1
|
Daniels S, El Mansari M, Blier P. AMPA receptors modulate enhanced dopamine neuronal activity induced by the combined administration of venlafaxine and brexpiprazole. Neuropsychopharmacology 2024; 49:2042-2051. [PMID: 39147870 PMCID: PMC11480427 DOI: 10.1038/s41386-024-01958-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/17/2024]
Abstract
Addition of dopamine (DA)/serotonin (5-HT) partial agonists to 5-HT/norepinephrine (NE) reuptake inhibitors are commonly used to enhance the antidepressant response. The simultaneous inhibition of 5-HT and NE transporters with venlafaxine and its combination of brexpiprazole, which blocks the α2-adrenergic autoreceptor on NE terminals, could constitute a superior strategy. Anesthetized rats received venlafaxine and brexpiprazole for 2 and 14 days, then the firing activity of dorsal raphe nucleus 5-HT, locus coeruleus NE, and ventral tegmental area DA neurons were assessed. Net 5-HT and NE neurotransmissions were evaluated by assessing the tonic activation of 5-HT1A, and α1- and α2-adrenergic receptors in the hippocampus. The combination of brexpiprazole with venlafaxine resulted in normalized 5-HT and NE neuron activity, which occurred earlier than that with venlafaxine alone. A significant enhancement of the tonic activation of 5-HT1A receptors and α2-adrenoceptors in the hippocampus was observed following administration of the combination for 14 days. The combination more than doubled the number of DA neurons per electrode descent, after both 2 and 14 days, while this increase was observed only after 14 days of venlafaxine administration. This increase in population activity was prevented by NBQX, an AMPA receptor antagonist. In conclusion, early during administration, the combination of venlafaxine with brexpiprazole normalized firing activity of 5-HT and NE neurons, and increased the population activity of DA neurons through AMPA receptors. In the hippocampus, there was an overall increase in both 5-HT and NE transmissions. These results imply that this strategy could be a rapid-acting approach to treat depression.
Collapse
Affiliation(s)
- Stephen Daniels
- University of Ottawa Institute of Mental Health Research, 1145 Carling Avenue, K1Z 7K4, Ottawa, Canada
| | - Mostafa El Mansari
- University of Ottawa Institute of Mental Health Research, 1145 Carling Avenue, K1Z 7K4, Ottawa, Canada.
| | - Pierre Blier
- University of Ottawa Institute of Mental Health Research, 1145 Carling Avenue, K1Z 7K4, Ottawa, Canada
| |
Collapse
|
2
|
Wang L, Zhao S, Shao J, Su C. The effect and mechanism of low-dose esketamine in neuropathic pain-related depression-like behavior in rats. Brain Res 2024; 1843:149117. [PMID: 38977235 DOI: 10.1016/j.brainres.2024.149117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/28/2024] [Accepted: 07/05/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND Clinical evidence suggests that Esketamine (ESK) is an effective treatment for depression. However, the effects of Esketamine in treating depression-like behavior induced by neuropathic pain is unclear. The underlying molecular mechanisms require further investigation to provide new therapeutic targets for the treatment of clinical neuropathic pain-related depression. METHODS A neuropathic pain-related depression model was established in rats with spared nerve injury (SNI). Male Sprague-Dawley rats were randomly divided into four groups: Sham Group, SNI group, SNI + Normal Saline (NS) Group and SNI + ESK5mg/kg Group. Mechanical pain thresholds were measured to assess pain sensitivity in SNI rats. On the 14th day after surgery a forced swim test and sucrose preference test were used to evaluate the depressive-like behavior of rats in each group. Further, a proteomic analysis was used to quantify differentially expressed proteins. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were analyzed to explore the main protein targets of SNI in the medial prefrontal cortex. The expression of proteins was detected by Western blotting. RESULTS A neuropathic pain-related depression model was established. Compared with the Sham group, the mechanical pain threshold was decreased significantly (13.2 ± 1.0 vs. 0.7 ± 0.01 g n = 8), while immobility on the forced swim test was also decreased (93.1 ± 7.4 vs. 169.5 ± 9.6 s n = 8), and sucrose preference rate was significantly increased (98.8 ± 0.3 vs. 73.1 ± 1.4n = 7) in SNI group rats. Compared with the SNI + NS group, the mechanical pain threshold was not statistically significant, while immobility on the forced swim test was clearly decreased (161.1 ± 11.6 vs. 77.9 ± 5.0 s n = 8), and sucrose preference rate was significantly increased (53.1 ± 8.9 vs. 96.1 ± 1.4n = 7) in SNI + ESK5mg/kg group rats. To further investigate the underlying mechanism, we employed proteomics to identify proteins exhibiting more than a 1.2-fold difference (P < 0.05) in expression levels within each group for subsequent analysis. Relative to the Sham group, 88 downregulated and 104 up-regulated proteins were identified in the SNI group, while 120 and 84 proteins were up- and down-regulated in the Esketamine treatment group compared with the SNI + NS group. Compared with Sham group, the expressions of mGluR5 and Homer1a were up-regulated in the medial prefrontal cortex (mPFC) in SNI group (mGluR5:0.97 ± 0.05 vs 1.47 ± 0.15, Homer1a:1.03 ± 0.06 vs 1.46 ± 0.16n = 6), and down-regulated after intervention with Esketamine (mGluR5:1.54 ± 0.11 vs 1.06 ± 0.07, Homer1a:1.51 ± 0.13 vs 1.12 ± 0.34n = 6). CONCLUSIONS Low-dose Esketamine appeared to relieve depression-like behavior induced by neuropathic pain. The Homer1a-mGluR5 signaling pathway might be the mechanism of antidepressant effect of Esketamine.
Collapse
Affiliation(s)
- Lijuan Wang
- Department of Anesthesiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha, Hunan, China; Department of Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Shuwu Zhao
- Department of Anesthesiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha, Hunan, China
| | - Jiali Shao
- Department of Anesthesiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha, Hunan, China
| | - Chen Su
- Department of Anesthesiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha, Hunan, China.
| |
Collapse
|
3
|
Tan Y, Hashimoto K. Therapeutic potential of ketamine in management of epilepsy: Clinical implications and mechanistic insights. Asian J Psychiatr 2024; 101:104246. [PMID: 39366036 DOI: 10.1016/j.ajp.2024.104246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 09/11/2024] [Accepted: 09/14/2024] [Indexed: 10/06/2024]
Abstract
Epilepsy, a widespread neurological disorder, affects approximately 50 million people worldwide. This disorder is typified by recurring seizures due to abnormal neuron communication in the brain. The seizures can lead to severe ischemia and hypoxia, potentially threatening patients' lives. However, with proper diagnosis and treatment, up to 70 % of patients can live without seizures. The causes of epilepsy are complex and multifactorial, encompassing genetic abnormalities, structural brain anomalies, ion channel dysfunctions, neurotransmitter imbalances, neuroinflammation, and immune system involvement. These factors collectively disrupt the crucial balance between excitation and inhibition within the brain, leading to epileptic seizures. The management of treatment-resistant epilepsy remains a considerable challenge, necessitating innovative therapeutic approaches. Among emerging potential treatments, ketamine-a drug traditionally employed for anesthesia and depression-has demonstrated efficacy in reducing seizures. It is noteworthy that, independent of its anti-epileptic effects, ketamine has been found to improve the balance between excitatory and inhibitory (E/I) activities in the brain. The balance is crucial for maintaining normal neural function, and its disruption is widely considered a key driver of epileptic seizures. By acting on N-methyl-D-aspartate (NMDA) receptors and other potential mechanisms, ketamine may regulate neuronal excitability, reduce excessive synchronized neural activity, and counteract epileptic seizures. This positive impact on E/I balance reinforces the potential of ketamine as a promising drug for treating epilepsy, especially in patients who are insensitive to traditional anti-epileptic drugs. This review aims to consolidate the current understanding of ketamine's therapeutic role in epilepsy. It will focus its impact on neuronal excitability and synaptic plasticity, its neuroprotective qualities, and elucidate the drug's potential mechanisms of action in treating epilepsy. By scrutinizing ketamine's impact and mechanisms in various types of epilepsy, we aspire to contribute to a more comprehensive and holistic approach to epilepsy management.
Collapse
Affiliation(s)
- Yunfei Tan
- Center for Rehabilitation Medicine, Department of Psychiatry, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Kenji Hashimoto
- Chiba University Center for Forensic Mental Health, Chiba 260-8677, Japan.
| |
Collapse
|
4
|
Zhen Z, Sun X, Yuan S, Zhang J. Psychoactive substances for the treatment of neuropsychiatric disorders. Asian J Psychiatr 2024; 101:104193. [PMID: 39243659 DOI: 10.1016/j.ajp.2024.104193] [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: 06/03/2024] [Revised: 08/04/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
In the contemporary landscape of psychiatric medicine, critical advancements have been noted in the utilization of psychoactive substances such as hallucinogens, 3,4-methylenedioxymethamphetamine (MDMA), and ketamine for the treatment of severe mental health disorders. This review provides a detailed evaluation of these substances, focusing on their mechanisms of action and the profound clinical outcomes observed in controlled environments. Hallucinogens like lysergic acid diethylamide and psilocybin primarily target the 5-HT2A receptor agonist-2 (5-HT2AR), inducing substantial perceptual and cognitive shifts that facilitate deep psychological introspection and significant therapeutic advances, particularly in patients suffering from depression and anxiety disorders. MDMA, influencing multiple neurotransmitter systems including 5-Hydroxytryptamine (5-HT), dopamine, and norepinephrine, has been demonstrated to effectively alleviate symptoms of post-traumatic stress disorder, enhancing patients' emotional engagement and resilience during psychotherapy. Meanwhile, ketamine, a glutamate receptor antagonist, rapidly alleviates depressive symptoms, offering a lifeline for individuals with treatment-resistant depression through its fast-acting antidepressant properties. The integration of these substances into psychiatric practice has shown promising results, fundamentally changing the therapeutic landscape for patients unresponsive to traditional treatment modalities. However, the potent effects of these agents also necessitate a cautious approach in clinical application, ensuring careful dosage control, monitoring, and risk management to prevent potential abuse and mitigate adverse effects.
Collapse
Affiliation(s)
- Zifan Zhen
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | - Xueqiang Sun
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | - Shiying Yuan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China.
| | - Jiancheng Zhang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China.
| |
Collapse
|
5
|
Kupferberg A, Hasler G. From antidepressants and psychotherapy to oxytocin, vagus nerve stimulation, ketamine and psychedelics: how established and novel treatments can improve social functioning in major depression. Front Psychiatry 2024; 15:1372650. [PMID: 39469469 PMCID: PMC11513289 DOI: 10.3389/fpsyt.2024.1372650] [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: 01/18/2024] [Accepted: 08/05/2024] [Indexed: 10/30/2024] Open
Abstract
Social cognitive deficits and social behavior impairments are common in major depressive disorder (MDD) and affect the quality of life and recovery of patients. This review summarizes the impact of standard and novel treatments on social functioning in MDD and highlights the potential of combining different approaches to enhance their effectiveness. Standard treatments, such as antidepressants, psychotherapies, and brain stimulation, have shown mixed results in improving social functioning, with some limitations and side effects. Newer treatments, such as intranasal oxytocin, mindfulness-based cognitive therapy, and psychedelic-assisted psychotherapy, have demonstrated positive effects on social cognition and behavior by modulating self-referential processing, empathy, and emotion regulation and through enhancement of neuroplasticity. Animal models have provided insights into the neurobiological mechanisms underlying these treatments, such as the role of neuroplasticity. Future research should explore the synergistic effects of combining different treatments and investigate the long-term outcomes and individual differences in response to these promising interventions.
Collapse
Affiliation(s)
- Aleksandra Kupferberg
- Molecular Psychiatry Lab, Faculty of Science and Medicine, University of Freiburg, Villars-sur-Glâne, Switzerland
| | - Gregor Hasler
- Molecular Psychiatry Lab, Faculty of Science and Medicine, University of Freiburg, Villars-sur-Glâne, Switzerland
- University Psychiatry Research Unit, Freiburg Mental Health Network, Villars-sur-Glâne, Switzerland
- Department of Neuropsychology, Lake Lucerne Institute, Vitznau, Switzerland
| |
Collapse
|
6
|
Kawczak P, Feszak I, Bączek T. Ketamine, Esketamine, and Arketamine: Their Mechanisms of Action and Applications in the Treatment of Depression and Alleviation of Depressive Symptoms. Biomedicines 2024; 12:2283. [PMID: 39457596 PMCID: PMC11505277 DOI: 10.3390/biomedicines12102283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Research over the past years has compared the enantiomers (S)-ketamine (esketamine) and (R)-ketamine (arketamine) of the previously known racemic mixture called ketamine (R/S-ketamine). Esketamine has been found to be more potent, offering three times stronger analgesic effects and 1.5 times greater anesthetic efficacy than arketamine. It provides smoother anesthesia with fewer side effects and is widely used in clinical settings due to its neuroprotective, bronchodilatory, and antiepileptic properties. Approved by the FDA and EMA in 2019, esketamine is currently used alongside SSRIs or SNRIs for treatment-resistant depression (TRD). On the other hand, arketamine has shown potential for treating neurological disorders such as Alzheimer's, Parkinson's, and multiple sclerosis, offering possible antidepressant effects and anti-inflammatory benefits. While esketamine is already in clinical use, arketamine's future depends on further research to address its safety, efficacy, and optimal dosing. Both enantiomers hold significant clinical value, with esketamine excelling in anesthesia, and arketamine showing promise in neurological and psychiatric treatments.
Collapse
Affiliation(s)
- Piotr Kawczak
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, 80-416 Gdańsk, Poland;
| | - Igor Feszak
- Institute of Health Sciences, Pomeranian University in Słupsk, 76-200 Słupsk, Poland;
| | - Tomasz Bączek
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, 80-416 Gdańsk, Poland;
- Department of Nursing and Medical Rescue, Institute of Health Sciences, Pomeranian University in Słupsk, 76-200 Słupsk, Poland
| |
Collapse
|
7
|
Ahmadi J, Mansoori A, Mosavat SH, Bazrafshan A. Comparison of ketamine with buprenorphine as adjunctive therapy in the treatment of comorbid major depressive disorder and opium use disorders: A randomized controlled trial. Int J Psychiatry Med 2024; 59:521-535. [PMID: 38140979 DOI: 10.1177/00912174231225087] [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] [Indexed: 12/24/2023]
Abstract
BACKGROUND Comorbid major depressive disorder (MDD) and opium use disorder (OUD) are known to increase the risk of suicide. The purpose of this study was to compare the efficacy and safety of adjunctive therapy with either ketamine or buprenorphine in patients with comorbid MDD and OUD. METHODS This was a randomized double-blind controlled trial in adults admitted to a hospital in Iran. Sixty-six participants were enrolled and received either ketamine or buprenorphine, along with current antidepressant therapy. The primary outcome was change in depressive symptoms assessed using the Beck Depression Inventory (BDI) after 2 hours, 24 hours, and 7 days following initiation of treatment. Secondary outcomes included changes in suicidal ideation, evaluated by the Beck Scale for Suicidal Ideation (BSSI). RESULTS Both groups experienced a significant decrease in the severity of depression following the interventions (p < .05). However, there was no significant difference in the between-group comparison (p > .05). Both groups also exhibited a significant reduction in suicidal ideation compared to before the study, with the decrease in severity being over 85% in both groups (p < .05). CONCLUSION Both ketamine and buprenorphine appear to be equally effective in reducing symptoms of depression and suicidal ideation among individuals with MDD and OUD.
Collapse
Affiliation(s)
- Jamshid Ahmadi
- Substance Abuse Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- IMCES (Institute for Multicultural Counseling and Educational Services), Los Angeles, CA, USA
| | - Arash Mansoori
- Research Center for Psychiatry and Behavioral Science, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Hamdollah Mosavat
- Research Center for Traditional Medicine and History of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Bazrafshan
- Substance Abuse Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
8
|
Das K, Sen J, Borode AS. Ketamine and α-Amino-3-Hydroxy-5-Methyl-4-Isoxazolepropionic Acid (AMPA) Receptor Potentiation in the Somatosensory Cortex: A Comprehensive Review. Cureus 2024; 16:e69261. [PMID: 39398836 PMCID: PMC11470829 DOI: 10.7759/cureus.69261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024] Open
Abstract
Ketamine, a dissociative anesthetic primarily recognized for its antagonism of N-methyl-D-aspartate (NMDA) receptors, has gained significant attention for its rapid antidepressant effects and potential in treating mood disorders. However, recent research indicates that ketamine's influence extends beyond NMDA receptor inhibition, affecting α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors and sensory processing. This review delves into ketamine's role in enhancing AMPA receptor function and its implications for sensory processing within the somatosensory cortex. AMPA receptors, essential for fast excitatory neurotransmission and synaptic plasticity, play a key role in sensory perception and integration. By examining preclinical and clinical studies, this review sheds light on how ketamine's modulation of AMPA receptors may improve sensory processing and contribute to its therapeutic effects. Additionally, the review explores the potential for ketamine-based therapies to treat sensory processing disorders and refine current treatment strategies. A deeper understanding of ketamine's complex effects on AMPA receptors and sensory processing could provide valuable insights for developing targeted interventions and advancing clinical applications.
Collapse
Affiliation(s)
- Kaustuv Das
- Anaesthesiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Jayshree Sen
- Anaesthesiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Aishwarya S Borode
- Anaesthesiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
9
|
Pettorruso M, Di Lorenzo G, Benatti B, d’Andrea G, Cavallotto C, Carullo R, Mancusi G, Di Marco O, Mammarella G, D’Attilio A, Barlocci E, Rosa I, Cocco A, Padula LP, Bubbico G, Perrucci MG, Guidotti R, D’Andrea A, Marzetti L, Zoratto F, Dell’Osso BM, Martinotti G. Overcoming treatment-resistant depression with machine-learning based tools: a study protocol combining EEG and clinical data to personalize glutamatergic and brain stimulation interventions (SelecTool Project). Front Psychiatry 2024; 15:1436006. [PMID: 39086731 PMCID: PMC11288917 DOI: 10.3389/fpsyt.2024.1436006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Treatment-Resistant Depression (TRD) poses a substantial health and economic challenge, persisting as a major concern despite decades of extensive research into novel treatment modalities. The considerable heterogeneity in TRD's clinical manifestations and neurobiological bases has complicated efforts toward effective interventions. Recognizing the need for precise biomarkers to guide treatment choices in TRD, herein we introduce the SelecTool Project. This initiative focuses on developing (WorkPlane 1/WP1) and conducting preliminary validation (WorkPlane 2/WP2) of a computational tool (SelecTool) that integrates clinical data, neurophysiological (EEG) and peripheral (blood sample) biomarkers through a machine-learning framework designed to optimize TRD treatment protocols. The SelecTool project aims to enhance clinical decision-making by enabling the selection of personalized interventions. It leverages multi-modal data analysis to navigate treatment choices towards two validated therapeutic options for TRD: esketamine nasal spray (ESK-NS) and accelerated repetitive Transcranial Magnetic Stimulation (arTMS). In WP1, 100 subjects with TRD will be randomized to receive either ESK-NS or arTMS, with comprehensive evaluations encompassing neurophysiological (EEG), clinical (psychometric scales), and peripheral (blood samples) assessments both at baseline (T0) and one month post-treatment initiation (T1). WP2 will utilize the data collected in WP1 to train the SelecTool algorithm, followed by its application in a second, out-of-sample cohort of 20 TRD subjects, assigning treatments based on the tool's recommendations. Ultimately, this research seeks to revolutionize the treatment of TRD by employing advanced machine learning strategies and thorough data analysis, aimed at unraveling the complex neurobiological landscape of depression. This effort is expected to provide pivotal insights that will promote the development of more effective and individually tailored treatment strategies, thus addressing a significant void in current TRD management and potentially reducing its profound societal and economic burdens.
Collapse
Affiliation(s)
- Mauro Pettorruso
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D’Annunzio, Chieti, Italy
- Department of Mental Health, ASL02 Lanciano-Vasto-Chieti, Chieti, Italy
- Institute for Advanced Biomedical Technologies (ITAB), “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Giorgio Di Lorenzo
- Laboratory of Psychophysiology and Cognitive Neuroscience, Chair of Psychiatry, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Institute of Hospitalization and Care With Scientific Character (IRCCS) Fondazione Santa Lucia, Rome, Italy
| | - Beatrice Benatti
- Department of Biomedical and Clinical Sciences Luigi Sacco and Aldo Ravelli Center for Neurotechnology and Brain Therapeutic, University of Milan, Milano, Italy
| | - Giacomo d’Andrea
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D’Annunzio, Chieti, Italy
- Department of Mental Health, ASL02 Lanciano-Vasto-Chieti, Chieti, Italy
| | - Clara Cavallotto
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D’Annunzio, Chieti, Italy
| | - Rosalba Carullo
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D’Annunzio, Chieti, Italy
| | - Gianluca Mancusi
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D’Annunzio, Chieti, Italy
| | - Ornella Di Marco
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D’Annunzio, Chieti, Italy
| | - Giovanna Mammarella
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D’Annunzio, Chieti, Italy
| | - Antonio D’Attilio
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D’Annunzio, Chieti, Italy
| | - Elisabetta Barlocci
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D’Annunzio, Chieti, Italy
| | - Ilenia Rosa
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D’Annunzio, Chieti, Italy
| | - Alessio Cocco
- Department of Mental Health, ASL02 Lanciano-Vasto-Chieti, Chieti, Italy
| | - Lorenzo Pio Padula
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D’Annunzio, Chieti, Italy
| | - Giovanna Bubbico
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D’Annunzio, Chieti, Italy
| | - Mauro Gianni Perrucci
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D’Annunzio, Chieti, Italy
- Institute for Advanced Biomedical Technologies (ITAB), “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Roberto Guidotti
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D’Annunzio, Chieti, Italy
- Department of Mental Health, ASL02 Lanciano-Vasto-Chieti, Chieti, Italy
- Institute for Advanced Biomedical Technologies (ITAB), “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Antea D’Andrea
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D’Annunzio, Chieti, Italy
| | - Laura Marzetti
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D’Annunzio, Chieti, Italy
- Institute for Advanced Biomedical Technologies (ITAB), “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Francesca Zoratto
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Bernardo Maria Dell’Osso
- Department of Biomedical and Clinical Sciences Luigi Sacco and Aldo Ravelli Center for Neurotechnology and Brain Therapeutic, University of Milan, Milano, Italy
| | - Giovanni Martinotti
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D’Annunzio, Chieti, Italy
- Department of Mental Health, ASL02 Lanciano-Vasto-Chieti, Chieti, Italy
- Institute for Advanced Biomedical Technologies (ITAB), “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
- Psychopharmacology, Drug Misuse and Novel Psychoactive Substances Research Unit, School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| |
Collapse
|
10
|
Gokalp D, Unal G. The role of mGluR5 on the therapeutic effects of ketamine in Wistar rats. Psychopharmacology (Berl) 2024; 241:1399-1415. [PMID: 38459971 PMCID: PMC11199271 DOI: 10.1007/s00213-024-06571-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
RATIONALE Ketamine produces dissociative, psychomimetic, anxiolytic, antidepressant, and anesthetic effects in a dose dependent manner. It has a complex mechanism of action that involve alterations in other glutamate receptors. The metabotropic glutamate receptor 5 (mGluR5) has been investigated in relation to the psychotic and anesthetic properties of ketamine, while its role in mediating the therapeutic effects of ketamine remains unknown. OBJECTIVES We investigated the role of mGluR5 on the antidepressant, anxiolytic and fear memory-related effects of ketamine in adult male Wistar rats. METHODS Two sets of experiments were conducted. We first utilized the positive allosteric modulator CDPPB to investigate how acute mGluR5 activation regulates the therapeutic effects of ketamine (10 mg/kg). We then tested the synergistic antidepressant effect of mGluR5 antagonism and ketamine by combining MTEP with a sub-effective dose of ketamine (1 mg/kg). Behavioral despair, locomotor activity, anxiety-like behavior, and fear memory were respectively assessed in the forced swim test (FST), open field test (OFT), elevated plus maze (EPM), and auditory fear conditioning. RESULTS Enhancing mGluR5 activity via CDPPB occluded the antidepressant effect of ketamine without changing locomotor activity. Furthermore, concomitant administration of MTEP and ketamine exhibited a robust synergistic antidepressant effect. The MTEP + ketamine treatment, however, blocked the anxiolytic effect observed by sole administration of MTEP or the low dose ketamine. CONCLUSIONS These findings suggest that suppressed mGluR5 activity is required for the antidepressant effects of ketamine. Consequently, the antagonism of mGluR5 enhances the antidepressant effectiveness of low dose ketamine, but eliminates its anxiolytic effects.
Collapse
Affiliation(s)
- Dilan Gokalp
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, 34342, Istanbul, Turkey
| | - Gunes Unal
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, 34342, Istanbul, Turkey.
| |
Collapse
|
11
|
Juneja K, Afroze S, Goti Z, Sahu S, Asawa S, Bhuchakra HP, Natarajan B. Beyond therapeutic potential: a systematic investigation of ketamine misuse in patients with depressive disorders. DISCOVER MENTAL HEALTH 2024; 4:23. [PMID: 38951348 PMCID: PMC11217219 DOI: 10.1007/s44192-024-00077-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 06/17/2024] [Indexed: 07/03/2024]
Abstract
Ketamine, a pharmacological agent that acts as an antagonist of the N-methyl-D-aspartate (NMDA) receptor, has garnered considerable interest because of its notable and expeditious antidepressant properties observed in individuals diagnosed with major depressive disorder (MDD) who exhibit resistance to conventional therapeutic interventions. A comprehensive and rigorous systematic review was undertaken to evaluate the prevalence of ketamine abuse undergoing ketamine treatment for depressive disorders. A comprehensive search was conducted across the electronic databases to identify pertinent studies published between 2021 and 2023. The present investigation incorporated a comprehensive range of studies encompassing the abuse or misuse of ketamine, including case reports, observational studies, and clinical trials. Data extraction and quality assessment were conducted in accordance with predetermined criteria. The findings of this systematic review demonstrate the importance of monitoring and addressing ketamine abuse in patients receiving ketamine treatment for depressive disorders like MDD. The wide range of reported prevalence rates highlights the need for standardized criteria and measures for defining and assessing ketamine abuse. This study presents a significant contribution to the field by introducing a novel screening questionnaire and assessment algorithm designed to identify and evaluate ketamine misuse among major depressive disorder (MDD) patients undergoing ketamine treatment. This innovative tool holds the potential to enhance clinical practice by providing healthcare professionals with a standardized approach to promptly detect and address ketamine misuse. The integration of this screening tool into routine care protocols can facilitate more effective monitoring and management of ketamine misuse in this population, ultimately leading to improved patient outcomes and safety.
Collapse
Affiliation(s)
| | - Sabah Afroze
- Shadan Hospital and Institute of Medical Sciences, Hyderabad, India
| | - Zeel Goti
- Government Medical College, Surat, India
| | | | | | | | | |
Collapse
|
12
|
Adam E, Kowalski M, Akeju O, Miller EK, Brown EN, McCarthy MM, Kopell N. Ketamine can produce oscillatory dynamics by engaging mechanisms dependent on the kinetics of NMDA receptors. Proc Natl Acad Sci U S A 2024; 121:e2402732121. [PMID: 38768339 PMCID: PMC11145256 DOI: 10.1073/pnas.2402732121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/22/2024] [Indexed: 05/22/2024] Open
Abstract
Ketamine is an N-methyl-D-aspartate (NMDA)-receptor antagonist that produces sedation, analgesia, and dissociation at low doses and profound unconsciousness with antinociception at high doses. At high and low doses, ketamine can generate gamma oscillations (>25 Hz) in the electroencephalogram (EEG). The gamma oscillations are interrupted by slow-delta oscillations (0.1 to 4 Hz) at high doses. Ketamine's primary molecular targets and its oscillatory dynamics have been characterized. However, how the actions of ketamine at the subcellular level give rise to the oscillatory dynamics observed at the network level remains unknown. By developing a biophysical model of cortical circuits, we demonstrate how NMDA-receptor antagonism by ketamine can produce the oscillatory dynamics observed in human EEG recordings and nonhuman primate local field potential recordings. We have identified how impaired NMDA-receptor kinetics can cause disinhibition in neuronal circuits and how a disinhibited interaction between NMDA-receptor-mediated excitation and GABA-receptor-mediated inhibition can produce gamma oscillations at high and low doses, and slow-delta oscillations at high doses. Our work uncovers general mechanisms for generating oscillatory brain dynamics that differs from ones previously reported and provides important insights into ketamine's mechanisms of action as an anesthetic and as a therapy for treatment-resistant depression.
Collapse
Affiliation(s)
- Elie Adam
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA02114
| | - Marek Kowalski
- Department of Mathematics and Statistics, Boston University, Boston, MA02215
| | - Oluwaseun Akeju
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA02114
- Department of Anesthesia, Harvard Medical School, Boston, MA02215
| | - Earl K. Miller
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Emery N. Brown
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA02114
- Department of Anesthesia, Harvard Medical School, Boston, MA02215
| | | | - Nancy Kopell
- Department of Mathematics and Statistics, Boston University, Boston, MA02215
| |
Collapse
|
13
|
Adam E, Kowalski M, Akeju O, Miller EK, Brown EN, McCarthy MM, Kopell N. Ketamine can produce oscillatory dynamics by engaging mechanisms dependent on the kinetics of NMDA receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587998. [PMID: 38617266 PMCID: PMC11014619 DOI: 10.1101/2024.04.03.587998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Ketamine is an NMDA-receptor antagonist that produces sedation, analgesia and dissociation at low doses and profound unconsciousness with antinociception at high doses. At high and low doses, ketamine can generate gamma oscillations (>25 Hz) in the electroencephalogram (EEG). The gamma oscillations are interrupted by slow-delta oscillations (0.1-4 Hz) at high doses. Ketamine's primary molecular targets and its oscillatory dynamics have been characterized. However, how the actions of ketamine at the subcellular level give rise to the oscillatory dynamics observed at the network level remains unknown. By developing a biophysical model of cortical circuits, we demonstrate how NMDA-receptor antagonism by ketamine can produce the oscillatory dynamics observed in human EEG recordings and non-human primate local field potential recordings. We have discovered how impaired NMDA-receptor kinetics can cause disinhibition in neuronal circuits and how a disinhibited interaction between NMDA-receptor-mediated excitation and GABA-receptor-mediated inhibition can produce gamma oscillations at high and low doses, and slow-delta oscillations at high doses. Our work uncovers general mechanisms for generating oscillatory brain dynamics that differs from ones previously reported, and provides important insights into ketamine's mechanisms of action as an anesthetic and as a therapy for treatment-resistant depression.
Collapse
Affiliation(s)
- Elie Adam
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114
| | - Marek Kowalski
- Department of Mathematics and Statistics, Boston University, Boston, MA 02215
| | - Oluwaseun Akeju
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114
- Department of Anesthesia, Harvard Medical School, Boston, MA 02215
| | - Earl K. Miller
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Emery N. Brown
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114
- Department of Anesthesia, Harvard Medical School, Boston, MA 02215
| | | | - Nancy Kopell
- Department of Mathematics and Statistics, Boston University, Boston, MA 02215
| |
Collapse
|
14
|
Uzungil V, Luza S, Opazo CM, Mees I, Li S, Ang CS, Williamson NA, Bush AI, Hannan AJ, Renoir T. Phosphoproteomics implicates glutamatergic and dopaminergic signalling in the antidepressant-like properties of the iron chelator deferiprone. Neuropharmacology 2024; 246:109837. [PMID: 38184274 DOI: 10.1016/j.neuropharm.2024.109837] [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: 08/29/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/08/2024]
Abstract
BACKGROUND Current antidepressants have limitations due to insufficient efficacy and delay before improvement in symptoms. Polymorphisms of the serotonin transporter (5-HTT) gene have been linked to depression (when combined with stressful life events) and altered response to selective serotonergic reuptake inhibitors. We have previously revealed the antidepressant-like properties of the iron chelator deferiprone in the 5-HTT knock-out (KO) mouse model of depression. Furthermore, deferiprone was found to alter neural activity in the prefrontal cortex of both wild-type (WT) and 5-HTT KO mice. METHODS In the current study, we examined the molecular effects of acute deferiprone treatment in the prefrontal cortex of both genotypes via phosphoproteomics analysis. RESULTS In WT mice treated with deferiprone, there were 22 differentially expressed phosphosites, with gene ontology analysis implicating cytoskeletal proteins. In 5-HTT KO mice treated with deferiprone, we found 33 differentially expressed phosphosites. Gene ontology analyses revealed phosphoproteins that were predominantly involved in synaptic and glutamatergic signalling. In a drug-naïve cohort (without deferiprone administration), the analysis revealed 21 differentially expressed phosphosites in 5-HTT KO compared to WT mice. We confirmed the deferiprone-induced increase in tyrosine hydroxylase serine 40 residue phosphorylation (pTH-Ser40) (initially revealed in our phosphoproteomics study) by Western blot analysis, with deferiprone increasing pTH-Ser40 expression in WT and 5-HTT KO mice. CONCLUSION As glutamatergic and synaptic signalling are dysfunctional in 5-HTT KO mice (and are the target of fast-acting antidepressant drugs such as ketamine), these molecular effects may underpin deferiprone's antidepressant-like properties. Furthermore, dopaminergic signalling may also be involved in deferiprone's antidepressant-like properties.
Collapse
Affiliation(s)
- Volkan Uzungil
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Sandra Luza
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia; Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
| | - Carlos M Opazo
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Isaline Mees
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Shanshan Li
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Ching-Seng Ang
- Bio21 Mass Spectrometry and Proteomics Facility, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Nicholas A Williamson
- Bio21 Mass Spectrometry and Proteomics Facility, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Anthony J Hannan
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
| | - Thibault Renoir
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia.
| |
Collapse
|
15
|
Kotzalidis GD, Fiaschè F, Alcibiade A, Monti L, Di Segni F, Mazza M, Sani G. New pharmacotherapies to tackle the unmet needs in bipolar disorder: a focus on acute suicidality. Expert Opin Pharmacother 2024; 25:435-446. [PMID: 38517222 DOI: 10.1080/14656566.2024.2334425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/20/2024] [Indexed: 03/23/2024]
Abstract
INTRODUCTION Suicidal behavior is relatively frequent in patients with bipolar disorder (BD) and constitutes their most frequent cause of death. Suicide rates remain high in patients with BD despite adherence to guidelines recommending lithium as first line, and/or antidepressants, antipsychotics, psychotherapy, psychosocial interventions, and electroconvulsive therapy. Hence the need to identify more effective and rapid anti-suicide interventions. AREAS COVERED To tackle the unmet needs of pharmacotherapy, we investigated the PubMed database on 24-25 January 2024 using strategies like ('acute suicid*'[ti] OR 'suicide crisis syndrome' OR 'acute suicidal affective disturbance') AND (lithium[ti] OR clozapine[ti]), which obtained 3 results, and ('acute suicid*'[ti] OR 'suicide crisis syndrome' OR 'acute suicidal affective disturbance') AND (ketamine[ti] OR esketamine[ti] OR NMDA[ti] OR glutamat*[ti]), which yielded 14 results. We explored glutamatergic abnormalities in BD and suicide and found alterations in both. The noncompetitive NMDS antagonist ketamine and its S-enantiomer esketamine reportedly decrease acute suicidality. EXPERT OPINION Intranasal esketamine or subcutaneous ketamine, single-bolus or intravenous, and possibly other glutamate receptor modulators may improve suicidal behavior in patients with unipolar and bipolar depression. This may be achieved through prompt remodulation of glutamate activity. The correct use of glutamatergic modulators could reduce acute suicidality and mortality in patients with BD.
Collapse
Affiliation(s)
- Georgios D Kotzalidis
- Department of Neurosciences, Mental Health, and Sensory Organs (NESMOS), Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy
- Department of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Federica Fiaschè
- ASL RM1, Presidio Ospedaliero San Filippo Neri, Servizio Psichiatrico di Diagnosi e Cura, Rome, Italy
| | - Alessandro Alcibiade
- Marina Militare Italiana (Italian Navy), Defense Ministry of Italy, Rome, Italy
- Psychiatry Residency Training Programme, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Laura Monti
- UOS Clinical Psychology, Clinical Government, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Federica Di Segni
- Psychiatry Residency Training Programme, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Marianna Mazza
- Department of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Gabriele Sani
- Department of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Neuroscience, Section of Psychiatry, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
16
|
Teng Y, Niu J, Liu Y, Wang H, Chen J, Kong Y, Wang L, Lian B, Wang W, Sun H, Yue K. Ketamine alleviates fear memory and spatial cognition deficits in a PTSD rat model via the BDNF signaling pathway of the hippocampus and amygdala. Behav Brain Res 2024; 459:114792. [PMID: 38048914 DOI: 10.1016/j.bbr.2023.114792] [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: 10/07/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/06/2023]
Abstract
BACKGROUND Post-traumatic stress disorder (PTSD) is associated with traumatic stress experiences. This condition can be accompanied by learning and cognitive deficits. Studies have demonstrated that ketamine can rapidly and significantly alleviate symptoms in patients with chronic PTSD. Nonetheless, the effects of ketamine on neurocognitive impairment and its mechanism of action in PTSD remain unclear. METHODS In this study, different concentrations of ketamine (5, 10, 15, and 20 mg/kg, i.p.) were evaluated in rat models of single prolonged stress and electrophonic shock (SPS&S). Expression levels of brain-derived neurotrophic factor (BDNF) and post-synaptic density-95 (PSD-95) in the hippocampus (HIP) and amygdala (AMG) were determined by Western blot analysis and immunohistochemistry. RESULTS The data showed that rats subjected to SPS&S exhibited significant PTSD-like cognitive impairment. The effect of ketamine on SPS&S-induced neurocognitive function showed a U-shaped dose effect in rats. A single administration of ketamine at a dosage of 10-15 mg/kg resulted in significant changes in behavioral outcomes. These manifestations of improvement in cognitive function and molecular changes were reversed at high doses (15-20 mg/kg). CONCLUSION Overall, ketamine reversed SPS&S-induced fear and spatial memory impairment and the down-regulation of BDNF and BDNF-related PSD-95 signaling in the HIP and AMG. A dose equal to 15 mg/kg rapidly reversed the behavioral and molecular changes and promoted the amelioration of cognitive dysfunction. The enhanced association of BDNF signaling with PSD-95 effects could be involved in the therapeutic efficiency of ketamine for PTSD.
Collapse
Affiliation(s)
- Yue Teng
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - JiaYao Niu
- School of Clinical Medicine, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Yang Liu
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Han Wang
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - JinHong Chen
- School of Continuing Education, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - YuJia Kong
- School of Public Health, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Ling Wang
- Clinical Competency Training Center, Medical experiment and training center, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Bo Lian
- Department of Bioscience and Technology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - WeiWen Wang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100864, PR China
| | - HongWei Sun
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China.
| | - KuiTao Yue
- The Medical imaging Center, Affiliated Hospital of Weifang Medical University, 2428# Yuhe Road, Weifang, Shandong 261053, PR China.
| |
Collapse
|
17
|
Viudez-Martínez A, Torregrosa AB, Navarrete F, García-Gutiérrez MS. Understanding the Biological Relationship between Migraine and Depression. Biomolecules 2024; 14:163. [PMID: 38397400 PMCID: PMC10886628 DOI: 10.3390/biom14020163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/25/2024] Open
Abstract
Migraine is a highly prevalent neurological disorder. Among the risk factors identified, psychiatric comorbidities, such as depression, seem to play an important role in its onset and clinical course. Patients with migraine are 2.5 times more likely to develop a depressive disorder; this risk becomes even higher in patients suffering from chronic migraine or migraine with aura. This relationship is bidirectional, since depression also predicts an earlier/worse onset of migraine, increasing the risk of migraine chronicity and, consequently, requiring a higher healthcare expenditure compared to migraine alone. All these data suggest that migraine and depression may share overlapping biological mechanisms. Herein, this review explores this topic in further detail: firstly, by introducing the common epidemiological and risk factors for this comorbidity; secondly, by focusing on providing the cumulative evidence of common biological aspects, with a particular emphasis on the serotoninergic system, neuropeptides such as calcitonin-gene-related peptide (CGRP), pituitary adenylate cyclase-activating polypeptide (PACAP), substance P, neuropeptide Y and orexins, sexual hormones, and the immune system; lastly, by remarking on the future challenges required to elucidate the etiopathological mechanisms of migraine and depression and providing updated information regarding new key targets for the pharmacological treatment of these clinical entities.
Collapse
Affiliation(s)
- Adrián Viudez-Martínez
- Hospital Pharmacy Service, Hospital General Dr. Balmis de Alicante, 03010 Alicante, Spain;
| | - Abraham B. Torregrosa
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - María Salud García-Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| |
Collapse
|
18
|
Yonezawa K, Uchida H, Yatomi T, Ohtani Y, Nomoto-Takahashi K, Nakajima S, Mimura M, Tani H. Factors Associated with Antidepressant Effects of Ketamine: A Reanalysis of Double-Blind Randomized Placebo-Controlled Trial of Intravenous Ketamine for Treatment-Resistant Depression. PHARMACOPSYCHIATRY 2024; 57:35-40. [PMID: 37846462 DOI: 10.1055/a-2179-8884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
INTRODUCTION Predictors of treatment response to intravenous ketamine remain unclear in patients with treatment-resistant depression (TRD); therefore, this study aimed to clarify these predictors using the US National Institutes of Health database of clinical trials. METHODS Data from a placebo-controlled, double-blind, randomized controlled trial were used to assess the efficacy of intravenous ketamine in adult patients with TRD (NCT01920555). For the analysis, data were used from the participants who had received therapeutic doses of intravenous ketamine (i. e., 0.5 and 1.0 mg/kg). Logistic and multivariable regression analyses were conducted to explore the demographic and clinical factors associated with response to treatment or changes in the Hamilton Depression Rating Scale 6 items (HAM-D-6) total score. RESULTS This study included 31 patients with TRD (13 women; mean±standard deviation age, 48.4±10.9 years). Logistic regression analysis showed that the age of onset was positively correlated with treatment response after three days of ketamine administration (β=0.08, p=0.037); however, no association was observed between treatment response and age, sex, baseline HAM-D-6 total score, or dissociative score assessed with the Clinician-Administered Dissociative States Scale 40 min after ketamine infusion. Multiple regression analysis showed that no factors were correlated significantly with the percentage change in the HAM-D-6 total score three days after ketamine administration. DISCUSSION Later disease onset correlates with a better treatment response three days after ketamine infusion in patients with TRD. Glutamatergic signal transmission may be impaired in patients with an earlier onset of depression, resulting in decreased neuroplasticity, which diminishes ketamine response.
Collapse
Affiliation(s)
- Kengo Yonezawa
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Hiroyuki Uchida
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Taisuke Yatomi
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Yohei Ohtani
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | | | - Shinichiro Nakajima
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Masaru Mimura
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Hideaki Tani
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
19
|
Zaretsky TG, Jagodnik KM, Barsic R, Antonio JH, Bonanno PA, MacLeod C, Pierce C, Carney H, Morrison MT, Saylor C, Danias G, Lepow L, Yehuda R. The Psychedelic Future of Post-Traumatic Stress Disorder Treatment. Curr Neuropharmacol 2024; 22:636-735. [PMID: 38284341 PMCID: PMC10845102 DOI: 10.2174/1570159x22666231027111147] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 01/30/2024] Open
Abstract
Post-traumatic stress disorder (PTSD) is a mental health condition that can occur following exposure to a traumatic experience. An estimated 12 million U.S. adults are presently affected by this disorder. Current treatments include psychological therapies (e.g., exposure-based interventions) and pharmacological treatments (e.g., selective serotonin reuptake inhibitors (SSRIs)). However, a significant proportion of patients receiving standard-of-care therapies for PTSD remain symptomatic, and new approaches for this and other trauma-related mental health conditions are greatly needed. Psychedelic compounds that alter cognition, perception, and mood are currently being examined for their efficacy in treating PTSD despite their current status as Drug Enforcement Administration (DEA)- scheduled substances. Initial clinical trials have demonstrated the potential value of psychedelicassisted therapy to treat PTSD and other psychiatric disorders. In this comprehensive review, we summarize the state of the science of PTSD clinical care, including current treatments and their shortcomings. We review clinical studies of psychedelic interventions to treat PTSD, trauma-related disorders, and common comorbidities. The classic psychedelics psilocybin, lysergic acid diethylamide (LSD), and N,N-dimethyltryptamine (DMT) and DMT-containing ayahuasca, as well as the entactogen 3,4-methylenedioxymethamphetamine (MDMA) and the dissociative anesthetic ketamine, are reviewed. For each drug, we present the history of use, psychological and somatic effects, pharmacology, and safety profile. The rationale and proposed mechanisms for use in treating PTSD and traumarelated disorders are discussed. This review concludes with an in-depth consideration of future directions for the psychiatric applications of psychedelics to maximize therapeutic benefit and minimize risk in individuals and communities impacted by trauma-related conditions.
Collapse
Affiliation(s)
- Tamar Glatman Zaretsky
- James J. Peters Veterans Affairs Medical Center, New York, NY, USA
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kathleen M. Jagodnik
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Barsic
- James J. Peters Veterans Affairs Medical Center, New York, NY, USA
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Josimar Hernandez Antonio
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Philip A. Bonanno
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carolyn MacLeod
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Charlotte Pierce
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hunter Carney
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Morgan T. Morrison
- James J. Peters Veterans Affairs Medical Center, New York, NY, USA
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Charles Saylor
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - George Danias
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lauren Lepow
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rachel Yehuda
- James J. Peters Veterans Affairs Medical Center, New York, NY, USA
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
20
|
Cao YY, Wu LL, Li XN, Yuan YL, Zhao WW, Qi JX, Zhao XY, Ward N, Wang J. Molecular Mechanisms of AMPA Receptor Trafficking in the Nervous System. Int J Mol Sci 2023; 25:111. [PMID: 38203282 PMCID: PMC10779435 DOI: 10.3390/ijms25010111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Synaptic plasticity enhances or reduces connections between neurons, affecting learning and memory. Postsynaptic AMPARs mediate greater than 90% of the rapid excitatory synaptic transmission in glutamatergic neurons. The number and subunit composition of AMPARs are fundamental to synaptic plasticity and the formation of entire neural networks. Accordingly, the insertion and functionalization of AMPARs at the postsynaptic membrane have become a core issue related to neural circuit formation and information processing in the central nervous system. In this review, we summarize current knowledge regarding the related mechanisms of AMPAR expression and trafficking. The proteins related to AMPAR trafficking are discussed in detail, including vesicle-related proteins, cytoskeletal proteins, synaptic proteins, and protein kinases. Furthermore, significant emphasis was placed on the pivotal role of the actin cytoskeleton, which spans throughout the entire transport process in AMPAR transport, indicating that the actin cytoskeleton may serve as a fundamental basis for AMPAR trafficking. Additionally, we summarize the proteases involved in AMPAR post-translational modifications. Moreover, we provide an overview of AMPAR transport and localization to the postsynaptic membrane. Understanding the assembly, trafficking, and dynamic synaptic expression mechanisms of AMPAR may provide valuable insights into the cognitive decline associated with neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi-Yang Cao
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Ling-Ling Wu
- School of Medicine, Shanghai University, Shanghai 200444, China;
| | - Xiao-Nan Li
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Yu-Lian Yuan
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Wan-Wei Zhao
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Jing-Xuan Qi
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Xu-Yu Zhao
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Natalie Ward
- Medical Laboratory, Exceptional Community Hospital, 19060 N John Wayne Pkwy, Maricopa, AZ 85139, USA;
| | - Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| |
Collapse
|
21
|
Osaka H, Kanazawa T. Emerging trends in antipsychotic and antidepressant drug development: Targeting nonmonoamine receptors and innovative mechanisms. PCN REPORTS : PSYCHIATRY AND CLINICAL NEUROSCIENCES 2023; 2:e157. [PMID: 38868733 PMCID: PMC11114387 DOI: 10.1002/pcn5.157] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/26/2023] [Accepted: 10/30/2023] [Indexed: 06/14/2024]
Abstract
The domain of psychiatric drug development is currently witnessing a notable transformation, with a paramount emphasis on targeting nonmonoamine receptors and exploring inventive mechanisms of action. This paper presents an overview of the ongoing advancements in antipsychotic and antidepressant drug development. Historically, antipsychotics predominantly targeted dopamine receptors, but there is now an escalating interest in drugs that act on alternative receptors, exemplified by the TAAR1 receptor. One noteworthy candidate is Ulotaront (SEP-363856), an agent acting as a TAAR1 agonist with 5-HT1A agonist activity, demonstrating promising outcomes in the treatment of schizophrenia, devoid of extrapyramidal symptoms or metabolic side-effects. Similarly, MIN-101 (Roluperidone) and KarXT are currently in development, with its focus on addressing the symptoms in schizophrenia. In the domain of antidepressants, novel therapeutic approaches have surfaced, such as Auvelity, a Food and Drug Administration (FDA)-approved NMDA receptor antagonist synergistically combined with Bupropion to enhance its effects. Another notable candidate is Zuranolone, operating as a GABA A receptor-positive allosteric modulator, showcasing efficacy in treating major depressive disorder (MDD) and postpartum depression. Additionally, TAK-653 (NBI-1065845) and MJI821 (Onfasprodil) have emerged as potential antidepressants targeting AMPA receptors and NMDA receptor 2B (NR2B) negative allosteric modulation, respectively. This paper underscores the transformative potential of these novel drug candidates in psychiatric treatment and their ability to address cases that were previously treatment-resistant. By focusing on nonmonoamine receptors and introducing innovative mechanisms, these drugs offer a promising prospect of improved outcomes for individuals suffering from schizophrenia and MDD. Thus, sustained attention and dedication to the development of such drugs are essential to augmenting the therapeutic options available for psychiatric patients.
Collapse
Affiliation(s)
- Hitoshi Osaka
- Department of NeuropsychiatryOsaka Medical and Pharmaceutical UniversityTakatsuki‐cityOsakaJapan
| | - Tetsufumi Kanazawa
- Department of NeuropsychiatryOsaka Medical and Pharmaceutical UniversityTakatsuki‐cityOsakaJapan
| |
Collapse
|
22
|
Bottemanne H, Berkovitch L, Gauld C, Balcerac A, Schmidt L, Mouchabac S, Fossati P. Storm on predictive brain: A neurocomputational account of ketamine antidepressant effect. Neurosci Biobehav Rev 2023; 154:105410. [PMID: 37793581 DOI: 10.1016/j.neubiorev.2023.105410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 08/24/2023] [Accepted: 09/26/2023] [Indexed: 10/06/2023]
Abstract
For the past decade, ketamine, an N-methyl-D-aspartate receptor (NMDAr) antagonist, has been considered a promising treatment for major depressive disorder (MDD). Unlike the delayed effect of monoaminergic treatment, ketamine may produce fast-acting antidepressant effects hours after a single administration at subanesthetic dose. Along with these antidepressant effects, it may also induce transient dissociative (disturbing of the sense of self and reality) symptoms during acute administration which resolve within hours. To understand ketamine's rapid-acting antidepressant effect, several biological hypotheses have been explored, but despite these promising avenues, there is a lack of model to understand the timeframe of antidepressant and dissociative effects of ketamine. In this article, we propose a neurocomputational account of ketamine's antidepressant and dissociative effects based on the Predictive Processing (PP) theory, a framework for cognitive and sensory processing. PP theory suggests that the brain produces top-down predictions to process incoming sensory signals, and generates bottom-up prediction errors (PEs) which are then used to update predictions. This iterative dynamic neural process would relies on N-methyl-D-aspartate (NMDAr) and α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic receptors (AMPAr), two major component of the glutamatergic signaling. Furthermore, it has been suggested that MDD is characterized by over-rigid predictions which cannot be updated by the PEs, leading to miscalibration of hierarchical inference and self-reinforcing negative feedback loops. Based on former empirical studies using behavioral paradigms, neurophysiological recordings, and computational modeling, we suggest that ketamine impairs top-down predictions by blocking NMDA receptors, and enhances presynaptic glutamate release and PEs, producing transient dissociative symptoms and fast-acting antidepressant effect in hours following acute administration. Moreover, we present data showing that ketamine may enhance a delayed neural plasticity pathways through AMPAr potentiation, triggering a prolonged antidepressant effect up to seven days for unique administration. Taken together, the two sides of antidepressant effects with distinct timeframe could constitute the keystone of antidepressant properties of ketamine. These PP disturbances may also participate to a ketamine-induced time window of mental flexibility, which can be used to improve the psychotherapeutic process. Finally, these proposals could be used as a theoretical framework for future research into fast-acting antidepressants, and combination with existing antidepressant and psychotherapy.
Collapse
Affiliation(s)
- Hugo Bottemanne
- Paris Brain Institute - Institut du Cerveau (ICM), UMR 7225 / UMRS 1127, Sorbonne University / CNRS / INSERM, Paris, France; Sorbonne University, Department of Philosophy, Science Norm Democracy Research Unit, UMR, 8011, Paris, France; Sorbonne University, Department of Psychiatry, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.
| | - Lucie Berkovitch
- Saclay CEA Centre, Neurospin, Gif-Sur-Yvette Cedex, France; Department of Psychiatry, GHU Paris Psychiatrie et Neurosciences, Service Hospitalo-Universitaire, Paris, France
| | - Christophe Gauld
- Department of Child Psychiatry, CHU de Lyon, F-69000 Lyon, France; Institut des Sciences Cognitives Marc Jeannerod, UMR 5229 CNRS & Université Claude Bernard Lyon 1, F-69000 Lyon, France
| | - Alexander Balcerac
- Paris Brain Institute - Institut du Cerveau (ICM), UMR 7225 / UMRS 1127, Sorbonne University / CNRS / INSERM, Paris, France; Sorbonne University, Department of Neurology, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Liane Schmidt
- Paris Brain Institute - Institut du Cerveau (ICM), UMR 7225 / UMRS 1127, Sorbonne University / CNRS / INSERM, Paris, France
| | - Stephane Mouchabac
- Paris Brain Institute - Institut du Cerveau (ICM), UMR 7225 / UMRS 1127, Sorbonne University / CNRS / INSERM, Paris, France; Sorbonne University, Department of Psychiatry, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Philippe Fossati
- Paris Brain Institute - Institut du Cerveau (ICM), UMR 7225 / UMRS 1127, Sorbonne University / CNRS / INSERM, Paris, France; Sorbonne University, Department of Philosophy, Science Norm Democracy Research Unit, UMR, 8011, Paris, France
| |
Collapse
|
23
|
Rech TDST, Strelow DN, Krüger LD, Neto JSS, Blödorn GB, Alves D, Brüning CA, Bortolatto CF. Pharmacological evidence for glutamatergic pathway involvement in the antidepressant-like effects of 2-phenyl-3-(phenylselanyl)benzofuran in male Swiss mice. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3033-3044. [PMID: 37160481 DOI: 10.1007/s00210-023-02508-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 04/20/2023] [Indexed: 05/11/2023]
Abstract
Depression is a multifactorial and heterogeneous disease with several neurobiological mechanisms underlying its pathophysiology, including dysfunctional glutamatergic neurotransmission, which makes the exploration of the glutamate pathway an interesting strategy for developing novel rapid-acting antidepressant treatments. In the present study, we aimed to evaluate the possible glutamatergic pathway relation in the antidepressant-like action of 2-phenyl-3-(phenylselanyl)benzofuran (SeBZF1) in Swiss mice employing the tail suspension test (TST). Male Swiss mice received drugs targeting glutamate receptors before acute SeBZF1 administration at effective (50 mg/kg) or subeffective (1 mg/kg) doses by intragastric route (ig). TST and the open-field test (OFT) were employed in all behavioral experiments. The pretreatment of mice with N-methyl-D-aspartate (NMDA) (0.1 pmol/site, intracerebroventricular, icv, a selective agonist of the NMDA receptors), D-serine (30 µg/site, icv, a co-agonist at the NMDA receptor), arcaine (1 mg/kg, intraperitoneal, ip, an antagonist of the polyamine-binding site at the NMDA receptor), and 6,7-dinitroquinoxaline-2,3-dione (DNQX) (2,5 µg/site, icv, an antagonist of the AMPA/kainate type of glutamate receptors) inhibited the antidepressant-like effects of SeBZF1 (50 mg/kg, ig) in the TST. Coadministration of a subeffective dose of SeBZF1 with low doses of MK-801 (0.001 mg/kg, ip, a non-competitive NMDA receptor antagonist) or ketamine (0.1 mg/kg, ip, a non-selective antagonist of the NMDA receptors) produced significant antidepressant-like effects (synergistic action). These findings suggest the involvement of the glutamatergic system, probably through modulation of ionotropic glutamate receptors, in the antidepressant-like action of SeBZF1 in mice and contribute to a better understanding of the mechanisms underlying its pharmacological effects.
Collapse
Affiliation(s)
- Taís da Silva Teixeira Rech
- Programa de Pós-Graduação em Bioquímica e Bioprospecção (PPGBBio), Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), RS, CEP 96010-900, Pelotas, Brazil
| | - Dianer Nornberg Strelow
- Programa de Pós-Graduação em Bioquímica e Bioprospecção (PPGBBio), Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), RS, CEP 96010-900, Pelotas, Brazil
| | - Letícia Devantier Krüger
- Programa de Pós-Graduação em Bioquímica e Bioprospecção (PPGBBio), Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), RS, CEP 96010-900, Pelotas, Brazil
| | | | - Gustavo Bierhals Blödorn
- Programa de Pós-Graduação em Química (PPGQ), Laboratório de Síntese Orgânica Limpa (LASOL), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), RS, CEP 96010-900, Pelotas, Brazil
| | - Diego Alves
- Programa de Pós-Graduação em Química (PPGQ), Laboratório de Síntese Orgânica Limpa (LASOL), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), RS, CEP 96010-900, Pelotas, Brazil
| | - César Augusto Brüning
- Programa de Pós-Graduação em Bioquímica e Bioprospecção (PPGBBio), Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), RS, CEP 96010-900, Pelotas, Brazil.
| | - Cristiani Folharini Bortolatto
- Programa de Pós-Graduação em Bioquímica e Bioprospecção (PPGBBio), Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), RS, CEP 96010-900, Pelotas, Brazil.
| |
Collapse
|
24
|
Coffelt CB, Gibson K, VanLandingham J. Ketamine for Traumatic Assault-Induced Depression: A Case Report. J Trauma Nurs 2023; 30:353-356. [PMID: 37937877 DOI: 10.1097/jtn.0000000000000755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
BACKGROUND This case report describes the use of ketamine as a rapid, effective treatment of depression in a 68-year-old female patient with no significant medical history of psychiatric disorders. Patients who experience intentional or unintentional traumas are at an increased risk for developing depression or posttraumatic stress disorder, and emerging evidence has supported the use of ketamine as an alternative treatment of depression. CASE PRESENTATION This is the case of a 68-year-old female patient who was assaulted, resulting in multiple stab wounds to both hands and the right upper quadrant. She underwent placement of a chest tube and surgical repair of the liver and was subsequently admitted to the intensive care unit. These events led to the development of severe depression symptoms, as evidenced by a Montgomery-Asberg Depression Rating Scale (MADRS) score of 37. As treatment of her acute depression, the patient received a single intravenous dose of ketamine (0.5 mg/kg) infused over 40 min and was monitored for side effects. The MADRS is a 10-item depression screening tool that assesses symptoms and changes over time. Within 4 hr of receiving ketamine, the patient reported a significant improvement in her mood and her MADRS score decreased to 16, classifying this patient as experiencing mild depression. The patient continued to improve, and 24 hr after receiving ketamine, her MADRS score was 4, indicating remission of her depression symptoms. CONCLUSION This case report aims to provide an account of the potential benefits of ketamine as a rapid treatment of depression in an adult trauma patient.
Collapse
Affiliation(s)
- Caitlyn B Coffelt
- University of Georgia College of Pharmacy, Athens, Georgia (Dr Coffelt); and Northeast Georgia Medical Center, Gainesville, Georgia (Ms Gibson and Dr VanLandingham)
| | | | | |
Collapse
|
25
|
Dębowska W, Więdłocha M, Dębowska M, Kownacka Z, Marcinowicz P, Szulc A. Transcranial magnetic stimulation and ketamine: implications for combined treatment in depression. Front Neurosci 2023; 17:1267647. [PMID: 37954877 PMCID: PMC10637948 DOI: 10.3389/fnins.2023.1267647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023] Open
Abstract
Drug-resistant mental disorders, particularly treatment-resistant depression, pose a significant medical and social problem. To address this challenge, modern psychiatry is constantly exploring the use of novel treatment methods, including biological treatments, such as transcranial magnetic stimulation (TMS), and novel rapid-acting antidepressants, such as ketamine. While both TMS and ketamine demonstrate high effectiveness in reducing the severity of depressive symptoms, some patients still do not achieve the desired improvement. Recent literature suggests that combining these two methods may yield even stronger and longer-lasting results. This review aims to consolidate knowledge in this area and elucidate the potential mechanisms of action underlying the increased efficacy of combined treatment, which would provide a foundation for the development and optimization of future treatment protocols.
Collapse
Affiliation(s)
- Weronika Dębowska
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | | | | | | | | | | |
Collapse
|
26
|
Zheng M, Zhu T, Chen B, Zhao H, Lu X, Lu Q, Ni M, Cheng L, Han H, Ye T, Ye Y, Liu H, Huang C. Intranasal Monophosphoryl Lipid a Administration Ameliorates depression-like Behavior in Chronically Stressed Mice Through Stimulation of Microglia. Neurochem Res 2023; 48:3160-3176. [PMID: 37358676 DOI: 10.1007/s11064-023-03974-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/08/2023] [Accepted: 06/16/2023] [Indexed: 06/27/2023]
Abstract
We and others have reported that systematic stimulation of the central innate immune system by a low dose of lipopolysaccharide (LPS) can improve depression-like behavior in chronically stressed animals. However, it is unclear whether similar stimulation by intranasal administration could improve depression-like behavior in animals. We investigated this question using monophosphoryl lipid A (MPL), a derivative of LPS that lacks the adverse effects of LPS but is still immuno-stimulatory. We found that a single intranasal administration of MPL at a dose of 10 or 20 µg/mouse, but not at a dose of 5 µg/mouse, ameliorated chronic unpredictable stress (CUS)-induced depression-like behavior in mice, as evidenced by the decrease in immobility time in tail suspension test and forced swimming test and the increase in sucrose intake in sucrose preference test. In the time-dependent analysis, the antidepressant-like effect of a single intranasal MPL administration (20 µg/mouse) was observed 5 and 8 h but not 3 h after drug administration and persisted for at least 7 days. Fourteen days after the first intranasal MPL administration, a second intranasal MPL administration (20 µg/mouse) still showed an antidepressant-like effect. The innate immune response mediated by microglia might mediate the antidepressant-like effect of intranasal MPL administration, because both inhibition of microglial activation by pretreatment with minocycline and depletion of microglia by pretreatment with PLX3397 prevented the antidepressant-like effect of intranasal MPL administration. These results suggest that intranasal administration of MPL can produce significant antidepressant-like effects in animals under chronic stress conditions via stimulation of microglia.
Collapse
Affiliation(s)
- Meng Zheng
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu Province, China
| | - Tao Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu Province, China
| | - Bingran Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu Province, China
| | - Hui Zhao
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu Province, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu Province, China
| | - Qun Lu
- Department of Pharmacy, Nantong Third Hospital Affiliated to Nantong University, #60 Middle Qingnian Road, Nantong, 226006, Jiangsu, China
| | - Mingxie Ni
- Department of Pharmacy, Changzhou Geriatric Hospital Affiliated to Soochow University, Changzhou No.7 People's Hospital, 288# Yanling East Road, Changzhou, 213000, Jiangsu, China
| | - Li Cheng
- Department of Pharmacy, Changzhou Geriatric Hospital Affiliated to Soochow University, Changzhou No.7 People's Hospital, 288# Yanling East Road, Changzhou, 213000, Jiangsu, China
| | - Han Han
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu Province, China
| | - Ting Ye
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu Province, China
| | - Ying Ye
- Department of Ultrasound, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong, 226001, Jiangsu, China
| | - Huijun Liu
- Department of Pharmacy, The First People's Hospital of Yancheng, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, #66 Renmin South Road, Yancheng, 224006, Jiangsu, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu Province, China.
| |
Collapse
|
27
|
Lullau APM, Haga EMW, Ronold EH, Dwyer GE. Antidepressant mechanisms of ketamine: a review of actions with relevance to treatment-resistance and neuroprogression. Front Neurosci 2023; 17:1223145. [PMID: 37614344 PMCID: PMC10442706 DOI: 10.3389/fnins.2023.1223145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/12/2023] [Indexed: 08/25/2023] Open
Abstract
Concurrent with recent insights into the neuroprogressive nature of depression, ketamine shows promise in interfering with several neuroprogressive factors, and has been suggested to reverse neuropathological patterns seen in depression. These insights come at a time of great need for novel approaches, as prevalence is rising and current treatment options remain inadequate for a large number of people. The rapidly growing literature on ketamine's antidepressant potential has yielded multiple proposed mechanisms of action, many of which have implications for recently elucidated aspects of depressive pathology. This review aims to provide the reader with an understanding of neuroprogressive aspects of depressive pathology and how ketamine is suggested to act on it. Literature was identified through PubMed and Google Scholar, and the reference lists of retrieved articles. When reviewing the evidence of depressive pathology, a picture emerges of four elements interacting with each other to facilitate progressive worsening, namely stress, inflammation, neurotoxicity and neurodegeneration. Ketamine acts on all of these levels of pathology, with rapid and potent reductions of depressive symptoms. Converging evidence suggests that ketamine works to increase stress resilience and reverse stress-induced dysfunction, modulate systemic inflammation and neuroinflammation, attenuate neurotoxic processes and glial dysfunction, and facilitate synaptogenesis rather than neurodegeneration. Still, much remains to be revealed about ketamine's antidepressant mechanisms of action, and research is lacking on the durability of effect. The findings discussed herein calls for more longitudinal approaches when determining efficacy and its relation to neuroprogressive factors, and could provide relevant considerations for clinical implementation.
Collapse
Affiliation(s)
- August P. M. Lullau
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Emily M. W. Haga
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Eivind H. Ronold
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Gerard E. Dwyer
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
- NORMENT Centre of Excellence, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
28
|
Chaib S, Bouillot C, Bouvard S, Vidal B, Zimmer L, Levigoureux E. Single subanesthetic dose of ketamine produces delayed impact on brain [ 18F]FDG PET imaging and metabolic connectivity in rats. Front Neurosci 2023; 17:1213941. [PMID: 37521685 PMCID: PMC10372660 DOI: 10.3389/fnins.2023.1213941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/23/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Ketamine, a glutamate NMDA receptor antagonist, is suggested to act very rapidly and durably on the depressive symptoms including treatment-resistant patients but its mechanisms of action remain unclear. There is a requirement for non-invasive biomarkers, such as imaging techniques, which hold promise in monitoring and elucidating its therapeutic impact. Methods We explored the glucose metabolism with [18F]FDG positron emission tomography (PET) in ten male rats in a longitudinal study designed to compare imaging patterns immediately after acute subanaesthetic ketamine injection (i.p. 10 mg/kg) with its sustained effects, 5 days later. Changes in [18F]FDG uptake following ketamine administration were estimated using a voxel-based analysis with SPM12 software, and a region of interest (ROI) analysis. A metabolic connectivity analysis was also conducted to estimate the immediate and delayed effects of ketamine on the inter-individual metabolic covariance between the ROIs. Results No significant difference was observed in brain glucose metabolism immediately following acute subanaesthetic ketamine injection. However, a significant decrease of glucose uptake appeared 5 days later, reflecting a sustained and delayed effect of ketamine in the frontal and the cingulate cortex. An increase in the raphe, caudate and cerebellum was also measured. Moreover, metabolic connectivity analyses revealed a significant decrease between the hippocampus and the thalamus at day 5 compared to the baseline. Discussion This study showed that the differences in metabolic profiles appeared belatedly, 5 days after ketamine administration, particularly in the cortical regions. Finally, this methodology will help to characterize the effects of future molecules for the treatment of treatment resistant depression.
Collapse
Affiliation(s)
- Sarah Chaib
- Université Claude Bernard Lyon 1, Lyon Neuroscience Research Center, CNRS, INSERM, Lyon, France
- Hospices Civils de Lyon, Lyon, France
| | | | - Sandrine Bouvard
- Université Claude Bernard Lyon 1, Lyon Neuroscience Research Center, CNRS, INSERM, Lyon, France
| | - Benjamin Vidal
- Université Claude Bernard Lyon 1, Lyon Neuroscience Research Center, CNRS, INSERM, Lyon, France
| | - Luc Zimmer
- Université Claude Bernard Lyon 1, Lyon Neuroscience Research Center, CNRS, INSERM, Lyon, France
- Hospices Civils de Lyon, Lyon, France
- CERMEP-Imaging Platform, Bron, France
| | - Elise Levigoureux
- Université Claude Bernard Lyon 1, Lyon Neuroscience Research Center, CNRS, INSERM, Lyon, France
- Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
29
|
Pepe M, Bartolucci G, Marcelli I, Simonetti A, Camardese G, Di Nicola M, Sani G. Reduction in Cognitive Symptoms Following Intranasal Esketamine Administration in Patients With Chronic Treatment-resistant Depression: A 12-Week Case Series. J Psychiatr Pract 2023; 29:325-332. [PMID: 37449831 DOI: 10.1097/pra.0000000000000723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
BACKGROUND Cognitive symptoms are a core feature of depressive disorders, interfere with full functional recovery and are prominent in patients with treatment-resistant depression (TRD), particularly in severe chronic cases. Intranasal (IN) esketamine was recently approved for the treatment of TRD; however, its effects on cognitive symptoms are unclear. In this article, we describe cognitive changes in 8 patients with chronic TRD who were treated with IN administration of esketamine. METHODS Eight outpatients with chronic TRD received IN esketamine over 3 months and were assessed at baseline and after 4, 8, and 12 weeks of treatment using the Montgomery-Åsberg Depression Rating Scale (MADRS), the Digit Symbol Substitution Test (DSST), the Trail Making Test-B (TMT-B), the Patient Deficits Questionnaire for Depression 5-item (PDQ-D5), the Hamilton Anxiety Rating Scale (HARS), and the Clinical Global Impressions Scale (CGI). FINDINGS We observed reductions in cognitive symptoms according to DSST, TMT-B, and PDQ-D5 scores within the first 2 months of treatment with IN esketamine. These improvements were observed before patients achieved clinical response (≥50% decrease in baseline MADRS scores), and they also occurred earlier than reductions in HARS scores. CONCLUSIONS A clinical response to IN esketamine was detected in severely ill patients with chronic TRD after 3 months of treatment. Interestingly, improvements on measures of cognitive symptoms were observed before patients achieved antidepressant response. These preliminary observations suggest an additional value to the antidepressant properties of IN esketamine. Clinical studies specifically investigating cognition as a primary outcome measure of IN esketamine in TRD are warranted.
Collapse
Affiliation(s)
- Maria Pepe
- PEPE, BARTOLUCCI, and MARCELLI: Department of Neuroscience, Section of Psychiatry, Università Cattolica del Sacro Cuore, Rome, Italy; SIMONETTI: Department of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy, and Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX; CAMARDESE, DI NICOLA, and SANI: Department of Neuroscience, Section of Psychiatry, Università Cattolica del Sacro Cuore, and Department of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
30
|
Cutler AJ, Mattingly GW, Maletic V. Understanding the mechanism of action and clinical effects of neuroactive steroids and GABAergic compounds in major depressive disorder. Transl Psychiatry 2023; 13:228. [PMID: 37365161 PMCID: PMC10293235 DOI: 10.1038/s41398-023-02514-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 05/12/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023] Open
Abstract
The pathophysiology of major depressive disorder (MDD) is thought to result from impaired connectivity between key brain networks. Gamma-aminobutyric acid (GABA) is the key inhibitory neurotransmitter in the brain, working primarily via GABAA receptors, with an important role in virtually all physiologic functions in the brain. Some neuroactive steroids (NASs) are positive allosteric modulators (PAMs) of GABAA receptors and potentiate phasic and tonic inhibitory responses via activation of synaptic and extrasynaptic GABAA receptors, respectively. This review first discusses preclinical and clinical data that support the association of depression with diverse defects in the GABAergic system of neurotransmission. Decreased levels of GABA and NASs have been observed in adults with depression compared with healthy controls, while treatment with antidepressants normalized the altered levels of GABA and NASs. Second, as there has been intense interest in treatment approaches for depression that target dysregulated GABAergic neurotransmission, we discuss NASs approved or currently in clinical development for the treatment of depression. Brexanolone, an intravenous NAS and a GABAA receptor PAM, is approved by the U.S. Food and Drug Administration for the treatment of postpartum depression (PPD) in patients 15 years and older. Other NASs include zuranolone, an investigational oral GABAA receptor PAM, and PH10, which acts on nasal chemosensory receptors; clinical data to date have shown improvement in depressive symptoms with these investigational NASs in adults with MDD or PPD. Finally, the review discusses how NAS GABAA receptor PAMs may potentially address the unmet need for novel and effective treatments with rapid and sustained antidepressant effects in patients with MDD.
Collapse
|
31
|
Rohn TT, Radin D, Brandmeyer T, Linder BJ, Andriambeloson E, Wagner S, Kehler J, Vasileva A, Wang H, Mee JL, Fallon JH. Genetic modulation of the HTR2A gene reduces anxiety-related behavior in mice. PNAS NEXUS 2023; 2:pgad170. [PMID: 37346271 PMCID: PMC10281383 DOI: 10.1093/pnasnexus/pgad170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 05/15/2023] [Indexed: 06/23/2023]
Abstract
The expanding field of precision gene editing using CRISPR/Cas9 has demonstrated its potential as a transformative technology in the treatment of various diseases. However, whether this genome-editing tool could be used to modify neural circuits in the central nervous system (CNS), which are implicated in complex behavioral traits, remains uncertain. In this study, we demonstrate the feasibility of noninvasive, intranasal delivery of adeno-associated virus serotype 9 (AAV9) vectors containing CRISPR/Cas9 cargo within the CNS resulting in modification of the HTR2A receptor gene. In vitro, exposure to primary mouse cortical neurons to AAV9 vectors targeting the HT2RA gene led to a concentration-dependent decrease in spontaneous electrical activity following multielectrode array (MEA) analysis. In vivo, at 5 weeks postintranasal delivery in mice, analysis of brain samples revealed single base pair deletions and nonsense mutations, leading to an 8.46-fold reduction in mRNA expression and a corresponding 68% decrease in the 5HT-2A receptor staining. Our findings also demonstrate a significant decrease in anxiety-like behavior in treated mice. This study constitutes the first successful demonstration of a noninvasive CRISPR/Cas9 delivery platform, capable of bypassing the blood-brain barrier and enabling modulation of neuronal 5HT-2A receptor pathways. The results of this study targeting the HTR2A gene provide a foundation for the development of innovative therapeutic strategies for a broad range of neurological disorders, including anxiety, depression, attentional deficits, and cognitive dysfunction.
Collapse
|
32
|
Liu B, Du Y, Xu C, Liu Q, Zhang L. Antidepressant effects of repeated s-ketamine administration as NMDAR Antagonist: Involvement of CaMKIIα and mTOR signaling in the hippocampus of CUMS mice. Brain Res 2023; 1811:148375. [PMID: 37146745 DOI: 10.1016/j.brainres.2023.148375] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/07/2023]
Abstract
With the approval of s-ketamine nasal spray as a novel antidepressant, its robust antidepressant effects have been intensively examined in clinical trials. However, the therapeutic efficacy and mechanisms of repeated intermittent drug administration remain unclear. In the present study, we applied a classic chronic unpredictable mild stress (CUMS) model to induce depressive-like behaviors of mice and evaluated the role of repeated s-ketamine administration (10 mg/kg, 7 consecutive days) in ameliorating depressive-like behaviors and modulating related molecular pathways. A battery of behavioral tests were performed to assess CUMS-induced depression. The protein expressions of GluN1, GluN2A, GluN2B, GluR1, CaMKIIα, phosphorylated CaMKIIα (p-CaMKIIα), BDNF, TrkB, phosphorylated TrkB (p-TrkB), mTOR, and phosphorylated mTOR (p-mTOR) as well as modification of synaptic ultrastructure was identified in hippocampal tissues. It turned out that s-ketamine manifested evident antidepressant effects with improved synaptic plasticity. Meanwhile, the results suggested that s-ketamine could differentially modulate glutamate receptors with upregulated GluN1 and GluR1 levels and downregulated GluN2B levels. CUMS-induced elevation of CaMKIIα phosphorylation and decline of BDNF, TrkB phosphorylation and mTOR could also be reversed through s-ketamine treatment. Together, our study provided evidence that selectively modulated glutamate receptors as well as CaMKIIα and mTOR signaling were involved in repeated s-ketamine administration.
Collapse
Affiliation(s)
- Bingjie Liu
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Yuxin Du
- Department of Anesthesiology, Jinling Hospital, the First School of Clinical Medicine, Southern Medical University, Nanjing, China
| | - Chang Xu
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Qingzhen Liu
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Lidong Zhang
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China.
| |
Collapse
|
33
|
Niquet J, Nguyen D, de Araujo Furtado M, Lumley L. Treatment of cholinergic-induced status epilepticus with polytherapy targeting GABA and glutamate receptors. Epilepsia Open 2023; 8 Suppl 1:S117-S140. [PMID: 36807554 PMCID: PMC10173853 DOI: 10.1002/epi4.12713] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 02/15/2023] [Indexed: 02/23/2023] Open
Abstract
Despite new antiseizure medications, the development of cholinergic-induced refractory status epilepticus (RSE) continues to be a therapeutic challenge as pharmacoresistance to benzodiazepines and other antiseizure medications quickly develops. Studies conducted by Epilepsia. 2005;46:142 demonstrated that the initiation and maintenance of cholinergic-induced RSE are associated with trafficking and inactivation of gamma-aminobutyric acid A receptors (GABAA R) thought to contribute to the development of benzodiazepine pharmacoresistance. In addition, Dr. Wasterlain's laboratory reported that increased N-methyl-d-aspartate receptors (NMDAR) and alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPAR) contribute to enhanced glutamatergic excitation (Neurobiol Dis. 2013;54:225; Epilepsia. 2013;54:78). Thus, Dr. Wasterlain postulated that targeting both maladaptive responses of reduced inhibition and increased excitation that is associated with cholinergic-induced RSE should improve therapeutic outcome. We currently review studies in several animal models of cholinergic-induced RSE that demonstrate that benzodiazepine monotherapy has reduced efficacy when treatment is delayed and that polytherapy with drugs that include a benzodiazepine (eg midazolam and diazepam) to counter loss of inhibition, concurrent with an NMDA antagonist (eg ketamine) to reduce excitation provide improved efficacy. Improved efficacy with polytherapy against cholinergic-induced seizure is demonstrated by reduction in (1) seizure severity, (2) epileptogenesis, and (3) neurodegeneration compared with monotherapy. Animal models reviewed include pilocarpine-induced seizure in rats, organophosphorus nerve agent (OPNA)-induced seizure in rats, and OPNA-induced seizure in two mouse models: (1) carboxylesterase knockout (Es1-/- ) mice which, similarly to humans, lack plasma carboxylesterase and (2) human acetylcholinesterase knock-in carboxylesterase knockout (KIKO) mice. We also review studies showing that supplementing midazolam and ketamine with a third antiseizure medication (valproate or phenobarbital) that targets a nonbenzodiazepine site rapidly terminates RSE and provides further protection against cholinergic-induced SE. Finally, we review studies on the benefits of simultaneous compared with sequential drug treatments and the clinical implications that lead us to predict improved efficacy of early combination drug therapies. The data generated from seminal rodent studies of efficacious treatment of cholinergic-induced RSE conducted under Dr. Wasterlain's guidance suggest that future clinical trials should treat the inadequate inhibition and temper the excess excitation that characterize RSE and that early combination therapies may provide improved outcome over benzodiazepine monotherapy.
Collapse
Affiliation(s)
- Jerome Niquet
- Department of NeurologyDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
- Epilepsy Research LaboratoryVeterans Affairs Greater Los Angeles Healthcare SystemLos AngelesCaliforniaUSA
| | - Donna Nguyen
- Neuroscience DepartmentU.S. Army Medical Research Institute of Chemical Defense (USAMRICD)Aberdeen Proving GroundMarylandUSA
| | | | - Lucille Lumley
- Neuroscience DepartmentU.S. Army Medical Research Institute of Chemical Defense (USAMRICD)Aberdeen Proving GroundMarylandUSA
| |
Collapse
|
34
|
Shad MU. Recent Developments in Pharmacotherapy of Depression: Bench to Bedside. J Pers Med 2023; 13:jpm13050773. [PMID: 37240943 DOI: 10.3390/jpm13050773] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/20/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
For the last 70 years, we did not move beyond the monoamine hypothesis of depression until the approval of the S-enantiomer of ketamine, an N-methyl-D-aspartate (NMDA) receptor blocker and the first non-monoaminergic antidepressant characterized by rapid antidepressant and antisuicidal effects. A similar profile has been reported with another NMDA receptor antagonist, dextromethorphan, which has also been approved to manage depression in combination with bupropion. More recently, the approval of a positive allosteric modulator of GABA-A receptors, brexanolone, has added to the list of recent breakthroughs with the relatively rapid onset of antidepressant efficacy. However, multiple factors have compromised the clinical utility of these exciting discoveries in the general population, including high drug acquisition costs, mandatory monitoring requirements, parenteral drug administration, lack of insurance coverage, indirect COVID-19 effects on healthcare systems, and training gaps in psychopharmacology. This narrative review aims to analyze the clinical pharmacology of recently approved antidepressants and discuss potential barriers to the bench-to-bedside transfer of knowledge and clinical application of exciting recent discoveries. Overall, clinically meaningful advances in the treatment of depression have not reached a large proportion of depressed patients, including those with treatment-resistant depression, who might benefit the most from the novel antidepressants.
Collapse
Affiliation(s)
- Mujeeb U Shad
- Valley Health System (VHS), Las Vegas, NV 89118, USA
- The Department of Psychiatry, University of Nevada, Las Vegas, School of Medicine, The Touro University of Nevada College of Osteopathic Medicine (TUNCOM), Henderson, NV 89014, USA
- The University of Nevada, Las Vegas, NV 89154, USA
| |
Collapse
|
35
|
Candee R, Wilkenson R, Schreiber M, DeCenzo M. The roles of neuroinflammation and glutamatergic excitotoxicity in treatment-resistant depression. JAAPA 2023; 36:12-17. [PMID: 36913608 DOI: 10.1097/01.jaa.0000921252.57819.4b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
ABSTRACT Major depressive disorder affects nearly 20% of people during their lifetime. A growing body of evidence supports the theory that neuroinflammation plays a prominent role in the neurobiology of depression, which implicates glutamate and gamma aminobutyric acid as key factors in the pathophysiology of the disease process. This article reviews the pathologic pathways of glutamate excess in the central nervous system and how they may be implicated in the underlying disorder of treatment-resistant depression and targeted for treatment.
Collapse
Affiliation(s)
- Ryan Candee
- Ryan Candee practices at Matthews-Vu Medical Group in Colorado Springs, Colo. Rachel Wilkenson is an integrated psychiatrist at Matthews-Vu Medical Group. Molly Schreiber practices at UCHealth in Woodland Park, Colo. Megan DeCenzo practices at Matthews-Vu Medical Group. The authors have disclosed no potential conflicts of interest, financial or otherwise
| | | | | | | |
Collapse
|
36
|
Wang G, Yang H, Zuo W, Mei X. Antidepressant-like effect of acute dose of Naringin involves suppression of NR1 and activation of protein kinase A/cyclic adenosine monophosphate response element-binding protein/brain-derived neurotrophic factor signaling in hippocampus. Behav Pharmacol 2023; 34:101-111. [PMID: 36503881 DOI: 10.1097/fbp.0000000000000713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Naringin (Nr) has been identified to have antidepressant-like effects through repeated treatment. However, the underlying mechanism of the rapid antidepressant-like effects of Nr was still unclear. The present study used behavioral tests, classic depressive model and pharmacological methods to reveal the rapid antidepressant-like potential of Nr. We found that a single dose of Nr (20 mg/kg) produced antidepressant-like action after 2 h in the tail suspension test (TST) and forced swimming test (FST). Moreover, ketamine-like effects were also demonstrated by using the chronic mild stress model (CMS) and learned helplessness (LH), and the results showed that Nr reversed all behavioral defects, TST, FST, source preference test (SPT) in CMS, and LH testing, TST, FST in LH model, at 2 h after a single administration. In addition, Nr (20 mg/kg) could improve the abnormal expressions of NMDA receptor NR1 and PKA/CREB/BDNF pathway in hippocampus 2 h after a single administration in CMS mice. Further investigation revealed that activation of NMDA receptors by NMDA (750 mg/kg) could block the antidepressant effects of acute administration of Nr (20 mg/kg). However, the inhibition of NMDA receptors by MK-801 (0.05 mg/kg) promoted the subdose of Nr (10 mg/kg) to have antidepressant effect, which was similar to the effective dose Nr (20 mg/kg). Taken together, acute dose of Nr produces rapid antidepressant-like action, and the underlying mechanism could be through inhibiting NMDA receptors in the hippocampus.
Collapse
Affiliation(s)
- Guangyao Wang
- Department of Basic Theory of Chinese Medicine, College of Chinese Medicine, Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine
| | - Haixia Yang
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenren Zuo
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoyun Mei
- Department of Basic Theory of Chinese Medicine, College of Chinese Medicine, Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine
| |
Collapse
|
37
|
Kwami Edem Kukuia K, Boakye Burns F, Kofi Adutwum-Ofosu K, Appiah F, Kwabena Amponsah S, Begyinah R, Efua Koomson A, Yaw Takyi F, Amatey Tagoe T, Amoateng P. Increased BDNF and hippocampal dendritic spine density are associated with the rapid antidepressant-like effect of iron-citalopram and iron-imipramine combinations in mice. Neuroscience 2023; 519:90-106. [PMID: 36948482 DOI: 10.1016/j.neuroscience.2023.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 03/07/2023] [Accepted: 03/13/2023] [Indexed: 03/24/2023]
Abstract
Iron supplementation previously demonstrated antidepressant-like effects in post-partum rats. The present study evaluates the possible synergistic antidepressant effect of sub-therapeutic dose of iron co-administered with citalopram or imipramine in female Institute of Cancer Research mice. Depression-like symptoms were induced in the forced swim (FST), tail suspension (TST), and open space swim (OSST) tests while open field test (OFT) was used to assess locomotor activity. Mice (n=8) received iron (0.8- 7.2 mg/kg), citalopram (3-30 mg/kg), imipramine (3-30 mg/kg), desferrioxamine (50 mg/kg) or saline in the single treatment phase of each model and subsequently a sub-therapeutic dose of iron co-administered with citalopram or imipramine. Assessment of serum BDNF and dendritic spine density was done using ELISA and Golgi staining techniques respectively. Iron, citalopram and imipramine, unlike desferrioxamine, reduced immobility score in the TST, FST and OSST without affecting locomotor activity, suggesting antidepressant-like effect. Sub-therapeutic dose of iron in combination with citalopram or imipramine further enhanced the antidepressant-like effect, producing a more rapid effect when compared to the iron, citalopram or imipramine alone. Iron, citalopram and imipramine or their combinations increased serum BDNF concentration, hippocampal neuronal count and dendritic spine densities. Our study provides experimental evidence that iron has antidepressant-like effect and sub-therapeutic dose of iron combined with citalopram or imipramine produces more rapid antidepressant-like effect. We further show that iron alone or its combination with citalopram or imipramine attenuates the neuronal loss associated with depressive conditions, increases dendritic spines density and BDNF levels. These finding suggest iron-induced neuronal plasticity in the mice brain.
Collapse
Affiliation(s)
- Kennedy Kwami Edem Kukuia
- Department of Medical Pharmacology, College of Health Sciences, University of Ghana, Korle Bu, Accra, Ghana.
| | - Frederick Boakye Burns
- Department of Pharmacology & Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, P.O Box LG 43, Legon, Accra, Ghana.
| | - Kevin Kofi Adutwum-Ofosu
- Department of Anatomy, University of Ghana Medical School, College of Health Sciences, University of Ghana, Accra, Ghana.
| | - Frimpong Appiah
- Department of Community Health and Medicine, School of Food and Health Sciences, Anglican University College of Technology, Nkoranza, Ghana.
| | - Seth Kwabena Amponsah
- Department of Medical Pharmacology, College of Health Sciences, University of Ghana, Korle Bu, Accra, Ghana.
| | - Richard Begyinah
- Department of Pharmacology & Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, P.O Box LG 43, Legon, Accra, Ghana.
| | - Awo Efua Koomson
- Department of Medical Pharmacology, College of Health Sciences, University of Ghana, Korle Bu, Accra, Ghana.
| | - Ferka Yaw Takyi
- Department of Pharmacology & Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, P.O Box LG 43, Legon, Accra, Ghana.
| | - Thomas Amatey Tagoe
- Department of Physiology, College of Health Sciences, University of Ghana, Korle Bu, Accra, Ghana.
| | - Patrick Amoateng
- Department of Pharmacology & Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, P.O Box LG 43, Legon, Accra, Ghana.
| |
Collapse
|
38
|
Podkowa K, Czarnacki K, Borończyk A, Borończyk M, Paprocka J. The NMDA receptor antagonists memantine and ketamine as anti-migraine agents. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023:10.1007/s00210-023-02444-2. [PMID: 36869904 DOI: 10.1007/s00210-023-02444-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 02/22/2023] [Indexed: 03/05/2023]
Abstract
Migraine is a debilitating disorder affecting females more frequently than males. There is some evidence that drugs targeting glutamate receptors: memantine and ketamine might be beneficial in the therapy of this entity. Therefore, the purpose of this work is to present NMDA receptor antagonists, memantine and ketamine, as potential anti-migraine agents. We searched PubMed/MEDLINE, Embase, and clinical trials submitted to ClinicalTrials.gov to find publications describing eligible trials published between database inception and December 31, 2021. This comprehensive literature review summarizes data on the use of the NMDA receptor antagonists memantine and ketamine in the pharmacotherapy of migraine. Results from 20 previous and recent preclinical experiments are discussed and correlated with 19 clinical trials (including case series, open-label, and randomized placebo-controlled trials). For the purposes of this review, the authors hypothesized that the propagation of SD is a major mechanism in the pathophysiology of migraine. In several animal studies and in vitro studies, memantine and ketamine inhibited or reduced propagation of the SD. In addition, the results of clinical trials suggest that memantine or ketamine may be an effective treatment option for migraine. However, most studies on these agents lack control group. Although further clinical trials are needed, the results suggest that ketamine or memantine may be promising molecules for the treatment of severe migraine. Particular attention should be paid to people who have a treatment-resistant form of migraine with aura or have exhausted existing treatment options. For them, the drugs under discussion could represent an interesting alternative in the future.
Collapse
Affiliation(s)
- Karolina Podkowa
- Department of Pathophysiology, Jagiellonian University Medical College, Kraków, Poland.
| | - Kamil Czarnacki
- Students' Scientific Society, Department of Pediatric Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Agnieszka Borończyk
- Students' Scientific Association, Department of Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Michał Borończyk
- Students' Scientific Association, Department of Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Justyna Paprocka
- Department of Pediatric Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
39
|
Beach KM, Hung LF, Lou L, Ostrin LA. Diurnal Variation and Effects of Dilation and Sedation on Intraocular Pressure in Infant Rhesus Monkeys. Curr Eye Res 2023; 48:289-296. [PMID: 36357337 PMCID: PMC10006333 DOI: 10.1080/02713683.2022.2141782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/26/2022] [Indexed: 11/12/2022]
Abstract
PURPOSE Intraocular pressure (IOP) is an important factor in numerous ocular conditions and research areas, including eye growth and myopia. In infant monkeys, IOP is typically measured under anesthesia. This study aimed to establish a method for awake IOP measurement in infant rhesus monkeys, determine diurnal variation, and assess the effects of dilation and sedation. METHODS Awake IOP (iCare TonoVet) was measured every 2 h from 7:30 am to 5:30 pm to assess potential diurnal variations in infant rhesus monkeys (age 3 weeks, n = 11). The following day, and every 2 weeks to age 15 weeks, IOP was measured under three conditions: (1) awake, (2) awake and dilated (tropicamide 0.5%), and (3) sedated (ketamine and acepromazine) and dilated. Intraclass correlation coefficient (ICC) was used to determine intersession repeatability, and repeated measures. ANOVA was used to determine effects of age and condition. RESULTS At age 3 weeks, mean (±SEM) awake IOP was 15.4 ± 0.6 and 15.2 ± 0.7 mmHg for right and left eyes, respectively (p=.59). The ICC between sessions was 0.63[-0.5 to 0.9], with a mean difference of 2.2 ± 0.3 mmHg. Diurnal IOP from 7:30 am to 5:30 pm showed no significant variation (p=.65). From 3 to 15 weeks of age, there was a significant effect of age (p=.01) and condition (p<.001). Across ages, IOP was 17.8 ± 0.7 mmHg while awake and undilated, 18.4 ± 0.2 mmHg awake and dilated, and 11.0 ± 0.3 mmHg after sedation and dilation. CONCLUSIONS Awake IOP measurement was feasible in young rhesus monkeys. No significant diurnal variations in IOP were observed between 7:30 am and 5:30 pm at age 3 weeks. In awake monkeys, IOP was slightly higher after mydriasis and considerably lower after sedation. Findings show that IOP under ketamine/acepromazine anesthesia is significantly different than awake IOP in young rhesus monkeys.
Collapse
Affiliation(s)
- Krista M Beach
- University of Houston College of Optometry, Houston, TX, United States
| | - Li-Fang Hung
- University of Houston College of Optometry, Houston, TX, United States
| | - Linjiang Lou
- University of Houston College of Optometry, Houston, TX, United States
| | - Lisa A Ostrin
- University of Houston College of Optometry, Houston, TX, United States
| |
Collapse
|
40
|
Sachdeva B, Sachdeva P, Ghosh S, Ahmad F, Sinha JK. Ketamine as a therapeutic agent in major depressive disorder and posttraumatic stress disorder: Potential medicinal and deleterious effects. IBRAIN 2023; 9:90-101. [PMID: 37786516 PMCID: PMC10528797 DOI: 10.1002/ibra.12094] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 10/04/2023]
Abstract
Major depressive disorder (MDD) and posttraumatic stress disorder (PTSD) are the most common causes of emotional distress that impair an individual's quality of life. MDD is a chronic mental illness that affects 300 million people across the world. Clinical manifestations of MDD include fatigue, loss of interest in routine tasks, psychomotor agitation, impaired ability to focus, suicidal ideation, hypersomnolence, altered psychosocial functioning, and appetite loss. Individuals with depression also demonstrate a reduced behavioral response while experiencing pleasure, a symptom known as anhedonia. Like MDD, PTSD is a prevalent and debilitating psychiatric disorder resulting from a traumatic incident such as sexual assault, war, severe accident, or natural disaster. Symptoms such as recalling event phases, hypervigilance, irritability, and anhedonia are common in PTSD. Both MDD and PTSD pose enormous socioeconomic burdens across the globe. The search for effective treatment with minimal side effects is still ongoing. Ketamine is known for its anesthetic and analgesic properties. Psychedelic and psychotropic effects of ketamine have been found on the nervous system, which highlights its toxicity. In this article, the effectiveness of ketamine as a potential therapeutic for PTSD and MDD along with its mechanisms of action, clinical trials, and possible side effects have been discussed.
Collapse
Affiliation(s)
- Bhuvi Sachdeva
- Department of Physics and Astrophysics, Bhagini Nivedita CollegeUniversity of DelhiDelhiIndia
| | | | - Shampa Ghosh
- GloNeuro AcademyNoidaUttar PradeshIndia
- ICMR—National Institute of NutritionTarnakaHyderabadIndia
| | - Faizan Ahmad
- Department of Medical Elementology and ToxicologyJamia HamdardDelhiIndia
| | | |
Collapse
|
41
|
The therapeutic role of ketamine and esketamine in treating psychopathological domains of depression. Neuropharmacology 2023; 223:109299. [PMID: 36336068 DOI: 10.1016/j.neuropharm.2022.109299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
Over the past two decades, ketamine has emerged as a novel effective and rapid-acting antidepressant. While the vast majority of studies on ketamine have focused on its ability to reduce the severity of depression broadly, its effectiveness in specific domains such as cognition, anhedonia, suicidality, and workplace/social/scholastic functionality has been neglected. Similarly, current treatments (e.g., SSRIs and SNRIs) aim to improve overall depression severity, which often results in the persistence of one or more residual symptom domains and prevents full recovery to premorbid functionality. In this review, we narratively synthesize the literature pertaining to the effectiveness of ketamine in treating key domains of depressive symptomatology (i.e., cognition, anhedonia, suicidality, and psychosocial functionality). Our findings suggest that ketamine is effective across domains varyingly, with the strongest evidence being for its ability to reduce suicidality. The rapid acting nature of ketamine further supports its use in treating suicidality and potentially preventing the completion of suicide. Evidence for the effectiveness of ketamine in other domains is weak, primarily due to a lack of robust studies specifically designed to assess these domains as primary outcomes. Future studies should scrutinize the effects of ketamine on specific domains of depression to optimize its implementation.
Collapse
|
42
|
Huang Y, Sun P, Wu Z, Guo X, Wu X, Chen J, Yang L, Wu X, Fang Y. Comparison on the clinical features in patients with or without treatment-resistant depression: A National Survey on Symptomatology of Depression report. Psychiatry Res 2023; 319:114972. [PMID: 36434937 DOI: 10.1016/j.psychres.2022.114972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 11/22/2022]
Abstract
Patients with treatment-resistant depression (TRD) have fewer treatment options and worse prognoses than those without TRD. Although the etiology or pathophysiology of TRD remains unclear, certain clinical variables have been found to be related to its severity and prognosis. Therefore, 1151 patients with recurrent depression were recruited from the National Survey on Symptomatology of Depression (NSSD) and their depressive symptoms were assessed by using the doctor-rating assessment questionnaire. Then, the differences between patients with or without TRD were compared by parametric or nonparametric tests and the risk factors for TRD were explored by logistic regression. The results showed there were differences in clinical variables between patients with and without TRD. Additionally, we found depression with more somatic symptoms had a higher risk for TRD. Further analysis by stepwise logistic regression showed that age, gender, religious belief, drinking habit, the total course of depression, the number of hospitalizations, characteristics of seasonal episode remission, depressed mood, hypersexuality, emotionally incoherent psychotic symptoms, psychomotor agitation, respiratory system symptoms and history of suicide attempts were strongly associated with TRD. So, it is crucial for clinicians to identify these clinical features and adjust treatments timely.
Collapse
Affiliation(s)
- Yingying Huang
- Department of Psychiatry and Mental Health, Jining Medical University, Shandong 272002, China; Department 2 of the Elderly, Qingdao Mental Health Center, Shandong 266034, China
| | - Ping Sun
- Department 2 of the Elderly, Qingdao Mental Health Center, Shandong 266034, China; Clinical Research Center & Division of Mood Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhiguo Wu
- Department of Psychiatry, Shanghai Yangpu District Mental Health Center, Shanghai 200093, China; Clinical Research Centre in Mental Health, Shanghai University of Medicine & Health Sciences, Shanghai 200030, China
| | - Xiaoyun Guo
- Clinical Research Center & Division of Mood Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xiaohui Wu
- Clinical Research Center & Division of Mood Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Jun Chen
- Clinical Research Center & Division of Mood Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai 200031, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai 201108, China
| | - Lu Yang
- Clinical Research Center & Division of Mood Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xiao Wu
- Department of Bacteriology and Immunology, Beijing Key Laboratory on Drug-Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing 101125, China
| | - Yiru Fang
- Clinical Research Center & Division of Mood Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai 200031, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai 201108, China.
| |
Collapse
|
43
|
Bhatt S, Devadoss T, Jha NK, Baidya M, Gupta G, Chellappan DK, Singh SK, Dua K. Targeting inflammation: a potential approach for the treatment of depression. Metab Brain Dis 2023; 38:45-59. [PMID: 36239867 DOI: 10.1007/s11011-022-01095-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 09/29/2022] [Indexed: 02/03/2023]
Abstract
Major depressive disorder (MDD) or Depression is one of the serious neuropsychiatric disorders affecting over 280 million people worldwide. It is 4th important cause of disability, poor quality of life, and economic burden. Women are more affected with the depression as compared to men and severe depression can lead to suicide. Most of the antidepressants predominantly work through the modulation on the availability of monoaminergic neurotransmitter (NTs) levels in the synapse. Current antidepressants have limited efficacy and tolerability. Moreover, treatment resistant depression (TRD) is one of the main causes for failure of standard marketed antidepressants. Recently, inflammation has also emerged as a crucial factor in pathological progression of depression. Proinflammatory cytokine levels are increased in depressive patients. Antidepressant treatment may attenuate depression via modulation of pathways of inflammation, transformation in structure of brain, and synaptic plasticity. Hence, targeting inflammation may be emerged as an effective approach for the treatment of depression. The present review article will focus on the preclinical and clinical studies that targets inflammation. In addition, it also concentrates on the therapeutic approaches' that targets depression via influence on the inflammatory signaling pathways. Graphical abstract demonstrate the role of various factors in the progression and neuroinflammation, oxidative stress. It also exhibits the association of neuroinflammation, oxidative stress with depression.
Collapse
Affiliation(s)
- Shvetank Bhatt
- School of Pharmacy, Dr. Vishwanath Karad MIT World Peace University, Maharashtra, 411038, Pune, India.
| | - Thangaraj Devadoss
- Department of Pharmaceutical Chemistry, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Mumbai Agra Highway, Maharashtra, 424001, Dhule, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, 201310, Greater Noida, Uttar Pradesh, India
| | - Moushumi Baidya
- Department of Pharmaceutical Technology, JIS University, 700109, Kolkata, West Bengal, India
- Department of Pharmaceutical Technology, Bharat Pharmaceutical Technology, 799130, Agartala, West Tripura, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India
- Department of Pharmacology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospitals, Saveetha University, Chennai, India
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, 248007, Dehradun, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
- Faculty of Health, Australian Research Centre in Complementary & Integrative Medicine, University of Technology Sydney, 2007, Ultimo, NSW, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary & Integrative Medicine, University of Technology Sydney, 2007, Ultimo, NSW, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, 2007, Ultimo, NSW, Australia
| |
Collapse
|
44
|
Halaris A, Cook J. The Glutamatergic System in Treatment-Resistant Depression and Comparative Effectiveness of Ketamine and Esketamine: Role of Inflammation? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:487-512. [PMID: 36949323 DOI: 10.1007/978-981-19-7376-5_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
The glutamatergic system is the primary excitatory pathway within the CNS and is responsible for cognition, memory, learning, emotion, and mood. Because of its significant importance in widespread nervous system function, it is tightly regulated through multiple mechanisms, such as glutamate recycling, microglial interactions, and inflammatory pathways. Imbalance within the glutamatergic system has been implicated in a wide range of pathological conditions including neurodegenerative conditions, neuromuscular conditions, and mood disorders including depression. Major depressive disorder (MDD) is the most common mood disorder worldwide, has a high prevalence rate, and afflicts approximately 280 million people. While there are numerous treatments for the disease, 30-40% of patients are unresponsive to treatment and deemed treatment resistant; approximately another third experience only partial improvement (World Health Organization, Depression fact sheet [Internet], 2020). Esketamine, the S-enantiomer of ketamine, was approved by the Food and Drug Administration for treatment-resistant depression (TRD) in 2019 and has offered new hope to patients. It is the first treatment targeting the glutamatergic system through a complex mechanism. Numerous studies have implicated imbalance in the glutamatergic system in depression and treatment resistance. Esketamine and ketamine principally work through inhibition of the NMDA receptor, though more recent studies have implicated numerous other mechanisms mediating the antidepressant efficacy of these agents. These mechanisms include increase in brain-derived neurotrophic factor (BDNF), activation of mammalian target of the rapamycin complex (mTORC), and reduction in inflammation. Esketamine and ketamine have been shown to decrease inflammation in numerous ways principally through reducing pro-inflammatory cytokines (e.g., TNF-α, IL-6) (Loix et al., Acta Anaesthesiol Belg 62(1):47-58, 2011; Chen et al., Psychiatry Res 269:207-11, 2018; Kopra et al., J Psychopharmacol 35(8):934-45, 2021). This anti-inflammatory effect has also been shown to be involved in the antidepressive properties of both ketamine and esketamine (Chen et al., Psychiatry Res 269:207-11, 2018; Kopra et al., J Psychopharmacol 35(8):934-45, 2021).
Collapse
Affiliation(s)
- Angelos Halaris
- Department of Psychiatry, Loyola University Stritch School of Medicine, Maywood, IL, USA.
| | - John Cook
- Department of Psychiatry, Loyola University Stritch School of Medicine, Maywood, IL, USA
| |
Collapse
|
45
|
García-Gutiérrez MS, Navarro D, Austrich-Olivares A, Manzanares J. Unveiling behavioral and molecular neuroadaptations related to the antidepressant action of cannabidiol in the unpredictable chronic mild stress model. Front Pharmacol 2023; 14:1171646. [PMID: 37144214 PMCID: PMC10151764 DOI: 10.3389/fphar.2023.1171646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/03/2023] [Indexed: 05/06/2023] Open
Abstract
Introduction: This study aims to further characterize cannabidiol's pharmacological and molecular profile as an antidepressant. Methods: Effects of cannabidiol (CBD), alone or combined with sertraline (STR), were evaluated in male CD1 mice (n = 48) exposed to an unpredictable chronic mild stress (UCMS) procedure. Once the model was established (4 weeks), mice received CBD (20 mg·kg-1, i.p.), STR (10 mg·kg-1, p.o.) or its combination for 28 days. The efficacy of CBD was evaluated using the light-dark box (LDB), elevated plus maze (EPM), tail suspension (TS), sucrose consumption (SC) and novel object recognition (NOR) tests. Gene expression changes in the serotonin transporter, 5-HT1A and 5-HT2A receptors, BDNF, VGlut1 and PPARdelta, were evaluated in the dorsal raphe, hippocampus (Hipp) and amygdala by real-time PCR. Besides, BDNF, NeuN and caspase-3 immunoreactivity were assessed in the Hipp. Results: CBD exerted anxiolytic and antidepressant-like effects at 4 and 7 days of treatment in the LDB and TS tests, respectively. In contrast, STR required 14 days of treatment to show efficacy. CBD improved cognitive impairment and anhedonia more significantly than STR. CBD plus STR showed a similar effect than CBD in the LBD, TST and EPM. However, a worse outcome was observed in the NOR and SI tests. CBD modulates all molecular disturbances induced by UCMS, whereas STR and the combination could not restore 5-HT1A, BDNF and PPARdelta in the Hipp. Discussion: These results pointed out CBD as a potential new antidepressant with faster action and efficiency than STR. Particular attention should be given to the combination of CBD with current SSRI since it appears to produce a negative impact on treatment.
Collapse
Affiliation(s)
- María Salud García-Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández, Alicante, Spain
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Daniela Navarro
- Instituto de Neurociencias, Universidad Miguel Hernández, Alicante, Spain
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | | | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernández, Alicante, Spain
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
- *Correspondence: Jorge Manzanares,
| |
Collapse
|
46
|
Golubeva EA, Lavrov MI, Radchenko EV, Palyulin VA. Diversity of AMPA Receptor Ligands: Chemotypes, Binding Modes, Mechanisms of Action, and Therapeutic Effects. Biomolecules 2022; 13:biom13010056. [PMID: 36671441 PMCID: PMC9856200 DOI: 10.3390/biom13010056] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/19/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
L-Glutamic acid is the main excitatory neurotransmitter in the central nervous system (CNS). Its associated receptors localized on neuronal and non-neuronal cells mediate rapid excitatory synaptic transmission in the CNS and regulate a wide range of processes in the brain, spinal cord, retina, and peripheral nervous system. In particular, the glutamate receptors selective to α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) also play an important role in numerous neurological disorders and attract close attention as targets for the creation of new classes of drugs for the treatment or substantial correction of a number of serious neurodegenerative and neuropsychiatric diseases. For this reason, the search for various types of AMPA receptor ligands and studies of their properties are attracting considerable attention both in academic institutions and in pharmaceutical companies around the world. This review focuses mainly on the advances in this area published since 2017. Particular attention is paid to the structural diversity of new chemotypes of agonists, competitive AMPA receptor antagonists, positive and negative allosteric modulators, transmembrane AMPA regulatory protein (TARP) dependent allosteric modulators, ion channel blockers as well as their binding sites. This review also presents the studies of the mechanisms of action of AMPA receptor ligands that mediate their therapeutic effects.
Collapse
|
47
|
Adjunctive dopaminergic enhancement of esketamine in treatment-resistant depression. Prog Neuropsychopharmacol Biol Psychiatry 2022; 119:110603. [PMID: 35842074 DOI: 10.1016/j.pnpbp.2022.110603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 07/10/2022] [Indexed: 11/22/2022]
Abstract
Esketamine is a novel treatment for treatment resistant depression (TRD) and was approved by the FDA in early 2019. It antagonizes the NMDA receptor providing rapid improvement in symptoms with a complex mechanism of action primarily mediated through glutamatergic activation. Significant barriers exist to widespread use of esketamine including durability of response, particularly in the maintenance phase. Since it must be administered in combination with an oral antidepressant, investigating appropriate treatments to be given concomitantly may further improve outcomes and response duration. Specifically, due to dysfunction in dopaminergic pathways in many patients with MDD and TRD, addition of a prodopaminergic agent, such as bupropion, may provide additional benefit and durability of response. Historically, the addition of a dopaminergic agent to traditional treatment (e.g., SSRI, SNRI) has been shown to improve response in TRD. While we have anecdotal evidence to support adjunctive dopaminergic enhancement of esketamine response, robust data are limited. There are case reports that exhibit efficacy with the use of a MAO-I in combination with ketamine supporting at least in part a dopaminergic component. Additionally, there is mechanistic rationale for the use of dopaminergic agents with a NMDA antagonist. This includes co-localization of NMDA and dopamine receptors as well as increased glutamatergic signaling due to dopamine-induced phosphorylation of AMPAR. Recently, AXS-05, an oral combination of dextromethorphan and bupropion, has shown promise in both MDD and TRD clinical trials highlighting the potential validity of this mechanism. This paper describes how dopaminergic enhancement may increase efficacy and durability of response with esketamine, encouraging further research into this treatment option.
Collapse
|
48
|
Martinotti G, Vita A, Fagiolini A, Maina G, Bertolino A, Dell'Osso B, Siracusano A, Clerici M, Bellomo A, Sani G, d'Andrea G, Chiaie RD, Conca A, Barlati S, Di Lorenzo G, De Fazio P, De Filippis S, Nicolò G, Rosso G, Valchera A, Nucifora D, Di Mauro S, Bassetti R, Martiadis V, Olivola M, Belletti S, Andriola I, Di Nicola M, Pettorruso M, McIntyre RS, di Giannantonio M. Real-world experience of esketamine use to manage treatment-resistant depression: A multicentric study on safety and effectiveness (REAL-ESK study). J Affect Disord 2022; 319:646-654. [PMID: 36167246 DOI: 10.1016/j.jad.2022.09.043] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/28/2022] [Accepted: 09/19/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND Treatment-resistant Depression (TRD) represents a widespread disorder with significant direct and indirect healthcare costs. esketamine, the S-enantiomer of ketamine, has been recently approved for TRD, but real-world studies are needed to prove its efficacy in naturalistic settings. OBJECTIVES Evaluate the effectiveness and safety of esketamine nasal spray in a clinical sample of patients with TRD from several Italian mental health services. METHODS REAL-ESK study is an observational, retrospective and multicentric study comprising a total of 116 TRD patients treated with esketamine nasal spray. Anamnestic data and psychometric assessment (MADRS, HAMD-21, HAM-A) were collected from medical records at baseline (T0), one month (T1) and three month (T2) follow-ups. RESULTS A significant reduction of depressive symptoms was found at T1 and T2 compared to T0. A dramatic increase in clinical response (64.2 %) and remission rates (40.6 %) was detected at T2 compared to T1. No unexpected safety concerns were observed, side effects rates were comparable to those reported in RCTs. No differences in efficacy have been found among patients with and without psychiatric comorbidities. LIMITATIONS The open design of the study and the absence of a placebo or active comparator group are limitations. The study lacks an inter-rater reliability evaluation of the assessments among the different centres. Side effects evaluation did not involve any specific scale. CONCLUSIONS Our findings support the safety and tolerability of esketamine in a real-world TRD sample. The later response and the non-inferiority in effectiveness in patients with comorbidities represent novel and interesting findings.
Collapse
Affiliation(s)
- Giovanni Martinotti
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D'Annunzio, Chieti, Italy; Psychopharmacology, Drug Misuse and Novel Psychoactive Substances Research Unit, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9AB, UK
| | - Antonio Vita
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Department of Mental Health and Addiction Services, ASST Spedali Civili of Brescia, Italy
| | - Andrea Fagiolini
- School of Medicine, Department of Molecular Medicine, University of Siena, Siena, Italy
| | - Giuseppe Maina
- Department of Neurosciences "Rita Levi Montalcini", University of Torino, Turin, Italy
| | | | - Bernardo Dell'Osso
- Department of Biomedical and Clinical Sciences Luigi Sacco and Aldo Ravelli Center for Neurotechnology and Brain Therapeutic, University of Milan, Milano, Italy
| | - Alberto Siracusano
- Chair of Psychiatry, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Massimo Clerici
- Dipartimento di Medicina e Chirurgia, Università degli studi Milano Bicocca, Italy; Dipartimento di Salute Mentale e Dipendenze ASST Monza, Italy
| | - Antonello Bellomo
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Gabriele Sani
- Department of Neuroscience, Section of Psychiatry, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giacomo d'Andrea
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D'Annunzio, Chieti, Italy.
| | - Roberto Delle Chiaie
- Department of Neuroscience and Mental Health-Policlinico Umberto I Hospital, Sapienza University of Rome, 00161 Rome, Italy
| | - Andreas Conca
- Psychiatric Service of the Health District of Bozen, Bozen-Bolzano, Italy
| | - Stefano Barlati
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Department of Mental Health and Addiction Services, ASST Spedali Civili of Brescia, Italy
| | - Giorgio Di Lorenzo
- Chair of Psychiatry, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Pasquale De Fazio
- Psychiatry Unit, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | | | - Giuseppe Nicolò
- Department of Mental Health and Addiction, ASL Roma 5, Rome, Italy
| | - Gianluca Rosso
- Department of Neurosciences "Rita Levi Montalcini", University of Torino, Turin, Italy
| | | | | | | | - Roberta Bassetti
- SPDC Frosinone - ASL, Frosinone, Italy; Department of Mental Health and Addiction Services, Niguarda Hospital, Milan, Italy
| | | | - Miriam Olivola
- Department of Brain and Behavioural Science, University of Pavia, Italy
| | - Sandro Belletti
- Mental Heath Department, Azienda Unità Sanitaria Locale (AUSL) Umbria 2, Italy
| | | | - Marco Di Nicola
- Department of Neuroscience, Section of Psychiatry, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Mauro Pettorruso
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D'Annunzio, Chieti, Italy
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, Toronto, ON, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada; Canadian Rapid Treatment Center of Excellence, Mississauga, ON, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Massimo di Giannantonio
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D'Annunzio, Chieti, Italy
| | | |
Collapse
|
49
|
Xu C, Xiong Q, Tian X, Liu W, Sun B, Ru Q, Shu X. Alcohol Exposure Induces Depressive and Anxiety-like Behaviors via Activating Ferroptosis in Mice. Int J Mol Sci 2022; 23:ijms232213828. [PMID: 36430312 PMCID: PMC9698590 DOI: 10.3390/ijms232213828] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
Alcohol use disorder (AUD) is a global public health problem and is frequently comorbid with mental disorders, including anxiety and depression. Ferroptosis is an iron-dependent cell death, which is involved in the pathological process of various diseases such as neurodegenerative diseases, but the role of ferroptosis in the mediation of AUD and its induced mental disorders is unclear. In this study, we aimed to investigate whether ferroptosis was involved in alcohol-induced depressive and anxiety-like behaviors in mice. Following an 8-week period of intermittent alcohol exposure, the alcohol group showed noticeable depressive and anxiety-like behaviors. In addition, nissl staining revealed that alcohol exposure induced neuron damage in the hippocampus (Hip) and prefrontal cortex (PFC) of mice. The levels of synapse-related proteins were significantly reduced in the alcohol group. Iron staining demonstrated that alcohol increased the number of iron-positive staining cells. The protein expression of the transferrin receptor (TFRC) was increased, and the expression of glutathione peroxidase 4 (GPX4) was decreased, respectively, in the alcohol group. Furthermore, the ferroptosis inhibitor ferrostatin-1 significantly prevented alcohol-induced neuron damage and enhanced the expression of N-methyl-d-aspartic acid (NMDA) receptor 2B (NR2B), α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) receptor 1 (GluA1) and GPX4 in vitro. These results indicated that alcohol exposure could induce depressive and anxiety-like behaviors, and that this effect may occur via activating ferroptosis.
Collapse
Affiliation(s)
- Congyue Xu
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430000, China
| | - Qi Xiong
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430000, China
| | - Xiang Tian
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430000, China
| | - Wei Liu
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430000, China
| | - Binlian Sun
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430000, China
| | - Qin Ru
- Department of Health and Physical Education, Jianghan University, Wuhan 430000, China
- Correspondence: (Q.R.); (X.S.); Tel.: +86-27-84225807 (X.S.)
| | - Xiji Shu
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430000, China
- Correspondence: (Q.R.); (X.S.); Tel.: +86-27-84225807 (X.S.)
| |
Collapse
|
50
|
Forcén FE, Marengo L, Behn M. Ketamine-assisted meaning-centered psychotherapy for a patient with severe suicidal behavior. Palliat Support Care 2022; 21:1-3. [PMID: 36193694 DOI: 10.1017/s1478951522001304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
There is a current debate in society as to whether depression can be a terminal illness. Meaning-centered psychotherapy (MCP) and psychedelic medicines have both been shown to treat existential distress in palliative care settings. We are reporting the case of a patient for whom MCP combined with ketamine was an effective treatment for his recurrent and severe depressive disorder with suicidal ideation. His complex assessment and management of suicide (CAMS) scores improved significantly with this treatment modality. Ketamine is generally well tolerated and can enhance treatment outcomes in patients undergoing MCP.
Collapse
Affiliation(s)
- Fernando Espí Forcén
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Laura Marengo
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Maya Behn
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|