1
|
Shao M, Botvinov J, Banerjee D, Girirajan S, Lüscher B. Transcriptome signatures of the medial prefrontal cortex underlying GABAergic control of resilience to chronic stress exposure. Mol Psychiatry 2025; 30:2197-2209. [PMID: 39550415 PMCID: PMC12014471 DOI: 10.1038/s41380-024-02832-x] [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: 07/10/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 11/18/2024]
Abstract
Analyses of postmortem human brains and preclinical studies of rodents have identified somatostatin (SST)-positive, dendrite-targeting GABAergic interneurons as key elements that regulate the vulnerability to stress-related psychiatric disorders. Conversely, genetically induced disinhibition of SST neurons (induced by Cre-mediated deletion of the γ2 GABAA receptor subunit gene selectively from SST neurons, SSTCre:γ2f/f mice) results in stress resilience. Similarly, chronic chemogenetic activation of SST neurons in the medial prefrontal cortex (mPFC) results in stress resilience but only in male and not in female mice. Here, we used RNA sequencing of the mPFC of SSTCre:γ2f/f mice to characterize the transcriptome changes underlying GABAergic control of stress resilience. We found that stress resilience of male but not female SSTCre:γ2f/f mice is characterized by resilience to chronic stress-induced transcriptome changes in the mPFC. Interestingly, the transcriptome of non-stressed SSTCre:γ2f/f (stress-resilient) male mice resembled that of chronic stress-exposed SSTCre (stress-vulnerable) mice. However, the behavior and the serum corticosterone levels of non-stressed SSTCre:γ2f/f mice showed no signs of physiological stress. Most strikingly, chronic stress exposure of SSTCre:γ2f/f mice was associated with an almost complete reversal of their chronic stress-like transcriptome signature, along with pathway changes suggesting stress-induced enhancement of mRNA translation. Behaviorally, the SSTCre:γ2f/f mice were not only resilient to chronic stress-induced anhedonia - they also showed an inversed, anxiolytic-like behavioral response to chronic stress exposure that mirrored the chronic stress-induced reversal of the chronic stress-like transcriptome signature. We conclude that GABAergic dendritic inhibition by SST neurons exerts bidirectional control over behavioral vulnerability and resilience to chronic stress exposure that is mirrored in bidirectional changes in the expression of putative stress resilience genes, through a sex-specific brain substrate.
Collapse
Affiliation(s)
- Meiyu Shao
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Julia Botvinov
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Deepro Banerjee
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Santhosh Girirajan
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Bernhard Lüscher
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA.
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA.
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
2
|
Essam RM, Mohamed YS, El-Sayed SS, Kamel NM. Linking KATP channel activation to p-AKT/mTORC1/eEF2/BDNF axis unravels Nicorandil's promise in countering acetaminophen-induced hepatic encephalopathy in mice. Life Sci 2025; 366-367:123477. [PMID: 39983823 DOI: 10.1016/j.lfs.2025.123477] [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: 11/12/2024] [Revised: 02/04/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025]
Abstract
Nicorandil (NIC), an antianginal agent that acts both as an opener of adenosine triphosphate-sensitive potassium (KATP) channels and a nitric oxide donor, has demonstrated protective and curative effects in various diseases. The predominance of these mechanisms varies based on the dose of NIC and the specific organ affected. This study scrutinized the possible beneficial effects of NIC in acetaminophen (APAP)-induced hepatic encephalopathy (HE) model through highlighting the role of KATP channels in mediating these effects. Forty-eight mice were randomly subdivided into four groups: control (saline), APAP model (1 g/kg, i.p.), NIC treatment (15 mg/kg/day p.o. for 14 days), and glibenclamide (GLIB "KATP blocker", 5 mg/kg/day, p.o. 1 h before NIC for 14 days). NIC significantly mitigated APAP-induced liver injury, hyperammonemia, and cognitive deficits, as evidenced by reduced serum alanine aminotransferase, aspartate aminotransferase, ammonia levels, and improved performance in Y-maze and Morris Water Maze tests. Mechanistically, NIC suppressed hippocampal glutamate, activated phosphoserine 473 protein kinase B (p-AKT(Ser473))/mammalian target of rapamycin complex 1 (mTORC1) pathway, lessened the inactive phosphorylation of eukaryotic elongation factor 2 (eEF2), upsurged brain-derived neurotrophic factor (BDNF), leading to reduced neuroinflammation proved by nuclear factor-kappa B and tumor necrosis factor-alpha suppression. Histopathological analyses revealed improved liver and hippocampal morphology, while immunohistochemistry showed reduced astrocyte activation with NIC treatment. These effects were abolished by GLIB pre-treatment, indicating the crucial role of KATP channel. Accordingly, NIC could alleviate APAP-induced liver injury and HE mainly dependent on KATP channel opening, with resultant inhibition of glutamate signaling, activation of p-AKT/mTORC1/eEF2/BDNF trajectory, and abating hippocampal inflammation.
Collapse
Affiliation(s)
- Reham M Essam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt; Biology Department, School of Pharmacy, Newgiza University, Giza 12577, Egypt.
| | - Yasmin S Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt.
| | - Sarah S El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt.
| | - Nada M Kamel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt.
| |
Collapse
|
3
|
Onisiforou A, Michael A, Apostolakis M, Mammadov E, Mitka A, Kalatta MA, Koumas M, Georgiou A, Chatzittofis A, Panayiotou G, Georgiou P, Zarate CA, Zanos P. Ketamine and Hydroxynorketamine as Novel Pharmacotherapies for the Treatment of Opioid Use Disorders. Biol Psychiatry 2025; 97:563-579. [PMID: 39293647 PMCID: PMC11839383 DOI: 10.1016/j.biopsych.2024.09.008] [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: 07/10/2024] [Revised: 08/30/2024] [Accepted: 09/11/2024] [Indexed: 09/20/2024]
Abstract
Opioid use disorder (OUD) has reached epidemic proportions, with many countries facing high levels of opioid use and related fatalities. Although currently prescribed medications for OUD are considered lifesaving, they inadequately address negative affect and cognitive impairment, resulting in high relapse rates to nonmedical opioid use even years after drug cessation (protracted abstinence). Evidence supports the notion that ketamine, an anesthetic and rapid-acting antidepressant drug, holds promise as a candidate for OUD treatment, including the management of acute withdrawal somatic symptoms, negative affect during protracted opioid abstinence, and prevention of retaking nonmedical opioids. In this review, we comprehensively discuss preclinical and clinical research that has evaluated ketamine and its metabolites as potential novel therapeutic strategies for treating OUD. Furthermore, we examine evidence that supports the relevance of the molecular targets of ketamine and its metabolites in relation to their potential effects and therapeutic outcomes in OUD. Overall, existing evidence demonstrates that ketamine and its metabolites can effectively modulate pathophysiological processes affected in OUD, suggesting a promising therapeutic role in the treatment of OUD and the prevention of return to opioid use during abstinence.
Collapse
Affiliation(s)
- Anna Onisiforou
- Department of Psychology, University of Cyprus, Nicosia, Cyprus; Center for Applied Neuroscience, University of Cyprus, Nicosia, Cyprus
| | - Andria Michael
- Department of Psychology, University of Cyprus, Nicosia, Cyprus; Center for Applied Neuroscience, University of Cyprus, Nicosia, Cyprus
| | - Markos Apostolakis
- Department of Psychology, University of Cyprus, Nicosia, Cyprus; Center for Applied Neuroscience, University of Cyprus, Nicosia, Cyprus
| | - Elmar Mammadov
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Angeliki Mitka
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
| | - Maria A Kalatta
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
| | - Morfeas Koumas
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | - Andrea Georgiou
- Department of Psychology, University of Cyprus, Nicosia, Cyprus; Center for Applied Neuroscience, University of Cyprus, Nicosia, Cyprus
| | - Andreas Chatzittofis
- Department of Clinical Sciences/Psychiatry, Umeå University, Umeå, Sweden; Medical School, University of Cyprus, Nicosia, Cyprus
| | - Georgia Panayiotou
- Department of Psychology, University of Cyprus, Nicosia, Cyprus; Center for Applied Neuroscience, University of Cyprus, Nicosia, Cyprus
| | - Polymnia Georgiou
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Panos Zanos
- Department of Psychology, University of Cyprus, Nicosia, Cyprus; Center for Applied Neuroscience, University of Cyprus, Nicosia, Cyprus.
| |
Collapse
|
4
|
Sołek P, Stępniowska A, Koszła O, Jankowski J, Ognik K. Antibiotics/coccidiostat exposure induces gut-brain axis remodeling for Akt/mTOR activation and BDNF-mediated neuroprotection in APEC-infected turkeys. Poult Sci 2025; 104:104636. [PMID: 39721265 PMCID: PMC11732450 DOI: 10.1016/j.psj.2024.104636] [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: 10/04/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
The poultry industry relies extensively on antibiotics and coccidiostats as essential tools for disease management and productivity enhancement. However, increasing concerns about antimicrobial resistance (AMR) and the toxicological safety of these substances have prompted a deeper examination of their broader impacts on animal and human health. This study investigates the toxicological effects of antibiotics and coccidiostats on the gut-brain axis and microbiota in turkeys, with a particular focus on molecular mechanisms that may influence neurochemical and inflammatory responses. Our findings reveal that enrofloxacin exposure leads to the upregulation of BDNF, suggesting a neuroprotective effect, while monensin treatment significantly increased eEF2 kinase expression, indicative enhanced neuronal activity. In turkeys infected with Avian Pathogenic Escherichia coli (APEC), early administration of doxycycline and monensin significantly upregulated the mTOR/BDNF and Akt/mTOR pathways, along with elevated histamine levels, underscoring their role in inflammatory responses modulation. However, treatments administered at 50 days post-hatch did not significantly alter protein levels, though both enrofloxacin and monensin increased serotonin and dopamine levels, suggesting potential neurotoxicological impacts on mood and cognitive functions. These results highlight the complex interactions between antibiotic use, gut microbiota alterations, and neurochemical pathways, with toxicological implications for environmental pollution and public health. This research provides critical insights into the potential toxic effects of prolonged antibiotic and coccidiostat exposure in poultry production, emphasizing the need for responsible use to mitigate risks to ecosystems and human health.
Collapse
Affiliation(s)
- Przemysław Sołek
- Department of Biochemistry and Toxicology, University of Life Sciences, Akademicka 13, 20-950 Lublin, Poland; Department of Biopharmacy, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland.
| | - Anna Stępniowska
- Department of Biochemistry and Toxicology, University of Life Sciences, Akademicka 13, 20-950 Lublin, Poland
| | - Oliwia Koszła
- Department of Biopharmacy, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Jan Jankowski
- Department of Poultry Science and Apiculture, University of Warmia and Mazury in Olsztyn, Oczapowskiego 5, 10-719 Olsztyn, Poland
| | - Katarzyna Ognik
- Department of Biochemistry and Toxicology, University of Life Sciences, Akademicka 13, 20-950 Lublin, Poland
| |
Collapse
|
5
|
Comai S, De Martin S, Mattarei A, Guidetti C, Pappagallo M, Folli F, Alimonti A, Manfredi PL. N-methyl-D-aspartate Receptors and Depression: Linking Psychopharmacology, Pathology and Physiology in a Unifying Hypothesis for the Epigenetic Code of Neural Plasticity. Pharmaceuticals (Basel) 2024; 17:1618. [PMID: 39770460 PMCID: PMC11728621 DOI: 10.3390/ph17121618] [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: 10/07/2024] [Revised: 11/03/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
Uncompetitive NMDAR (N-methyl-D-aspartate receptor) antagonists restore impaired neural plasticity, reverse depressive-like behavior in animal models, and relieve major depressive disorder (MDD) in humans. This review integrates recent findings from in silico, in vitro, in vivo, and human studies of uncompetitive NMDAR antagonists into the extensive body of knowledge on NMDARs and neural plasticity. Uncompetitive NMDAR antagonists are activity-dependent channel blockers that preferentially target hyperactive GluN2D subtypes because these subtypes are most sensitive to activation by low concentrations of extracellular glutamate and are more likely activated by certain pathological agonists and allosteric modulators. Hyperactivity of GluN2D subtypes in specific neural circuits may underlie the pathophysiology of MDD. We hypothesize that neural plasticity is epigenetically regulated by precise Ca2+ quanta entering cells via NMDARs. Stimuli reach receptor cells (specialized cells that detect specific types of stimuli and convert them into electrical signals) and change their membrane potential, regulating glutamate release in the synaptic cleft. Free glutamate binds ionotropic glutamatergic receptors regulating NMDAR-mediated Ca2+ influx. Quanta of Ca2+ via NMDARs activate enzymatic pathways, epigenetically regulating synaptic protein homeostasis and synaptic receptor expression; thereby, Ca2+ quanta via NMDARs control the balance between long-term potentiation and long-term depression. This NMDAR Ca2+ quantal hypothesis for the epigenetic code of neural plasticity integrates recent psychopharmacology findings into established physiological and pathological mechanisms of brain function.
Collapse
Affiliation(s)
- Stefano Comai
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35121 Padua, Italy; (S.C.); (S.D.M.); (A.M.)
- Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
- Department of Psychiatry, McGill University, Montreal, QC H3A 1A1, Canada
- IRCSS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Sara De Martin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35121 Padua, Italy; (S.C.); (S.D.M.); (A.M.)
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35121 Padua, Italy; (S.C.); (S.D.M.); (A.M.)
| | - Clotilde Guidetti
- Child Neuropsychiatry Unit, Department of Neuroscience, IRCCS Bambino Gesù Pediatric Hospital, 00165 Rome, Italy;
- Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - Marco Pappagallo
- Relmada Therapeutics, Inc., Coral Gables, FL 33134, USA;
- MGGM LLC, 85 Baker Road, Kerhonkson, NY 12446, USA
| | - Franco Folli
- Department of Health Sciences, University of Milan, 20141 Milan, Italy;
| | - Andrea Alimonti
- The Institute of Oncology Research, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland;
- Veneto Institute of Molecular Medicine, 35129 Padua, Italy
- Department of Medicine, Zurich University, 8006 Zurich, Switzerland
- Department of Medicine, University of Padua, 35122 Padua, Italy
| | - Paolo L. Manfredi
- Relmada Therapeutics, Inc., Coral Gables, FL 33134, USA;
- MGGM LLC, 85 Baker Road, Kerhonkson, NY 12446, USA
| |
Collapse
|
6
|
Sandoval KE, Witt KA. Somatostatin: Linking Cognition and Alzheimer Disease to Therapeutic Targeting. Pharmacol Rev 2024; 76:1291-1325. [PMID: 39013601 PMCID: PMC11549939 DOI: 10.1124/pharmrev.124.001117] [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: 02/13/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024] Open
Abstract
Over 4 decades of research support the link between Alzheimer disease (AD) and somatostatin [somatotropin-releasing inhibitory factor (SRIF)]. SRIF and SRIF-expressing neurons play an essential role in brain function, modulating hippocampal activity and memory formation. Loss of SRIF and SRIF-expressing neurons in the brain rests at the center of a series of interdependent pathological events driven by amyloid-β peptide (Aβ), culminating in cognitive decline and dementia. The connection between the SRIF and AD further extends to the neuropsychiatric symptoms, seizure activity, and inflammation, whereas preclinical AD investigations show SRIF or SRIF receptor agonist administration capable of enhancing cognition. SRIF receptor subtype-4 activation in particular presents unique attributes, with the potential to mitigate learning and memory decline, reduce comorbid symptoms, and enhance enzymatic degradation of Aβ in the brain. Here, we review the links between SRIF and AD along with the therapeutic implications. SIGNIFICANCE STATEMENT: Somatostatin and somatostatin-expressing neurons in the brain are extensively involved in cognition. Loss of somatostatin and somatostatin-expressing neurons in Alzheimer disease rests at the center of a series of interdependent pathological events contributing to cognitive decline and dementia. Targeting somatostatin-mediated processes has significant therapeutic potential for the treatment of Alzheimer disease.
Collapse
Affiliation(s)
- Karin E Sandoval
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| | - Ken A Witt
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| |
Collapse
|
7
|
Qin H, Yu M, Han N, Zhu M, Li X, Zhou J. Antidepressant effects of esketamine via the BDNF/AKT/mTOR pathway in mice with postpartum depression and their offspring. Prog Neuropsychopharmacol Biol Psychiatry 2024; 132:110992. [PMID: 38484929 DOI: 10.1016/j.pnpbp.2024.110992] [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: 09/09/2023] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Postpartum depression (PPD) is a serious mental health problem that can negatively affect future generations. BDNF/AKT/mTOR signaling in the frontal lobe and hippocampus in mice is associated with depression, but its role in mice with PPD and their offspring is unknown. This study was aimed at investigating the effects of esketamine (ESK), a drug approved for treatment of refractory depression, on the BDNF/AKT/mTOR pathway in mice with PPD and their offspring. A model of chronic unpredictable mild stress with pregnancy was used. ESK was injected into postpartum mice, and behavioral tests were conducted to predict the severity of symptoms at the end of lactation and in the offspring after adulthood. Both mice with PPD and their offspring showed significant anxiety- and depression-like behaviors that were ameliorated with the ESK intervention. ESK enhanced exploratory behavior in unfamiliar environments, increased the preference for sucrose, and ameliorated the impaired BDNF/AKT/mTOR signaling in the frontal and hippocampal regions in mice. Thus, ESK may have great potential in treating PPD and decreasing the incidence of depression in offspring.
Collapse
Affiliation(s)
- Han Qin
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Miao Yu
- Department of Science Experiment Center, China Medical University, Shenyang, China
| | - Nianjiao Han
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Meilin Zhu
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Xia Li
- Department of Gynecology, The First Hospital, China Medical University, Shenyang, China.
| | - Jing Zhou
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
8
|
Guo Y, Chen N, Zhao M, Cao B, Zhu F, Guo C, Shi Y, Wang Q, Li Y, Zhang L. D-arabinose acts as antidepressant by activating the ACSS2-PPARγ/TFEB axis and CRTC1 transcription. Pharmacol Res 2024; 202:107136. [PMID: 38460778 DOI: 10.1016/j.phrs.2024.107136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024]
Abstract
CREB-regulated transcription coactivator 1 (CRTC1), a pivotal synaptonuclear messenger, regulates synaptic plasticity and transmission to prevent depression. Despite exhaustive investigations into CRTC1 mRNA reductions in the depressed mice, the regulatory mechanisms governing its transcription remain elusive. Consequently, exploring rapid but non-toxic CRTC1 inducers at the transcriptional level is important for resisting depression. Here, we demonstrate the potential of D-arabinose, a unique monosaccharide prevalent in edible-medicinal plants, to rapidly enter the brain and induce CRTC1 expression, thereby eliciting rapid-acting and persistent antidepressant responses in chronic restrain stress (CRS)-induced depressed mice. Mechanistically, D-arabinose induces the expressions of peroxisome proliferator-activated receptor gamma (PPARγ) and transcription factor EB (TFEB), thereby activating CRTC1 transcription. Notably, we elucidate the pivotal role of the acetyl-CoA synthetase short-chain family member 2 (ACSS2) as an obligatory mediator for PPARγ and TFEB to potentiate CRTC1 transcription. Furthermore, D-arabinose augments ACSS2-dependent CRTC1 transcription by activating AMPK through lysosomal AXIN-LKB1 pathway. Correspondingly, the hippocampal down-regulations of ACSS2, PPARγ or TFEB alone failed to reverse CRTC1 reductions in CRS-exposure mice, ultimately abolishing the anti-depressant efficacy of D-arabinose. In summary, our study unveils a previously unexplored role of D-arabinose in activating the ACSS2-PPARγ/TFEB-CRTC1 axis, presenting it as a promising avenue for the prevention and treatment of depression.
Collapse
Affiliation(s)
- Yaxin Guo
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Nuo Chen
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ming Zhao
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Baihui Cao
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Faliang Zhu
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chun Guo
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yongyu Shi
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qun Wang
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yan Li
- Department of Pathogen Biology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Lining Zhang
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
9
|
Zhang L, Zheng J, Liu SY, Hou LL, Zhang B, Tian SW. Acute Administration of Lactate Exerts Antidepressant-like Effect Through cAMP-dependent Protein Synthesis. Neuroscience 2024; 542:11-20. [PMID: 38336096 DOI: 10.1016/j.neuroscience.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/16/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
Lactate acts as an important metabolic substrate and signalling molecule modulating neural activities in the brain, and recent preclinical and clinical studies have revealed its antidepressant effect after acute or chronic peripheral administration. However, the neural mechanism underlying the antidepressant effect of lactate, in particular when lactate is acutely administered remains largely unknown. In the current study, we focused on forced swimming test (FST) to elucidate the neural mechanisms through which acute intracerebroventricular (ICV) infusion of lactate exerts antidepressant-like effect. A total of 238 male Sprague Dawley rats were used as experimental subjects. Results showed lactate produced antidepressant-like effect, as indicated by reduced immobility, in a dose- and time-dependent manner. Moreover, the antidepressant-like effect of lactate was dependent of new protein synthesis but not new gene expression, lactate's metabolic effect or hydroxy-carboxylic acid receptor 1 (HCAR1) activation. Furthermore, lactate rapidly promoted dephosphorylation of eukaryotic elongation factor 2 (eEF2) and increased brain-derived neurotrophic factor (BDNF) protein synthesis in the hippocampus in a cyclic adenosine monophosphate (cAMP)-dependent manner. Finally, inhibition of cAMP production blocked the antidepressant-like effect of lactate. These findings suggest that acute administration of lactate exerts antidepressant-like effect through cAMP-dependent protein synthesis.
Collapse
Affiliation(s)
- Liang Zhang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin, Guangxi 541199, China; Department of Anesthesiology, National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong 518112, China; Department of Anesthesiology, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan 421001, China
| | - Jing Zheng
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Shi-Yan Liu
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Li-Li Hou
- Department of Anesthesiology, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan 421001, China
| | - Bo Zhang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Shao-Wen Tian
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin, Guangxi 541199, China.
| |
Collapse
|
10
|
Mitsi V, Ruiz A, Polizu C, Farzinpour Z, Ramakrishnan A, Serafini RA, Parise EM, Floodstrand M, Sial OK, Gaspari S, Tang CY, Nestler EJ, Schmidt EF, Shen L, Zachariou V. RGS4 Actions in Mouse Prefrontal Cortex Modulate Behavioral and Transcriptomic Responses to Chronic Stress and Ketamine. Mol Pharmacol 2024; 105:272-285. [PMID: 38351270 PMCID: PMC10949159 DOI: 10.1124/molpharm.123.000753] [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: 07/06/2023] [Accepted: 01/16/2024] [Indexed: 03/16/2024] Open
Abstract
The signal transduction protein, regulator of G protein signaling 4 (RGS4), plays a prominent role in physiologic and pharmacological responses by controlling multiple intracellular pathways. Our earlier work identified the dynamic but distinct roles of RGS4 in the efficacy of monoamine-targeting versus fast-acting antidepressants. Using a modified chronic variable stress (CVS) paradigm in mice, we demonstrate that stress-induced behavioral abnormalities are associated with the downregulation of RGS4 in the medial prefrontal cortex (mPFC). Knockout of RGS4 (RGS4KO) increases susceptibility to CVS, as mutant mice develop behavioral abnormalities as early as 2 weeks after CVS resting-state functional magnetic resonance imaging I (rs-fMRI) experiments indicate that stress susceptibility in RGS4KO mice is associated with changes in connectivity between the mediodorsal thalamus (MD-THL) and the mPFC. Notably, RGS4KO also paradoxically enhances the antidepressant efficacy of ketamine in the CVS paradigm. RNA-sequencing analysis of naive and CVS samples obtained from mPFC reveals that RGS4KO triggers unique gene expression signatures and affects several intracellular pathways associated with human major depressive disorder. Our analysis suggests that ketamine treatment in the RGS4KO group triggers changes in pathways implicated in synaptic activity and responses to stress, including pathways associated with axonal guidance and myelination. Overall, we show that reducing RGS4 activity triggers unique gene expression adaptations that contribute to chronic stress disorders and that RGS4 is a negative modulator of ketamine actions. SIGNIFICANCE STATEMENT: Chronic stress promotes robust maladaptation in the brain, but the exact intracellular pathways contributing to stress vulnerability and mood disorders have not been thoroughly investigated. In this study, the authors used murine models of chronic stress and multiple methodologies to demonstrate the critical role of the signal transduction modulator regulator of G protein signaling 4 in the medial prefrontal cortex in vulnerability to chronic stress and the efficacy of the fast-acting antidepressant ketamine.
Collapse
Affiliation(s)
- Vasiliki Mitsi
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Anne Ruiz
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Claire Polizu
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Zahra Farzinpour
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Randal A Serafini
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Eric M Parise
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Madeline Floodstrand
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Omar K Sial
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Sevasti Gaspari
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Cheuk Y Tang
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Eric F Schmidt
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Li Shen
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Venetia Zachariou
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| |
Collapse
|
11
|
Ren L. The mechanistic basis for the rapid antidepressant-like effects of ketamine: From neural circuits to molecular pathways. Prog Neuropsychopharmacol Biol Psychiatry 2024; 129:110910. [PMID: 38061484 DOI: 10.1016/j.pnpbp.2023.110910] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
Conventional antidepressants that target monoaminergic receptors require several weeks to be efficacious. This lag represents a significant problem in the currently available treatments for serious depression. Ketamine, acting as an N-methyl-d-aspartate receptor antagonist, was shown to have rapid antidepressant-like effects, marking a significant advancement in the study of mood disorders. However, serious side effects and adverse reactions limit its clinical use. Considering the limitations of ketamine, it is crucial to further define the network targets of ketamine. The rapid action of ketamine an as antidepressant is thought to be mediated by the glutamate system. It is believed that synaptic plasticity is essential for the rapid effects of ketamine as an antidepressant. Other mechanisms include the involvement of the γ-aminobutyric acidergic (GABAergic), 5-HTergic systems, and recent studies have linked astrocytes to ketamine's rapid antidepressant-like effects. The interactions between these systems exert a synergistic rapid antidepressant effect through neural circuits and molecular mechanisms. Here, we discuss the neural circuits and molecular mechanisms underlying the action of ketamine. This work will help explain how molecular and neural targets are responsible for the effects of rapidly acting antidepressants and will aid in the discovery of new therapeutic approaches for major depressive disorder.
Collapse
Affiliation(s)
- Li Ren
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu 611137, China.
| |
Collapse
|
12
|
Liu K, Qu Y, Li B, Zeng N, Yao G, Wu X, Xu H, Yan C, Wu L. GRP94 in cerebrospinal fluid may contribute to a potential biomarker of depression: Based on proteomics. J Psychiatr Res 2024; 169:328-340. [PMID: 38081093 DOI: 10.1016/j.jpsychires.2023.11.028] [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: 05/07/2023] [Revised: 11/03/2023] [Accepted: 11/15/2023] [Indexed: 01/15/2024]
Abstract
The present study was designed to investigate potential biomarkers of depression and targets of antidepressants from the perspective of hippocampal endoplasmic reticulum stress (ERS) based on cerebrospinal fluid (CSF) proteomics. Firstly, a six-week depression model was established and treated with fluoxetine (FLX). We found antidepressant-FLX could ameliorate depression-like behaviors and cognition in depressed rats caused by chronic unpredictable mild stress (CUMS). FLX significantly increased neuronal numbers in dentate gyrus (DG) and CA3 regions of hippocampus. CSF proteome data revealed thirty-seven differentially expressed proteins (DEPs) co-regulated by CUMS and FLX, including GRP94 and EIF2α. Results of Gene Oncology (GO) annotation and KEGG pathway enrichment for DEPs mainly included PERK-mediated unfolded protein response, endoplasmic reticulum, and translational initiation. The expression levels of GRP94, p-PERK, p-EIF2α, CHOP and Caspase-12 were increased in hippocampus of CUMS rats, and FLX worked the opposite way. FLX had strong affinity and binding activity with GRP94 protein, and four key proteins on the PERK pathway (PERK, EIF2α, p-EIF2α, CHOP). We proposed that FLX may exert antidepressant effects and neuroprotective action by alleviating excessive activation of the hippocampal PERK pathway and reducing neuronal deficits in depressed rats. PERK, EIF2α, p-EIF2α, and CHOP may be potential targets for antidepressant-FLX. GRP94 in CSF may be a potential biomarker of depression and the therapeutic effects of antidepressants.
Collapse
Affiliation(s)
- Kaige Liu
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yue Qu
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Bozhi Li
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Ningxi Zeng
- Department of Rehabilitation Medicine, The People's Hospital of Longhua District, Shenzhen, 518109, China
| | - Gaolei Yao
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xiaofeng Wu
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Hanfang Xu
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Can Yan
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Lili Wu
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
13
|
Zhu Y, Feng W, Kong Q, Sheng F, Li Z, Xu W, Li Q, Han Y, Wu X, Jia C, Guo J, Zhao Y. Evaluating the effects of S-ketamine on postoperative delirium in elderly patients following total hip or knee arthroplasty under intraspinal anesthesia: a single-center randomized, double-blind, placebo-controlled, pragmatic study protocol. Front Aging Neurosci 2023; 15:1298661. [PMID: 38099265 PMCID: PMC10720081 DOI: 10.3389/fnagi.2023.1298661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/14/2023] [Indexed: 12/17/2023] Open
Abstract
Introduction Postoperative delirium (POD) is an acute, transient brain disorder associated with decreased postoperative quality of life, dementia, neurocognitive changes, and mortality. A small number of trials have explored the role of S-ketamine in the treatment of POD due to its neuroprotective effects. Surprisingly, these trials have failed to yield supportive results. However, heterogeneity in delirium assessment methodologies, sample sizes, and outcome settings as well as deficiencies in S-ketamine use methods make the evidence provided by these studies less persuasive. Given the severe impact of POD on the health of elderly patients and the potential for S-ketamine to prevent it, we believe that designing a large sample size, and rigorous randomized controlled trial for further evaluation is necessary. Methods This is a single-center, randomized, double-blind, placebo-controlled, pragmatic study. Subjects undergoing total hip or knee arthroplasty will be randomized in a 1:1 ratio to intervention (n = 186) and placebo (n = 186) groups. This trial aims to explore the potential role of S-ketamine in the prevention of POD. Its primary outcome is the incidence of POD within 3 postoperative days. Secondary outcomes include the number of POD episodes, the onset and duration of POD, the severity and subtype of POD, pain scores and opioid consumption, sleep quality, clinical outcomes, and safety outcomes. Discussion To our knowledge, this is the first pragmatic study that proposes to use S-ketamine to prevent POD. We reviewed a large body of literature to identify potential preoperative confounding variables that may bias associations between the intervention and primary outcome. We will use advanced statistical methods to correct potential confounding variables, improving the test's power and external validity of test results. Of note, the patient population included in this trial will undergo intraspinal anesthesia. Although large, multicenter, randomized controlled studies have found no considerable difference in the effects of regional and general anesthesia on POD, patients receiving intraspinal anesthesia have less exposure to at-risk drugs, such as sevoflurane, propofol, and benzodiazepines, than patients receiving general anesthesia. At-risk drugs have been shown to negatively interfere with the neuroprotective effects of S-ketamine, which may be the reason for the failure of a large number of previous studies. There is currently a lack of randomized controlled studies evaluating S-ketamine for POD prevention, and our trial helps to fill a gap in this area.Trial registration: http://www.chictr.org.cn, identifier ChiCTR2300075796.
Collapse
Affiliation(s)
- Youzhuang Zhu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Wei Feng
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Qinghan Kong
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Fang Sheng
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Zhichao Li
- Department of Anesthesiology, Cancer Hospital Chinese Academy of Medical Science, Beijing, China
| | - Weilong Xu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Qun Li
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Yan Han
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xiuyun Wu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Changxin Jia
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Jie Guo
- Department of Anesthesiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yang Zhao
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
14
|
Chen T, Cheng L, Ma J, Yuan J, Pi C, Xiong L, Chen J, Liu H, Tang J, Zhong Y, Zhang X, Liu Z, Zuo Y, Shen H, Wei Y, Zhao L. Molecular mechanisms of rapid-acting antidepressants: New perspectives for developing antidepressants. Pharmacol Res 2023; 194:106837. [PMID: 37379962 DOI: 10.1016/j.phrs.2023.106837] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 06/11/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023]
Abstract
Major depressive disorder (MDD) is a chronic relapsing psychiatric disorder. Conventional antidepressants usually require several weeks of continuous administration to exert clinically significant therapeutic effects, while about two-thirds of the patients are prone to relapse of symptoms or are completely ineffective in antidepressant treatment. The recent success of the N-methyl-D-aspartic acid (NMDA) receptor antagonist ketamine as a rapid-acting antidepressant has propelled extensive research on the action mechanism of antidepressants, especially in relation to its role in synaptic targets. Studies have revealed that the mechanism of antidepressant action of ketamine is not limited to antagonism of postsynaptic NMDA receptors or GABA interneurons. Ketamine produces powerful and rapid antidepressant effects by affecting α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptors, adenosine A1 receptors, and the L-type calcium channels, among others in the synapse. More interestingly, the 5-HT2A receptor agonist psilocybin has demonstrated potential for rapid antidepressant effects in depressed mouse models and clinical studies. This article focuses on a review of new pharmacological target studies of emerging rapid-acting antidepressant drugs such as ketamine and hallucinogens (e.g., psilocybin) and briefly discusses the possible strategies for new targets of antidepressants, with a view to shed light on the direction of future antidepressant research.
Collapse
Affiliation(s)
- Tao Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou 646000 China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000 China; Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan 646000, China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ling Cheng
- Hospital-Acquired Infection Control Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jingwen Ma
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou 646000 China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000 China; Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan 646000, China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jiyuan Yuan
- Clinical trial center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chao Pi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou 646000 China
| | - Linjin Xiong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou 646000 China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000 China; Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan 646000, China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jinglin Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou 646000 China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000 China; Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan 646000, China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Huiyang Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou 646000 China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000 China; Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan 646000, China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jia Tang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou 646000 China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000 China; Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan 646000, China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yueting Zhong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou 646000 China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000 China; Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan 646000, China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaomei Zhang
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, Institute of medicinal chemistry of Chinese Medicine, Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China
| | - Zerong Liu
- Central Nervous System Drug Key Laboratory of Sichuan Province, Sichuan Credit Pharmaceutical CO., Ltd., Luzhou, Sichuan 646000, China; Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Ying Zuo
- Department of Comprehensive Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University; Luzhou, Sichuan 646000, China
| | - Hongping Shen
- Clinical trial center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Yumeng Wei
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou 646000 China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Ling Zhao
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000 China; Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan 646000, China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
15
|
Mingardi J, Ndoj E, Bonifacino T, Misztak P, Bertoli M, La Via L, Torazza C, Russo I, Milanese M, Bonanno G, Popoli M, Barbon A, Musazzi L. Functional and Molecular Changes in the Prefrontal Cortex of the Chronic Mild Stress Rat Model of Depression and Modulation by Acute Ketamine. Int J Mol Sci 2023; 24:10814. [PMID: 37445990 DOI: 10.3390/ijms241310814] [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: 06/01/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Stress is a primary risk factor in the onset of neuropsychiatric disorders, including major depressive disorder (MDD). We have previously used the chronic mild stress (CMS) model of depression in male rats to show that CMS induces morphological, functional, and molecular changes in the hippocampus of vulnerable animals, the majority of which were recovered using acute subanesthetic ketamine in just 24 h. Here, we focused our attention on the medial prefrontal cortex (mPFC), a brain area regulating emotional and cognitive functions, and asked whether vulnerability/resilience to CMS and ketamine antidepressant effects were associated with molecular and functional changes in the mPFC of rats. We found that most alterations induced by CMS in the mPFC were selectively observed in stress-vulnerable animals and were rescued by acute subanesthetic ketamine, while others were found only in resilient animals or were induced by ketamine treatment. Importantly, only a few of these modifications were also previously demonstrated in the hippocampus, while most are specific to mPFC. Overall, our results suggest that acute antidepressant ketamine rescues brain-area-specific glutamatergic changes induced by chronic stress.
Collapse
Affiliation(s)
- Jessica Mingardi
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Elona Ndoj
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy
| | - Tiziana Bonifacino
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, 16148 Genoa, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy
| | - Paulina Misztak
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Matteo Bertoli
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy
| | - Luca La Via
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy
| | - Carola Torazza
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, 16148 Genoa, Italy
| | - Isabella Russo
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy
- Genetics Unit, IRCCS Istituto Centro S. Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Marco Milanese
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, 16148 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Giambattista Bonanno
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, 16148 Genoa, Italy
| | - Maurizio Popoli
- Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, 20133 Milano, Italy
| | - Alessandro Barbon
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy
| | - Laura Musazzi
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
16
|
Shram MJ, Henningfield JE, Apseloff G, Gorodetzky CW, De Martin S, Vocci FL, Sapienza FL, Kosten TR, Huston J, Buchhalter A, Ashworth J, Lanier R, Folli F, Mattarei A, Guidetti C, Comai S, O'Gorman C, Traversa S, Inturrisi CE, Manfredi PL, Pappagallo M. The novel uncompetitive NMDA receptor antagonist esmethadone (REL-1017) has no meaningful abuse potential in recreational drug users. Transl Psychiatry 2023; 13:192. [PMID: 37286536 PMCID: PMC10247777 DOI: 10.1038/s41398-023-02473-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/02/2023] [Accepted: 05/12/2023] [Indexed: 06/09/2023] Open
Abstract
Esmethadone (REL-1017) is the opioid-inactive dextro-isomer of methadone and a low-affinity, low-potency uncompetitive NMDA receptor antagonist. In a Phase 2, randomized, double-blind, placebo-controlled trial, esmethadone showed rapid, robust, and sustained antidepressant effects. Two studies were conducted to evaluate the abuse potential of esmethadone. Each study utilized a randomized, double-blind, active-, and placebo-controlled crossover design to assess esmethadone compared with oxycodone (Oxycodone Study) or ketamine (Ketamine Study) in healthy recreational drug users. Esmethadone 25 mg (proposed therapeutic daily dose), 75 mg (loading dose), and 150 mg (Maximum Tolerated Dose) were evaluated in each study. Positive controls were oral oxycodone 40 mg and intravenous ketamine 0.5 mg/kg infused over 40 min. The Ketamine study included oral dextromethorphan 300 mg as an exploratory comparator. The primary endpoint was maximum effect (Emax) for Drug Liking, assessed using a bipolar 100-point visual analog scale (VAS). A total of 47 and 51 participants completed the Oxycodone Study and the Ketamine Study, respectively (Completer Population). In both studies, esmethadone doses ranging from therapeutic (25 mg) to 6 times therapeutic (150 mg) had a meaningful and statistically significantly (p < 0.001) lower Drug Liking VAS Emax compared with the positive control. Results were consistent for all secondary endpoints in both studies. In both studies, all doses of esmethadone were statistically equivalent to placebo on Drug Liking VAS Emax (p < 0.05). In the Ketamine Study, Drug Liking VAS Emax scores for esmethadone at all tested doses were significantly lower vs. dextromethorphan (p < 0.05) (exploratory endpoint). These studies indicate no meaningful abuse potential for esmethadone at all tested doses.
Collapse
Affiliation(s)
| | | | | | - Charles W Gorodetzky
- Relmada Therapeutics, Coral Gables, FL, USA
- Consultant in Pharmaceutical Medicine, Kansas City, MO, USA
| | - Sara De Martin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Frank L Vocci
- Relmada Therapeutics, Coral Gables, FL, USA
- Friends Research Institute, Baltimore, MD, USA
| | - Frank L Sapienza
- Relmada Therapeutics, Coral Gables, FL, USA
- The Drug and Chemical Advisory Group LLC, Fairfax, VA, USA
| | - Thomas R Kosten
- Relmada Therapeutics, Coral Gables, FL, USA
- Baylor College of Medicine, MD Anderson Cancer Center, University of Houston, Houston, TX, USA
| | | | | | | | | | - Franco Folli
- Department of Health Science, University of Milan, Milan, Italy
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Clotilde Guidetti
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Pediatric Hospital, IRCCS, Rome, Italy
| | - Stefano Comai
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | | | | | | | | | | |
Collapse
|
17
|
Borsellino P, Krider RI, Chea D, Grinnell R, Vida TA. Ketamine and the Disinhibition Hypothesis: Neurotrophic Factor-Mediated Treatment of Depression. Pharmaceuticals (Basel) 2023; 16:ph16050742. [PMID: 37242525 DOI: 10.3390/ph16050742] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Ketamine is a promising alternative to traditional pharmacotherapies for major depressive disorder, treatment-resistant depression, and other psychiatric conditions that heavily contribute to the global disease burden. In contrast to the current standard of care medications for these disorders, ketamine offers rapid onset, enduring clinical efficacy, and unique therapeutic potential for use in acute, psychiatric emergencies. This narrative presents an alternative framework for understanding depression, as mounting evidence supports a neuronal atrophy and synaptic disconnection theory, rather than the prevailing monoamine depletion hypothesis. In this context, we describe ketamine, its enantiomers, and various metabolites in a range of mechanistic actions through multiple converging pathways, including N-methyl-D-aspartate receptor (NMDAR) inhibition and the enhancement of glutamatergic signaling. We describe the disinhibition hypothesis, which posits that ketamine's pharmacological action ultimately results in excitatory cortical disinhibition, causing the release of neurotrophic factors, the most important of which is brain-derived neurotrophic factor (BDNF). BDNF-mediated signaling along with vascular endothelial growth factor (VEGF) and insulin-like growth factor 1 (IGF-1) subsequently give rise to the repair of neuro-structural abnormalities in patients with depressive disorders. Ketamine's efficacious amelioration of treatment-resistant depression is revolutionizing psychiatric treatment and opening up fresh vistas for understanding the underlying causes of mental illness.
Collapse
Affiliation(s)
- Philip Borsellino
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA
| | - Reese I Krider
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA
| | - Deanna Chea
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA
| | - Ryan Grinnell
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA
| | - Thomas A Vida
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA
| |
Collapse
|
18
|
VanderZwaag J, Halvorson T, Dolhan K, Šimončičová E, Ben-Azu B, Tremblay MÈ. The Missing Piece? A Case for Microglia's Prominent Role in the Therapeutic Action of Anesthetics, Ketamine, and Psychedelics. Neurochem Res 2023; 48:1129-1166. [PMID: 36327017 DOI: 10.1007/s11064-022-03772-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 08/25/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
There is much excitement surrounding recent research of promising, mechanistically novel psychotherapeutics - psychedelic, anesthetic, and dissociative agents - as they have demonstrated surprising efficacy in treating central nervous system (CNS) disorders, such as mood disorders and addiction. However, the mechanisms by which these drugs provide such profound psychological benefits are still to be fully elucidated. Microglia, the CNS's resident innate immune cells, are emerging as a cellular target for psychiatric disorders because of their critical role in regulating neuroplasticity and the inflammatory environment of the brain. The following paper is a review of recent literature surrounding these neuropharmacological therapies and their demonstrated or hypothesized interactions with microglia. Through investigating the mechanism of action of psychedelics, such as psilocybin and lysergic acid diethylamide, ketamine, and propofol, we demonstrate a largely under-investigated role for microglia in much of the emerging research surrounding these pharmacological agents. Among others, we detail sigma-1 receptors, serotonergic and γ-aminobutyric acid signalling, and tryptophan metabolism as pathways through which these agents modulate microglial phagocytic activity and inflammatory mediator release, inducing their therapeutic effects. The current review includes a discussion on future directions in the field of microglial pharmacology and covers bidirectional implications of microglia and these novel pharmacological agents in aging and age-related disease, glial cell heterogeneity, and state-of-the-art methodologies in microglial research.
Collapse
Affiliation(s)
- Jared VanderZwaag
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Torin Halvorson
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Kira Dolhan
- Department of Psychology, University of Victoria, Vancouver, BC, Canada
- Department of Biology, University of Victoria, Vancouver, BC, Canada
| | - Eva Šimončičová
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Benneth Ben-Azu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Marie-Ève Tremblay
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Département de médecine moléculaire, Université Laval, Québec City, QC, Canada.
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada.
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
19
|
Chronic orexin-1 receptor blockage attenuates depressive behaviors and provokes PSD-95 expression in a rat model of depression. Behav Brain Res 2023; 437:114123. [PMID: 36154849 DOI: 10.1016/j.bbr.2022.114123] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/21/2022]
Abstract
Depression is a devastating mood disorder affecting more than 300 million people worldwide. Almost 30 % of patients still suffer from treatment resistant depression. Although many reports support the involvement of orexin in the pathophysiology of depression, the precise role of orexin is still unclear. In this study, we evaluated the role of the orexin 1 receptor (Orx1R) on depressive behaviors and the alterations in postsynaptic density-95 (PSD-95) protein in the chronic mild stress (CMS) model of depression. Fifty-four male Wistar rats were randomly allocated to 6 groups; Control, CMS, acute SB-334867 (SB), CMS+SB, chronic SB (CSB) and CMS+CSB. Rats were exposed to one or two unpredictable stressors each day for three weeks for the induction of CMS. Intracerebroventricular (icv) injection of SB-334867, a selective Orx1R antagonist, was performed either 30 min before behavioral tests (acute) or once daily for 14 days (chronic). Behavioral despair was assessed by immobility time in the forced swim test (FST), sucrose consumption in sucrose preference test (SPT), and the number of crosses in the open field test (OFT) on days 1, 11, and 22 of the experiment. Finally, rats were decapitated, and brain tissue of the hippocampus (HPC) and prefrontal cortex (PFC) were collected, and the relative expression of PSD-95 was evaluated by western blotting. The CMS model rats showed a significant increase in FST immobility time (P = 0.001) and a decrease in locomotion (P = 0.04) and sucrose preference (P = 0.039). Chronic application of SB decreased immobility time to the control values (P = 0.001) and diminished locomotion (P = 0.047) and sucrose preference (P = 0.042) in comparison to the CMS group. Acute SB reversed just the immobility time (P ≤ 0.006). Chronic SB treatment increased the relative PSD-95 expression in PFC (P = 0.001). Hence, chronic antagonism of Orx1R alleviates depressive behaviors induced by CMS and improves PSD-95 expression in PFC.
Collapse
|
20
|
Onisiforou A, Georgiou P, Zanos P. Role of group II metabotropic glutamate receptors in ketamine's antidepressant actions. Pharmacol Biochem Behav 2023; 223:173531. [PMID: 36841543 DOI: 10.1016/j.pbb.2023.173531] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/08/2023] [Accepted: 02/15/2023] [Indexed: 02/26/2023]
Abstract
Major Depressive Disorder (MDD) is a serious neuropsychiatric disorder afflicting around 16-17 % of the global population and is accompanied by recurrent episodes of low mood, hopelessness and suicidal thoughts. Current pharmacological interventions take several weeks to even months for an improvement in depressive symptoms to emerge, with a significant percentage of individuals not responding to these medications at all, thus highlighting the need for rapid and effective next-generation treatments for MDD. Pre-clinical studies in animals have demonstrated that antagonists of the metabotropic glutamate receptor subtype 2/3 (mGlu2/3 receptor) exert rapid antidepressant-like effects, comparable to the actions of ketamine. Therefore, it is possible that mGlu2 or mGlu3 receptors to have a regulatory role on the unique antidepressant properties of ketamine, or that convergent intracellular mechanisms exist between mGlu2/3 receptor signaling and ketamine's effects. Here, we provide a comprehensive and critical evaluation of the literature on these convergent processes underlying the antidepressant action of mGlu2/3 receptor inhibitors and ketamine. Importantly, combining sub-threshold doses of mGlu2/3 receptor inhibitors with sub-antidepressant ketamine doses induce synergistic antidepressant-relevant behavioral effects. We review the evidence supporting these combinatorial effects since sub-effective dosages of mGlu2/3 receptor antagonists and ketamine could reduce the risk for the emergence of significant adverse events compared with taking normal dosages. Overall, deconvolution of ketamine's pharmacological targets will give critical insights to influence the development of next-generation antidepressant treatments with rapid actions.
Collapse
Affiliation(s)
- Anna Onisiforou
- Department of Psychology, University of Cyprus, Nicosia 2109, Cyprus
| | - Polymnia Georgiou
- Department of Biological Sciences, University of Cyprus, Nicosia 2109, Cyprus; Department of Psychology, University of Wisconsin Milwaukee, WI 53211, USA
| | - Panos Zanos
- Department of Psychology, University of Cyprus, Nicosia 2109, Cyprus.
| |
Collapse
|
21
|
Asch RH, Hillmer AT, Baldassarri SR, Esterlis I. The metabotropic glutamate receptor 5 as a biomarker for psychiatric disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:265-310. [PMID: 36868631 DOI: 10.1016/bs.irn.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The role of glutamate system in the etiology and pathophysiology of psychiatric disorders has gained considerable attention in the past two decades, including dysregulation of the metabotropic glutamatergic receptor subtype 5 (mGlu5). Thus, mGlu5 may represent a promising therapeutic target for psychiatric conditions, particularly stress-related disorders. Here, we describe mGlu5 findings in mood disorders, anxiety, and trauma disorders, as well as substance use (specifically nicotine, cannabis, and alcohol use). We highlight insights gained from positron emission tomography (PET) studies, where possible, and discuss findings from treatment trials, when available, to explore the role of mGlu5 in these psychiatric disorders. Through the research evidence reviewed in this chapter, we make the argument that, not only is dysregulation of mGlu5 evident in numerous psychiatric disorders, potentially functioning as a disease "biomarker," the normalization of glutamate neurotransmission via changes in mGlu5 expression and/or modulation of mGlu5 signaling may be a needed component in treating some psychiatric disorders or symptoms. Finally, we hope to demonstrate the utility of PET as an important tool for investigating mGlu5 in disease mechanisms and treatment response.
Collapse
Affiliation(s)
- Ruth H Asch
- Department of Psychiatry, Yale University, New Haven, CT, United States.
| | - Ansel T Hillmer
- Department of Psychiatry, Yale University, New Haven, CT, United States; Department of Radiology and Biomedical Imaging, New Haven, CT, United States
| | - Stephen R Baldassarri
- Yale Program in Addiction Medicine, Yale University, New Haven, CT, United States; Department of Internal Medicine, Yale University, New Haven, CT, United States
| | - Irina Esterlis
- Department of Psychiatry, Yale University, New Haven, CT, United States; Department of Psychology, Yale University, New Haven, CT, United States; Clinical Neurosciences Division, U.S. Department of Veterans Affairs National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| |
Collapse
|
22
|
Tatar G, Taskin Tok T, Ozpolat B, Ay M. Structure prediction of eukaryotic elongation factor-2 kinase and identification of the binding mechanisms of its inhibitors: homology modeling, molecular docking, and molecular dynamics simulation. J Biomol Struct Dyn 2022; 40:13355-13365. [PMID: 30880628 DOI: 10.1080/07391102.2019.1592024] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Protein kinases emerged as one of the most successful families of drug targets due to their increased activity and involvement in mediating critical signal transduction pathways in cancer cells. Recent evidence suggests that eukaryotic elongation factor 2 kinase (eEF-2K) is a potential therapeutic target for treating some highly aggressive solid cancers, including lung, pancreatic and triple-negative breast cancers. Thus, several compounds have been developed for the inhibition of the enzyme activity, but they are not sufficiently specific and potent. Besides, the crystal structure of this kinase remains unknown. Hence, the functional organization and regulation of eEF-2K remain poorly characterized. For this purpose, we constructed a homology model of eEF-2K and then used docking methodology to better understanding the binding mechanism of eEF-2K with 58 compounds that have been proposed as existing inhibitors. The results of this analysis were compared with the experimental results and the compounds effective against eEF-2K were determined against eEF-2K as a result of both studies. And finally, molecular dynamics (MD) simulations were performed for the stability of eEF-2K with these compounds. According to these study defined that the binding mechanism of eEF-2K with inhibitors at the molecular level and elucidated the residues of eEF-2K that play an important role in enzyme selectivity and ligand affinity. This information may lead to new selective and potential drug molecules to be for inhibition of eEF-2K.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Gizem Tatar
- Department of Bioinformatics and Computational Biology, Institute of Health Sciences, Gaziantep University, Gaziantep, Turkey
| | - Tugba Taskin Tok
- Department of Bioinformatics and Computational Biology, Institute of Health Sciences, Gaziantep University, Gaziantep, Turkey.,Department of Chemistry, Faculty of Arts and Sciences, Gaziantep University, Gaziantep, Turkey
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas-Houston MD Anderson Cancer Center, Houston, USA
| | - Mehmet Ay
- Natural Products and Drug Research Laboratory, Department of Chemistry, Faculty of Science and Arts, Çanakkale Onsekiz Mart University Çanakkale, TURKEY
| |
Collapse
|
23
|
Beiser T, Lisniansky E, Weitz M, Bingor A, Grad E, Rosenblum K, Thornton C, Yaka R. A functional eEF2K-eEF2 pathway in the NAc is critical for the expression of cocaine-induced psychomotor sensitisation and conditioned place preference. Transl Psychiatry 2022; 12:460. [PMID: 36319619 PMCID: PMC9626485 DOI: 10.1038/s41398-022-02232-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 01/24/2023] Open
Abstract
Recent evidence links synaptic plasticity and mRNA translation, via the eukaryotic elongation factor 2 kinase (eEF2K) and its only known substrate, eEF2. However, the involvement of the eEF2 pathway in cocaine-induced neuroadaptations and cocaine-induced behaviours is not known. Knock-in (KI) mice and shRNA were used to globally and specifically reduce eEF2K expression. Cocaine psychomotor sensitization and conditioned place preference were used to evaluate behavioural outcome. Changes in eEF2 phosphorylation were determined by western blot analyses. No effect was observed on the AMPA/NMDA receptor current ratio in the ventral tegmental area, 24 h after cocaine injection in eEF2K-KI mice compared with WT. However, development and expression of cocaine psychomotor sensitization were decreased in KI mice. Phosphorylated eEF2 was decreased one day after psychomotor sensitization and returned to baseline at seven days in the nucleus accumbens (NAc) of WT mice, but not in eEF2K-KI mice. However, one day following cocaine challenge, phosphorylated eEF2 decreased in WT but not KI mice. Importantly, specific targeting of eEF2K expression by shRNA in the NAc decreased cocaine condition place preference. These results suggest that the eEF2 pathway play a role in cocaine-induced locomotor sensitization and conditioned place preference.
Collapse
Affiliation(s)
- Tehila Beiser
- grid.9619.70000 0004 1937 0538Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Elvira Lisniansky
- grid.9619.70000 0004 1937 0538Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Moriya Weitz
- grid.9619.70000 0004 1937 0538Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Alexey Bingor
- grid.9619.70000 0004 1937 0538Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Etty Grad
- grid.9619.70000 0004 1937 0538Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Kobi Rosenblum
- grid.18098.380000 0004 1937 0562Sagol Department of Neuroscience, University of Haifa, Haifa, Israel
| | - Claire Thornton
- grid.20931.390000 0004 0425 573XDepartment of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Rami Yaka
- Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
24
|
Esmethadone (REL-1017) and Other Uncompetitive NMDAR Channel Blockers May Improve Mood Disorders via Modulation of Synaptic Kinase-Mediated Signaling. Int J Mol Sci 2022; 23:ijms232012196. [PMID: 36293063 PMCID: PMC9602945 DOI: 10.3390/ijms232012196] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 11/30/2022] Open
Abstract
This article presents a mechanism of action hypothesis to explain the rapid antidepressant effects of esmethadone (REL-1017) and other uncompetitive N-methyl-D-aspartate receptor (NMDAR) antagonists and presents a corresponding mechanism of disease hypothesis for major depressive disorder (MDD). Esmethadone and other uncompetitive NMDAR antagonists may restore physiological neural plasticity in animal models of depressive-like behavior and in patients with MDD via preferential tonic block of pathologically hyperactive GluN2D subtypes. Tonic Ca2+ currents via GluN2D subtypes regulate the homeostatic availability of synaptic proteins. MDD and depressive behaviors may be determined by reduced homeostatic availability of synaptic proteins, due to upregulated tonic Ca2+ currents through GluN2D subtypes. The preferential activity of low-potency NMDAR antagonists for GluN2D subtypes may explain their rapid antidepressant effects in the absence of dissociative side effects.
Collapse
|
25
|
Bettini E, Stahl SM, De Martin S, Mattarei A, Sgrignani J, Carignani C, Nola S, Locatelli P, Pappagallo M, Inturrisi CE, Bifari F, Cavalli A, Alimonti A, Pani L, Fava M, Traversa S, Folli F, Manfredi PL. Pharmacological Comparative Characterization of REL-1017 (Esmethadone-HCl) and Other NMDAR Channel Blockers in Human Heterodimeric N-Methyl-D-Aspartate Receptors. Pharmaceuticals (Basel) 2022; 15:ph15080997. [PMID: 36015145 PMCID: PMC9414551 DOI: 10.3390/ph15080997] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 12/19/2022] Open
Abstract
Excessive Ca2+ currents via N-methyl-D-aspartate receptors (NMDARs) have been implicated in many disorders. Uncompetitive NMDAR channel blockers are an emerging class of drugs in clinical use for major depressive disorder (MDD) and other neuropsychiatric diseases. The pharmacological characterization of uncompetitive NMDAR blockers in clinical use may improve our understanding of NMDAR function in physiology and pathology. REL-1017 (esmethadone-HCl), a novel uncompetitive NMDAR channel blocker in Phase 3 trials for the treatment of MDD, was characterized together with dextromethorphan, memantine, (±)-ketamine, and MK-801 in cell lines over-expressing NMDAR subtypes using fluorometric imaging plate reader (FLIPR), automated patch-clamp, and manual patch-clamp electrophysiology. In the absence of Mg2+, NMDAR subtypes NR1-2D were most sensitive to low, sub-μM glutamate concentrations in FLIPR experiments. FLIPR Ca2+ determination demonstrated low μM affinity of REL-1017 at NMDARs with minimal subtype preference. In automated and manual patch-clamp electrophysiological experiments, REL-1017 exhibited preference for the NR1-2D NMDAR subtype in the presence of 1 mM Mg2+ and 1 μM L-glutamate. Tau off and trapping characteristics were similar for (±)-ketamine and REL-1017. Results of radioligand binding assays in rat cortical neurons correlated with the estimated affinities obtained in FLIPR assays and in automated and manual patch-clamp assays. In silico studies of NMDARs in closed and open conformation indicate that REL-1017 has a higher preference for docking and undocking the open-channel conformation compared to ketamine. In conclusion, the pharmacological characteristics of REL-1017 at NMDARs, including relatively low affinity at the NMDAR, NR1-2D subtype preference in the presence of 1 mM Mg2+, tau off and degree of trapping similar to (±)-ketamine, and preferential docking and undocking of the open NMDAR, could all be important variables for understanding the rapid-onset antidepressant effects of REL-1017 without psychotomimetic side effects.
Collapse
Affiliation(s)
- Ezio Bettini
- In Vitro Pharmacology Department, Aptuit, An Evotec Company, 37135 Verona, Italy
| | - Stephen M. Stahl
- Department of Psychiatry, VAMC (SD), University of California, San Diego, CA 92093, USA
- Neuroscience Education Institute, Carlsbad, CA 92008, USA
| | - Sara De Martin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35122 Padua, Italy
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35122 Padua, Italy
| | - Jacopo Sgrignani
- Institute for Research in Biomedicine (IRB), Università della Svizzera Italiana (USI), 6500 Bellinzona, Switzerland
| | - Corrado Carignani
- In Vitro Pharmacology Department, Aptuit, An Evotec Company, 37135 Verona, Italy
| | - Selena Nola
- In Vitro Pharmacology Department, Aptuit, An Evotec Company, 37135 Verona, Italy
| | - Patrizia Locatelli
- Institute for Research in Biomedicine (IRB), Università della Svizzera Italiana (USI), 6500 Bellinzona, Switzerland
| | - Marco Pappagallo
- Department of Anesthesiology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Relmada Therapeutics, Coral Gables, FL 33134, USA
| | | | - Francesco Bifari
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20122 Milan, Italy
| | - Andrea Cavalli
- Institute for Research in Biomedicine (IRB), Università della Svizzera Italiana (USI), 6500 Bellinzona, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Andrea Alimonti
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
- Institute of Oncology Research, Southern Switzerland, 6500 Bellinzona, Switzerland
- The Institute of Oncology Research, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
- Veneto Institute of Molecular Medicine, 35129 Padua, Italy
- Department of Medicine, Zurich University, 8006 Zurich, Switzerland
- Department of Medicine—DIMED, University of Padua, 35122 Padua, Italy
| | - Luca Pani
- Relmada Therapeutics, Coral Gables, FL 33134, USA
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Maurizio Fava
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Franco Folli
- Department of Health Sciences, University of Milan, 20122 Milan, Italy
| | - Paolo L. Manfredi
- Relmada Therapeutics, Coral Gables, FL 33134, USA
- Correspondence: ; Tel.: +1-786-629-1376
| |
Collapse
|
26
|
Moaddel R, Zanos P, Farmer CA, Kadriu B, Morris PJ, Lovett J, Acevedo-Diaz EE, Cavanaugh GW, Yuan P, Yavi M, Thomas CJ, Park LT, Ferrucci L, Gould TD, Zarate CA. Comparative metabolomic analysis in plasma and cerebrospinal fluid of humans and in plasma and brain of mice following antidepressant-dose ketamine administration. Transl Psychiatry 2022; 12:179. [PMID: 35501309 PMCID: PMC9061764 DOI: 10.1038/s41398-022-01941-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/13/2022] [Accepted: 04/20/2022] [Indexed: 11/25/2022] Open
Abstract
Subanesthetic-dose racemic (R,S)-ketamine (ketamine) produces rapid, robust, and sustained antidepressant effects in major depressive disorder (MDD) and bipolar disorder (BD) and has also been shown to effectively treat neuropathic pain, complex regional pain syndrome, and post-traumatic stress disorder (PTSD). However, to date, its mechanism of action remains unclear. Preclinical studies found that (2 R,6 R;2 S,6 S)-hydroxynorketamine (HNK), a major circulating metabolite of ketamine, elicits antidepressant effects similar to those of ketamine. To help determine how (2 R,6 R)-HNK contributes to ketamine's mechanism of action, an exploratory, targeted, metabolomic analysis was carried out on plasma and CSF of nine healthy volunteers receiving a 40-minute ketamine infusion (0.5 mg/kg). A parallel targeted metabolomic analysis in plasma, hippocampus, and hypothalamus was carried out in mice receiving either 10 mg/kg of ketamine, 10 mg/kg of (2 R,6 R)-HNK, or saline. Ketamine and (2 R,6 R)-HNK both affected multiple pathways associated with inflammatory conditions. In addition, several changes were unique to either the healthy human volunteers and/or the mouse arm of the study, indicating that different pathways may be differentially involved in ketamine's effects in mice and humans. Mechanisms of action found to consistently underlie the effects of ketamine and/or (2 R,6 R)-HNK across both the human metabolome in plasma and CSF and the mouse arm of the study included LAT1, IDO1, NAD+, the nitric oxide (NO) signaling pathway, and sphingolipid rheostat.
Collapse
Affiliation(s)
- Ruin Moaddel
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, 21224, MD, USA.
| | - Panos Zanos
- Departments of Psychiatry, Pharmacology, and Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, 21201, MD, USA
- Department of Psychology, University of Cyprus, 2109, Nicosia, Cyprus
| | - Cristan A Farmer
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Bashkim Kadriu
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Patrick J Morris
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Jacqueline Lovett
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, 21224, MD, USA
| | - Elia E Acevedo-Diaz
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Grace W Cavanaugh
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Peixiong Yuan
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Mani Yavi
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Lawrence T Park
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Luigi Ferrucci
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, 21224, MD, USA
| | - Todd D Gould
- Departments of Psychiatry, Pharmacology, and Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, 21201, MD, USA
- Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
27
|
Wang CS, Kavalali ET, Monteggia LM. BDNF signaling in context: From synaptic regulation to psychiatric disorders. Cell 2022; 185:62-76. [PMID: 34963057 PMCID: PMC8741740 DOI: 10.1016/j.cell.2021.12.003] [Citation(s) in RCA: 309] [Impact Index Per Article: 103.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/10/2021] [Accepted: 12/02/2021] [Indexed: 01/09/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is a neuropeptide that plays numerous important roles in synaptic development and plasticity. While its importance in fundamental physiology is well established, studies of BDNF often produce conflicting and unclear results, and the scope of existing research makes the prospect of setting future directions daunting. In this review, we examine the importance of spatial and temporal factors on BDNF activity, particularly in processes such as synaptogenesis, Hebbian plasticity, homeostatic plasticity, and the treatment of psychiatric disorders. Understanding the fundamental physiology of when, where, and how BDNF acts and new approaches to control BDNF signaling in time and space can contribute to improved therapeutics and patient outcomes.
Collapse
Affiliation(s)
- Camille S Wang
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232-2050, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA
| | - Ege T Kavalali
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232-2050, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA
| | - Lisa M Monteggia
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232-2050, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA.
| |
Collapse
|
28
|
Beretta S, Gritti L, Ponzoni L, Scalmani P, Mantegazza M, Sala M, Verpelli C, Sala C. Rescuing epileptic and behavioral alterations in a Dravet syndrome mouse model by inhibiting eukaryotic elongation factor 2 kinase (eEF2K). Mol Autism 2022; 13:1. [PMID: 34980259 PMCID: PMC8722032 DOI: 10.1186/s13229-021-00484-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/23/2021] [Indexed: 11/30/2022] Open
Abstract
Background Dravet Syndrome is a severe childhood pharmaco-resistant epileptic disorder mainly caused by mutations in the SCN1A gene, which encodes for the α1 subunit of the type I voltage-gated sodium channel (NaV1.1), that causes imbalance between excitation and inhibition in the brain. We recently found that eEF2K knock out mice displayed enhanced GABAergic transmission and tonic inhibition and were less susceptible to epileptic seizures. Thus, we investigated the effect of inhibition of eEF2K on the epileptic and behavioral phenotype of Scn1a ± mice, a murine model of Dravet Syndrome. Methods To elucidate the role of eEF2K pathway in the etiopathology of Dravet syndrome we generated a new mouse model deleting the eEF2K gene in Scn1a ± mice. By crossing Scn1a ± mice with eEF2K−/− mice we obtained the three main genotypes needed for our studies, Scn1a+/+ eEF2K+/+ (WT mice), Scn1a ± eEF2K+/+ mice (Scn1a ± mice) and Scn1a ± eEF2K−/− mice, that were fully characterized for EEG and behavioral phenotype. Furthermore, we tested the ability of a pharmacological inhibitor of eEF2K in rescuing EEG alterations of the Scn1a ± mice. Results We showed that the activity of eEF2K/eEF2 pathway was enhanced in Scn1a ± mice. Then, we demonstrated that both genetic deletion and pharmacological inhibition of eEF2K were sufficient to ameliorate the epileptic phenotype of Scn1a ± mice. Interestingly we also found that motor coordination defect, memory impairments, and stereotyped behavior of the Scn1a ± mice were reverted by eEF2K deletion. The analysis of spontaneous inhibitory postsynaptic currents (sIPSCs) suggested that the rescue of the pathological phenotype was driven by the potentiation of GABAergic synapses. Limitations Even if we found that eEF2K deletion was able to increase inhibitory synapses function, the molecular mechanism underlining the inhibition of eEF2K/eEF2 pathway in rescuing epileptic and behavioral alterations in the Scn1a ± needs further investigations. Conclusions Our data indicate that pharmacological inhibition of eEF2K could represent a novel therapeutic intervention for treating epilepsy and related comorbidities in the Dravet syndrome. Supplementary Information The online version contains supplementary material available at 10.1186/s13229-021-00484-0.
Collapse
Affiliation(s)
- Stefania Beretta
- CNR Neuroscience Institute, Milan, and NeuroMi Milan Center for Neuroscience, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy
| | - Laura Gritti
- CNR Neuroscience Institute, Milan, and NeuroMi Milan Center for Neuroscience, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy
| | - Luisa Ponzoni
- CNR Neuroscience Institute, Milan, and NeuroMi Milan Center for Neuroscience, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy
| | - Paolo Scalmani
- L'Unità Operativa Complessa di Epilettologia Clinica e Sperimentale, Foundation Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Neurological Institute Carlo Besta, 20133, Milan, Italy
| | - Massimo Mantegazza
- CNRS UMR 7275, Institut National de La Santé Et de La Recherche Médicale, LabEx ICST, Institute of Molecular and Cellular Pharmacology (IPMC), Université Côte d'Azur (UCA), 06560, Valbonne-Sophia Antipolis, France
| | - Mariaelvina Sala
- CNR Neuroscience Institute, Milan, and NeuroMi Milan Center for Neuroscience, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy
| | - Chiara Verpelli
- CNR Neuroscience Institute, Milan, and NeuroMi Milan Center for Neuroscience, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy.
| | - Carlo Sala
- CNR Neuroscience Institute, Milan, and NeuroMi Milan Center for Neuroscience, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy.
| |
Collapse
|
29
|
Wei Y, Chang L, Hashimoto K. Molecular mechanisms underlying the antidepressant actions of arketamine: beyond the NMDA receptor. Mol Psychiatry 2022; 27:559-573. [PMID: 33963284 PMCID: PMC8960399 DOI: 10.1038/s41380-021-01121-1] [Citation(s) in RCA: 160] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/01/2021] [Accepted: 04/13/2021] [Indexed: 02/08/2023]
Abstract
The discovery of robust antidepressant actions exerted by the N-methyl-D-aspartate receptor (NMDAR) antagonist (R,S)-ketamine has been a crucial breakthrough in mood disorder research. (R,S)-ketamine is a racemic mixture of equal amounts of (R)-ketamine (arketamine) and (S)-ketamine (esketamine). In 2019, an esketamine nasal spray from Johnson & Johnson was approved in the United States of America and Europe for treatment-resistant depression. However, an increasing number of preclinical studies show that arketamine has greater potency and longer-lasting antidepressant-like effects than esketamine in rodents, despite the lower binding affinity of arketamine for the NMDAR. In clinical trials, non-ketamine NMDAR-related compounds did not exhibit ketamine-like robust antidepressant actions in patients with depression, despite these compounds showing antidepressant-like effects in rodents. Thus, the rodent data do not necessarily translate to humans due to the complexity of human psychiatric disorders. Collectively, the available studies indicate that it is unlikely that NMDAR plays a major role in the antidepressant action of (R,S)-ketamine and its enantiomers, although the precise molecular mechanisms underlying antidepressant actions of (R,S)-ketamine and its enantiomers remain unclear. In this paper, we review recent findings on the molecular mechanisms underlying the antidepressant actions of (R,S)-ketamine and its potent enantiomer arketamine. Furthermore, we discuss the possible role of the brain-gut-microbiota axis and brain-spleen axis in stress-related psychiatric disorders and in the antidepressant-like action of arketamine. Finally, we discuss the potential of arketamine as a treatment for cognitive impairment in psychiatric disorders, Parkinson's disease, osteoporosis, inflammatory bowel diseases, and stroke.
Collapse
Affiliation(s)
- Yan Wei
- grid.411500.1Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan ,grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan China
| | - Lijia Chang
- grid.411500.1Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan.
| |
Collapse
|
30
|
Kang MJY, Hawken E, Vazquez GH. The Mechanisms Behind Rapid Antidepressant Effects of Ketamine: A Systematic Review With a Focus on Molecular Neuroplasticity. Front Psychiatry 2022; 13:860882. [PMID: 35546951 PMCID: PMC9082546 DOI: 10.3389/fpsyt.2022.860882] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/18/2022] [Indexed: 12/25/2022] Open
Abstract
The mechanism of action underlying ketamine's rapid antidepressant effects in patients with depression, both suffering from major depressive disorder (MDD) and bipolar disorder (BD), including treatment resistant depression (TRD), remains unclear. Of the many speculated routes that ketamine may act through, restoring deficits in neuroplasticity may be the most parsimonious mechanism in both human patients and preclinical models of depression. Here, we conducted a literature search using PubMed for any reports of ketamine inducing neuroplasticity relevant to depression, to identify cellular and molecular events, relevant to neuroplasticity, immediately observed with rapid mood improvements in humans or antidepressant-like effects in animals. After screening reports using our inclusion/exclusion criteria, 139 publications with data from cell cultures, animal models, and patients with BD or MDD were included (registered on PROSPERO, ID: CRD42019123346). We found accumulating evidence to support that ketamine induces an increase in molecules involved in modulating neuroplasticity, and that these changes are paired with rapid antidepressant effects. Molecules or complexes of high interest include glutamate, AMPA receptors (AMPAR), mTOR, BDNF/TrkB, VGF, eEF2K, p70S6K, GSK-3, IGF2, Erk, and microRNAs. In summary, these studies suggest a robust relationship between improvements in mood, and ketamine-induced increases in molecular neuroplasticity, particularly regarding intracellular signaling molecules.
Collapse
Affiliation(s)
- Melody J Y Kang
- Center of Neuroscience Studies (CNS), Queen's University, Kingston, ON, Canada
| | - Emily Hawken
- Department of Psychiatry, Queen's University School of Medicine, Kingston, ON, Canada.,Providence Care Hospital, Kingston, ON, Canada
| | - Gustavo Hector Vazquez
- Center of Neuroscience Studies (CNS), Queen's University, Kingston, ON, Canada.,Department of Psychiatry, Queen's University School of Medicine, Kingston, ON, Canada.,Providence Care Hospital, Kingston, ON, Canada
| |
Collapse
|
31
|
Ponton E, Turecki G, Nagy C. Sex Differences in the Behavioral, Molecular, and Structural Effects of Ketamine Treatment in Depression. Int J Neuropsychopharmacol 2021; 25:75-84. [PMID: 34894233 PMCID: PMC8756094 DOI: 10.1093/ijnp/pyab082] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/08/2021] [Accepted: 11/15/2021] [Indexed: 12/14/2022] Open
Abstract
Major depressive disorder (MDD) is a common psychiatric illness that manifests in sex-influenced ways. Men and women may experience depression differently and also respond to various antidepressant treatments in sex-influenced ways. Ketamine, which is now being used as a rapid-acting antidepressant, is likely the same. To date, the majority of studies investigating treatment outcomes in MDD do not disaggregate the findings in males and females, and this is also true for ketamine. This review aims to highlight that gap by exploring pre-clinical data-at a behavioral, molecular, and structural level-and recent clinical trials. Sex hormones, particularly estrogen and progesterone, influence the response at all levels examined, and sex is therefore a critical factor to examine when looking at ketamine response. Taken together, the data show females are more sensitive to ketamine than males, and it might be possible to monitor the phase of the menstrual cycle to mitigate some risks associated with the use of ketamine for females with MDD. Based on the studies reviewed in this article, we suggest that ketamine should be administered adhering to sex-specific considerations.
Collapse
Affiliation(s)
- Ethan Ponton
- Department of Anatomy and Cell Biology, McGill University, Montreal, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Montreal, Canada
- Department of Psychiatry, McGill University, Montreal, Canada
| | - Corina Nagy
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Montreal, Canada
- Department of Psychiatry, McGill University, Montreal, Canada
- Correspondence: Corina Nagy, PhD, 6875 LaSalle Blvd, Verdun, Québec, Canada H4H 1R3 ()
| |
Collapse
|
32
|
Kowalczyk M, Kowalczyk E, Kwiatkowski P, Łopusiewicz Ł, Sienkiewicz M, Talarowska M. Ketamine-New Possibilities in the Treatment of Depression: A Narrative Review. Life (Basel) 2021; 11:1186. [PMID: 34833062 PMCID: PMC8619908 DOI: 10.3390/life11111186] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/31/2021] [Accepted: 10/31/2021] [Indexed: 12/11/2022] Open
Abstract
The SARS-CoV-2 coronavirus epidemic has led to an increase in the number of people with depression. Symptoms related to the mental sphere (mainly depression and anxiety) may be experienced by one third of the worldwide population. This entails the need for the effective and rapid treatment of depressive episodes. An effective drug seems to be s-ketamine, which was accepted in March 2019 by the Food and Drug Administration (FDA) for the treatment of drug-resistant depression. This drug provides a quick antidepressant effect with maximum effectiveness achieved after 24 h. It also appears to reduce the occurrence of suicidal thoughts. However, research into undesirable effects, especially in groups of people susceptible to psychotic episodes or those who use alcohol or psychoactive substances, is necessary.
Collapse
Affiliation(s)
- Mateusz Kowalczyk
- Babinski Memorial Hospital, Aleksandrowska St. 159, 91-229 Lodz, Poland;
| | - Edward Kowalczyk
- Department of Pharmacology and Toxicology, Medical University of Lodz, Żeligowskiego St. 7/9, 90-752 Lodz, Poland;
| | - Paweł Kwiatkowski
- Department of Diagnostic Immunology, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Łukasz Łopusiewicz
- Center of Bioimmobilisation and Innovative Packaging Materials, Faculty of Food Sciences and Fisheries, West Pomeranian University of Technology Szczecin, Janickiego 35, 71-270 Szczecin, Poland;
| | - Monika Sienkiewicz
- Department of Pharmaceutical Microbiology and Microbiological Diagnostic, Medical University of Lodz, Muszyńskiego St. 1, 90-151 Lodz, Poland
| | - Monika Talarowska
- Department of Clinical Psychology and Psychopathology, Institute of Psychology, University of Lodz, Smugowa St. 10/12, 91-433 Lodz, Poland;
| |
Collapse
|
33
|
Choudhury D, Autry AE, Tolias KF, Krishnan V. Ketamine: Neuroprotective or Neurotoxic? Front Neurosci 2021; 15:672526. [PMID: 34566558 PMCID: PMC8461018 DOI: 10.3389/fnins.2021.672526] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 07/12/2021] [Indexed: 12/20/2022] Open
Abstract
Ketamine, a non-competitive N-methyl-D-aspartate receptor (NMDAR) antagonist, has been employed clinically as an intravenous anesthetic since the 1970s. More recently, ketamine has received attention for its rapid antidepressant effects and is actively being explored as a treatment for a wide range of neuropsychiatric syndromes. In model systems, ketamine appears to display a combination of neurotoxic and neuroprotective properties that are context dependent. At anesthetic doses applied during neurodevelopmental windows, ketamine contributes to inflammation, autophagy, apoptosis, and enhances levels of reactive oxygen species. At the same time, subanesthetic dose ketamine is a powerful activator of multiple parallel neurotrophic signaling cascades with neuroprotective actions that are not always NMDAR-dependent. Here, we summarize results from an array of preclinical studies that highlight a complex landscape of intracellular signaling pathways modulated by ketamine and juxtapose the somewhat contrasting neuroprotective and neurotoxic features of this drug.
Collapse
Affiliation(s)
- Divya Choudhury
- Department of BioSciences, Rice University, Houston, TX, United States
| | - Anita E. Autry
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Kimberley F. Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Vaishnav Krishnan
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
- Department of Neurology, Baylor College of Medicine, Houston, TX, United States
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
34
|
Ballard DJ, Peng HY, Das JK, Kumar A, Wang L, Ren Y, Xiong X, Ren X, Yang JM, Song J. Insights Into the Pathologic Roles and Regulation of Eukaryotic Elongation Factor-2 Kinase. Front Mol Biosci 2021; 8:727863. [PMID: 34532346 PMCID: PMC8438118 DOI: 10.3389/fmolb.2021.727863] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/16/2021] [Indexed: 12/14/2022] Open
Abstract
Eukaryotic Elongation Factor-2 Kinase (eEF2K) acts as a negative regulator of protein synthesis, translation, and cell growth. As a structurally unique member of the alpha-kinase family, eEF2K is essential to cell survival under stressful conditions, as it contributes to both cell viability and proliferation. Known as the modulator of the global rate of protein translation, eEF2K inhibits eEF2 (eukaryotic Elongation Factor 2) and decreases translation elongation when active. eEF2K is regulated by various mechanisms, including phosphorylation through residues and autophosphorylation. Specifically, this protein kinase is downregulated through the phosphorylation of multiple sites via mTOR signaling and upregulated via the AMPK pathway. eEF2K plays important roles in numerous biological systems, including neurology, cardiology, myology, and immunology. This review provides further insights into the current roles of eEF2K and its potential to be explored as a therapeutic target for drug development.
Collapse
Affiliation(s)
- Darby J. Ballard
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Hao-Yun Peng
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, United States
| | - Jugal Kishore Das
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Anil Kumar
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Liqing Wang
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, United States
| | - Yijie Ren
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Xiaofang Xiong
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Xingcong Ren
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Jin-Ming Yang
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Jianxun Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| |
Collapse
|
35
|
Blaze J, Navickas A, Phillips HL, Heissel S, Plaza-Jennings A, Miglani S, Asgharian H, Foo M, Katanski CD, Watkins CP, Pennington ZT, Javidfar B, Espeso-Gil S, Rostandy B, Alwaseem H, Hahn CG, Molina H, Cai DJ, Pan T, Yao WD, Goodarzi H, Haghighi F, Akbarian S. Neuronal Nsun2 deficiency produces tRNA epitranscriptomic alterations and proteomic shifts impacting synaptic signaling and behavior. Nat Commun 2021; 12:4913. [PMID: 34389722 PMCID: PMC8363735 DOI: 10.1038/s41467-021-24969-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 07/16/2021] [Indexed: 02/07/2023] Open
Abstract
Epitranscriptomic mechanisms linking tRNA function and the brain proteome to cognition and complex behaviors are not well described. Here, we report bi-directional changes in depression-related behaviors after genetic disruption of neuronal tRNA cytosine methylation, including conditional ablation and transgene-derived overexpression of Nsun2 in the mouse prefrontal cortex (PFC). Neuronal Nsun2-deficiency was associated with a decrease in tRNA m5C levels, resulting in deficits in expression of 70% of tRNAGly isodecoders. Altogether, 1488/5820 proteins changed upon neuronal Nsun2-deficiency, in conjunction with glycine codon-specific defects in translational efficiencies. Loss of Gly-rich proteins critical for glutamatergic neurotransmission was associated with impaired synaptic signaling at PFC pyramidal neurons and defective contextual fear memory. Changes in the neuronal translatome were also associated with a 146% increase in glycine biosynthesis. These findings highlight the methylation sensitivity of glycinergic tRNAs in the adult PFC. Furthermore, they link synaptic plasticity and complex behaviors to epitranscriptomic modifications of cognate tRNAs and the proteomic homeostasis associated with specific amino acids.
Collapse
Affiliation(s)
- J Blaze
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - A Navickas
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - H L Phillips
- Departments of Psychiatry and Behavioral Sciences, Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, USA
| | - S Heissel
- The Rockefeller University Proteomics Resource Center, The Rockefeller University, New York, NY, USA
| | - A Plaza-Jennings
- Department of Psychiatry, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - S Miglani
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - H Asgharian
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - M Foo
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - C D Katanski
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - C P Watkins
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Z T Pennington
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - B Javidfar
- Friedman Brain Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - S Espeso-Gil
- Friedman Brain Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - B Rostandy
- The Rockefeller University Proteomics Resource Center, The Rockefeller University, New York, NY, USA
| | - H Alwaseem
- The Rockefeller University Proteomics Resource Center, The Rockefeller University, New York, NY, USA
| | - C G Hahn
- Department of Neurosciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - H Molina
- The Rockefeller University Proteomics Resource Center, The Rockefeller University, New York, NY, USA
| | - D J Cai
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - T Pan
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - W D Yao
- Departments of Psychiatry and Behavioral Sciences, Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, USA
| | - H Goodarzi
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - F Haghighi
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
- Research and Development Service, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
| | - S Akbarian
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mt. Sinai, New York, NY, USA.
| |
Collapse
|
36
|
Shinohara R, Aghajanian GK, Abdallah CG. Neurobiology of the Rapid-Acting Antidepressant Effects of Ketamine: Impact and Opportunities. Biol Psychiatry 2021; 90:85-95. [PMID: 33568318 DOI: 10.1016/j.biopsych.2020.12.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/13/2020] [Accepted: 12/04/2020] [Indexed: 12/28/2022]
Abstract
The discovery of the rapid-acting antidepressant effects of ketamine has 1) led to a paradigm shift in our perception of what is possible in treating severe depression; 2) spurred a wave of basic, translation, and clinical research; and 3) provided an unprecedented investigational tool to conduct longitudinal mechanistic studies that may capture behavioral changes as complex as clinical remission and relapse within hours and days of treatment. Unfortunately, these advances did not yet translate into clinical biomarkers or novel treatments, beyond ketamine. In contrast to slow-acting antidepressants, in which targeting monoaminergic receptors identified several efficacious drugs with comparable mechanisms, the focus on the receptor targets of ketamine has failed in several clinical trials over the past decade. Thus, it is becoming increasingly crucial that we concentrate our effort on the downstream molecular mechanisms of ketamine and their effects on the brain circuitry and networks. Honoring the legacy of our mentor, friend, and colleague Ron Duman, we provide a historical note on the discovery of ketamine and its putative mechanisms. We then detail the molecular and circuits effect of ketamine based on preclinical findings, followed by a summary of the impact of this work on our understanding of chronic stress pathology across psychiatric disorders, with particular emphasis on the role of synaptic connectivity and its brain network effects in the pathology and treatment of clinical depression.
Collapse
Affiliation(s)
- Ryota Shinohara
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - George K Aghajanian
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Chadi G Abdallah
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut; VA National Center for PTSD-Clinical Neuroscience Division, West Haven, Connecticut; Michael E. DeBakey VA Medical Center, Houston, Texas; Menninger Department of Psychiatry, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
37
|
Gilbert JR, Galiano CS, Nugent AC, Zarate CA. Ketamine and Attentional Bias Toward Emotional Faces: Dynamic Causal Modeling of Magnetoencephalographic Connectivity in Treatment-Resistant Depression. Front Psychiatry 2021; 12:673159. [PMID: 34220581 PMCID: PMC8249755 DOI: 10.3389/fpsyt.2021.673159] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/25/2021] [Indexed: 12/11/2022] Open
Abstract
The glutamatergic modulator ketamine rapidly reduces depressive symptoms in individuals with treatment-resistant major depressive disorder (TRD) and bipolar disorder. While its underlying mechanism of antidepressant action is not fully understood, modulating glutamatergically-mediated connectivity appears to be a critical component moderating antidepressant response. This double-blind, crossover, placebo-controlled study analyzed data from 19 drug-free individuals with TRD and 15 healthy volunteers who received a single intravenous infusion of ketamine hydrochloride (0.5 mg/kg) as well as an intravenous infusion of saline placebo. Magnetoencephalographic recordings were collected prior to the first infusion and 6-9 h after both drug and placebo infusions. During scanning, participants completed an attentional dot probe task that included emotional faces. Antidepressant response was measured across time points using the Montgomery-Asberg Depression Rating Scale (MADRS). Dynamic causal modeling (DCM) was used to measure changes in parameter estimates of connectivity via a biophysical model that included realistic local neuronal architecture and receptor channel signaling, modeling connectivity between the early visual cortex, fusiform cortex, amygdala, and inferior frontal gyrus. Clinically, ketamine administration significantly reduced depressive symptoms in TRD participants. Within the model, ketamine administration led to faster gamma aminobutyric acid (GABA) and N-methyl-D-aspartate (NMDA) transmission in the early visual cortex, faster NMDA transmission in the fusiform cortex, and slower NMDA transmission in the amygdala. Ketamine administration also led to direct and indirect changes in local inhibition in the early visual cortex and inferior frontal gyrus and to indirect increases in cortical excitability within the amygdala. Finally, reductions in depressive symptoms in TRD participants post-ketamine were associated with faster α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) transmission and increases in gain control of spiny stellate cells in the early visual cortex. These findings provide additional support for the GABA and NMDA inhibition and disinhibition hypotheses of depression and support the role of AMPA throughput in ketamine's antidepressant effects. Clinical Trial Registration: https://clinicaltrials.gov/ct2/show/NCT00088699?term=NCT00088699&draw=2&rank=1, identifier NCT00088699.
Collapse
Affiliation(s)
- Jessica R. Gilbert
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | | | | | | |
Collapse
|
38
|
Abstract
After participating in this activity, learners should be better able to:• Identify the effects of dysregulated opioid signalling in depression• Evaluate the use of opioid compounds and ketamine in patients with depression ABSTRACT: Major depressive disorder (MDD) remains one of the leading causes of disability and functional impairment worldwide. Current antidepressant therapeutics require weeks to months of treatment prior to the onset of clinical efficacy on depressed mood but remain ineffective in treating suicidal ideation and cognitive impairment. Moreover, 30%-40% of individuals fail to respond to currently available antidepressant medications. MDD is a heterogeneous disorder with an unknown etiology; novel strategies must be developed to treat MDD more effectively. Emerging evidence suggests that targeting one or more of the four opioid receptors-mu (MOR), kappa (KOR), delta (DOR), and the nociceptin/orphanin FQ receptor (NOP)-may yield effective therapeutics for stress-related psychiatric disorders. Furthermore, the effects of the rapidly acting antidepressant ketamine may involve opioid receptors. This review highlights dysregulated opioid signaling in depression, evaluates clinical trials with opioid compounds, and considers the role of opioid mechanisms in rapidly acting antidepressants.
Collapse
|
39
|
Fagerholm ED, Leech R, Williams S, Zarate CA, Moran RJ, Gilbert JR. Fine-tuning neural excitation/inhibition for tailored ketamine use in treatment-resistant depression. Transl Psychiatry 2021; 11:335. [PMID: 34052834 PMCID: PMC8164631 DOI: 10.1038/s41398-021-01442-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/04/2021] [Accepted: 05/12/2021] [Indexed: 12/26/2022] Open
Abstract
The glutamatergic modulator ketamine has been shown to rapidly reduce depressive symptoms in patients with treatment-resistant major depressive disorder (TRD). Although its mechanisms of action are not fully understood, changes in cortical excitation/inhibition (E/I) following ketamine administration are well documented in animal models and could represent a potential biomarker of treatment response. Here, we analyse neuromagnetic virtual electrode time series collected from the primary somatosensory cortex in 18 unmedicated patients with TRD and in an equal number of age-matched healthy controls during a somatosensory 'airpuff' stimulation task. These two groups were scanned as part of a clinical trial of ketamine efficacy under three conditions: (a) baseline; (b) 6-9 h following subanesthetic ketamine infusion; and (c) 6-9 h following placebo-saline infusion. We obtained estimates of E/I interaction strengths by using dynamic causal modelling (DCM) on the time series, thereby allowing us to pinpoint, under each scanning condition, where each subject's dynamics lie within the Poincaré diagram-as defined in dynamical systems theory. We demonstrate that the Poincaré diagram offers classification capability for TRD patients, in that the further the patients' coordinates were shifted (by virtue of ketamine) toward the stable (top-left) quadrant of the Poincaré diagram, the more their depressive symptoms improved. The same relationship was not observed by virtue of a placebo effect-thereby verifying the drug-specific nature of the results. We show that the shift in neural dynamics required for symptom improvement necessitates an increase in both excitatory and inhibitory coupling. We present accompanying MATLAB code made available in a public repository, thereby allowing for future studies to assess individually tailored treatments of TRD.
Collapse
Affiliation(s)
| | - Robert Leech
- Department of Neuroimaging, King's College London, London, UK
| | - Steven Williams
- Department of Neuroimaging, King's College London, London, UK
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, NIMH, NIH, Bethesda, MD, USA
| | - Rosalyn J Moran
- Department of Neuroimaging, King's College London, London, UK
| | - Jessica R Gilbert
- Experimental Therapeutics and Pathophysiology Branch, NIMH, NIH, Bethesda, MD, USA.
| |
Collapse
|
40
|
Piserchio A, Long K, Lee K, Kumar EA, Abzalimov R, Dalby KN, Ghose R. Structural dynamics of the complex of calmodulin with a minimal functional construct of eukaryotic elongation factor 2 kinase and the role of Thr348 autophosphorylation. Protein Sci 2021; 30:1221-1234. [PMID: 33890716 DOI: 10.1002/pro.4087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 04/15/2021] [Indexed: 12/31/2022]
Abstract
The calmodulin (CaM) activated α-kinase, eukaryotic elongation factor 2 kinase (eEF-2K), plays a central role in regulating translational elongation by phosphorylating eukaryotic elongation factor 2 (eEF-2), thereby reducing its ability to associate with the ribosome and suppressing global protein synthesis. Using TR (for truncated), a minimal functional construct of eEF-2K, and utilizing hydrogen/deuterium exchange mass spectrometry (HXMS), solution-state nuclear magnetic resonance (NMR) and biochemical approaches, we investigate the conformational changes accompanying complex formation between Ca2+ -CaM and TR and the effects of autophosphorylation of the latter at Thr348, its primary regulatory site. Our results suggest that a CaM C-lobe surface, complementary to the one involved in recognizing the calmodulin-binding domain (CBD) of TR, provides a secondary TR-interaction platform. CaM helix F, which is part of this secondary surface, responds to both Thr348 phosphorylation and pH changes, indicating its integration into an allosteric network that encompasses both components of the Ca2+ -CaM•TR complex. Solution NMR data suggest that CaMH107K , which carries a helix F mutation, is compromised in its ability to drive the conformational changes in TR necessary to enable efficient Thr348 phosphorylation. Biochemical studies confirm the diminished capacity of CaMH107K to induce TR autophosphorylation compared to wild-type CaM.
Collapse
Affiliation(s)
- Andrea Piserchio
- Department of Chemistry and Biochemistry, The City College of New York, New York, New York, USA
| | - Kimberly Long
- Division of Chemical Biology and Medicinal Chemistry, University of Texas, Austin, Texas, USA
| | - Kwangwoon Lee
- Department of Chemistry and Biochemistry, The City College of New York, New York, New York, USA.,Graduate Programs in Biochemistry, The Graduate Center of CUNY, New York, New York, USA.,Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Eric A Kumar
- Division of Chemical Biology and Medicinal Chemistry, University of Texas, Austin, Texas, USA
| | - Rinat Abzalimov
- Biomolecular Mass Spectrometry Facility, CUNY ASRC, New York, New York, USA
| | - Kevin N Dalby
- Division of Chemical Biology and Medicinal Chemistry, University of Texas, Austin, Texas, USA.,Graduate Program in Cell and Molecular Biology, University of Texas, Austin, Texas, USA
| | - Ranajeet Ghose
- Department of Chemistry and Biochemistry, The City College of New York, New York, New York, USA.,Graduate Programs in Biochemistry, The Graduate Center of CUNY, New York, New York, USA.,Graduate Programs in Chemistry, The Graduate Center of CUNY, New York, New York, USA.,Graduate Programs in Physics, The Graduate Center of CUNY, New York, New York, USA
| |
Collapse
|
41
|
Mathew AS, Gorick CM, Thim EA, Garrison WJ, Klibanov AL, Miller GW, Sheybani ND, Price RJ. Transcriptomic response of brain tissue to focused ultrasound-mediated blood-brain barrier disruption depends strongly on anesthesia. Bioeng Transl Med 2021; 6:e10198. [PMID: 34027087 PMCID: PMC8126816 DOI: 10.1002/btm2.10198] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/19/2020] [Accepted: 10/21/2020] [Indexed: 12/16/2022] Open
Abstract
Focused ultrasound (FUS) mediated blood-brain barrier disruption (BBBD) targets the delivery of systemically-administered therapeutics to the central nervous system. Preclinical investigations of BBBD have been performed on different anesthetic backgrounds; however, the influence of the choice of anesthetic on the molecular response to BBBD is unknown, despite its potential to critically affect interpretation of experimental therapeutic outcomes. Here, using bulk RNA sequencing, we comprehensively examined the transcriptomic response of both normal brain tissue and brain tissue exposed to FUS-induced BBBD in mice anesthetized with either isoflurane with medical air (Iso) or ketamine/dexmedetomidine (KD). In normal murine brain tissue, Iso alone elicited minimal differential gene expression (DGE) and repressed pathways associated with neuronal signaling. KD alone, however, led to massive DGE and enrichment of pathways associated with protein synthesis. In brain tissue exposed to BBBD (1 MHz, 0.5 Hz pulse repetition frequency, 0.4 MPa peak-negative pressure), we systematically evaluated the relative effects of anesthesia, microbubbles, and FUS on the transcriptome. Of particular interest, we observed that gene sets associated with sterile inflammatory responses and cell-cell junctional activity were induced by BBBD, regardless of the choice of anesthesia. Meanwhile, gene sets associated with metabolism, platelet activity, tissue repair, and signaling pathways, were differentially affected by BBBD, with a strong dependence on the anesthetic. We conclude that the underlying transcriptomic response to FUS-mediated BBBD may be powerfully influenced by anesthesia. These findings raise considerations for the translation of FUS-BBBD delivery approaches that impact, in particular, metabolism, tissue repair, and intracellular signaling.
Collapse
Affiliation(s)
- Alexander S. Mathew
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Catherine M. Gorick
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - E. Andrew Thim
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - William J. Garrison
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Radiology & Medical ImagingUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Alexander L. Klibanov
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Internal Medicine, Cardiovascular DivisionUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - G. Wilson Miller
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Radiology & Medical ImagingUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Natasha D. Sheybani
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Richard J. Price
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Radiology & Medical ImagingUniversity of VirginiaCharlottesvilleVirginiaUSA
| |
Collapse
|
42
|
Abstract
Over the last two decades, the dissociative anaesthetic agent ketamine, an uncompetitive N-Methyl-D-Aspartate (NMDA) receptor antagonist, has emerged as a novel therapy for treatment-resistant depression (TRD), demonstrating rapid and robust antidepressant effects within hours of administration. Ketamine is a racemic mixture composed of equal amounts of (S)-ketamine and (R)-ketamine. Although ketamine currently remains an off-label treatment for TRD, an (S)-ketamine nasal spray has been approved for use in TRD (in conjunction with an oral antidepressant) in the United States and Europe. Despite the promise of ketamine, key challenges including how to maintain response, concerns regarding short and long-term side-effects and the potential for abuse remain. This review provides an overview of the history of ketamine, its use in psychiatry and its basic pharmacology. The clinical evidence for the use of ketamine in depression and potential adverse effects associated with treatment are summarized. A synopsis of some of the putative neurobiological mechanisms underlying ketamine's rapid-acting antidepressant effects is provided before finally outlining future research directions, including the need to identify biomarkers for predicting response and treatment targets that may be used in the development of next-generation rapid-acting antidepressants that may lack ketamine's side-effects or abuse potential.
Collapse
Affiliation(s)
- Luke A Jelen
- Centre for Affective Disorders, Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - James M Stone
- Centre for Affective Disorders, Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,South London and Maudsley NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
43
|
Pizzo L, Lasser M, Yusuff T, Jensen M, Ingraham P, Huber E, Singh MD, Monahan C, Iyer J, Desai I, Karthikeyan S, Gould DJ, Yennawar S, Weiner AT, Pounraja VK, Krishnan A, Rolls MM, Lowery LA, Girirajan S. Functional assessment of the "two-hit" model for neurodevelopmental defects in Drosophila and X. laevis. PLoS Genet 2021; 17:e1009112. [PMID: 33819264 PMCID: PMC8049494 DOI: 10.1371/journal.pgen.1009112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 04/15/2021] [Accepted: 03/16/2021] [Indexed: 12/13/2022] Open
Abstract
We previously identified a deletion on chromosome 16p12.1 that is mostly inherited and associated with multiple neurodevelopmental outcomes, where severely affected probands carried an excess of rare pathogenic variants compared to mildly affected carrier parents. We hypothesized that the 16p12.1 deletion sensitizes the genome for disease, while "second-hits" in the genetic background modulate the phenotypic trajectory. To test this model, we examined how neurodevelopmental defects conferred by knockdown of individual 16p12.1 homologs are modulated by simultaneous knockdown of homologs of "second-hit" genes in Drosophila melanogaster and Xenopus laevis. We observed that knockdown of 16p12.1 homologs affect multiple phenotypic domains, leading to delayed developmental timing, seizure susceptibility, brain alterations, abnormal dendrite and axonal morphology, and cellular proliferation defects. Compared to genes within the 16p11.2 deletion, which has higher de novo occurrence, 16p12.1 homologs were less likely to interact with each other in Drosophila models or a human brain-specific interaction network, suggesting that interactions with "second-hit" genes may confer higher impact towards neurodevelopmental phenotypes. Assessment of 212 pairwise interactions in Drosophila between 16p12.1 homologs and 76 homologs of patient-specific "second-hit" genes (such as ARID1B and CACNA1A), genes within neurodevelopmental pathways (such as PTEN and UBE3A), and transcriptomic targets (such as DSCAM and TRRAP) identified genetic interactions in 63% of the tested pairs. In 11 out of 15 families, patient-specific "second-hits" enhanced or suppressed the phenotypic effects of one or many 16p12.1 homologs in 32/96 pairwise combinations tested. In fact, homologs of SETD5 synergistically interacted with homologs of MOSMO in both Drosophila and X. laevis, leading to modified cellular and brain phenotypes, as well as axon outgrowth defects that were not observed with knockdown of either individual homolog. Our results suggest that several 16p12.1 genes sensitize the genome towards neurodevelopmental defects, and complex interactions with "second-hit" genes determine the ultimate phenotypic manifestation.
Collapse
Affiliation(s)
- Lucilla Pizzo
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States of America
| | - Micaela Lasser
- Department of Biology, Boston College, Chestnut Hill, MA, United States of America
| | - Tanzeen Yusuff
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States of America
| | - Matthew Jensen
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States of America
| | - Phoebe Ingraham
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States of America
| | - Emily Huber
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States of America
| | - Mayanglambam Dhruba Singh
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States of America
| | - Connor Monahan
- Department of Biology, Boston College, Chestnut Hill, MA, United States of America
| | - Janani Iyer
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States of America
| | - Inshya Desai
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States of America
| | - Siddharth Karthikeyan
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States of America
| | - Dagny J. Gould
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States of America
| | - Sneha Yennawar
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States of America
| | - Alexis T. Weiner
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States of America
| | - Vijay Kumar Pounraja
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States of America
| | - Arjun Krishnan
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI, United States of America
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States of America
| | - Melissa M. Rolls
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States of America
| | - Laura Anne Lowery
- Department of Medicine, Boston University Medical Center, Boston, MA, United States of America
| | - Santhosh Girirajan
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States of America
- Department of Anthropology, The Pennsylvania State University, University Park, PA, United States of America
| |
Collapse
|
44
|
Li W, Ali T, Zheng C, Liu Z, He K, Shah FA, Ren Q, Rahman SU, Li N, Yu ZJ, Li S. Fluoxetine regulates eEF2 activity (phosphorylation) via HDAC1 inhibitory mechanism in an LPS-induced mouse model of depression. J Neuroinflammation 2021; 18:38. [PMID: 33526073 PMCID: PMC7852137 DOI: 10.1186/s12974-021-02091-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/19/2021] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND Selective serotonin reuptaker inhibitors, including fluoxetine, are widely studied and prescribed antidepressants, while their exact molecular and cellular mechanism are yet to be defined. We investigated the involvement of HDAC1 and eEF2 in the antidepressant mechanisms of fluoxetine using a lipopolysaccharide (LPS)-induced depression-like behavior model. METHODS For in vivo analysis, mice were treated with LPS (2 mg/kg BW), fluoxetine (20 mg/kg BW), HDAC1 activator (Exifone: 54 mg/kg BW) and NH125 (1 mg/kg BW). Depressive-like behaviors were confirmed via behavior tests including OFT, FST, SPT, and TST. Cytokines were measured by ELISA while Iba-1 and GFAP expression were determined by immunofluorescence. Further, the desired gene expression was measured by immunoblotting. For in vitro analysis, BV2 cell lines were cultured; treated with LPS, exifone, and fluoxetine; collected; and analyzed. RESULTS Mice treated with LPS displayed depression-like behaviors, pronounced neuroinflammation, increased HDAC1 expression, and reduced eEF2 activity, as accompanied by altered synaptogenic factors including BDNF, SNAP25, and PSD95. Fluoxetine treatment exhibited antidepressant effects and ameliorated the molecular changes induced by LPS. Exifone, a selective HDAC1 activator, reversed the antidepressant and anti-inflammatory effects of fluoxetine both in vivo and in vitro, supporting a causing role of HDAC1 in neuroinflammation allied depression. Further molecular mechanisms underlying HDAC1 were explored with NH125, an eEF2K inhibitor, whose treatment reduced immobility time, altered pro-inflammatory cytokines, and NLRP3 expression. Moreover, NH125 treatment enhanced eEF2 and GSK3β activities, BDNF, SNAP25, and PSD95 expression, but had no effects on HDAC1. CONCLUSIONS Our results showed that the antidepressant effects of fluoxetine may involve HDAC1-eEF2 related neuroinflammation and synaptogenesis.
Collapse
Affiliation(s)
- Weifen Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055 China
| | - Tahir Ali
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055 China
| | - Chengyou Zheng
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055 China
| | - Zizhen Liu
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055 China
| | - Kaiwu He
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055 China
| | - Fawad Ali Shah
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055 China
- Riphah Institute of Pharmaceutical Sciences, Riphah International University Islamabad, Islamabad, Pakistan
| | - Qingguo Ren
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Shafiq Ur Rahman
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055 China
- Department of Pharmacy, Shaheed Benazir Bhutto University, Sheringal, Dir, 18000, Pakistan
| | - Ningning Li
- Tomas Lindahl Nobel Laureate Laboratory, Precision Medicine Research Centre, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107 China
| | - Zhi-Jian Yu
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, The 6th Affiliated Hospital of Shenzhen University Health Science Center, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052 China
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055 China
- Campbell Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario Canada
| |
Collapse
|
45
|
Muscat SA, Hartelius G, Crouch CR, Morin KW. An Integrative Approach to Ketamine Therapy May Enhance Multiple Dimensions of Efficacy: Improving Therapeutic Outcomes With Treatment Resistant Depression. Front Psychiatry 2021; 12:710338. [PMID: 34899408 PMCID: PMC8653702 DOI: 10.3389/fpsyt.2021.710338] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 10/21/2021] [Indexed: 12/31/2022] Open
Abstract
Research over the last two decades has established ketamine as a safe, effective, fast-acting, and sustained antidepressant that significantly reduces adverse symptoms associated with depression, even in patients who are treatment resistant. Much of this research has evolved within the framework of several independent branches of scientific inquiry: in addition to the study of ketamine is a non-selective NMDAR antagonist with rapid antidepressant effects, it has also been found effective as a psychoplastogen that stimulates synaptogenesis and increases neuroplasticity, as a powerful anti-inflammatory that may improve inflammation-related depressive symptoms, as a substance that induces beneficial high entropy brain states, and as a subjectively impactful psychedelic agent. Each branch of inquiry has generated independent evidence of ketamine's efficacy but has advanced without substantive coordination or communication with other lines of inquiry. Integrative research that considers these branches of research together may lead toward a better understanding of ketamine's effects and improved treatment protocols and clinical outcomes. Such an overview can inform more comprehensive patient care through: (a) informed patient psychoeducation that encompasses all of ketamine's mechanisms of action; (b) calibration of optimal dosage to ensure induction and maintenance of high entropy brain states during each ketamine session utilizing EEG measurement; (c) Improved management of emergence side effects through proper care for set and setting; (d) inclusion of pre-selected appropriate music to enhance the emotional experience; (e) increased monitoring of ketamine effects on cortical activity, inter-hemispheric imbalance, and inflammation-related levels of cytokines to further improvements in ketamine protocols; and (f) appropriate timing of any adjunctive psychotherapy sessions to coincide with peak neurogenesis at 24-48 h post ketamine treatment.
Collapse
Affiliation(s)
- Sherry-Anne Muscat
- Youth Forensic Psychiatry, Alberta Hospital, Alberta Health Services, Edmonton, AB, Canada.,Integral and Transpersonal Psychology, California Institute of Integral Studies, San Francisco, CA, United States
| | - Glenn Hartelius
- Integral and Transpersonal Psychology, California Institute of Integral Studies, San Francisco, CA, United States
| | - Courtenay Richards Crouch
- Integral and Transpersonal Psychology, California Institute of Integral Studies, San Francisco, CA, United States
| | - Kevin W Morin
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada.,Acute Adult Psychiatry, Alberta Hospital, Alberta Health Services, Edmonton, AB, Canada
| |
Collapse
|
46
|
Haarsma J, Harmer CJ, Tamm S. A continuum hypothesis of psychotomimetic rapid antidepressants. Brain Neurosci Adv 2021; 5:23982128211007772. [PMID: 34017922 PMCID: PMC8114748 DOI: 10.1177/23982128211007772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/08/2021] [Indexed: 01/10/2023] Open
Abstract
Ketamine, classical psychedelics and sleep deprivation are associated with rapid effects on depression. Interestingly, these interventions also have common psychotomimetic actions, mirroring aspects of psychosis such as an altered sense of self, perceptual distortions and distorted thinking. This raises the question whether these interventions might be acute antidepressants through the same mechanisms that underlie some of their psychotomimetic effects. That is, perhaps some symptoms of depression can be understood as occupying the opposite end of a spectrum where elements of psychosis can be found on the other side. This review aims at reviewing the evidence underlying a proposed continuum hypothesis of psychotomimetic rapid antidepressants, suggesting that a range of psychotomimetic interventions are also acute antidepressants as well as trying to explain these common features in a hierarchical predictive coding framework, where we hypothesise that these interventions share a common mechanism by increasing the flexibility of prior expectations. Neurobiological mechanisms at play and the role of different neuromodulatory systems affected by these interventions and their role in controlling the precision of prior expectations and new sensory evidence will be reviewed. The proposed hypothesis will also be discussed in relation to other existing theories of antidepressants. We also suggest a number of novel experiments to test the hypothesis and highlight research areas that could provide further insights, in the hope to better understand the acute antidepressant properties of these interventions.
Collapse
Affiliation(s)
- Joost Haarsma
- Wellcome Centre for Human Neuroimaging, University College London, London, UK
| | - Catherine J Harmer
- Department of Psychiatry and Oxford Health NHS Foundation Trust, Warneford Hospital, University of Oxford, Oxford, UK
| | - Sandra Tamm
- Department of Psychiatry and Oxford Health NHS Foundation Trust, Warneford Hospital, University of Oxford, Oxford, UK
- Stress Research Institute, Department of Psychology, Stockholm University, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
47
|
Alshammari TK. The Ketamine Antidepressant Story: New Insights. Molecules 2020; 25:molecules25235777. [PMID: 33297563 PMCID: PMC7730956 DOI: 10.3390/molecules25235777] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/02/2020] [Accepted: 12/06/2020] [Indexed: 12/12/2022] Open
Abstract
Ketamine is a versatile agent primarily utilized as a dissociative anesthetic, which acts by blocking the excitatory receptor N-methyl-d-aspartate receptor (NMDA). It functions to inhibit the current of both Na+ and K+ voltage-gated channels, thus preventing serotonin and dopamine reuptake. Studies have indicated that administering a single subanesthetic dose of ketamine relieves depression rapidly and that the effect is sustained. For decades antidepressant agents were based on the monoamine theory. Although ketamine may not be the golden antidepressant, it has opened new avenues toward mechanisms involved in the pathology of treatment-resistant depression and achieving rapid antidepressant effects. Thus, preclinical studies focusing on deciphering the molecular mechanisms involved in the antidepressant action of ketamine will assist in the development of a new antidepressant. This review was conducted to elucidate the emerging pathways that can explain the complex dose-dependent mechanisms achieved by administering ketamine to treat major depressive disorders. Special attention was paid to reviewing the literature on hydroxynorketamines, which are ketamine metabolites that have recently attracted attention in the context of depression.
Collapse
Affiliation(s)
- Tahani K Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2475, Riyadh 11451, Saudi Arabia
| |
Collapse
|
48
|
Neis VB, Moretti M, Rosa PB, Dalsenter YDO, Werle I, Platt N, Kaufmann FN, Rosado AF, Besen MH, Rodrigues ALS. The involvement of PI3K/Akt/mTOR/GSK3β signaling pathways in the antidepressant-like effect of AZD6765. Pharmacol Biochem Behav 2020; 198:173020. [DOI: 10.1016/j.pbb.2020.173020] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/10/2020] [Accepted: 08/26/2020] [Indexed: 12/31/2022]
|
49
|
Woelfer M, Li M, Colic L, Liebe T, Di X, Biswal B, Murrough J, Lessmann V, Brigadski T, Walter M. Ketamine-induced changes in plasma brain-derived neurotrophic factor (BDNF) levels are associated with the resting-state functional connectivity of the prefrontal cortex. World J Biol Psychiatry 2020; 21:696-710. [PMID: 31680600 DOI: 10.1080/15622975.2019.1679391] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVES Synaptic plasticity and brain-derived neurotrophic factor (BDNF) signalling are proposed to play key roles in antidepressant drug action. Ketamine, an N-methyl-D-aspartate receptor antagonist and putative antidepressant, may increase synaptic plasticity in prefrontal cortex through higher expression of BDNF. Furthermore, ketamine was shown to change resting-state functional connectivity (RSFC) of dorsomedial prefrontal cortex (dmPFC). METHODS In a randomised, placebo-controlled study, we investigated acutely (100 min) and at 24 h following subanesthetic ketamine infusion which dmPFC seeded RSFC changes are most strongly associated with plasma BDNF level changes in 53 healthy participants (21 females, age: 24.4 ± 2.9 years) using 7 T-fMRI. RESULTS We observed higher relative levels of BDNF 2 h and 24 h after ketamine compared to placebo. Whole-brain regression revealed that the change in BDNF after 24 h was associated with RSFC decreases from dmPFC to posterior cingulate cortex and ventromedial PFC at 24 h and exploratively also at the 100 min measurement point. Follow-up analyses revealed that RSFC reductions following ketamine were restricted to subjects showing increased BDNF levels at 24 h. CONCLUSIONS Our findings indicate BDNF level dynamics following ketamine are related to acute and 24 h RSFC changes. Particularly when BDNF increases are observed after ketamine infusion, a disconnection from dmPFC after 24 h is seen and may reflect synaptic plasticity effects.
Collapse
Affiliation(s)
- Marie Woelfer
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany.,Leibniz Institute for Neurobiology, Magdeburg, Germany.,Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Meng Li
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany.,Department of Psychiatry and Psychotherapy, University Tuebingen, Tuebingen, Germany
| | - Lejla Colic
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany.,Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Thomas Liebe
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany
| | - Xin Di
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Bharat Biswal
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA.,School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - James Murrough
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Volkmar Lessmann
- Institute of Physiology, Otto-von-Guericke-University, Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Tanja Brigadski
- Institute of Physiology, Otto-von-Guericke-University, Magdeburg, Germany.,Department of Informatics and Microsystems Technology, University of Applied Science Kaiserslautern, Zweibrücken, Germany
| | - Martin Walter
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany.,Department of Psychiatry and Psychotherapy, University Tuebingen, Tuebingen, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| |
Collapse
|
50
|
Gladulich LFH, Peixoto-Rodrigues MC, Campello-Costa P, Paes-de-Carvalho R, Cossenza M. NMDA-induced nitric oxide generation and CREB activation in central nervous system is dependent on eukaryotic elongation factor 2 kinase. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118783. [DOI: 10.1016/j.bbamcr.2020.118783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/11/2020] [Accepted: 06/17/2020] [Indexed: 01/28/2023]
|