Muñoz Zamora A, Deng SX, Xu X, Landry DW, Denny CA. Prophylactic (R,S)-ketamine and (2S,6S)-hydroxynorketamine Decrease Fear Expression by Differentially Modulating Fear Neural Ensembles. Biol Psychiatry 2024:S0006-3223(24)01649-4. [PMID: 39389408 DOI: 10.1016/j.biopsych.2024.09.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 09/20/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND We previously reported that a single injection of (R,S)-ketamine or its metabolite (2S,6S)-hydroxynorketamine (HNK) prior to stress attenuated learned fear. However, whether these drugs attenuate learned fear through divergent or convergent effects on neural activity remains to be determined. METHODS 129S6/SvEv male mice were injected with saline, (R,S)-ketamine, or (2S,6S)-HNK 1 week before a 3-shock contextual fear conditioning paradigm. Five days later, mice were re-exposed to the aversive context and euthanized 1 hour later to quantify active cells. Brains were processed for c-fos immunoreactivity, and neural networks were built with a novel, wide-scale imaging pipeline. RESULTS We found that (R,S)-ketamine and (2S,6S)-HNK attenuated learned fear. Fear-related neural activity was altered in dorsal CA3 following (2S,6S)-HNK; ventral CA3 and CA1, infralimbic and prelimbic regions, insular cortex, retrosplenial cortex, piriform cortex, nucleus reuniens, and periaqueductal gray following both (R,S)-ketamine and (2S,6S)-HNK; and in the paraventricular nucleus of the thalamus (PVT) following (R,S)-ketamine. Dorsal CA3 and ventral hippocampus activation correlated with freezing in the (R,S)-ketamine group, and retrosplenial cortex activation correlated with freezing in both (R,S)-ketamine and (2S,6S)-HNK groups. (R,S)-ketamine increased connectivity between cortical and subcortical regions while (2S,6S)-HNK increased connectivity within these regions. CONCLUSIONS This work identifies novel nodes in fear networks that involve the nucleus reuniens, piriform cortex, insular cortex, periaqueductal gray, and retrosplenial cortex that can be targeted with neuromodulatory strategies or pharmaceutical compounds to treat fear-induced disorders. This approach could be used to optimize target engagement and dosing strategies of existing medications.
Collapse
Affiliation(s)
- Alessia Mastrodonato
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York; Division of Systems Neuroscience, Area Neuroscience, Research Foundation for Mental Hygiene, Inc./New York State Psychiatric Institute, New York, New York; MIND Area, Research Foundation for Mental Hygiene, Inc./New York State Psychiatric Institute, New York, New York.
| | - Michelle Jin
- Neurobiology and Behavior Graduate Program, Columbia University, New York, New York; Medical Scientist Training Program, Columbia University Irving Medical Center, New York, New York
| | - Noelle Kee
- Department of Neuroscience and Behavior, Barnard College, New York, New York
| | - Marcos Lanio
- Neurobiology and Behavior Graduate Program, Columbia University, New York, New York; Medical Scientist Training Program, Columbia University Irving Medical Center, New York, New York
| | - Juliana Tapia
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York; Division of Systems Neuroscience, Area Neuroscience, Research Foundation for Mental Hygiene, Inc./New York State Psychiatric Institute, New York, New York
| | - Liliette Quintana
- Department of Neuroscience and Behavior, Barnard College, New York, New York
| | - Andrea Muñoz Zamora
- Division of Systems Neuroscience, Area Neuroscience, Research Foundation for Mental Hygiene, Inc./New York State Psychiatric Institute, New York, New York
| | - Shi-Xian Deng
- Department of Medicine, Columbia University Irving Medical Center, New York, New York; Organic Chemistry Collaborative Center, Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Xiaoming Xu
- Department of Medicine, Columbia University Irving Medical Center, New York, New York; Organic Chemistry Collaborative Center, Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Donald W Landry
- Department of Medicine, Columbia University Irving Medical Center, New York, New York; Organic Chemistry Collaborative Center, Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Christine A Denny
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York; Division of Systems Neuroscience, Area Neuroscience, Research Foundation for Mental Hygiene, Inc./New York State Psychiatric Institute, New York, New York.
| |
Collapse
|
9
|
Yip E, Fleck D. The Use of Prophylactic Ketamine to Mitigate Postoperative Depressive Symptoms: A Systematic Review. J Perianesth Nurs 2024:S1089-9472(24)00210-7. [PMID: 39365203 DOI: 10.1016/j.jopan.2024.05.026] [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: 09/06/2023] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 10/05/2024]
Abstract
PURPOSE Depression is predicted to be the leading burden of disease worldwide by 2030 with a prevalence of 10% to 60% in the surgical population. Depressive symptoms in the perioperative population are associated with a myriad of grave complications, including higher morbidity and mortality. This systematic review aims to determine whether a single dose of intravenous ketamine can be used prophylactically as a routine resilience-enhancing agent in all high-risk adult patients undergoing surgery to mitigate depressive symptoms postoperatively by appraising evidence of existing literature. DESIGN An evidence-based systematic review. METHODS The databases PubMed, CINAHL, and Scopus were searched, adhering to the Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines. Search and Medical Subject Headings (MeSH) terms used: "ketamine," "surgical procedures," "surgical," "surgery," "depression," and "depressive disorder." FINDINGS In 9 randomized controlled trials (RCTs), bolus ketamine doses of 0.1 mg/kg to 1 mg/kg were administered intraoperatively or in the postanesthesia care unit. The primary outcome was the subjective feelings of depression as evaluated by different validated depression screening tools and seven RCTs observed statistically significant findings between the control and intervention (ketamine) groups with postoperative depression scores. Two RCTs did not find a significant change in depression scores. CONCLUSIONS Research on the use of prophylactic ketamine as an antidepressant in the perioperative population offers insight into changing routine practices and can shape how mental health is viewed in hospitals. Further research is warranted on the safety profile of ketamine, and risk stratification with careful consideration of baseline depressive symptoms, timing, and its use across a wider variety of surgical subspecialties.
Collapse
Affiliation(s)
- Eleanor Yip
- University of Pennsylvania School of Nursing, Philadelphia, PA.
| | - Desiree Fleck
- University of Pennsylvania School of Nursing, Philadelphia, PA
| |
Collapse
|
10
|
Xia R, Yang L, Liang C, Lyu D, Zang W, Sun G, Yan J. Research on aerobic fitness in children and adolescents: a bibliometric analysis based on the 100 most-cited articles. Front Med (Lausanne) 2024; 11:1409532. [PMID: 39386747 PMCID: PMC11461214 DOI: 10.3389/fmed.2024.1409532] [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: 03/30/2024] [Accepted: 08/28/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction This study aims to conduct a bibliometric analysis of the 100 most-cited articles to examine research trends, hot topics, and gaps in aerobic fitness research in children and adolescents, addressing the lack of evidence synthesis. Methods The Web of Science Core Collection database was used for literature search, and bibliometric characteristics of the included research articles were imported and calculated. Descriptive statistics and visualizations by the VOS viewer were used for the presentation of bibliometric characteristics. Results The 100 most cited articles received an average of 104 citations. British Journal of Sports Medicine and Medicine and Science in Sports and Exercise were the two top journals that published aerobic fitness research in children and adolescents. The United States was the top country that contributed to the most-cited research articles. Three top research topics were identified from the analysis, such as neuroscience, developmental psychology, and aerobic health. Conclusion Aerobic fitness research in children and adolescents has received much attention and interest since 2000. The most contributing authors in this research field were from developed countries, such as the United States, and cognition and health-related research were priorities.
Collapse
Affiliation(s)
- Rui Xia
- School of Physical Education, Chaohu University, Hefei, China
| | - Liu Yang
- School of Education, College of Human and Social Futures, University of Newcastle, Newcastle, NSW, Australia
| | | | - Dongye Lyu
- College of Education Sciences, The Hong Kong University of Science and Technology, Guangzhou, Guangzhou, China
| | - Wanli Zang
- Postgraduate School, University of Harbin Sport, Harbin, China
| | - Guanrong Sun
- Department of Public Physical and Art Education, Zhejiang University, Hangzhou, China
| | - Jin Yan
- School of Physical Education and Sports Science, Soochow University, Suzhou, China
| |
Collapse
|
11
|
Caraci F, Drago F. Ketamine and new targets for treatment-resistant depression: A role for transforming growth factor-β1? Eur Neuropsychopharmacol 2024; 86:65-66. [PMID: 39018790 DOI: 10.1016/j.euroneuro.2024.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 07/19/2024]
Affiliation(s)
- Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, Catania, Italy; Oasi Research Institute-IRCCS, Unit of Translational Neuropharmacology and Translational Neurosciences, Troina, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| |
Collapse
|
12
|
Kosuge A, Kunisawa K, Iida T, Wulaer B, Kawai T, Tanabe M, Saito K, Nabeshima T, Mouri A. Chronic social defeat stress induces the down-regulation of the Nedd4L-GLT-1 ubiquitination pathway in the prefrontal cortex of mice. J Neurochem 2024; 168:2479-2494. [PMID: 38497582 DOI: 10.1111/jnc.16100] [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: 08/22/2023] [Revised: 02/25/2024] [Accepted: 02/29/2024] [Indexed: 03/19/2024]
Abstract
Stressful life events contribute to the onset of major depressive disorder (MDD). We recently demonstrated abnormalities in ubiquitination in the pathophysiology of MDD. However, the underlying molecular mechanisms remain unclear. We investigated the involvement of the ubiquitination system-mediated glutamatergic dysfunction in social impairment induced by chronic social defeat stress (CSDS). Adult C57BL/6J mice were exposed to aggressor ICR male mice for 10 consecutive days. Social impairment was induced by CSDS in the social interaction test 1 days after the last stress exposure. In terms of brain microdialysis, CSDS reduced depolarization-evoked glutamate release in the prefrontal cortex (PFC), which was reversed by a glutamate transporter 1 (GLT-1) inhibitor. Interestingly, the expression of ubiquitinated, but not total GLT-1, was decreased in the PFC of mice exposed to CSDS. The expression of neural precursor cells expressing developmentally downregulated gene 4-like (Nedd4L: E3 ligase for GLT-1), and ubiquitin-conjugating enzyme E2D2 (Ube2d2: E2 ubiquitin-conjugating enzyme for Nedd4L) was also reduced in CSDS mice. Furthermore, the downregulation of the Nedd4L-GLT-1 ubiquitination pathway decreased SIT ratio, but up-regulation increased it even in non-CSDS mice. Taken together, the decrease in GLT-1 ubiquitination may reduce the release of extracellular glutamate induced by high-potassium stimulation, which may lead to social impairment, while we could not find differences in GLT-1 ubiquitination between susceptible and resistant CSDS mice. In conclusion, GLT-1 ubiquitination could play a crucial role in the pathophysiology of MDD and is an attractive target for the development of novel antidepressants.
Collapse
Affiliation(s)
- Aika Kosuge
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Sciences, Toyoake, Aichi, Japan
| | - Kazuo Kunisawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Sciences, Toyoake, Aichi, Japan
| | - Tsubasa Iida
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Sciences, Toyoake, Aichi, Japan
| | - Bolati Wulaer
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Health Sciences, Toyoake, Aichi, Japan
| | - Tomoki Kawai
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Sciences, Toyoake, Aichi, Japan
| | - Moeka Tanabe
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Sciences, Toyoake, Aichi, Japan
| | - Kuniaki Saito
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Health Sciences, Toyoake, Aichi, Japan
- Laboratory of Health and Medical Science Innovation, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research, Toyoake, Aichi, Japan
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
- Laboratory of Health and Medical Science Innovation, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research, Toyoake, Aichi, Japan
| | - Akihiro Mouri
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Sciences, Toyoake, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research, Toyoake, Aichi, Japan
| |
Collapse
|
13
|
Hess EM, Greenstein DK, Hutchinson OL, Zarate CA, Gould TD. Entactogen Effects of Ketamine: A Reverse-Translational Study. Am J Psychiatry 2024; 181:815-823. [PMID: 38982828 PMCID: PMC11492270 DOI: 10.1176/appi.ajp.20230980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
OBJECTIVE The authors sought to assess the prosocial, entactogen effects of ketamine. METHODS Pleasure from social situations was assessed in a sample of participants with treatment-resistant depression from randomized, double-blind, placebo-controlled studies, using four items of the Snaith-Hamilton Pleasure Scale (SHAPS) at five time points over 1 week following treatment with ketamine (0.5 mg/kg intravenously) or placebo. The primary endpoint was postinfusion self-reported pleasure on the four SHAPS items pertaining to social situations, including the item on helping others, between the ketamine and placebo groups. In a rodent experiment, the impact of ketamine on helping behavior in rats was assessed using the harm aversion task. The primary endpoint was a reduction in lever response rate relative to baseline, which indicated the willingness of rats to forgo obtaining sucrose to help protect their cage mate from electric shock. RESULTS Relative to placebo, ketamine increased ratings of feeling pleasure from being with family or close friends, seeing other people's smiling faces, helping others, and receiving praise, for 1 week following treatment. In the rodent experiment, during the harm aversion task, ketamine-treated rats maintained lower response rates relative to baseline to a greater extent than what was observed in vehicle-treated rats for 6 days posttreatment and delivered fewer shocks overall. CONCLUSIONS In patients with treatment-resistant depression, ketamine treatment was associated with increased pleasure from social situations, such as feeling pleasure from helping others. Ketamine-treated rats were more likely to protect their cage mate from harm, at the cost of obtaining sucrose. These findings suggest that ketamine has entactogen effects.
Collapse
Affiliation(s)
- Evan M Hess
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dede K Greenstein
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Olivia L Hutchinson
- Department of Psychiatry, University of Maryland School of Medicine, 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
| | - Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Pharmacology, 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
| |
Collapse
|
14
|
Sathappan A, Yudkoff B. Empowering understanding: navigating consent to ketamine treatment in adolescent mental health. Front Psychiatry 2024; 15:1433348. [PMID: 39193578 PMCID: PMC11347886 DOI: 10.3389/fpsyt.2024.1433348] [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: 05/15/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
The rising prevalence of mental health disorders in adolescents, compounded by treatment resistance, underscores the need for innovative interventions. Ketamine, recognized for its rapid antidepressant and anti-suicidal effects in adults, has recently emerged as a potential treatment for adolescents with treatment-resistant depression and suicidality. This paper aims to highlight key elements of the informed consent process, including obtaining parental consent and adolescent assent, and discussing the nature of ketamine treatment, its benefits, and potential risks. Obtaining informed consent for ketamine treatment in this demographic poses unique challenges. During the informed consent process, clinicians should balance an adolescent's growing autonomy with parental consent and address the distinct features of treatment, including ketamine's potential to induce psychedelic-like effects. Additionally, clinicians should highlight the "off-label" use in this population and the uncertainty inherent to treatment at this time, including the lack of data on repeated ketamine exposure on the developing brain. This paper also addresses challenging scenarios related to informed consent for this treatment, such as instances when parents are willing to consent but the adolescent refuses. Alternative treatment strategies such as transcranial magnetic stimulation (TMS) and electroconvulsive therapy (ECT) are also considered. In conclusion, while an emerging body of evidence suggests that ketamine shows potential for the acute treatment of adolescents with severe depression and suicidality, adherence to informed consent principles is paramount to ensure best clinical practices and uphold ethical standards amidst the current landscape of ongoing research.
Collapse
Affiliation(s)
- Aakash Sathappan
- Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Benjamin Yudkoff
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Lumin Health, Boston, MA, United States
| |
Collapse
|
15
|
Patarroyo-Rodriguez L, Cavalcanti S, Vande Voort JL, Singh B. The Use of Ketamine for the Treatment of Anhedonia in Depression. CNS Drugs 2024; 38:583-596. [PMID: 38910222 DOI: 10.1007/s40263-024-01099-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/29/2024] [Indexed: 06/25/2024]
Abstract
Anhedonia, a complex symptom rooted in deficits across reward processes, is primarily linked to depression and schizophrenia but transcends diagnostic boundaries across various mental disorders. Its presence correlates with poorer clinical outcomes, including an increased risk of suicide and diminished response to treatment. The neurobiological underpinnings of anhedonia remain incompletely understood despite advancements in biomarkers and imaging that contribute to deeper insights. Ketamine, known for its rapid-acting antidepressant properties, appears to possess antianhedonic effects through a mechanism of action not fully elucidated. This effect appears to be independent of its antidepressant properties. Explorations into alternative antianhedonic treatments have been underway, yet lingering questions persist, underscoring the imperative need for ongoing research to advance the field.
Collapse
Affiliation(s)
| | - Stefanie Cavalcanti
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jennifer L Vande Voort
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Balwinder Singh
- Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
16
|
Brenna CTA, Goldstein BI, Zarate CA, Orser BA. Repurposing General Anesthetic Drugs to Treat Depression: A New Frontier for Anesthesiologists in Neuropsychiatric Care. Anesthesiology 2024; 141:222-237. [PMID: 38856663 DOI: 10.1097/aln.0000000000005037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
During the last 100 years, the role of anesthesiologists in psychiatry has focused primarily on facilitating electroconvulsive therapy and mitigating postoperative delirium and other perioperative neurocognitive disorders. The discovery of the rapid and sustained antidepressant properties of ketamine, and early results suggesting that other general anesthetic drugs (including nitrous oxide, propofol, and isoflurane) have antidepressant properties, has positioned anesthesiologists at a new frontier in the treatment of neuropsychiatric disorders. Moreover, shared interest in understanding the biologic underpinnings of anesthetic drugs as psychotropic agents is eroding traditional academic boundaries between anesthesiology and psychiatry. This article presents a brief overview of anesthetic drugs as novel antidepressants and identifies promising future candidates for the treatment of depression. The authors issue a call to action and outline strategies to foster collaborations between anesthesiologists and psychiatrists as they work toward the common goals of repurposing anesthetic drugs as antidepressants and addressing mood disorders in surgical patients.
Collapse
Affiliation(s)
- Connor T A Brenna
- Department of Anesthesiology & Pain Medicine and Department of Physiology, University of Toronto, Toronto, Canada; Perioperative Brain Health Centre, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Benjamin I Goldstein
- Centre for Addiction and Mental Health, Toronto, Canada; Department of Psychiatry and Department of Pharmacology, University of Toronto, Toronto, Canada
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Beverley A Orser
- Department of Anesthesiology & Pain Medicine and Department of Physiology, University of Toronto, Toronto, Canada; Perioperative Brain Health Centre, Sunnybrook Health Sciences Centre, Toronto, Canada
| |
Collapse
|
17
|
Zang D, Yang X, Wang H, Li Z, Ma Y, Liu J, Mei X, Li S, Feng J, Shi X, Tan Z. Study on the antidepressant activity of (2R,6R; 2S,6S)-Hydroxynorketamine (HNK) and its derivatives. Clinics (Sao Paulo) 2024; 79:100435. [PMID: 38996724 PMCID: PMC11301192 DOI: 10.1016/j.clinsp.2024.100435] [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: 03/20/2024] [Revised: 05/23/2024] [Accepted: 06/12/2024] [Indexed: 07/14/2024] Open
Abstract
OBJECTIVE This study mainly explores (2R,6R; 2S,6S)-HNK and its compounds whether there are antidepressant effects. METHODS Four HNK compounds were obtained from 2-(Chlorophenyl) Cyclopentylmethanone. Forced swimming test, locomotor sensitization test, and conditioned location preference test were used to screen the antidepressant activity of the synthesized target compounds. RESULTS In the case of 10 mg HNK treatment, compared with saline, the immobile time of mice in the HNK group, I5 group and I6 group at 1 h and 7 days had statistical significance. In the case of 10 mg HNK treatment, compared with saline, the immobile time of compound C and D groups in the glass cylinder area was significantly different. In the locomotor sensitization test, the movement distance of compound C and D groups on day 15 and day 7 mice increased significantly compared with the first day. In the conditioned place preference experiment, compound C and compound D induced conditioned place preference in mice compared with the Veh group. CONCLUSION The results of the forced swimming test, locomotor sensitization test, and conditioned location preference test showed that compounds C and D may have certain anti-depressant activity. However, HNK exerts a rapid and significant antidepressant effect within 1 week, but the duration is short.
Collapse
Affiliation(s)
- Dongdong Zang
- Department of Neonatology, Shenzhen Children's Hospital, Shenzhen City, Guangdong Province, China
| | - Xuemei Yang
- Health Management Center, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen City, Guangdong Province, China
| | - Hao Wang
- Department of Neurosurgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen City, Guangdong Province, China; Guangdong Engineering Technological Research Center for Nervous anatomy and Related Clinical Applications, Shenzhen City, Guangdong Province, China
| | - Zhenxing Li
- Health Management Center, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen City, Guangdong Province, China
| | - Yanjun Ma
- Shenzhen Ruijian Biotechnology Co., LTD, Shenzhen City, Guangdong Province, China
| | - Jianxi Liu
- Shenzhen Ruijian Biotechnology Co., LTD, Shenzhen City, Guangdong Province, China
| | - Xi Mei
- Zhuhai Pengkun Biomedicine Technology Co. LTD, Zhuhai City, Guangdong Province, China
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University, Shenzhen City, Guangdong Province, China
| | - Jinxing Feng
- Department of Neonatology, Shenzhen Children's Hospital, Shenzhen City, Guangdong Province, China
| | - Xin Shi
- Department of Neurosurgery, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Centre, Shenzhen University, Shenzhen City, Guangdong Province, China
| | - Zhen Tan
- Department of Neonatology, Shenzhen Children's Hospital, Shenzhen City, Guangdong Province, China; Health Management Center, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen City, Guangdong Province, China.
| |
Collapse
|
18
|
Chai Z, Zheng J, Shen J. Mechanism of ferroptosis regulating ischemic stroke and pharmacologically inhibiting ferroptosis in treatment of ischemic stroke. CNS Neurosci Ther 2024; 30:e14865. [PMID: 39042604 PMCID: PMC11265528 DOI: 10.1111/cns.14865] [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: 04/11/2024] [Revised: 06/27/2024] [Accepted: 07/05/2024] [Indexed: 07/25/2024] Open
Abstract
Ferroptosis is a newly discovered form of programmed cell death that is non-caspase-dependent and is characterized by the production of lethal levels of iron-dependent lipid reactive oxygen species (ROS). In recent years, ferroptosis has attracted great interest in the field of cerebral infarction because it differs morphologically, physiologically, and genetically from other forms of cell death such as necrosis, apoptosis, autophagy, and pyroptosis. In addition, ROS is considered to be an important prognostic factor for ischemic stroke, making it a promising target for stroke treatment. This paper summarizes the induction and defense mechanisms associated with ferroptosis, and explores potential treatment strategies for ischemic stroke in order to lay the groundwork for the development of new neuroprotective drugs.
Collapse
Affiliation(s)
- Zhaohui Chai
- Department of NeurosurgeryFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou CityChina
| | - Jiesheng Zheng
- Department of NeurosurgeryFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou CityChina
| | - Jian Shen
- Department of NeurosurgeryFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou CityChina
| |
Collapse
|
19
|
Shafique H, Demers JC, Biesiada J, Golani LK, Cerne R, Smith JL, Szostak M, Witkin JM. ( R)-(-)-Ketamine: The Promise of a Novel Treatment for Psychiatric and Neurological Disorders. Int J Mol Sci 2024; 25:6804. [PMID: 38928508 PMCID: PMC11203826 DOI: 10.3390/ijms25126804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
NMDA receptor antagonists have potential for therapeutics in neurological and psychiatric diseases, including neurodegenerative diseases, epilepsy, traumatic brain injury, substance abuse disorder (SUD), and major depressive disorder (MDD). (S)-ketamine was the first of a novel class of antidepressants, rapid-acting antidepressants, to be approved for medical use. The stereoisomer, (R)-ketamine (arketamine), is currently under development for treatment-resistant depression (TRD). The compound has demonstrated efficacy in multiple animal models. Two clinical studies disclosed efficacy in TRD and bipolar depression. A study by the drug sponsor recently failed to reach a priori clinical endpoints but post hoc analysis revealed efficacy. The clinical value of (R)-ketamine is supported by experimental data in humans and rodents, showing that it is less sedating, does not produce marked psychotomimetic or dissociative effects, has less abuse potential than (S)-ketamine, and produces efficacy in animal models of a range of neurological and psychiatric disorders. The mechanisms of action of the antidepressant effects of (R)-ketamine are hypothesized to be due to NMDA receptor antagonism and/or non-NMDA receptor mechanisms. We suggest that further clinical experimentation with (R)-ketamine will create novel and improved medicines for some of the neurological and psychiatric disorders that are underserved by current medications.
Collapse
Affiliation(s)
- Hana Shafique
- Duke University School of Medicine, Durham, NC 27710, USA
| | - Julie C. Demers
- Indiana University-Purdue University, Indianapolis, IN 46202, USA; (J.C.D.); (J.B.)
| | - Julia Biesiada
- Indiana University-Purdue University, Indianapolis, IN 46202, USA; (J.C.D.); (J.B.)
| | - Lalit K. Golani
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, USA;
| | - Rok Cerne
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN 46260, USA; (R.C.); (J.L.S.)
| | - Jodi L. Smith
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN 46260, USA; (R.C.); (J.L.S.)
| | - Marta Szostak
- Department of Psychology, SWPS University, 03-815 Warsaw, Poland;
| | - Jeffrey M. Witkin
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN 46260, USA; (R.C.); (J.L.S.)
- Departments of Neuroscience and Trauma Research, Ascension St. Vincent Hospital, Indianapolis, IN 46260, USA
| |
Collapse
|
20
|
Magny R, Mégarbane B, Chevillard L, Roulland E, Bardèche-Trystram B, Dumestre-Toulet V, Labat L, Houzé P. A combined toxicokinetic and metabolic approach to investigate deschloro-N-ethylketamine exposure in a multidrug user. J Pharm Biomed Anal 2024; 243:116086. [PMID: 38518457 DOI: 10.1016/j.jpba.2024.116086] [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: 12/12/2023] [Revised: 02/27/2024] [Accepted: 03/05/2024] [Indexed: 03/24/2024]
Abstract
The use of new psychoactive substances derived from ketamine is rarely reported in France. A chronic GHB, 3-MMC, and methoxetamine consumer presented a loss of consciousness in a chemsex context and was referred to the intensive care unit with a rapid and favorable outcome. To investigate the chemicals responsible for the intoxication, a comprehensive analysis was conducted on the ten plasma samples collected over a 29.5-hour period, urine obtained upon admission, a 2-cm hair strand sample, and a seized crystal. These analyses were performed using liquid chromatography hyphenated to high resolution tandem mass spectrometry operating in targeted and untargeted modes. Additionally, analyses using gas chromatography coupled to mass spectrometry and nuclear magnetic resonance were conducted to probe the composition of the seized crystal. The molecular network-based approach was employed for data processing in non-targeted analyses. It allowed to confirm a multidrug exposure encompassing GHB, methyl-(aminopropyl)benzofuran (MAPB), (aminopropyl)benzofuran (APB), methylmethcathinone, chloromethcathinone, and a new psychoactive substance belonging to the arylcyclohexylamine family namely deschloro-N-ethyl-ketamine (O-PCE). Molecular network analysis facilitated the annotation of 27 O-PCE metabolites, including phase II compounds not previously reported. Plasma kinetics of O-PCE allowed the estimation of the elimination half-life of ∼5 hours. Kinetics of O-PCE metabolites was additionally characterized, possibly useful as surrogate biomarkers of consumption. We also observed marked alterations in lipid metabolism related to poly consumption of drugs. In conclusion, this case report provides a comprehensive analysis of exposure to O-PCE in a multidrug user including kinetic and metabolism data in human.
Collapse
Affiliation(s)
- Romain Magny
- Laboratoire de Toxicologie Biologique, Fédération de Toxicologie, Hôpital Lariboisière, AP-HP, Paris 75010, France; INSERM UMRS-1144, Université Paris Cité, Paris 75006, France
| | - Bruno Mégarbane
- INSERM UMRS-1144, Université Paris Cité, Paris 75006, France; Réanimation Médicale et Toxicologique, Fédération de Toxicologie, Hôpital Lariboisière, AP-HP, Paris 75010, France.
| | | | | | - Benoit Bardèche-Trystram
- Laboratoire de Toxicologie Biologique, Fédération de Toxicologie, Hôpital Lariboisière, AP-HP, Paris 75010, France
| | | | - Laurence Labat
- Laboratoire de Toxicologie Biologique, Fédération de Toxicologie, Hôpital Lariboisière, AP-HP, Paris 75010, France; INSERM UMRS-1144, Université Paris Cité, Paris 75006, France
| | - Pascal Houzé
- Laboratoire de Toxicologie Biologique, Fédération de Toxicologie, Hôpital Lariboisière, AP-HP, Paris 75010, France; INSERM UMRS-1144, Université Paris Cité, Paris 75006, France.
| |
Collapse
|
21
|
Benneh CK, Abotsi WKM, Biney RP, Mante PK, Abeka MK, Tandoh A, Woode E. Synergistic antidepressant-like effect of xylopic acid co-administered with selected antidepressants. IBRO Neurosci Rep 2024; 16:329-335. [PMID: 38390234 PMCID: PMC10882107 DOI: 10.1016/j.ibneur.2024.01.011] [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: 05/04/2023] [Revised: 01/24/2024] [Accepted: 01/27/2024] [Indexed: 02/24/2024] Open
Abstract
Background Xylopic acid (XA), a kaurene diterpene from the dried fruits of Xylopia aethiopica, has anxiolytic- and antidepressant-like activity in mice and zebrafish. We aimed to assess the potential synergistic antidepressant-like effects of XA when combined with selected antidepressants in the mouse forced-swim test. Materials and methods The antidepressant-like effect of xylopic acid (XA) (10, 30, 100 mgkg-1), fluoxetine (Flx) (3, 10, 30 mgkg-1), sertraline (Sert) (3, 10, 30 mgkg-1), imipramine (Imi) (10, 30, 100 mgkg-1) and ketamine (Ket) (0.1, 0.3, 1.0 mgkg-1), was evaluated in forced swim test. The dose (ED50) that achieved a 50% reduction in immobility time was determined from the respective log-dose response curves. XA and the selected antidepressants were co-administered in fixed-dose ratio combinations (1/2:1/2, 1/4:1/4, 1/8:1/8) of the ED50 to identify the experimental ED50 (ED50mix). The theoretical ED50(ED50add), of all combinations was determined using isobolograms and compared with the ED50mix to identify the nature of the interaction. The effect of dose combinations on general locomotor activity was assessed in the open-field test. Results The interaction index (γ) for the following XA combinations, XA/Flx, XA/Sert, XA/Imi and XA/Ket were 0.42, 0.41, 0.31 and 0.34. An independent sample t-test revealed that the experimental ED50 (ED50mix) was significantly lower than the theoretical ED50 (ED50add) in all combinations of XA, indicative of a synergistic antidepressant-like effect. However, combinations of XA with ketamine significantly reduced general locomotor activity at all dose combinations. Conclusion The co-administration of xylopic acid and fluoxetine, imipramine, sertraline and ketamine produces a synergistic antidepressant-like effect in mice.
Collapse
Affiliation(s)
- Charles Kwaku Benneh
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Health and Allied Sciences, Ho, Ghana
| | - Wonder Kofi Mensah Abotsi
- Department of Pharmacotherapeutics and Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Robert Peter Biney
- Department of Pharmacology and Toxicology, School of Pharmacy and Pharmaceutical Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Priscilla Kolibea Mante
- Department of Pharmacotherapeutics and Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Mustapha Kobina Abeka
- Department of Pharmaceutical Chemistry, School of Pharmacy and Pharmaceutical Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Augustine Tandoh
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Health and Allied Sciences, Ho, Ghana
| | - Eric Woode
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Health and Allied Sciences, Ho, Ghana
| |
Collapse
|
22
|
Huang C, Wu Z, Wang D, Qu Y, Zhang J, Jiang R, Xu X, Xu X, Wang Y, Liu H, He T, Liu C, Chen G, Yang JJ, Hashimoto K, Yang C. Myelin-associated oligodendrocytic basic protein-dependent myelin repair confers the long-lasting antidepressant effect of ketamine. Mol Psychiatry 2024; 29:1741-1753. [PMID: 37848708 DOI: 10.1038/s41380-023-02288-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/19/2023]
Abstract
Ketamine exhibits rapid and sustained antidepressant effects. As decreased myelination has been linked to depression pathology, changes in myelination may be a pivotal mechanism underlying ketamine's long-lasting antidepressant effects. Although ketamine has a long-lasting facilitating effect on myelination, the precise roles of myelination in ketamine's sustained antidepressant effects remain unknown. In this study, we employed spatial transcriptomics (ST) to examine ketamine's lasting effects in the medial prefrontal cortex (mPFC) and hippocampus of mice subjected to chronic social defeat stress and identified several differentially expressed myelin-related genes. Ketamine's ability to restore impaired myelination in the brain by promoting the differentiation of oligodendrocyte precursor cells (OPCs) into mature oligodendrocytes was demonstrated. Moreover, we showed that inhibiting the expression of myelin-associated oligodendrocytic basic protein (Mobp) blocked ketamine's long-lasting antidepressant effects. We also illustrated that α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) signaling mediated ketamine's facilitation on myelination. In addition, we found that the (R)-stereoisomer of ketamine showed stronger effects on myelination than (S)-ketamine, which may explain its longer-lasting antidepressant effects. These findings reveal novel mechanisms underlying the sustained antidepressant effects of ketamine and the differences in antidepressant effects between (R)-ketamine and (S)-ketamine, providing new insights into the role of myelination in antidepressant mechanisms.
Collapse
Affiliation(s)
- Chaoli Huang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
| | - Zifeng Wu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Di Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Youge Qu
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Jichun Zhang
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Riyue Jiang
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiangqing Xu
- Nhwa Institute of Pharmaceutical Research, Jiangsu Nhwa Pharmaceutical Co., Ltd and Jiangsu Key Laboratory of Central Nervous System Drug Research and Development, Xuzhou, 221116, China
| | - Xiangyang Xu
- Nhwa Institute of Pharmaceutical Research, Jiangsu Nhwa Pharmaceutical Co., Ltd and Jiangsu Key Laboratory of Central Nervous System Drug Research and Development, Xuzhou, 221116, China
| | - Yuanyuan Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hanyu Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Teng He
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Cunming Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Guiquan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan.
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
23
|
Ren L, Chen Q, Gao J, Liu Y, Tao Y, Li X, Luo Q, Lv F, Min S. Clinical efficacy of adjunctive esketamine anesthesia in electroconvulsive therapy for major depressive disorders: A pragmatic, randomized, controlled trial. Psychiatry Res 2024; 335:115843. [PMID: 38461645 DOI: 10.1016/j.psychres.2024.115843] [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: 11/01/2023] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/12/2024]
Abstract
Electroconvulsive therapy (ECT) is an effective treatment for depression, and esketamine has been shown to have antidepressant effects. However, it is currently unclear whether adjunctive esketamine can enhance the clinical efficacy of ECT in real-world clinical practice. In this pragmatic clinical trial, patients with major depression were randomly assigned into two groups: patients received 0.25 mg/kg esketamine plus propofol (esketamine group) or the same volume of saline (control group) plus propofol. Results indicated that there was no difference in response and remission rates between the two groups. However, patients receiving esketamine had a higher remission rate of SI and lower psychotic scores. Patients receiving esketamine also required a lower electric dose, but the seizure duration and cognitive function were comparable between the two groups. Diastolic blood pressure increased after esketamine injection, but there was no increased risk of hypertension. Furthermore, incidence of delirium and confusion were comparable between the groups. Conclusively, adjunctive esketamine anesthesia does not provide any advantage in improving the response and remission rates of ECT. However, it can improve remission of SI and alleviate accompanying psychotic symptoms in depressive patients. With adjunctive usage, the adverse cardiovascular and neuropsychiatric events associated with esketamine appear to be tolerable.
Collapse
Affiliation(s)
- Li Ren
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Qibin Chen
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Jin Gao
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Yuanyuan Liu
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Yi Tao
- Department of Phase I Clinical Trial Ward, the First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Xiao Li
- Department of Psychiatry, the First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Qinghua Luo
- Department of Psychiatry, the First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Feng Lv
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China.
| | - Su Min
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
24
|
Więdłocha M, Marcinowicz P, Komarnicki J, Tobiaszewska M, Dębowska W, Dębowska M, Szulc A. Depression with comorbid borderline personality disorder - could ketamine be a treatment catalyst? Front Psychiatry 2024; 15:1398859. [PMID: 38742125 PMCID: PMC11089186 DOI: 10.3389/fpsyt.2024.1398859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 04/15/2024] [Indexed: 05/16/2024] Open
Abstract
Borderline personality disorder (BPD) is diagnosed in 10-30% of patients with major depressive disorder (MDD), and the frequency of MDD among individuals with BPD reaches over 80%. The comorbidity of MDD and BPD is associated with more severe depressive symptoms and functional impairment, higher risk of treatment resistance and increased suicidality. The effectiveness of ketamine usage in treatment resistant depression (TRD) has been demonstrated in numerous studies. In most of these studies, individuals with BPD were not excluded, thus given the high co-occurrence of these disorders, it is possible that the beneficial effects of ketamine also extend to the subpopulation with comorbid TRD and BPD. However, no protocols were developed that would account for comorbidity. Moreover, psychotherapeutic interventions, which may be crucial for achieving a lasting therapeutic effect in TRD and BPD comorbidity, were not included. In the article, we discuss the results of a small number of existing studies and case reports on the use of ketamine in depressive disorders with comorbid BPD. We elucidate how, at the molecular and brain network levels, ketamine can impact the neurobiology and symptoms of BPD. Furthermore, we explore whether ketamine-induced neuroplasticity, augmented by psychotherapy, could be of use in alleviating core BPD-related symptoms such as emotional dysregulation, self-identity disturbances and self-harming behaviors. We also discuss the potential of ketamine-assisted psychotherapy (KAP) in BPD treatment. As there is no standard approach to the application of ketamine or KAP in individuals with comorbid TRD and BPD, we consider further research in the field as imperative. The priorities should include development of dedicated protocols, distinguishing subpopulations that may benefit most from such treatment and investigating factors that may influence its effectiveness and safety.
Collapse
Affiliation(s)
- Magdalena Więdłocha
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Pruszkow, Masovian, Poland
- KeyClinic, Warsaw, Poland
| | - Piotr Marcinowicz
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Pruszkow, Masovian, Poland
- KeyClinic, Warsaw, Poland
| | - Jan Komarnicki
- Leszek Giec Upper-Silesian Medical Centre of the Medical University of Silesia, Katowice, Poland
| | | | - Weronika Dębowska
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Pruszkow, Masovian, Poland
| | - Marta Dębowska
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Pruszkow, Masovian, Poland
| | - Agata Szulc
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Pruszkow, Masovian, Poland
- MindHealth, Warsaw, Poland
| |
Collapse
|
25
|
Sikiric P, Boban Blagaic A, Strbe S, Beketic Oreskovic L, Oreskovic I, Sikiric S, Staresinic M, Sever M, Kokot A, Jurjevic I, Matek D, Coric L, Krezic I, Tvrdeic A, Luetic K, Batelja Vuletic L, Pavic P, Mestrovic T, Sjekavica I, Skrtic A, Seiwerth S. The Stable Gastric Pentadecapeptide BPC 157 Pleiotropic Beneficial Activity and Its Possible Relations with Neurotransmitter Activity. Pharmaceuticals (Basel) 2024; 17:461. [PMID: 38675421 PMCID: PMC11053547 DOI: 10.3390/ph17040461] [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: 02/15/2024] [Revised: 03/24/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
We highlight the particular aspects of the stable gastric pentadecapeptide BPC 157 pleiotropic beneficial activity (not destroyed in human gastric juice, native and stable in human gastric juice, as a cytoprotection mediator holds a response specifically related to preventing or recovering damage as such) and its possible relations with neurotransmitter activity. We attempt to resolve the shortage of the pleiotropic beneficial effects of BPC 157, given the general standard neurotransmitter criteria, in classic terms. We substitute the lack of direct conclusive evidence (i.e., production within the neuron or present in it as a precursor molecule, released eliciting a response on the receptor on the target cells on neurons and being removed from the site of action once its signaling role is complete). This can be a network of interconnected evidence, previously envisaged in the implementation of the cytoprotection effects, consistent beneficial particular evidence that BPC 157 therapy counteracts dopamine, serotonin, glutamate, GABA, adrenalin/noradrenalin, acetylcholine, and NO-system disturbances. This specifically includes counteraction of those disturbances related to their receptors, both blockade and over-activity, destruction, depletion, tolerance, sensitization, and channel disturbances counteraction. Likewise, BPC 157 activates particular receptors (i.e., VGEF and growth hormone). Furthermore, close BPC 157/NO-system relations with the gasotransmitters crossing the cell membrane and acting directly on molecules inside the cell may envisage particular interactions with receptors on the plasma membrane of their target cells. Finally, there is nerve-muscle relation in various muscle disturbance counteractions, and nerve-nerve relation in various encephalopathies counteraction, which is also exemplified specifically by the BPC 157 therapy application.
Collapse
Affiliation(s)
- Predrag Sikiric
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Alenka Boban Blagaic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Sanja Strbe
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Lidija Beketic Oreskovic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Ivana Oreskovic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Suncana Sikiric
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Mario Staresinic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Surgery, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Marko Sever
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Surgery, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Antonio Kokot
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Anatomy and Neuroscience, School of Medicine, J.J. Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Ivana Jurjevic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Danijel Matek
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Luka Coric
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Ivan Krezic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Ante Tvrdeic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Kresimir Luetic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Lovorka Batelja Vuletic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Predrag Pavic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Surgery, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Tomislav Mestrovic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Anatomy and Neuroscience, School of Medicine, J.J. Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Ivica Sjekavica
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Diagnostic and Interventional Radiology, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia
| | - Anita Skrtic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Sven Seiwerth
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
26
|
Li H, Luo X, Qi K, Lv Y, Kan J, Yang C, Lin X, Tao J, Zhang W, Liu Y, Rong K, Wang A, Jiang Z, Li X. Glutamate Chemical Exchange Saturation Transfer (GluCEST) MRI to Evaluate the Rapid Antidepressant Effects of Ketamine in the Hippocampus of Rat Depression Model. J Magn Reson Imaging 2024; 59:1373-1381. [PMID: 37496196 DOI: 10.1002/jmri.28921] [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: 05/05/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/28/2023] Open
Abstract
BACKGROUND Ketamine is a quick acting antidepressant drug, and an accurate detection method is lacking. Ketamine's effects in a rat depression model have not previously been well explored using glutamate chemical exchange saturation transfer (GluCEST). PURPOSE To investigate the GluCEST changes of chronic unpredictable mild stress (CUMS) rats after receiving either ketamine or saline injection. STUDY TYPE Randomized animal model trial. ANIMAL MODEL 12 CUMS and 6 Sprague-Dawley rats. Divided into three groups: ketamine (N = 6), saline (N = 6), and control (N = 6). FIELD STRENGTH/SEQUENCE 7.0 T/the sequence is GluCEST and 1 H MR spectroscopy (MRS). ASSESSMENT The CUMS rats were exposed to different stress factors for 8 weeks. The glutamate concentration in the hippocampus was assessed by the GluCEST,1 H MRS, and the high-performance liquid chromatography (HPLC). STATISTICAL TESTS The t-test, Mann-Whitney U test, and Pearson's correlation. RESULTS In depression conditions, GluCEST signals were lower in the bilateral hippocampus than in control group. Thirty minutes after ketamine injection, the GluCEST signals in the bilateral hippocampus were higher compared with the saline group (left: 2.99 ± 0.34 [Control] vs. 2.44 ± 0.20 [Saline] vs. 2.85 ± 0.11 [Ketamine]; right: 2.97 ± 0.28 [Control] vs. 2.49 ± 0.25 [Saline] vs. 2.86 ± 0.19 [Ketamine]). In 1 H MRS, significant changes were only observed in the left hippocampus (2.00 ± 0.16 [Control] vs. 1.81 ± 0.09 [Saline] vs. 2.04 ± 0.14 [Ketamine]). Furthermore, HPLC results showed similar trends to those observed in the GluCEST results (left: 2.32 ± 0.22 [Control] vs. 1.96 ± 0.11 [Saline] vs. 2.18 ± 0.11 [Ketamine]; right: 2.35 ± 0.18 [Control] vs. 1.87 ± 0.16 [Saline] vs. 2.09 ± 0.08 [Ketamine]). DATA CONCLUSION GluCEST can sensitively evaluate the ketamine's antidepressant effects by detecting the fast increase in glutamate concentration. LEVEL OF EVIDENCE 1 TECHNICAL EFFICACY STAGE: 1.
Collapse
Affiliation(s)
- Hao Li
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Xunrong Luo
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing University, Chongqing, China
| | - Kai Qi
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Yijie Lv
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Junnan Kan
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Changfeng Yang
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Xiaoqian Lin
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Jin Tao
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Wei Zhang
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Yan Liu
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Kang Rong
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Ailing Wang
- Department of Clinical Laboratory, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Zhongde Jiang
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Xianglin Li
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| |
Collapse
|
27
|
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
|
28
|
Li M, Yang X, Zhu K, Shen L, Xie C. Effects of perioperative intravenous lidocaine and esketamine on the quality of recovery and emotional state of patients after thyroidectomy: A randomised, double-blind, controlled trial. Indian J Anaesth 2024; 68:340-347. [PMID: 38586266 PMCID: PMC10993934 DOI: 10.4103/ija.ija_1010_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 04/09/2024] Open
Abstract
Background and Aims Perioperative intravenous (IV) infusions of lidocaine and esketamine reduce postoperative pain, but there are few studies on the quality of recovery and patients' emotional states postoperatively. We aimed to explore the effects of perioperative IV lidocaine and esketamine on the quality of recovery and emotional state after thyroidectomy. Methods In this randomised trial, 137 patients undergoing thyroidectomy were randomly assigned to three groups: a lidocaine group (Group L), an esketamine group (Group E) and a normal saline placebo group (Group C). The primary outcome was the Quality of Recovery 40 (QoR-40) on postoperative days (PODs) 1 and 2. The secondary outcomes included Self-Rating Anxiety Scale (SAS) and Self-Rating Depression Scale (SDS) scores on days 1 and 2 after surgery, pain scores, opioid consumption and incidence of postoperative nausea and vomiting (PONV). Statistical analysis was performed using the one-way analysis of variance (ANOVA), the Kruskal-Wallis and Chi-square tests. Results The global QoR-40 scores in groups L and E on POD 1 and POD 2 were significantly higher than in group C (P < 0.001). The SAS and SDS scores on POD 1 and POD 2 in groups L and E were significantly lower than in group C (P < 0.05). There were statistically significant differences in Numerical Rating Scale (NRS) scores among the three groups at 1 h, 2 h, 6 h and 12 h (P < 0.05). Conclusion Perioperative IV lidocaine and esketamine improve the quality of postoperative recovery and the emotional state of patients undergoing thyroidectomy.
Collapse
Affiliation(s)
- Mengci Li
- Department of Anaesthesiology, Affiliated Huaian Hospital of Xuzhou Medical University, Huaian, Jiangsu, China
| | - Xiaoqian Yang
- Department of Anaesthesiology, Affiliated Huaian Hospital of Xuzhou Medical University, Huaian, Jiangsu, China
| | - Kairun Zhu
- Department of Anaesthesiology, Affiliated Huaian Hospital of Xuzhou Medical University, Huaian, Jiangsu, China
| | - Lulu Shen
- Department of Anaesthesiology, Affiliated Huaian Hospital of Xuzhou Medical University, Huaian, Jiangsu, China
| | - Chenglan Xie
- Department of Anaesthesiology, Affiliated Huaian Hospital of Xuzhou Medical University, Huaian, Jiangsu, China
| |
Collapse
|
29
|
Pomrenze MB, Vaillancourt S, Llorach P, Rijsketic DR, Casey AB, Gregory N, Salgado JS, Malenka RC, Heifets BD. Opioid receptor expressing neurons of the central amygdala gate behavioral effects of ketamine in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.03.583196. [PMID: 38496451 PMCID: PMC10942405 DOI: 10.1101/2024.03.03.583196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Ketamine has anesthetic, analgesic, and antidepressant properties which may involve multiple neuromodulatory systems. In humans, the opioid receptor (OR) antagonist naltrexone blocks the antidepressant effect of ketamine. It is unclear whether naltrexone blocks a direct effect of ketamine at ORs, or whether normal functioning of the OR system is required to realize the full antidepressant effects of treatment. In mice, the effect of ketamine on locomotion, but not analgesia or the forced swim test, was sensitive to naltrexone and was therefore used as a behavioral readout to localize the effect of naltrexone in the brain. We performed whole-brain imaging of cFos expression in ketamine-treated mice, pretreated with naltrexone or vehicle, and identified the central amygdala (CeA) as the area with greatest difference in cFos intensity. CeA neurons expressing both μOR (MOR) and PKCμ were strongly activated by naltrexone but not ketamine, and selectively interrupting MOR function in the CeA either pharmacologically or genetically blocked the locomotor effects of ketamine. These data suggest that MORs expressed in CeA neurons gate behavioral effects of ketamine but are not direct targets of ketamine.
Collapse
Affiliation(s)
- Matthew B. Pomrenze
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Sam Vaillancourt
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Pierre Llorach
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Daniel Ryskamp Rijsketic
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Austen B. Casey
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Nicholas Gregory
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Juliana S. Salgado
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Robert C. Malenka
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Boris D. Heifets
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
30
|
Yamagishi A, Ikekubo Y, Mishina M, Ikeda K, Ide S. Loss of the sustained antidepressant-like effect of (2R,6R)-hydroxynorketamine in NMDA receptor GluN2D subunit knockout mice. J Pharmacol Sci 2024; 154:203-208. [PMID: 38395521 DOI: 10.1016/j.jphs.2024.01.008] [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: 11/27/2023] [Revised: 01/17/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Ketamine, an N-methyl-d-aspartate (NMDA) receptor antagonist, has attracted attention for its acute and sustained antidepressant effects in patients with depression. Hydroxynorketamine (HNK), a metabolite of ketamine, exerts antidepressant effects without exerting ketamine's side effects and has attracted much attention in recent years. However, the detailed pharmacological mechanism of action of HNK remains unclear. We previously showed that the GluN2D NMDA receptor subunit is important for sustained antidepressant-like effects of (R)-ketamine. Therefore, we investigated whether the GluN2D subunit is involved in antidepressant-like effects of (2R,6R)-HNK and (2S,6S)-HNK. Treatment with (2R,6R)-HNK but not (2S,6S)-HNK exerted acute and sustained antidepressant-like effects in the tail-suspension test in wildtype mice. Interestingly, sustained antidepressant-like effects of (2R,6R)-HNK were abolished in GluN2D-knockout mice, whereas acute antidepressant-like effects were maintained in GluN2D-knockout mice. When expression levels of GluN2A and GluN2B subunits were evaluated, a decrease in GluN2B protein expression in the nucleus accumbens was found in stressed wildtype mice but not in stressed GluN2D-knockout mice. These results suggest that the GluN2D subunit and possibly the GluN2B subunit are involved in the sustained antidepressant-like effect of (2R,6R)-HNK.
Collapse
Affiliation(s)
- Aimi Yamagishi
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-Ku, Tokyo, Japan; Molecular and Cellular Medicine, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yuiko Ikekubo
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-Ku, Tokyo, Japan
| | - Masayoshi Mishina
- Brain Science Laboratory, The Research Organization of Science and Technology, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, Japan
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-Ku, Tokyo, Japan; Molecular and Cellular Medicine, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Soichiro Ide
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-Ku, Tokyo, Japan.
| |
Collapse
|
31
|
Zaccarelli-Magalhães J, Abreu GR, Fukushima AR, Pantaleon LP, Ribeiro BB, Munhoz C, Manes M, de Lima MA, Miglioli J, Flório JC, Lebrun I, Ricci EL, Spinosa HS. Ketamine causes poor maternal care in rats with postpartum depression and leads to few behavioral and neurochemical alterations on male offspring. Behav Brain Res 2024; 459:114799. [PMID: 38065224 DOI: 10.1016/j.bbr.2023.114799] [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: 10/05/2023] [Revised: 12/01/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023]
Abstract
Ketamine is an anesthetic drug that also has antidepressant properties, with quick action. Despite the great number of studies showing its effectiveness as a treatment for major depression, there is little information about its effects on postpartum depression, as pharmacological treatments bring risks to the health of both mother and child. Thus, this study aimed to evaluate the effects of prolonged treatment with subanesthetic doses of ketamine in a rat model of postpartum depression. Female dams were induced to postpartum depression by the maternal separation model from lactating day (LD) 2-12. They were divided into four groups: one control and three experimental groups, which were treated with different doses of ketamine (5, 10 or 20 mg/kg) from LD 2-21 i.p. Maternal studies were conducted from LD5 to LD21 and the offspring studies from postnatal day 2 through 90. Ketamine causes poor maternal care, with few neurochemical alterations. However, the highest dose used in this study had an antidepressant effect. Regarding the male offspring, indirect exposure to ketamine through breast milk caused few behavioral changes during infancy, but they were not permanent, as they faded in adulthood. Nevertheless, this exposure was able to cause alterations in their monoaminergic neurotransmission systems that were found in both infancy and adulthood periods.
Collapse
Affiliation(s)
- Julia Zaccarelli-Magalhães
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Avenida Professor Doutor Orlando Marques de Paiva, 87, 05508-270 São Paulo, Brazil.
| | - Gabriel R Abreu
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Avenida Professor Doutor Orlando Marques de Paiva, 87, 05508-270 São Paulo, Brazil
| | - André R Fukushima
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Avenida Professor Doutor Orlando Marques de Paiva, 87, 05508-270 São Paulo, Brazil; School of Health Sciences IGESP, Rua da Consolação, 1025, 01301-000 São Paulo, Brazil; Centro Universitário das Américas, Rua Augusta, 1508, 01304-001 São Paulo, Brazil
| | - Lorena P Pantaleon
- Health Science Institute, Presbyterian Mackenzie University, Rua da Consolação, 930, 01302-907 São Paulo, Brazil
| | - Beatriz B Ribeiro
- Health Science Institute, Presbyterian Mackenzie University, Rua da Consolação, 930, 01302-907 São Paulo, Brazil
| | - Camila Munhoz
- Health Science Institute, Presbyterian Mackenzie University, Rua da Consolação, 930, 01302-907 São Paulo, Brazil
| | - Marianna Manes
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Avenida Professor Doutor Orlando Marques de Paiva, 87, 05508-270 São Paulo, Brazil
| | - Mayara A de Lima
- Centro Universitário das Américas, Rua Augusta, 1508, 01304-001 São Paulo, Brazil
| | - Júlia Miglioli
- Centro Universitário das Américas, Rua Augusta, 1508, 01304-001 São Paulo, Brazil
| | - Jorge C Flório
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Avenida Professor Doutor Orlando Marques de Paiva, 87, 05508-270 São Paulo, Brazil
| | - Ivo Lebrun
- Laboratory of Biochemistry and Biophysics, Butantan Institute, Avenida Vital Brazil, 1500, 05503-900 São Paulo, Brazil
| | - Esther L Ricci
- School of Health Sciences IGESP, Rua da Consolação, 1025, 01301-000 São Paulo, Brazil; Health Science Institute, Presbyterian Mackenzie University, Rua da Consolação, 930, 01302-907 São Paulo, Brazil
| | - Helenice S Spinosa
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Avenida Professor Doutor Orlando Marques de Paiva, 87, 05508-270 São Paulo, Brazil
| |
Collapse
|
32
|
Ali DN, Ali HM, Lopez MR, Kang S, Choi DS. Astrocytic GABAergic Regulation in Alcohol Use and Major Depressive Disorders. Cells 2024; 13:318. [PMID: 38391931 PMCID: PMC10887002 DOI: 10.3390/cells13040318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/26/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
Gamma-aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the central nervous system (CNS). Most GABAergic neurons synthesize GABA from glutamate and release it in the synaptic cleft in the CNS. However, astrocytes can also synthesize and release GABA, activating GABA receptors in the neighboring neurons in physiological and pathological conditions. As the primary homeostatic glial cells in the brain, astrocytes play a crucial role in regulating GABA homeostasis and synaptic neurotransmission. Accumulating evidence demonstrates that astrocytic GABA dysregulation is implicated in psychiatric disorders, including alcohol use disorder (AUD) and major depressive disorder (MDD), the most prevalent co-occurring psychiatric disorders. Several current medications and emerging pharmacological agents targeting GABA levels are in clinical trials for treating AUD and MDD. This review offers a concise summary of the role of astrocytic GABA regulation in AUD and MDD. We also provide an overview of the current understanding and areas of debate regarding the mechanisms by which astrocytes regulate GABA in the CNS and their potential significance in the molecular basis of AUD and MDD, paving the way toward future research directions and potential therapeutic target areas within this field.
Collapse
Affiliation(s)
- Dina N. Ali
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
| | - Hossam M. Ali
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
| | - Matthew R. Lopez
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
| | - Shinwoo Kang
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
- Neuroscience Program, Rochester, MN 55905, USA
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| |
Collapse
|
33
|
Tiliwaerde M, Gao N, Yang Y, Jin Z. A novel NMDA receptor modulator: the antidepressant effect and mechanism of GW043. CNS Neurosci Ther 2024; 30:e14598. [PMID: 38332552 PMCID: PMC10853642 DOI: 10.1111/cns.14598] [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: 12/18/2023] [Accepted: 12/28/2023] [Indexed: 02/10/2024] Open
Abstract
AIMS The N-methyl-D-aspartate (NMDA) receptor (NMDAR) has been proven to be strongly correlated with rapid antidepressant effects. Here, GW043, as a new compound targeting NMDAR, we explored its antidepressant effects and its mechanism of action. METHODS Our study utilized electrophysiological techniques to confirm the effect of GW043 on NMDAR currents. Additionally, we assessed the selectivity of GW043 through high-throughput receptor-ligand binding experiments. The antidepressant properties of GW043 were examined using rodent behavioral models including the Forced Swim Test (FST), Tail Suspension Test (TST), and Chronic Unpredictable Mild Stress (CUMS). Mechanistic insight into GW043's onset was gained through western blot analysis, BrdU staining, Golgi staining, and electrophysiological techniques. RESULTS Electrophysiological studies indicated that GW043 acts as a partial agonist of NMDAR. Behavioral experiments confirmed the antidepressant effect of GW043 in rodents. Mechanistic investigations revealed that GW043 modulates synaptic plasticity through the LTP and BDNF-mTOR pathways, consequently leading to an increase in the number of newborn neurons and subsequent antidepressant effects. CONCLUSION Our findings disclose that GW043, as a partial agonist of NMDAR, can reverse depression-like behaviors in rats by modulating synaptic plasticity, indicating its potential as an antidepressant agent.
Collapse
Affiliation(s)
- Murezati Tiliwaerde
- Department of Pharmacology, School of Basic Medical SciencesCapital Medical UniversityBeijingChina
| | - Nana Gao
- Department of Gastrointestinal Surgery and Clinical Nutrition, Beijing Shijitan HospitalCaptial Medical UniversityBeijingChina
| | - Yaqi Yang
- Department of Pharmacology, School of Basic Medical SciencesCapital Medical UniversityBeijingChina
- Department of Pharmacy, Beijing Chaoyang HospitalCapital Medical UniversityBeijingChina
| | - Zengliang Jin
- Department of Pharmacology, School of Basic Medical SciencesCapital Medical UniversityBeijingChina
| |
Collapse
|
34
|
Chen Y, Guan W, Wang ML, Lin XY. PI3K-AKT/mTOR Signaling in Psychiatric Disorders: A Valuable Target to Stimulate or Suppress? Int J Neuropsychopharmacol 2024; 27:pyae010. [PMID: 38365306 PMCID: PMC10888523 DOI: 10.1093/ijnp/pyae010] [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: 09/22/2023] [Accepted: 02/08/2024] [Indexed: 02/18/2024] Open
Abstract
Economic development and increased stress have considerably increased the prevalence of psychiatric disorders in recent years, which rank as some of the most prevalent diseases globally. Several factors, including chronic social stress, genetic inheritance, and autogenous diseases, lead to the development and progression of psychiatric disorders. Clinical treatments for psychiatric disorders include psychotherapy, chemotherapy, and electric shock therapy. Although various achievements have been made researching psychiatric disorders, the pathogenesis of these diseases has not been fully understood yet, and serious adverse effects and resistance to antipsychotics are major obstacles to treating patients with psychiatric disorders. Recent studies have shown that the mammalian target of rapamycin (mTOR) is a central signaling hub that functions in nerve growth, synapse formation, and plasticity. The PI3K-AKT/mTOR pathway is a critical target for mediating the rapid antidepressant effects of these pharmacological agents in clinical and preclinical research. Abnormal PI3K-AKT/mTOR signaling is closely associated with the pathogenesis of several neurodevelopmental disorders. In this review, we focused on the role of mTOR signaling and the related aberrant neurogenesis in psychiatric disorders. Elucidating the neurobiology of the PI3K-AKT/mTOR signaling pathway in psychiatric disorders and its actions in response to antidepressants will help us better understand brain development and quickly identify new therapeutic targets for the treatment of these mental illnesses.
Collapse
Affiliation(s)
- Yan Chen
- Department of Neurology, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| | - Wei Guan
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, Jiangsu, China
| | - Mei-Lan Wang
- Department of Neurology, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| | - Xiao-Yun Lin
- Department of Neurology, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
35
|
Di Ianni T, Ewbank SN, Levinstein MR, Azadian MM, Budinich RC, Michaelides M, Airan RD. Sex dependence of opioid-mediated responses to subanesthetic ketamine in rats. Nat Commun 2024; 15:893. [PMID: 38291050 PMCID: PMC10828511 DOI: 10.1038/s41467-024-45157-7] [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: 04/23/2023] [Accepted: 01/17/2024] [Indexed: 02/01/2024] Open
Abstract
Subanesthetic ketamine is increasingly used for the treatment of varied psychiatric conditions, both on- and off-label. While it is commonly classified as an N-methyl D-aspartate receptor (NMDAR) antagonist, our picture of ketamine's mechanistic underpinnings is incomplete. Recent clinical evidence has indicated, controversially, that a component of the efficacy of subanesthetic ketamine may be opioid dependent. Using pharmacological functional ultrasound imaging in rats, we found that blocking opioid receptors suppressed neurophysiologic changes evoked by ketamine, but not by a more selective NMDAR antagonist, in limbic regions implicated in the pathophysiology of depression and in reward processing. Importantly, this opioid-dependent response was strongly sex-dependent, as it was not evident in female subjects and was fully reversed by surgical removal of the male gonads. We observed similar sex-dependent effects of opioid blockade affecting ketamine-evoked postsynaptic density and behavioral sensitization, as well as in opioid blockade-induced changes in opioid receptor density. Together, these results underscore the potential for ketamine to induce its affective responses via opioid signaling, and indicate that this opioid dependence may be strongly influenced by subject sex. These factors should be more directly assessed in future clinical trials.
Collapse
Affiliation(s)
- Tommaso Di Ianni
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Departments of Psychiatry & Behavioral Sciences and Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 94143, USA.
| | - Sedona N Ewbank
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Marjorie R Levinstein
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA
| | - Matine M Azadian
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Reece C Budinich
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Raag D Airan
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Materials Science and Engineering, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
36
|
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
|
37
|
Halvorson CS, Sánchez-Lafuente CL, Johnston JN, Kalynchuk LE, Caruncho HJ. Molecular Mechanisms of Reelin in the Enteric Nervous System and the Microbiota-Gut-Brain Axis: Implications for Depression and Antidepressant Therapy. Int J Mol Sci 2024; 25:814. [PMID: 38255890 PMCID: PMC10815176 DOI: 10.3390/ijms25020814] [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: 11/29/2023] [Revised: 12/30/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Current pharmacological treatments for depression fail to produce adequate remission in a significant proportion of patients. Increasingly, other systems, such as the microbiome-gut-brain axis, are being looked at as putative novel avenues for depression treatment. Dysbiosis and dysregulation along this axis are highly comorbid with the severity of depression symptoms. The endogenous extracellular matrix protein reelin is present in all intestinal layers as well as in myenteric and submucosal ganglia, and its receptors are also present in the gut. Reelin secretion from subepithelial myofibroblasts regulates cellular migration along the crypt-villus axis in the small intestine and colon. Reelin brain expression is downregulated in mood and psychotic disorders, and reelin injections have fast antidepressant-like effects in animal models of depression. This review seeks to discuss the roles of reelin in the gastrointestinal system and propose a putative role for reelin actions in the microbiota-gut-brain axis in the pathogenesis and treatment of depression, primarily reflecting on alterations in gut epithelial cell renewal and in the clustering of serotonin transporters.
Collapse
Affiliation(s)
- Ciara S. Halvorson
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Rd., Victoria, BC V8P 5C2, Canada; (C.S.H.); (C.L.S.-L.); (L.E.K.)
| | - Carla Liria Sánchez-Lafuente
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Rd., Victoria, BC V8P 5C2, Canada; (C.S.H.); (C.L.S.-L.); (L.E.K.)
| | - Jenessa N. Johnston
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Lisa E. Kalynchuk
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Rd., Victoria, BC V8P 5C2, Canada; (C.S.H.); (C.L.S.-L.); (L.E.K.)
| | - Hector J. Caruncho
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Rd., Victoria, BC V8P 5C2, Canada; (C.S.H.); (C.L.S.-L.); (L.E.K.)
| |
Collapse
|
38
|
Jóźwiak-Bębenista M, Sokołowska P, Wiktorowska-Owczarek A, Kowalczyk E, Sienkiewicz M. Ketamine - A New Antidepressant Drug with Anti-Inflammatory Properties. J Pharmacol Exp Ther 2024; 388:134-144. [PMID: 37977808 DOI: 10.1124/jpet.123.001823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/04/2023] [Accepted: 10/04/2023] [Indexed: 11/19/2023] Open
Abstract
Ketamine is a new, potent and rapid-acting antidepressant approved for therapy of treatment-resistant depression, which has a different mechanism of action than currently-available antidepressant therapies. It owes its uniquely potent antidepressant properties to a complex mechanism of action, which currently remains unclear. However, it is thought that it acts by modulating the functioning of the glutamatergic system, which plays an important role in the process of neuroplasticity associated with depression. However, preclinical and clinical studies have also found ketamine to reduce inflammation, either directly or indirectly (by activating neuroprotective branches of the kynurenine pathway), among patients exhibiting higher levels of inflammation. Inflammation and immune system activation are believed to play key roles in the development and course of depression. Therefore, the present work examines the role of the antidepressant effect of ketamine and its anti-inflammatory properties in the treatment of depression. SIGNIFICANCE STATEMENT: The present work examines the relationship between the antidepressant effect of ketamine and its anti-inflammatory properties, and the resulting benefits in treatment-resistant depression (TRD). The antidepressant mechanism of ketamine remains unclear, and there is an urgent need to develop new therapeutic strategies for treatment of depression, particularly TRD.
Collapse
Affiliation(s)
- Marta Jóźwiak-Bębenista
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| | - Paulina Sokołowska
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| | - Anna Wiktorowska-Owczarek
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| | - Edward Kowalczyk
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| | - Monika Sienkiewicz
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| |
Collapse
|
39
|
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
|
40
|
Hanson JE, Yuan H, Perszyk RE, Banke TG, Xing H, Tsai MC, Menniti FS, Traynelis SF. Therapeutic potential of N-methyl-D-aspartate receptor modulators in psychiatry. Neuropsychopharmacology 2024; 49:51-66. [PMID: 37369776 PMCID: PMC10700609 DOI: 10.1038/s41386-023-01614-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/24/2023] [Accepted: 05/15/2023] [Indexed: 06/29/2023]
Abstract
N-methyl-D-aspartate (NMDA) receptors mediate a slow component of excitatory synaptic transmission, are widely distributed throughout the central nervous system, and regulate synaptic plasticity. NMDA receptor modulators have long been considered as potential treatments for psychiatric disorders including depression and schizophrenia, neurodevelopmental disorders such as Rett Syndrome, and neurodegenerative conditions such as Alzheimer's disease. New interest in NMDA receptors as therapeutic targets has been spurred by the findings that certain inhibitors of NMDA receptors produce surprisingly rapid and robust antidepressant activity by a novel mechanism, the induction of changes in the brain that well outlast the presence of drug in the body. These findings are driving research into an entirely new paradigm for using NMDA receptor antagonists in a host of related conditions. At the same time positive allosteric modulators of NMDA receptors are being pursued for enhancing synaptic function in diseases that feature NMDA receptor hypofunction. While there is great promise, developing the therapeutic potential of NMDA receptor modulators must also navigate the potential significant risks posed by the use of such agents. We review here the emerging pharmacology of agents that target different NMDA receptor subtypes, offering new avenues for capturing the therapeutic potential of targeting this important receptor class.
Collapse
Affiliation(s)
- Jesse E Hanson
- Department of Neuroscience, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Hongjie Yuan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Riley E Perszyk
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Tue G Banke
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Hao Xing
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ming-Chi Tsai
- Department of Neuroscience, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Frank S Menniti
- MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA.
| | - Stephen F Traynelis
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
41
|
Duan C, Zhu Y, Zhang Z, Wu T, Shen M, Xu J, Gao W, Pan J, Wei L, Su H, Shi C. Esketamine inhibits the c-Jun N-terminal kinase pathway in the spinal dorsal horn to relieve bone cancer pain in rats. Mol Pain 2024; 20:17448069241239231. [PMID: 38417838 PMCID: PMC10938627 DOI: 10.1177/17448069241239231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/04/2024] [Accepted: 02/25/2024] [Indexed: 03/01/2024] Open
Abstract
Cancer-induced bone pain (CIBP) is one of the most common and feared symptoms in patients with advanced tumors. The X-C motif chemokine ligand 12 (CXCL12) and the CXCR4 receptor have been associated with glial cell activation in bone cancer pain. Moreover, mitogen-activated protein kinases (MAPKs), as downstream CXCL12/CXCR4 signals, and c-Jun, as activator protein AP-1 components, contribute to the development of various types of pain. However, the specific CIBP mechanisms remain unknown. Esketamine is a non-selective N-methyl-d-aspartic acid receptor (NMDA) inhibitor commonly used as an analgesic in the clinic, but its analgesic mechanism in bone cancer pain remains unclear. We used a tumor cell implantation (TCI) model and explored that CXCL12/CXCR4, p-MAPKs, and p-c-Jun were stably up-regulated in the spinal cord. Immunofluorescence images showed activated microglia in the spinal cord on day 14 after TCI and co-expression of CXCL12/CXCR4, p-MAPKs (p-JNK, p-ERK, p-p38 MAPK), and p-c-Jun in microglia. Intrathecal injection of the CXCR4 inhibitor AMD3100 reduced JNK and c-Jun phosphorylations, and intrathecal injection of the JNK inhibitor SP600125 and esketamine also alleviated TCI-induced pain and reduced the expression of p-JNK and p-c-Jun in microglia. Overall, our data suggest that the CXCL12/CXCR4-JNK-c-Jun signaling pathway of microglia in the spinal cord mediates neuronal sensitization and pain hypersensitivity in cancer-induced bone pain and that esketamine exerts its analgesic effect by inhibiting the JNK-c-Jun pathway.
Collapse
Affiliation(s)
- Chenxia Duan
- Department of Anesthesiology, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yi Zhu
- Department of Anesthesiology, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Zhuoliang Zhang
- Department of Anesthesiology, Suzhou Municipal Hospital, Xuzhou Medical University, Suzhou, China
| | - Tiantian Wu
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Mengwei Shen
- Department of Anesthesiology, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jinfu Xu
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Wenxin Gao
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Jianhua Pan
- Department of Anesthesiology, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Lei Wei
- Department of Anesthesiology, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Huibin Su
- Department of Anesthesiology, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Chenghuan Shi
- Department of Anesthesiology, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| |
Collapse
|
42
|
Nguyen TML, Jollant F, Tritschler L, Colle R, Corruble E, Gardier AM. [Ketamine and suicidal behavior: Contribution of animal models of aggression-impulsivity to understanding its mechanism of action]. ANNALES PHARMACEUTIQUES FRANÇAISES 2024; 82:3-14. [PMID: 37890717 DOI: 10.1016/j.pharma.2023.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023]
Abstract
More than two-thirds of suicides occur during a major depressive episode. Acting out prevention measures and therapeutic options to manage the suicidal crisis are limited. The impulsive-aggressive dimensions are vulnerability factors associated with suicide in patients suffering from a characterized depressive episode: this can be a dimension involved in animals. Impulsive and aggressive rodent models can help analyze, at least in part, the neurobiology of suicide and the beneficial effects of treatments. Ketamine, a glutamatergic antagonist, by rapidly improving the symptoms of depressive episodes, would help reduce suicidal thoughts in the short term. Animal models share with humans impulsive and aggressive endophenotypes modulated by the serotonergic system (5-HTB receptor, MAO-A enzyme), neuroinflammation or the hypothalamic-pituitary-adrenal axis and stress. Significant effects of ketamine on these endophenotypes remain to be demonstrated.
Collapse
Affiliation(s)
- Thi Mai Loan Nguyen
- Inserm CESP/UMR 1018, équipe MOODS, faculté de pharmacie, université Paris-Saclay, 91400 Orsay, France
| | - Fabrice Jollant
- Inserm CESP/UMR 1018, équipe MOODS, faculté de médecine, université Paris-Saclay, 94270 Le Kremin-Bicêtre, France; Service hospitalo-universitaire de psychiatrie, hôpital de Bicêtre, hôpitaux universitaires Paris-Saclay, Assistance publique-Hôpitaux de Paris (AP-HP), 94275 Le Kremlin-Bicêtre, France; Pôle de psychiatrie, CHU de Nîmes, Nîmes, France; Département de psychiatrie, Université McGill et Groupe McGill d'études sur le suicide, Montréal, Canada
| | - Laurent Tritschler
- Inserm CESP/UMR 1018, équipe MOODS, faculté de pharmacie, université Paris-Saclay, 91400 Orsay, France
| | - Romain Colle
- Inserm CESP/UMR 1018, équipe MOODS, faculté de médecine, université Paris-Saclay, 94270 Le Kremin-Bicêtre, France; Service hospitalo-universitaire de psychiatrie, hôpital de Bicêtre, hôpitaux universitaires Paris-Saclay, Assistance publique-Hôpitaux de Paris (AP-HP), 94275 Le Kremlin-Bicêtre, France
| | - Emmanuelle Corruble
- Inserm CESP/UMR 1018, équipe MOODS, faculté de médecine, université Paris-Saclay, 94270 Le Kremin-Bicêtre, France; Service hospitalo-universitaire de psychiatrie, hôpital de Bicêtre, hôpitaux universitaires Paris-Saclay, Assistance publique-Hôpitaux de Paris (AP-HP), 94275 Le Kremlin-Bicêtre, France
| | - Alain M Gardier
- Inserm CESP/UMR 1018, équipe MOODS, faculté de pharmacie, université Paris-Saclay, 91400 Orsay, France.
| |
Collapse
|
43
|
Zaretsky TG, Jagodnik KM, Barsic R, Antonio JH, Bonanno PA, MacLeod C, Pierce C, Carney H, Morrison MT, Saylor C, Danias G, Lepow L, Yehuda R. The Psychedelic Future of Post-Traumatic Stress Disorder Treatment. Curr Neuropharmacol 2024; 22:636-735. [PMID: 38284341 PMCID: PMC10845102 DOI: 10.2174/1570159x22666231027111147] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 01/30/2024] Open
Abstract
Post-traumatic stress disorder (PTSD) is a mental health condition that can occur following exposure to a traumatic experience. An estimated 12 million U.S. adults are presently affected by this disorder. Current treatments include psychological therapies (e.g., exposure-based interventions) and pharmacological treatments (e.g., selective serotonin reuptake inhibitors (SSRIs)). However, a significant proportion of patients receiving standard-of-care therapies for PTSD remain symptomatic, and new approaches for this and other trauma-related mental health conditions are greatly needed. Psychedelic compounds that alter cognition, perception, and mood are currently being examined for their efficacy in treating PTSD despite their current status as Drug Enforcement Administration (DEA)- scheduled substances. Initial clinical trials have demonstrated the potential value of psychedelicassisted therapy to treat PTSD and other psychiatric disorders. In this comprehensive review, we summarize the state of the science of PTSD clinical care, including current treatments and their shortcomings. We review clinical studies of psychedelic interventions to treat PTSD, trauma-related disorders, and common comorbidities. The classic psychedelics psilocybin, lysergic acid diethylamide (LSD), and N,N-dimethyltryptamine (DMT) and DMT-containing ayahuasca, as well as the entactogen 3,4-methylenedioxymethamphetamine (MDMA) and the dissociative anesthetic ketamine, are reviewed. For each drug, we present the history of use, psychological and somatic effects, pharmacology, and safety profile. The rationale and proposed mechanisms for use in treating PTSD and traumarelated disorders are discussed. This review concludes with an in-depth consideration of future directions for the psychiatric applications of psychedelics to maximize therapeutic benefit and minimize risk in individuals and communities impacted by trauma-related conditions.
Collapse
Affiliation(s)
- Tamar Glatman Zaretsky
- James J. Peters Veterans Affairs Medical Center, New York, NY, USA
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kathleen M. Jagodnik
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Barsic
- James J. Peters Veterans Affairs Medical Center, New York, NY, USA
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Josimar Hernandez Antonio
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Philip A. Bonanno
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carolyn MacLeod
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Charlotte Pierce
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hunter Carney
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Morgan T. Morrison
- James J. Peters Veterans Affairs Medical Center, New York, NY, USA
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Charles Saylor
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - George Danias
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lauren Lepow
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rachel Yehuda
- James J. Peters Veterans Affairs Medical Center, New York, NY, USA
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
44
|
Savić Vujović K, Jotić A, Medić B, Srebro D, Vujović A, Žujović J, Opanković A, Vučković S. Ketamine, an Old-New Drug: Uses and Abuses. Pharmaceuticals (Basel) 2023; 17:16. [PMID: 38276001 PMCID: PMC10820504 DOI: 10.3390/ph17010016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 01/27/2024] Open
Abstract
Ketamine as an old-new drug has a variety of clinical implications. In the last 30 years, ketamine has become popular for acute use in humans. Ketamine in standard doses is principally utilized for the induction and maintenance of surgical procedures. Besides its use in anesthesia and analgesia, recent studies have shown that ketamine has found a place in the treatment of asthma, epilepsy, depression, bipolar affective disorders, alcohol and heroin addiction. Ketamine primarily functions as a noncompetitive antagonist targeting the N-methyl-D-aspartate (NMDA) receptor, but its mechanism of action is complex. It is generally regarded as safe, with low doses and short-term use typically not leading to significant adverse effects. Also, ketamine is known as a powerful psychostimulant. During the past decade, ketamine has been one of the commonly abused drugs.
Collapse
Affiliation(s)
- Katarina Savić Vujović
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (B.M.); (D.S.); (S.V.)
| | - Ana Jotić
- Clinic for Otorhinolaryngology and Maxillofacial Surgery, University Clinical Center of Serbia, Pasterova 2, 11000 Belgrade, Serbia;
| | - Branislava Medić
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (B.M.); (D.S.); (S.V.)
| | - Dragana Srebro
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (B.M.); (D.S.); (S.V.)
| | | | - Janko Žujović
- Clinical Centre of Montenegro, Centre for Abdominal Surgery, 81000 Podgorica, Montenegro;
| | - Ana Opanković
- Clinical Centre of Serbia, Clinic for Psychiatry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Sonja Vučković
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (B.M.); (D.S.); (S.V.)
| |
Collapse
|
45
|
Hilal F, Jeanblanc J, Naassila M. [Interest and mechanisms of action of ketamine in alcohol addiction- A review of clinical and preclinical studies]. Biol Aujourdhui 2023; 217:161-182. [PMID: 38018944 DOI: 10.1051/jbio/2023028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Indexed: 11/30/2023]
Abstract
Alcohol Use Disorder (AUD) is a psychiatric condition characterized by chronic and excessive drinking despite negative consequences on overall health and social or occupational functioning. There are currently limited treatment options available for AUD, and the effects size and the response rates to these treatments are often low to moderate. The World Health Organization has identified the development of medications to treat AUD as one of its 24 priorities. This past decade was marked by a renewed interest in psychedelic use in psychiatry. At the centre of this renaissance, ketamine, an atypical psychedelic already used in the treatment of major depression, is an NMDA receptor antagonist that exists as a racemic compound made of two enantiomers, S-ketamine, and R-ketamine. Each form can be metabolized into different metabolites, some of which having antidepressant properties. In this article, we review both clinical and preclinical studies on ketamine and its metabolites in the treatment of AUD. Preclinical as well as clinical studies have revealed that ketamine is effective in reducing withdrawal symptoms and alcohol craving. Convergent data showed that antidepressant properties of ketamine largely contribute to the decreased likelihood of alcohol relapse, especially in patients undergoing ketamine-assisted psychotherapies. Its effectiveness is believed to be linked with its ability to regulate the glutamatergic pathway, enhance neuroplasticity, rewire brain resting state network functional connectivity and decrease depressive-like states. However, it remains to further investigate (i) why strong differences exist between male and female responses in preclinical studies and (ii) the respective roles of each of the metabolites in the ketamine effects in both genders. Interestingly, current studies are also focusing on ketamine addiction and the comorbidity between alcohol addiction and depression occurring more frequently in females.
Collapse
Affiliation(s)
- Fahd Hilal
- Groupe de recherche sur l'alcool et les pharmacodépendances, INSERM U1247, CURS, Amiens, France
| | - Jérôme Jeanblanc
- Groupe de recherche sur l'alcool et les pharmacodépendances, INSERM U1247, CURS, Amiens, France
| | - Mickaël Naassila
- Groupe de recherche sur l'alcool et les pharmacodépendances, INSERM U1247, CURS, Amiens, France
| |
Collapse
|
46
|
Vecera CM, C. Courtes A, Jones G, Soares JC, Machado-Vieira R. Pharmacotherapies Targeting GABA-Glutamate Neurotransmission for Treatment-Resistant Depression. Pharmaceuticals (Basel) 2023; 16:1572. [PMID: 38004437 PMCID: PMC10675154 DOI: 10.3390/ph16111572] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/22/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Treatment-resistant depression (TRD) is a term used to describe a particular type of major depressive disorder (MDD). There is no consensus about what defines TRD, with various studies describing between 1 and 4 failures of antidepressant therapies, with or without electroconvulsive therapy (ECT). That is why TRD is such a growing concern among clinicians and researchers, and it explains the necessity for investigating novel therapeutic targets beyond conventional monoamine pathways. An imbalance between two primary central nervous system (CNS) neurotransmitters, L-glutamate and γ-aminobutyric acid (GABA), has emerged as having a key role in the pathophysiology of TRD. In this review, we provide an evaluation and comprehensive review of investigational antidepressants targeting these two systems, accessing their levels of available evidence, mechanisms of action, and safety profiles. N-methyl-D-aspartate (NMDA) receptor antagonism has shown the most promise amongst the glutamatergic targets, with ketamine and esketamine (Spravato) robustly generating responses across trials. Two specific NMDA-glycine site modulators, D-cycloserine (DCS) and apimostinel, have also generated promising initial safety and efficacy profiles, warranting further investigation. Combination dextromethorphan-bupropion (AXS-05/Auvelity) displays a unique mechanism of action and demonstrated positive results in particular applicability in subpopulations with cognitive dysfunction. Currently, the most promising GABA modulators appear to be synthetic neurosteroid analogs with positive GABAA receptor modulation (such as brexanolone). Overall, advances in the last decade provide exciting perspectives for those who do not improve with conventional therapies. Of the compounds reviewed here, three are approved by the Food and Drug Administration (FDA): esketamine (Spravato) for TRD, Auvelity (dextromethorphan-bupropion) for major depressive disorder (MDD), and brexanolone (Zulresso) for post-partum depression (PPD). Notably, some concerns have arisen with esketamine and brexanolone, which will be detailed in this study.
Collapse
Affiliation(s)
- Courtney M. Vecera
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center, Houston, TX 77054, USA
| | - Alan C. Courtes
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center, Houston, TX 77054, USA
| | - Gregory Jones
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center, Houston, TX 77054, USA
| | - Jair C. Soares
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center, Houston, TX 77054, USA
| | - Rodrigo Machado-Vieira
- John S. Dunn Behavioral Sciences Center at UTHealth Houston, 5615 H.Mark Crosswell Jr St, Houston, TX 77021, USA
| |
Collapse
|
47
|
Lii TR, Smith AE, Flohr JR, Okada RL, Nyongesa CA, Cianfichi LJ, Hack LM, Schatzberg AF, Heifets BD. Randomized trial of ketamine masked by surgical anesthesia in patients with depression. NATURE MENTAL HEALTH 2023; 1:876-886. [PMID: 38188539 PMCID: PMC10769130 DOI: 10.1038/s44220-023-00140-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/14/2023] [Indexed: 01/09/2024]
Abstract
Ketamine may have antidepressant properties, but its acute psychoactive effects complicate successful masking in placebo-controlled trials. We present a single-center, parallel-arm, triple-masked, randomized, placebo-controlled trial assessing the antidepressant efficacy of intravenous ketamine masked by surgical anesthesia (ClinicalTrials.gov, NCT03861988). Forty adult patients with major depressive disorder who were scheduled for routine surgery were randomized to a single infusion of ketamine (0.5 mg/kg) or placebo (saline) during usual anesthesia. All participants, investigators, and direct patient care staff were masked to treatment allocation. The primary outcome was depression severity measured by the Montgomery-Åsberg Depression Rating Scale (MADRS) at 1, 2, and 3 days post-infusion. After all follow-up visits, participants were asked to guess which intervention they received. A mixed-effects model showed no evidence of effect of treatment assignment on the primary outcome (-5.82, 95% CI -13.3 to 1.64, p=0.13). 36.8% of participants guessed their treatment assignment correctly; both groups allocated their guesses in similar proportions. In conclusion, a single dose of intravenous ketamine delivered during surgical anesthesia had no greater effect than placebo in acutely reducing the severity of depressive symptoms in adults with major depressive disorder. This trial successfully masked treatment allocation in moderate-to-severely depressed patients using surgical anesthesia. Although this masking strategy is impractical for most placebo-controlled trials, future studies of novel antidepressants with acute psychoactive effects should make efforts to fully mask treatment assignment in order to minimize subject-expectancy bias.
Collapse
Affiliation(s)
- Theresa R Lii
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ashleigh E Smith
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Josephine R Flohr
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Robin L Okada
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Cynthia A Nyongesa
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Lisa J Cianfichi
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura M Hack
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Sierra-Pacific Mental Illness Research, Education, and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Alan F Schatzberg
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Boris D Heifets
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
48
|
Wu G, Xu H. A synopsis of multitarget therapeutic effects of anesthetics on depression. Eur J Pharmacol 2023; 957:176032. [PMID: 37660970 DOI: 10.1016/j.ejphar.2023.176032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/05/2023]
Abstract
Depression is a profound mental disorder that dampens the mood and undermines volition, which exhibited an increased incidence over the years. Although drug-based interventions remain the primary approach for depression treatment, the available medications still can't satisfy the patients. In recent years, the newly discovered therapeutic targets such as N-methyl-D-aspartate (NMDA) receptor, α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) receptor, and tyrosine kinase B (TrkB) have brought new breakthroughs in the development of antidepressant drugs. Moreover, it has come to light that certain anesthetics possess pharmacological mechanisms intricately linked to the aforementioned therapeutic targets for depression. At present, numerous preclinical and clinical studies have explored the therapeutic effects of anesthetic drugs such as ketamine, isoflurane, N2O, and propofol, on depression. These investigations suggested that these drugs can swiftly ameliorate patients' depression symptoms and engender long-term effects. In this paper, we provide a comprehensive review of the research progress and potential molecular mechanisms of various anesthetic drugs for depression treatment. By shedding light on this subject, we aim to facilitate the development and clinical implementation of new antidepressant drugs based on anesthetic medications.
Collapse
Affiliation(s)
- Guowei Wu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Hongwei Xu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China.
| |
Collapse
|
49
|
Chen Y, Yan P, Wei S, Zhu Y, Lai J, Zhou Q. Ketamine metabolite alleviates morphine withdrawal-induced anxiety via modulating nucleus accumbens parvalbumin neurons in male mice. Neurobiol Dis 2023; 186:106279. [PMID: 37661023 DOI: 10.1016/j.nbd.2023.106279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/20/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023] Open
Abstract
Opioid withdrawal generates extremely unpleasant physical symptoms and negative affective states. A rapid relief of opioid withdrawal-induced anxiety has obvious clinical relevance but has been rarely reported. We have shown that injection of ketamine metabolite (2R,6R)-hydroxynorketamine (HNK) leads to a rapid alleviation of anxiety-like behaviors in male mice undergoing chronic morphine withdrawal. Here we investigated the contribution of nucleus accumbens shell (sNAc) parvalbumin (PV)-neurons to this process. Chronic morphine withdrawal was associated with higher intrinsic excitability of sNAc PV-neurons via reduced voltage-dependent potassium currents. Chemogenetic inhibition of sNAc PV-neurons reversed the enhanced excitability of PV-neurons and anxiety-like behaviors in these morphine withdrawal male mice, while activation of sNAc PV-neurons induced anxiety-like behaviors in naive male mice. (2R,6R)-HNK reversed the altered potassium currents and intrinsic excitability of sNAc PV-neurons. Our findings demonstrate an important contribution of sNAc PV-neurons to modulating morphine withdrawal-induced anxiety-like behaviors and rapid relief of anxiety-like behaviors by (2R,6R)-HNK, this newly identified target may have therapeutic potentials in treating opioid addiction and anxiety disorders.
Collapse
Affiliation(s)
- Yuanyuan Chen
- College of Forensic Science, Xi'an Jiaotong University, Xi'an, China; School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Peng Yan
- College of Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Shuguang Wei
- College of Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Yongsheng Zhu
- College of Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Jianghua Lai
- College of Forensic Science, Xi'an Jiaotong University, Xi'an, China.
| | - Qiang Zhou
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China.
| |
Collapse
|
50
|
Fonseca C, Ettcheto M, Bicker J, Fernandes MJ, Falcão A, Camins A, Fortuna A. Under the umbrella of depression and Alzheimer's disease physiopathology: Can cannabinoids be a dual-pleiotropic therapy? Ageing Res Rev 2023; 90:101998. [PMID: 37414155 DOI: 10.1016/j.arr.2023.101998] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/17/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
Depression and Alzheimer´s disease (AD) are two disorders highly prevalent worldwide. Depression affects more than 300 million people worldwide while AD affects 60-80% of the 55 million cases of dementia. Both diseases are affected by aging with high prevalence in elderly and share not only the main brain affected areas but also several physiopathological mechanisms. Depression disease is already ascribed as a risk factor to the development of AD. Despite the wide diversity of pharmacological treatments currently available in clinical practice for depression management, they remain associated to a slow recovery process and to treatment-resistant depression. On the other hand, AD treatment is essentially based in symptomatology relieve. Thus, the need for new multi-target treatments arises. Herein, we discuss the current state-of-art regarding the contribution of the endocannabinoid system (ECS) in synaptic transmission processes, synapses plasticity and neurogenesis and consequently the use of exogenous cannabinoids in the treatment of depression and on delaying the progression of AD. Besides the well-known imbalance of neurotransmitter levels, including serotonin, noradrenaline, dopamine and glutamate, recent scientific evidence highlights aberrant spine density, neuroinflammation, dysregulation of neurotrophic factor levels and formation of amyloid beta (Aβ) peptides, as the main physiopathological mechanisms compromised in depression and AD. The contribution of the ECS in these mechanisms is herein specified as well as the pleiotropic effects of phytocannabinoids. At the end, it became evident that Cannabinol, Cannabidiol, Cannabigerol, Cannabidivarin and Cannabichromene may act in novel therapeutic targets, presenting high potential in the pharmacotherapy of both diseases.
Collapse
Affiliation(s)
- Carla Fonseca
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal; Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain
| | - Joana Bicker
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Maria José Fernandes
- Departamento de Neurologia/Neurocirurgia, Escola Paulista de Medicina, Universidade Federal de São Paulo-UNIFESP, Rua Pedro de Toledo, 669, CEP, São Paulo 04039-032, Brazil
| | - Amílcar Falcão
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain
| | - Ana Fortuna
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|