1
|
Acero-Castillo MC, Correia MBM, Caixeta FV, Motta V, Barros M, Maior RS. Is the antidepressant effect of ketamine separate from its psychotomimetic effect? A review of rodent models. Neuropharmacology 2024; 258:110088. [PMID: 39032814 DOI: 10.1016/j.neuropharm.2024.110088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/09/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Ketamine is an NMDA (N-methyl-d-aspartate) glutamate receptor antagonist, which has a myriad of dose-dependent pharmacological and behavioral effects, including anesthetic, sedative, amnestic, analgesic, and anti-inflammatory properties. Intriguingly, ketamine at subanesthetic doses displays a relevant profile both in mimicking symptoms of schizophrenia and also as the first fast-acting treatment for depression. Here, we present an overview of the state-of-the-art knowledge about ketamine as an antidepressant as well as a pharmacological model of schizophrenia in animal models and human participants. Ketamine's dual effect appears to arise from its mechanism of action involving NMDA receptors, with both immediate and downstream consequences being triggered as a result. Finally, we discuss the feasibility of a unified approach linking the glutamatergic hypothesis of schizophrenia to the promising preclinical and clinical success of ketamine in the treatment of refractory depression.
Collapse
Affiliation(s)
- M C Acero-Castillo
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - M B M Correia
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil; Department of Anthropology, Emory University, Atlanta GA, ZIP 30322, USA
| | - F V Caixeta
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - V Motta
- Department of Basic Psychological Processes, Institute of Psychology, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - M Barros
- Department of Pharmacy, School of Health Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - R S Maior
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil.
| |
Collapse
|
2
|
Cavaleri D, Riboldi I, Crocamo C, Paglia G, Carrà G, Bartoli F. Evidence from preclinical and clinical metabolomics studies on the antidepressant effects of ketamine and esketamine. Neurosci Lett 2024; 831:137791. [PMID: 38670523 DOI: 10.1016/j.neulet.2024.137791] [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: 03/15/2024] [Revised: 04/15/2024] [Accepted: 04/23/2024] [Indexed: 04/28/2024]
Abstract
The antidepressant effects of ketamine and esketamine are well-documented. Nonetheless, most of the underlying molecular mechanisms have to be uncovered yet. In the last decade, metabolomics has emerged as a useful means to investigate the metabolic phenotype associated with depression as well as changes induced by antidepressant treatments. This mini-review aims at summarizing the main findings from preclinical and clinical studies that used metabolomics to investigate the metabolic effects of subanesthetic, antidepressant doses of ketamine and esketamine and their relationship with clinical response. Both animal and human studies report alterations in several metabolic pathways - including the tricarboxylic acid cycle, glycolysis, the pentose phosphate pathway, lipid metabolism, amino acid metabolism, the kynurenine pathway, and the urea cycle - following the administration of ketamine or its enantiomers. Although more research is needed to clarify commonalities and differences in molecular mechanisms of action between the racemic compound and its enantiomers, these findings comprehensively support an influence of ketamine and esketamine on mitochondrial and cellular energy production, membrane homeostasis, neurotransmission, and signaling. Metabolomics may thus represent a promising strategy to clarify molecular mechanisms underlying treatment-resistant depression and related markers of clinical response to ketamine and esketamine. This body of preclinical and clinical evidence, if further substantiated, has the potential to guide clinicians towards personalized approaches, contributing to new paradigms in the clinical management of depression.
Collapse
Affiliation(s)
- Daniele Cavaleri
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy
| | - Ilaria Riboldi
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy
| | - Cristina Crocamo
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy
| | - Giuseppe Paglia
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy
| | - Giuseppe Carrà
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy; Division of Psychiatry, University College London, 149 Tottenham Ct Rd, London W1T 7NF, United Kingdom
| | - Francesco Bartoli
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy.
| |
Collapse
|
3
|
Lineham A, Avila-Quintero VJ, Bloch MH, Dwyer J. Exploring Predictors of Ketamine Response in Adolescent Treatment-Resistant Depression. J Child Adolesc Psychopharmacol 2024; 34:73-79. [PMID: 38170185 PMCID: PMC11262580 DOI: 10.1089/cap.2023.0047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Objective: Ketamine has proved effective as a rapid-acting antidepressant agent, but treatment is not effective for everyone (approximately a quarter to a half of patients). Some adult studies have begun to investigate predictors of ketamine's antidepressant response, but no studies have examined this in adolescents with depression. Methods: We conducted a secondary data analysis of adolescents who participated in a randomized, single-dose, midazolam-controlled crossover trial of ketamine for adolescents with treatment-resistant depression. We examined the relationship between 19 exploratory demographic and clinical variables and depression symptom improvement (using the Montgomery-Åsberg Depression Rating Scale [MADRS]) at 1 and 7 days postinfusion. Results: Subjects who had fewer medication trials of both antidepressant medications and augmentation treatments were more likely to experience depression symptom improvement with ketamine. Subjects with shorter duration of their current depressive episode were more likely to experience depression symptom improvement with ketamine. Subjects currently being treated with selective serotonin reuptake inhibitor medications, and not being treated with serotonin-norepinephrine reuptake inhibitor medications, also experienced greater symptom improvement with ketamine. When receiving the midazolam control, less severe depressive symptoms, as measured by the Children's Depression Rating Scale (CDRS) (but not MADRS), and a comorbid attention-deficit/hyperactivity disorder diagnosis were associated with increased response. Conclusions: Findings should be viewed as preliminary and exploratory given the small sample size and multiple secondary analyses. Identifying meaningful predictors of ketamine response is important to inform future therapeutic use of this compound, however, considerably more research is warranted before such clinical guidance is established. The trial was registered in clinicaltrials.gov with the identifier NCT02579928.
Collapse
Affiliation(s)
- Alice Lineham
- Child Study Center, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Michael H. Bloch
- Child Study Center, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Psychiatry and Yale School of Medicine, New Haven, Connecticut, USA
| | - Jennifer Dwyer
- Child Study Center, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
4
|
Baj J, Bargieł J, Cabaj J, Skierkowski B, Hunek G, Portincasa P, Flieger J, Smoleń A. Trace Elements Levels in Major Depressive Disorder-Evaluation of Potential Threats and Possible Therapeutic Approaches. Int J Mol Sci 2023; 24:15071. [PMID: 37894749 PMCID: PMC10606638 DOI: 10.3390/ijms242015071] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
The multifactorial etiology of major depressive disorder (MDD) includes biological, environmental, genetic, and psychological aspects. Recently, there has been an increasing interest in metallomic studies in psychiatry, aiming to evaluate the role of chosen trace elements in the MDD etiology as well as the progression of symptoms. This narrative review aims to summarize the available literature on the relationship between the concentration of chosen elements in the serum of patients with MDD and the onset and progression of this psychiatric condition. The authors reviewed PubMed, Web of Science, and Scopus databases searching for elements that had been investigated so far and further evaluated them in this paper. Ultimately, 15 elements were evaluated, namely, zinc, magnesium, selenium, iron, copper, aluminium, cadmium, lead, mercury, arsenic, calcium, manganese, chromium, nickel, and phosphorus. The association between metallomic studies and psychiatry has been developing dynamically recently. According to the results of current research, metallomics might act as a potential screening tool for patients with MDD while at the same time providing an assessment of the severity of symptoms. Either deficiencies or excessive amounts of chosen elements might be associated with the progression of depressive symptoms or even the onset of the disease among people predisposed to MDD.
Collapse
Affiliation(s)
- Jacek Baj
- Department of Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland
| | - Julia Bargieł
- Student Research Group of Department of Epidemiology and Clinical Research Methodology, Medical University of Lublin, Radziwiłłowska 11, 20-080 Lublin, Poland; (J.B.); (J.C.); (B.S.)
| | - Justyna Cabaj
- Student Research Group of Department of Epidemiology and Clinical Research Methodology, Medical University of Lublin, Radziwiłłowska 11, 20-080 Lublin, Poland; (J.B.); (J.C.); (B.S.)
| | - Bartosz Skierkowski
- Student Research Group of Department of Epidemiology and Clinical Research Methodology, Medical University of Lublin, Radziwiłłowska 11, 20-080 Lublin, Poland; (J.B.); (J.C.); (B.S.)
| | - Gabriela Hunek
- Student Research Group of Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland;
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy;
| | - Jolanta Flieger
- Department of Analytical Chemistry, Medical University of Lublin, Chodźki 4A, 20-093 Lublin, Poland;
| | - Agata Smoleń
- Department of Epidemiology and Clinical Research Methodology, Medical University of Lublin, 20-080 Lublin, Poland;
| |
Collapse
|
5
|
Wang X, Zhu X, Ji X, Yang J, Zhou J. Group-Based Symptom Trajectory of Intramuscular Administration of Scopolamine Augmentation in Moderate to Severe Major Depressive Disorder: A Post-Hoc Analysis. Neuropsychiatr Dis Treat 2023; 19:1043-1053. [PMID: 37153351 PMCID: PMC10162387 DOI: 10.2147/ndt.s408794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/21/2023] [Indexed: 05/09/2023] Open
Abstract
Objective Developing new strategies for rapid and sustained relief of depressive symptom has been the focus of research in the field of major depressive disorder (MDD). Scopolamine exerts rapid antidepressant effect in recent years but is controversial. Therefore, we aimed to identify a sensitive patient who may respond to intramuscular injections of scopolamine added to antidepressants based on distinct trajectory patterns. Methods We analyzed longitudinal post hoc data collected from 66 MDD patients at Beijing Anding Hospital, Capital Medical University, over a 4-week period. In addition to demographics, depressive symptoms were assessed using the 16-item Quick Inventory of Depressive Symptomatology and Self-Report (QIDS-SR16) Scale and 17-item Hamilton Rating Scale for Depression (HRSD-17) following an i.m. injection of scopolamine. We explored different longitudinal patterns of depressive symptoms using a group-based trajectory model (GBTM). We used multiple logistic regression models to help identify predictors of different depressive symptom trajectories. Results A two-class GBTM was identified as optimal for classifying depressive symptoms: high/rapidly declining (39.4%) and moderate/gradually declining depression trajectories (60.6%) were distinguished based on the HRSD-17. The high/rapidly declining depression trajectory was characterized by high initial depression followed by a rapid decrease at the end of the study. The moderate/gradual decline trajectory was dominated by moderate depression and gradual decline over 4 weeks. There were no significant associations of age, gender, education, or age of onset with the two trajectory groups. Conclusion Scopolamine added to antidepressants can effectively relieve the symptoms of patients with severe depression, and it decreases faster than patients with moderate depression.
Collapse
Affiliation(s)
- Xiao Wang
- The National Clinical Research Center for Mental Disorders & Beijing Anding Hospital of Capital Medical University, Beijing, People’s Republic of China
- Beijing Key Laboratory of Mental Disorders & Beijing Anding Hospital of Capital Medical University, Beijing, People’s Republic of China
| | - Xuequan Zhu
- The National Clinical Research Center for Mental Disorders & Beijing Anding Hospital of Capital Medical University, Beijing, People’s Republic of China
- Beijing Key Laboratory of Mental Disorders & Beijing Anding Hospital of Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| | - Xiao Ji
- The National Clinical Research Center for Mental Disorders & Beijing Anding Hospital of Capital Medical University, Beijing, People’s Republic of China
- Beijing Key Laboratory of Mental Disorders & Beijing Anding Hospital of Capital Medical University, Beijing, People’s Republic of China
| | - Jian Yang
- The National Clinical Research Center for Mental Disorders & Beijing Anding Hospital of Capital Medical University, Beijing, People’s Republic of China
- Beijing Key Laboratory of Mental Disorders & Beijing Anding Hospital of Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
- Correspondence: Jian Yang; Jingjing Zhou, The National Clinical Research Center for Mental Disorders & Beijing Anding Hospital of Capital Medical University, 5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088, People’s Republic of China, Email ;
| | - Jingjing Zhou
- The National Clinical Research Center for Mental Disorders & Beijing Anding Hospital of Capital Medical University, Beijing, People’s Republic of China
- Beijing Key Laboratory of Mental Disorders & Beijing Anding Hospital of Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
6
|
Melloni EMT, Bravi B, Poletti S, Dallaspezia S, Barbini B, Zanardi R, Benedetti F. Antidepressant chronotherapeutics normalizes prefrontal 1H-MRS glutamate in bipolar depression. Prog Neuropsychopharmacol Biol Psychiatry 2022; 119:110606. [PMID: 35843368 DOI: 10.1016/j.pnpbp.2022.110606] [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: 03/02/2022] [Revised: 07/05/2022] [Accepted: 07/12/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND Dysfunctional glutamatergic neurotransmission has been proposed both, as a biological underpinning of mood disorder and as a target for rapid-acting antidepressant treatments. Total sleep deprivation and light therapy (TSD + LT) can prompt antidepressant response in drug-resistant bipolar depression. Here we explored the effects of TSD + LT on dorsolateral prefrontal cortex (DLPFC) glutamate and/or glutamine+glutamate (Glx) levels. METHODS We studied single voxel 1H-MRS measures of DLPFC Glu and Glx levels of 48 healthy participants and 55 inpatients with a major depressive episode in course of Bipolar Disorder, a subset of which (N = 23) underwent three cycles of repeated TSD + LT and were evaluated before and after treatment. Treatment effects of mood and on Glu and Glx concentrations were analyzed in the context of the Generalized Linear Model (GLM), correcting for age, sex and ongoing lithium treatment. RESULTS Higher concentration of Glu (adjusted Z = -2189, p = 0,0285) and Glx (adjusted Z = -3,13, p = 0,0017) were observed in BD patients compared to HC. Treatment caused a significant rapid reduction of depressive symptom severity over time (F = 63.98, p < 0.01). Change in depression levels after TSD + LT treatment was significantly influenced by delta change in Glu levels (LR χ2 = 4.619, p = 0.0316) and in Glx levels (LR χ2 = 4.486, p = 0.0341). CONCLUSION A reduction in Glu and Glx levels associated with depression could contribute to the mechanism of action of TSD + LT, directly acting on glutamatergic neurons, or to the interaction between the glutamatergic system and dopamine (DA) and serotonin (5-HT) levels, known to be targeted by TSD. This is in line with several studies showing a glutamatergic modulation effects of antidepressants and mood stabilizing agents. This finding deepens our understanding of antidepressant effect of chronoterapeutics.
Collapse
Affiliation(s)
- Elisa M T Melloni
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy.
| | - Beatrice Bravi
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy
| | - Sara Poletti
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy
| | - Sara Dallaspezia
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Barbara Barbini
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Raffaella Zanardi
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Francesco Benedetti
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy
| |
Collapse
|
7
|
Medeiros GC, Gould TD, Prueitt WL, Nanavati J, Grunebaum MF, Farber NB, Singh B, Selvaraj S, Machado-Vieira R, Achtyes ED, Parikh SV, Frye MA, Zarate CA, Goes FS. Blood-based biomarkers of antidepressant response to ketamine and esketamine: A systematic review and meta-analysis. Mol Psychiatry 2022; 27:3658-3669. [PMID: 35760879 PMCID: PMC9933928 DOI: 10.1038/s41380-022-01652-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 05/17/2022] [Accepted: 05/31/2022] [Indexed: 02/08/2023]
Abstract
(R,S)-ketamine (ketamine) and its enantiomer (S)-ketamine (esketamine) can produce rapid and substantial antidepressant effects. However, individual response to ketamine/esketamine is variable, and there are no well-accepted methods to differentiate persons who are more likely to benefit. Numerous potential peripheral biomarkers have been reported, but their current utility is unclear. We conducted a systematic review/meta-analysis examining the association between baseline levels and longitudinal changes in blood-based biomarkers, and response to ketamine/esketamine. Of the 5611 citations identified, 56 manuscripts were included (N = 2801 participants), and 26 were compatible with meta-analytical calculations. Random-effect models were used, and effect sizes were reported as standardized mean differences (SMD). Our assessments revealed that more than 460 individual biomarkers were examined. Frequently studied groups included neurotrophic factors (n = 15), levels of ketamine and ketamine metabolites (n = 13), and inflammatory markers (n = 12). There were no consistent associations between baseline levels of blood-based biomarkers, and response to ketamine. However, in a longitudinal analysis, ketamine responders had statistically significant increases in brain-derived neurotrophic factor (BDNF) when compared to pre-treatment levels (SMD [95% CI] = 0.26 [0.03, 0.48], p = 0.02), whereas non-responders showed no significant changes in BDNF levels (SMD [95% CI] = 0.05 [-0.19, 0.28], p = 0.70). There was no consistent evidence to support any additional longitudinal biomarkers. Findings were inconclusive for esketamine due to the small number of studies (n = 2). Despite a diverse and substantial literature, there is limited evidence that blood-based biomarkers are associated with response to ketamine, and no current evidence of clinical utility.
Collapse
Affiliation(s)
- Gustavo C. Medeiros
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA.,Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Todd D. Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Departments of Pharmacology and Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA.,Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | | | - Julie Nanavati
- Welch Medical Library, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael F. Grunebaum
- Columbia University Irving Medical Center and New York State Psychiatric Institute, New York City, NY, USA
| | - Nuri B. Farber
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Balwinder Singh
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Sudhakar Selvaraj
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Rodrigo Machado-Vieira
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Eric D. Achtyes
- Division of Psychiatry and Behavioral Medicine, Michigan State University College of Human Medicine, Grand Rapids, MI, USA.,Pine Rest Christian Mental Health Services, Grand Rapids, MI, USA
| | - Sagar V. Parikh
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Mark A. Frye
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Carlos A. Zarate
- Experimental Therapeutics & Pathophysiology Branch, NIMH-NIH, Bethesda, MD, USA
| | - Fernando S. Goes
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA.,Correspondence and requests for materials should be addressed to Fernando S. Goes.,
| |
Collapse
|
8
|
Cano M, Cardoner N. Biomarkers of response to rapid-acting antidepressants. Eur Neuropsychopharmacol 2021; 53:101-103. [PMID: 34536713 DOI: 10.1016/j.euroneuro.2021.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/08/2021] [Accepted: 06/11/2021] [Indexed: 11/29/2022]
Affiliation(s)
- Marta Cano
- Mental Health Department, Unitat de Neurociència Traslacional, Parc Tauli University Hospital, Institut d'Investigació i Innovació Sanitària Parc Taulí (I3PT), Barcelona, Spain; CIBERSAM, Carlos III Health Institute, Madrid, Spain; Department of Psychobiology and Methodology of Health Sciences, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Narcís Cardoner
- Mental Health Department, Unitat de Neurociència Traslacional, Parc Tauli University Hospital, Institut d'Investigació i Innovació Sanitària Parc Taulí (I3PT), Barcelona, Spain; CIBERSAM, Carlos III Health Institute, Madrid, Spain; Department of Psychiatry and Forensic Medicine, School of Medicine Bellaterra, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
9
|
Benedetti F, Branchi I, Poletti S, Lorenzi C, Bigai G, Colombo C, Zanardi R. Adiponectin predicts poor response to antidepressant drugs in major depressive disorder. Hum Psychopharmacol 2021; 36:e2793. [PMID: 33945186 DOI: 10.1002/hup.2793] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/26/2021] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Produced by adipocytes, adiponectin crosses the blood-brain barrier to bind with specific receptors in the hypothalamus, brainstem, hippocampus, and cortex. In patients with major depressive disorder (MDD), circulating levels of adiponectin inversely related with antidepressant response to ketamine, and predicted a better response to multi-target drug combinations than to escitalopram. We investigated the effect of adiponectin on response to antidepressants in a naturalistic setting. METHODS We assessed baseline plasma levels of adiponectin in 121 MDD inpatients, treated with antidepressant drug monotherapy based on clinical need (selective serotonin reuptake inhibitors, venlafaxine, duloxetine) in a specialized hospital setting. Severity of depression was weekly assessed with Hamilton scale ratings. RESULTS Adiponectin plasma levels were higher in patients with MDD compared with healthy controls, and negatively influenced the pattern of antidepressant response (higher baseline levels, worse response) independent of the drug class and of the baseline severity of depression, and of age, sex, and body mass index. CONCLUSIONS The identification of adiponectin as a predictor of antidepressant response to drugs of different mechanism of action, such as ketamine, SSRIs, and SNRIs, and both in experimental and in clinical settings, warrants interest for further study of its pathways to search for novel biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Francesco Benedetti
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy.,Vita-Salute San Raffaele University, Milano, Italy
| | - Igor Branchi
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Roma, Italy
| | - Sara Poletti
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy.,Vita-Salute San Raffaele University, Milano, Italy
| | - Cristina Lorenzi
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | | | - Cristina Colombo
- Vita-Salute San Raffaele University, Milano, Italy.,Mood Disorders Unit, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Raffaella Zanardi
- Vita-Salute San Raffaele University, Milano, Italy.,Mood Disorders Unit, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| |
Collapse
|
10
|
Williams RJ, Brown EC, Clark DL, Pike GB, Ramasubbu R. Early post-treatment blood oxygenation level-dependent responses to emotion processing associated with clinical response to pharmacological treatment in major depressive disorder. Brain Behav 2021; 11:e2287. [PMID: 34333866 PMCID: PMC8413787 DOI: 10.1002/brb3.2287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 06/21/2021] [Accepted: 07/02/2021] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION Pre-treatment blood oxygenation level-dependent (BOLD) functional magnetic resonance imaging (fMRI) has been used for the early identification of patients with major depressive disorder (MDD) who later respond or fail to respond to medication. However, BOLD responses early after treatment initiation may offer insight into early neural changes associated with later clinical response. The present study evaluated both pre-treatment and early post-treatment fMRI responses to an emotion processing task, to further our understanding of neural changes associated with a successful response to pharmacological intervention. METHODS MDD patients who responded (n = 22) and failed to respond (n = 12) after 8 weeks of treatment with either citalopram or quetiapine extended release, and healthy controls (n = 18) underwent two fMRI scans, baseline (pre-treatment), and early post-treatment (one week after treatment commencement). Participants completed an emotional face matching task at both scans. RESULTS Using threshold-free cluster enhancement (TFCE) and non-parametric permutation testing, fMRI activation maps showed that after one week of treatment, responders demonstrated increased activation in the left parietal lobule, precentral gyrus, and bilateral insula (all P < 0.05 threshold-free cluster enhancement (TFCE) family-wise error-corrected) to negative facial expressions. Non-responders showed some small increases in the precentral gyrus, while controls showed no differences between scans. Compared to non-responders, responders showed some increased activation in the superior parietal lobule and middle temporal gyrus at the post-treatment scan. There were no group differences between responders, non-responders, and controls at baseline. CONCLUSIONS One week after treatment commencement, BOLD signal changes in the parietal lobules, insula, and middle temporal gyrus were related to clinical response to pharmacological treatment.
Collapse
Affiliation(s)
- Rebecca J Williams
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Department of Radiology, University of Calgary, Calgary, Alberta, Canada
| | - Elliot C Brown
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta, Canada.,Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada.,Department of Psychiatry, University of Calgary, Calgary, Alberta, Canada.,Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Neuroscience Research Center, Berlin, Germany
| | - Darren L Clark
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta, Canada.,Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada.,Department of Psychiatry, University of Calgary, Calgary, Alberta, Canada
| | - G Bruce Pike
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Department of Radiology, University of Calgary, Calgary, Alberta, Canada.,Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Rajamannar Ramasubbu
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta, Canada.,Department of Psychiatry, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
11
|
Meyer T, Brunovsky M, Horacek J, Novak T, Andrashko V, Seifritz E, Olbrich S. Predictive value of heart rate in treatment of major depression with ketamine in two controlled trials. Clin Neurophysiol 2021; 132:1339-1346. [PMID: 33888426 DOI: 10.1016/j.clinph.2021.01.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 12/30/2020] [Accepted: 01/09/2021] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Ketamine has been shown to be effective in treatment of episodes of major depressive disorder (MDD). This controlled study aimed to analyse the predictive and discriminative power of heart rate (HR) and heart rate variability (HRV) for ketamine treatment in MDD. METHODS In 51 patients, HR and HRV were assessed at baseline before and during ketamine infusion and 24 hours post ketamine infusion. Montgomery-Åsberg Depression Rating Scale (MADRS) was used to assess changes of depressive symptoms. A 30% or 50% reduction of symptoms after 24 hours or within 7 days was defined as response. A linear mixed model was used for analysis. RESULTS Ketamine infusion increased HR and HRV power during and after infusion. Responders to ketamine showed a higher HR during the whole course of investigation, including at baseline with medium effect sizes (Cohen's d = 0.47-0.67). Furthermore, HR and HRV power discriminated between responders and non-responders, while normalized low and high frequencies did not. CONCLUSION The findings show a predictive value of HR and HRV power for ketamine treatment. This further underlines the importance of the autonomous nervous system (ANS) and its possible malfunctions in MDD. SIGNIFICANCE The predictive power of HR and HRV markers should be studied in prospective studies. Neurophysiological markers could improve treatment for MDD via optimizing the choice of treatments.
Collapse
Affiliation(s)
- Torsten Meyer
- Department for Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry Zurich, Switzerland
| | - Martin Brunovsky
- National Institute of Mental Health, Klecany, Czech Republic; Charles University, Third Faculty of Medicine, Prague, Czech Republic
| | - Jiri Horacek
- National Institute of Mental Health, Klecany, Czech Republic; Charles University, Third Faculty of Medicine, Prague, Czech Republic
| | - Tomas Novak
- National Institute of Mental Health, Klecany, Czech Republic; Charles University, Third Faculty of Medicine, Prague, Czech Republic
| | - Veronika Andrashko
- National Institute of Mental Health, Klecany, Czech Republic; Charles University, Third Faculty of Medicine, Prague, Czech Republic
| | - Erich Seifritz
- Department for Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry Zurich, Switzerland
| | - Sebastian Olbrich
- Department for Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry Zurich, Switzerland.
| |
Collapse
|
12
|
Hoepner CT, McIntyre RS, Papakostas GI. Impact of Supplementation and Nutritional Interventions on Pathogenic Processes of Mood Disorders: A Review of the Evidence. Nutrients 2021; 13:nu13030767. [PMID: 33652997 PMCID: PMC7996954 DOI: 10.3390/nu13030767] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/10/2021] [Accepted: 02/20/2021] [Indexed: 02/06/2023] Open
Abstract
This narrative review was conducted using searches of the PubMed/Medline and Google Scholar databases from inception to November 2019. Clinical trials and relevant articles were identified by cross-referencing major depressive disorder (and/or variants) with the following terms: folate, homocysteine, S-adenosylmethionine (SAMe), L-acetylcarnitine, alpha-lipoic acid, N-acetylcysteine, L-tryptophan, zinc, magnesium, vitamin D, omega-3 fatty acids, coenzyme Q10, and inositol. Manual reviews of references were also performed using article reference lists. Abnormal levels of folate, homocysteine, and SAMe have been shown to be associated with a higher risk of depression. Numerous studies have demonstrated antidepressant activity with L-methylfolate and SAMe supplementation in individuals with depression. Additionally, the amino acids L-acetylcarnitine, alpha-lipoic acid, N-acetylcysteine, and L-tryptophan have been implicated in the development of depression and shown to exert antidepressant effects. Other agents with evidence for improving depressive symptoms include zinc, magnesium, omega-3 fatty acids, and coenzyme Q10. Potential biases and differences in study designs within and amongst the studies and reviews selected may confound results. Augmentation of antidepressant medications with various supplements targeting nutritional and physiological factors can potentiate antidepressant effects. Medical foods, particularly L-methylfolate, and other supplements may play a role in managing depression in patients with inadequate response to antidepressant therapies.
Collapse
Affiliation(s)
- Cara T. Hoepner
- Bay Area Psychiatric, A Nursing Corporation, San Francisco, CA 94111, USA
- Correspondence:
| | - Roger S. McIntyre
- Mood Disorders Psychopharmacology Unit, University of Toronto, Toronto, ON M5T 2S8, Canada;
| | | |
Collapse
|
13
|
Astrocyte Intracellular Ca 2+and TrkB Signaling in the Hippocampus Could Be Involved in the Beneficial Behavioral Effects of Antidepressant Treatment. Neurotox Res 2021; 39:860-871. [PMID: 33616872 DOI: 10.1007/s12640-021-00334-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/18/2021] [Accepted: 01/21/2021] [Indexed: 12/11/2022]
Abstract
Although monoaminergic-based antidepressant drugs are largely used to treat major depressive disorder (MDD), their mechanisms are still incompletely understood. Intracellular Ca2+ (iCa2+) and Calmodulin 1(CaM-1) homeostasis have been proposed to participate in the therapeutic effects of these compounds. We investigated whether intra-hippocampal inhibition of CaM-1 would modulate the behavioral responses to chronic treatment with imipramine (IMI) or 7-nitroindazole (7-NI), a selective inhibitor of the neuronal nitric oxide synthase 1 (NOS1) enzyme that shows antidepressant-like effects. We also investigated the interactions of IMI and CaM-1 on transient astrocyte iCa2+ evoked by glutamate stimuli. Intra-hippocampal microinjection of the lentiviral delivered (LV) short hairpin iRNA-driven against the CaM-1 mRNA (LV-shRNA-CaM-1) or the CaM-1 inhibitor N-(6-aminohexyl)-5-chloro-1-naphthalene sulphonamide (W-7) blocked the antidepressant-like effect of chronic treatment with IMI or 7-NI. The shRNA also inhibited the mRNA expression of the tropomyosin receptor kinase B (TrkB) in the microinjection region. The iCa2+ in ex vivo hippocampus slices stained with fluorescent Ca2+indicator Oregon Green 488 BAPTA-1 revealed that IMI increased the intensity and duration of iCa2+ oscillation and reduced the number of events evoked by glutamate stimuli, evaluated by using CCD imaging and the % ΔF/Fo parameters. The pre-treatment with W-7 fully antagonized this effect. The present results indicate that the behavioral benefits of chronic antidepressant treatment might be associated with astrocyte intracellular Ca2+dynamics and TrkB mRNA expression in the hippocampus.
Collapse
|
14
|
Benedetti F, Poletti S, Vai B, Mazza MG, Lorenzi C, Brioschi S, Aggio V, Branchi I, Colombo C, Furlan R, Zanardi R. Higher baseline interleukin-1β and TNF-α hamper antidepressant response in major depressive disorder. Eur Neuropsychopharmacol 2021; 42:35-44. [PMID: 33191075 DOI: 10.1016/j.euroneuro.2020.11.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/18/2020] [Accepted: 11/06/2020] [Indexed: 01/06/2023]
Abstract
Raised pro-inflammatory immune/inflammatory setpoints, leading to an increased production of peripheral cytokines, have been associated with Major Depressive Disorder (MDD) and with failure to respond to first-line antidepressant drugs. However, the usefulness of these biomarkers in clinical psychopharmacology has been questioned because single findings did not translate into the clinical practice, where patients are prescribed treatments upon clinical need. We studied a panel of 27 inflammatory biomarkers in a sample of 108 inpatients with MDD, treated with antidepressant monotherapy for 4 weeks upon clinical need in a specialized hospital setting, and assessed the predictive effect of baseline peripheral measures of inflammation on antidepressing efficacy (response rates and time-lagged pattern of decrease of depression severity) using a machine-learning approach with elastic net penalized regression, and multivariate analyses in the context of the general linear model. When considering both categorical and continuous measures of response, baseline levels of IL-1β predicted non-response to antidepressants, with the predicted probability to respond being highly dispersed at low levels of IL-1β, and stratifying toward non-response when IL-1β is high. Significant negative effects were also detected for TNF-α, while IL-12 weakly predicted response. These findings support the usefulness of inflammatory biomarkers in the clinical psychopharmacology of depression, and add to ongoing research efforts aiming at defining reliable cutoff values to identify depressed patients in clinical settings with high inflammation, and low probability to respond.
Collapse
Affiliation(s)
- Francesco Benedetti
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy.
| | - Sara Poletti
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy
| | - Benedetta Vai
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy; Fondazione Centro San Raffaele, Milano, Italy
| | - Mario Gennaro Mazza
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy
| | - Cristina Lorenzi
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Silvia Brioschi
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Veronica Aggio
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy
| | - Igor Branchi
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Cristina Colombo
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy
| | - Roberto Furlan
- Vita-Salute San Raffaele University, Milano, Italy; Clinical Neuroimmunology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Raffaella Zanardi
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy
| |
Collapse
|
15
|
Deng ZD, Luber B, Balderston NL, Velez Afanador M, Noh MM, Thomas J, Altekruse WC, Exley SL, Awasthi S, Lisanby SH. Device-Based Modulation of Neurocircuits as a Therapeutic for Psychiatric Disorders. Annu Rev Pharmacol Toxicol 2020; 60:591-614. [PMID: 31914895 DOI: 10.1146/annurev-pharmtox-010919-023253] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Device-based neuromodulation of brain circuits is emerging as a promising new approach in the study and treatment of psychiatric disorders. This work presents recent advances in the development of tools for identifying neurocircuits as therapeutic targets and in tools for modulating neurocircuits. We review clinical evidence for the therapeutic efficacy of circuit modulation with a range of brain stimulation approaches, including subthreshold, subconvulsive, convulsive, and neurosurgical techniques. We further discuss strategies for enhancing the precision and efficacy of neuromodulatory techniques. Finally, we survey cutting-edge research in therapeutic circuit modulation using novel paradigms and next-generation devices.
Collapse
Affiliation(s)
- Zhi-De Deng
- Noninvasive Neuromodulation Unit, Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892, USA; .,Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Bruce Luber
- Noninvasive Neuromodulation Unit, Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Nicholas L Balderston
- Section on Neurobiology of Fear and Anxiety, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Melbaliz Velez Afanador
- Noninvasive Neuromodulation Unit, Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Michelle M Noh
- Noninvasive Neuromodulation Unit, Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Jeena Thomas
- Noninvasive Neuromodulation Unit, Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - William C Altekruse
- Noninvasive Neuromodulation Unit, Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Shannon L Exley
- Noninvasive Neuromodulation Unit, Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Shriya Awasthi
- Noninvasive Neuromodulation Unit, Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Sarah H Lisanby
- Noninvasive Neuromodulation Unit, Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892, USA; .,Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina 27710, USA
| |
Collapse
|
16
|
Melloni EMT, Poletti S, Dallaspezia S, Bollettini I, Vai B, Barbini B, Zanardi R, Colombo C, Benedetti F. Changes of white matter microstructure after successful treatment of bipolar depression. J Affect Disord 2020; 274:1049-1056. [PMID: 32663931 DOI: 10.1016/j.jad.2020.05.146] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/22/2020] [Accepted: 05/27/2020] [Indexed: 01/31/2023]
Abstract
BACKGROUND Diffusion tensor imaging (DTI) measures suggest a widespread alteration of white matter (WM) microstructure in patients with bipolar disorder (BD). The chronotherapeutic combination of repeated total sleep deprivation and morning light therapy (TSD+LT) can acutely reverse depressive symptoms in approximately 60% of patients, and it has been confirmed as a model antidepressant treatment to investigate the neurobiological correlates of rapid antidepressant response. METHODS We tested if changes in DTI measures of WM microstructure could parallel antidepressant response in a sample of 44 patients with a major depressive episode in course of BD, treated with chronoterapeutics for one week. We used both a tract-wise and a voxel-wise approach for the whole-brain extraction of DTI measures of WM microstructure: axial (AD), radial (RD), and mean diffusivity (MD), and fractional anisotropy (FA). RESULTS Compared to baseline level, at one-week follow up we observed a significant increase in average FA measures paralleled by a significant decrease in MD measures of several WM tracts including cingulum, corpus callosum, corona radiata, cortico-spinal tract, internal capsule, fornix and uncinate fasciculus. The degree of change was associated to clinical response. CONCLUSIONS This is the first study to show changes of individual DTI measures of WM microstructure in response to antidepressant treatment in BD. Our results add new evidence to warrant a role for chronotherapeutics as a first-line treatment for bipolar depression and contribute identifying generalizable neuroimaging-based biomarkers of antidepressant response.
Collapse
Affiliation(s)
- Elisa M T Melloni
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy.
| | - Sara Poletti
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy
| | - Sara Dallaspezia
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Irene Bollettini
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Benedetta Vai
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy; Fondazione Centro San Raffaele, Milano, Italy
| | - Barbara Barbini
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Raffaella Zanardi
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Cristina Colombo
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy
| | - Francesco Benedetti
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy
| |
Collapse
|
17
|
Dimick MK, Omrin D, MacIntosh BJ, Mitchell RHB, Riegert D, Levitt A, Schaffer A, Belo S, Iazzetta J, Detzler G, Choi M, Choi S, Orser BA, Goldstein BI. Nitrous oxide as a putative novel dual-mechanism treatment for bipolar depression: Proof-of-concept study design and methodology. Contemp Clin Trials Commun 2020; 19:100600. [PMID: 32637725 PMCID: PMC7327241 DOI: 10.1016/j.conctc.2020.100600] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 06/10/2020] [Accepted: 06/21/2020] [Indexed: 12/23/2022] Open
Abstract
Introduction Depressive symptoms predominate in the course of bipolar disorder (BD) and there is an urgent need to evaluate novel application of repurposed compounds that act on pre-specified treatment targets. Several lines of reasoning suggest that nitrous oxide (N2O) is an ideal medication to study as a potential treatment and as a strategy to identify the underlying pathophysiology of bipolar depression. N2O is a potent cerebral vasodilator and there is compelling evidence of reduced frontal cerebral blood flow (CBF; i.e. hypoperfusion) in depression. Therefore, N2O may increase CBF and thereby improve symptoms of depression. The goal of this randomized, double-blind trial is to study the effect of a single administration of N2O versus the active comparator midazolam on mood and CBF in adults with treatment-resistant bipolar depression. Methods Participants with BD-I/-II currently experiencing a major depressive episode will be randomized to one of two conditions (n = 20/group): 1) inhaled N2O plus intravenous saline, or 2) inhaled room air plus intravenous midazolam. Montgomery-Asberg Depression Rating Scale scores will serve as the primary endpoint. CBF will be measured via arterial spin labelling magnetic resonance imaging. Conclusions N2O is a potential novel treatment for bipolar depression, as it causes cerebral vasodilation. This proof-of-concept study will provide valuable information regarding the acute impact of N2O on mood and on CBF. If N2O proves to be efficacious in future larger-scale trials, its ubiquity, safety, low cost, and ease of use suggest that it has great potential to become a game-changing acute treatment for bipolar depression.
Collapse
Affiliation(s)
- Mikaela K Dimick
- Pharmacology and Toxicology Department, University of Toronto, Toronto, Ontario, Canada.,Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Danielle Omrin
- Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Bradley J MacIntosh
- Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Rachel H B Mitchell
- Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Riegert
- Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.,Department of Anesthesia, University of Toronto, Toronto, Ontario, Canada
| | - Anthony Levitt
- Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Ayal Schaffer
- Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Susan Belo
- Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.,Department of Anesthesia, University of Toronto, Toronto, Ontario, Canada
| | - John Iazzetta
- Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.,Pharmacy Department, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | | | - Mabel Choi
- Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.,Department of Anesthesia, University of Toronto, Toronto, Ontario, Canada
| | - Stephen Choi
- Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.,Department of Anesthesia, University of Toronto, Toronto, Ontario, Canada
| | - Beverley A Orser
- Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.,Department of Anesthesia, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin I Goldstein
- Pharmacology and Toxicology Department, University of Toronto, Toronto, Ontario, Canada.,Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
18
|
Abstract
LEARNING OBJECTIVE After participating in this activity, learners should be better able to evaluate the evidence supporting the antidepressant effects of glutamatergic modulators.Both preclinical and clinical studies have implicated glutamatergic system dysfunction in the pathophysiology of mood disorders such as bipolar depression and major depressive disorder. In particular, rapid reductions in depressive symptoms have been noted in response to subanesthetic doses of the glutamatergic modulator ketamine in subjects with major depressive disorder or bipolar depression. These results have prompted the repurposing or development of other glutamatergic modulators, both as monotherapy or adjunctive to other therapies. Here, we highlight the evidence supporting the antidepressant effects of various glutamatergic modulators, including (1) broad glutamatergic modulators (ketamine, esketamine, dextromethorphan, dextromethorphan-quinidine [Nuedexta], AVP-786, nitrous oxide [N2O], AZD6765), (2) subunit (NR2B)-specific N-methyl-D-aspartate (NMDA) receptor antagonists (CP-101,606/traxoprodil, MK-0657 [CERC-301]), (3) glycine-site partial agonists (D-cycloserine, GLYX-13, sarcosine, AV-101), and (4) metabotropic glutamate receptor modulators (AZD2066, RO4917523/basimglurant, JNJ40411813/ADX71149, R04995819 [RG1578]).
Collapse
Affiliation(s)
- Ioline D Henter
- From the Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD
| | | | | |
Collapse
|
19
|
Park L, Furey M, Nugent AC, Farmer C, Ellis J, Szczepanik J, Lener MS, Zarate CA. Neurophysiological Changes Associated with Antidepressant Response to Ketamine Not Observed in a Negative Trial of Scopolamine in Major Depressive Disorder. Int J Neuropsychopharmacol 2019; 22:10-18. [PMID: 30184133 PMCID: PMC6313153 DOI: 10.1093/ijnp/pyy051] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 05/09/2018] [Accepted: 07/27/2018] [Indexed: 12/11/2022] Open
Abstract
Background This randomized, placebo-controlled, crossover trial examined the antidepressant efficacy of the muscarinic antagonist scopolamine in major depressive disorder subjects with more severe and refractory forms of major depressive disorder relative to previous reports. Methods Participants included 23 medication-free major depressive disorder subjects (12 F/11 M, 20-55 years) currently experiencing a major depressive episode. Subjects had scored ≥20 on the Montgomery-Asberg Depression Rating Scale. Following a single-blind, placebo lead-in, participants were randomized to receive 2 counterbalanced blocks of 3 i.v. infusions of scopolamine (4 μg/kg) and placebo in a double-blind manner. The primary and secondary outcomes were the Montgomery-Asberg Depression Rating Scale and the Hamilton Anxiety Rating Scale, respectively. Magnetoencephalography and plasma brain-derived neurotrophic factor concentrations were obtained prior to and after each treatment phase. Results As assessed by both the Montgomery-Asberg Depression Rating Scale and Hamilton Anxiety Rating Scale, scopolamine had no significant antidepressant or anxiolytic effects relative to placebo. No significant drug vs placebo effects were seen in magnetoencephalography gamma power or brain-derived neurotrophic factor plasma concentrations, and brain-derived neurotrophic factor changes did not correlate with change in Montgomery-Asberg Depression Rating Scale score in response to scopolamine. Conclusions These results do not support the efficacy of scopolamine for more severe or refractory forms of depression. No pre- to post-infusion changes in plasma brain-derived neurotrophic factor were detected, and magnetoencephalography gamma power changed only in the placebo lead-in, suggesting that these biomarker measures were not affected by scopolamine in this cohort. While difficult to interpret given the lack of antidepressant response, the findings suggest that the neurobiological effects of ketamine and scopolamine are at least partly distinct.
Collapse
Affiliation(s)
- Lawrence Park
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Maura Furey
- Janssen Research and Development, LLC, La Jolla, California
| | - Allison C Nugent
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Cristan Farmer
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Jessica Ellis
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Joanna Szczepanik
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Marc S Lener
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
20
|
Guo W, Machado-Vieira R, Mathew S, Murrough JW, Charney DS, Grunebaum M, Oquendo MA, Kadriu B, Akula N, Henter I, Yuan P, Merikangas K, Drevets W, Furey M, Mann JJ, McMahon FJ, Zarate CA, Shugart YY. Exploratory genome-wide association analysis of response to ketamine and a polygenic analysis of response to scopolamine in depression. Transl Psychiatry 2018; 8:280. [PMID: 30552317 PMCID: PMC6294748 DOI: 10.1038/s41398-018-0311-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/30/2018] [Accepted: 09/07/2018] [Indexed: 12/13/2022] Open
Abstract
Growing evidence suggests that the glutamatergic modulator ketamine has rapid antidepressant effects in treatment-resistant depressed subjects. The anticholinergic agent scopolamine has also shown promise as a rapid-acting antidepressant. This study applied genome-wide markers to investigate the role of genetic variants in predicting acute antidepressant response to both agents. The ketamine-treated sample included 157 unrelated European subjects with major depressive disorder (MDD) or bipolar disorder (BD). The scopolamine-treated sample comprised 37 unrelated European subjects diagnosed with either MDD or BD who had a current Major Depressive Episode (MDE), and had failed at least two adequate treatment trials for depression. Change in Montgomery-Asberg Depression Rating Scale (MADRS) or the 17-item Hamilton Depression Rating Scale (HAM-D) scale scores at day 1 (24 h post-treatment) was considered the primary outcome. Here, we conduct pilot genome-wide association study (GWAS) analyses to identify potential markers of ketamine response and dissociative side effects. Polygenic risk score analysis of SNPs ranked by the strength of their association with ketamine response was then calculated in order to assess whether common genetic markers from the ketamine study could predict response to scopolamine. Findings require replication in larger samples in light of low power of analyses of these small samples. Neverthless, these data provide a promising illustration of our future potential to identify genetic variants underlying rapid treatment response in mood disorders and may ultimately guide individual patient treatment selection in the future.
Collapse
Affiliation(s)
- Wei Guo
- Statistical Genomics and Data Analysis Core, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Rodrigo Machado-Vieira
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Sanjay Mathew
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - James W Murrough
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dennis S Charney
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew Grunebaum
- Columbia University Medical Center/New York State Psychiatric Institute, New York, NY, USA
| | - Maria A Oquendo
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bashkim Kadriu
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Nirmala Akula
- Human Genetics Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Ioline Henter
- Section on PET Neuroimaging Sciences, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Peixiong Yuan
- Human Genetics Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Kathleen Merikangas
- Genetic Epidemiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Wayne Drevets
- Janssen Pharmaceuticals, Neuroscience Research and Development, La Jolla, CA, USA
| | - Maura Furey
- Janssen Pharmaceuticals, Neuroscience Research and Development, La Jolla, CA, USA
| | - J John Mann
- Departments of Psychiatry and Radiology, College of Physicians and Surgeons, Columbia University, New York State Psychiatric Institute, New York, NY, USA
| | - Francis J McMahon
- Human Genetics Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Yin Yao Shugart
- Statistical Genomics and Data Analysis Core, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
21
|
Shalbaf R, Brenner C, Pang C, Blumberger DM, Downar J, Daskalakis ZJ, Tham J, Lam RW, Farzan F, Vila-Rodriguez F. Non-linear Entropy Analysis in EEG to Predict Treatment Response to Repetitive Transcranial Magnetic Stimulation in Depression. Front Pharmacol 2018; 9:1188. [PMID: 30425640 PMCID: PMC6218964 DOI: 10.3389/fphar.2018.01188] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 09/28/2018] [Indexed: 12/12/2022] Open
Abstract
Background: Biomarkers that predict clinical outcomes in depression are essential for increasing the precision of treatments and clinical outcomes. The electroencephalogram (EEG) is a non-invasive neurophysiological test that has promise as a biomarker sensitive to treatment effects. The aim of our study was to investigate a novel non-linear index of resting state EEG activity as a predictor of clinical outcome, and compare its predictive capacity to traditional frequency-based indices. Methods: EEG was recorded from 62 patients with treatment resistant depression (TRD) and 25 healthy comparison (HC) subjects. TRD patients were treated with excitatory repetitive transcranial magnetic stimulation (rTMS) to the dorsolateral prefrontal cortex (DLPFC) for 4 to 6 weeks. EEG signals were first decomposed using the empirical mode decomposition (EMD) method into band-limited intrinsic mode functions (IMFs). Subsequently, Permutation Entropy (PE) was computed from the obtained second IMF to yield an index named PEIMF2. Receiver Operator Characteristic (ROC) curve analysis and ANOVA test were used to evaluate the efficiency of this index (PEIMF2) and were compared to frequency-band based methods. Results: Responders (RP) to rTMS exhibited an increase in the PEIMF2 index compared to non-responders (NR) at F3, FCz and FC3 sites (p < 0.01). The area under the curve (AUC) for ROC analysis was 0.8 for PEIMF2 index for the FC3 electrode. The PEIMF2 index was superior to ordinary frequency band measures. Conclusion: Our data show that the PEIMF2 index, yields superior outcome prediction performance compared to traditional frequency band indices. Our findings warrant further investigation of EEG-based biomarkers in depression; specifically entropy indices applied in band-limited EEG components. Registration in ClinicalTrials.Gov; identifiers NCT02800226 and NCT01887782.
Collapse
Affiliation(s)
- Reza Shalbaf
- Non-Invasive Neurostimulation Therapies (NINET) Laboratory, Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Colleen Brenner
- Department of Psychology, Loma Linda University, Loma Linda, CA, United States
| | - Christopher Pang
- Non-Invasive Neurostimulation Therapies (NINET) Laboratory, Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Daniel M Blumberger
- Temerty Centre for Therapeutic Brain Intervention and Campbell Family Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Jonathan Downar
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,MRI-Guided rTMS Clinic and Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Zafiris J Daskalakis
- Temerty Centre for Therapeutic Brain Intervention and Campbell Family Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Joseph Tham
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Raymond W Lam
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Faranak Farzan
- School of Mechatronic Systems Engineering, Simon Fraser University, Surrey, BC, Canada
| | - Fidel Vila-Rodriguez
- Non-Invasive Neurostimulation Therapies (NINET) Laboratory, Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
22
|
Saland SK, Kabbaj M. Sex Differences in the Pharmacokinetics of Low-dose Ketamine in Plasma and Brain of Male and Female Rats. J Pharmacol Exp Ther 2018; 367:393-404. [PMID: 30213876 DOI: 10.1124/jpet.118.251652] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 09/10/2018] [Indexed: 11/22/2022] Open
Abstract
Recent work from our group and others has revealed a higher sensitivity of female rodents to the antidepressant-like effects of the N-methyl d-aspartate receptor antagonist ketamine strongly influenced by circulating estrogen and progesterone levels. However, in the absence of any preclinical studies of pharmacokinetic sex differences using low-dose ketamine in rats, it is unclear whether the effects of sex and hormonal milieu on ketamine's behavioral actions are influenced by differences in ketamine metabolism between male and female rats. Therefore, this work examined whether sex and hormonal status affect ketamine metabolism and distribution in male and female rats using a low antidepressant-like dose selectively effective in females. Intact male rats and female rats in either diestrus (low estrogen, progesterone) or proestrus (high estrogen, progesterone) were administered low-dose ketamine, and their plasma and brains were collected to analyze levels of ketamine and its metabolites norketamine (NK) and dehydronorketamine. Females exhibited greater concentrations of ketamine and NK over the first 30 min following treatment in both brain and plasma, largely accounted for by slower clearance rates and longer half-lives. Interestingly, despite the impact of ovarian hormones on behavioral sensitivity to ketamine, no appreciable differences in pharmacokinetic parameters existed between proestrus and diestrus female rats. This work is the first to demonstrate sex differences in ketamine pharmacokinetics in rats, and suggests that while sex differences in metabolism may influence the amount of ketamine and NK reaching target areas in the brain, the impact of circulating hormone levels here is negligible.
Collapse
Affiliation(s)
- Samantha K Saland
- Department of Biomedical Sciences, Program in Neuroscience, College of Medicine, Florida State University, Tallahassee, Florida
| | - Mohamed Kabbaj
- Department of Biomedical Sciences, Program in Neuroscience, College of Medicine, Florida State University, Tallahassee, Florida
| |
Collapse
|
23
|
Parsing the heterogeneity of depression: An exploratory factor analysis across commonly used depression rating scales. J Affect Disord 2018; 231:51-57. [PMID: 29448238 PMCID: PMC5852677 DOI: 10.1016/j.jad.2018.01.027] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 12/15/2017] [Accepted: 01/29/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Due to the heterogeneity of depressive symptoms-which can include depressed mood, anhedonia, negative cognitive biases, and altered activity levels-researchers often use a combination of depression rating scales to assess symptoms. This study sought to identify unidimensional constructs measured across rating scales for depression and to evaluate these constructs across clinical trials of a rapid-acting antidepressant (ketamine). METHODS Exploratory factor analysis (EFA) was conducted on baseline ratings from the Beck Depression Inventory (BDI), the Hamilton Depression Rating Scale (HAM-D), the Montgomery-Asberg Depression Rating Scale (MADRS), and the Snaith-Hamilton Pleasure Rating Scale (SHAPS). Inpatients with major depressive disorder (n = 76) or bipolar depression (n = 43) were participating in clinical ketamine trials. The trajectories of the resulting unidimensional scores were evaluated in 41 subjects with bipolar depression who participated in clinical ketamine trials. RESULTS The best solution, which exhibited excellent fit to the data, comprised eight factors: Depressed Mood, Tension, Negative Cognition, Impaired Sleep, Suicidal Thoughts, Reduced Appetite, Anhedonia, and Amotivation. Various response patterns were observed across the clinical trial data, both in treatment effect (ketamine versus placebo) and in degree of placebo response, suggesting that use of these unidimensional constructs may reveal patterns not observed with traditional scoring of individual instruments. LIMITATIONS Limitations include: 1) small sample (and related inability to confirm measurement invariance); 2) absence of an independent sample for confirmation of factor structure; and 3) the treatment-resistant nature of the population, which may limit generalizability. CONCLUSIONS The empirical identification of unidimensional constructs creates more refined scores that may elucidate the connection between specific symptoms and underlying pathophysiology.
Collapse
|
24
|
Abstract
Traditional pharmacological treatments for depression have a delayed therapeutic onset, ranging from several weeks to months, and there is a high percentage of individuals who never respond to treatment. In contrast, ketamine produces rapid-onset antidepressant, anti-suicidal, and anti-anhedonic actions following a single administration to patients with depression. Proposed mechanisms of the antidepressant action of ketamine include N-methyl-D-aspartate receptor (NMDAR) modulation, gamma aminobutyric acid (GABA)-ergic interneuron disinhibition, and direct actions of its hydroxynorketamine (HNK) metabolites. Downstream actions include activation of the mechanistic target of rapamycin (mTOR), deactivation of glycogen synthase kinase-3 and eukaryotic elongation factor 2 (eEF2), enhanced brain-derived neurotrophic factor (BDNF) signaling, and activation of α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptors (AMPARs). These putative mechanisms of ketamine action are not mutually exclusive and may complement each other to induce potentiation of excitatory synapses in affective-regulating brain circuits, which results in amelioration of depression symptoms. We review these proposed mechanisms of ketamine action in the context of how such mechanisms are informing the development of novel putative rapid-acting antidepressant drugs. Such drugs that have undergone pre-clinical, and in some cases clinical, testing include the muscarinic acetylcholine receptor antagonist scopolamine, GluN2B-NMDAR antagonists (i.e., CP-101,606, MK-0657), (2R,6R)-HNK, NMDAR glycine site modulators (i.e., 4-chlorokynurenine, pro-drug of the glycineB NMDAR antagonist 7-chlorokynurenic acid), NMDAR agonists [i.e., GLYX-13 (rapastinel)], metabotropic glutamate receptor 2/3 (mGluR2/3) antagonists, GABAA receptor modulators, and drugs acting on various serotonin receptor subtypes. These ongoing studies suggest that the future acute treatment of depression will typically occur within hours, rather than months, of treatment initiation.
Collapse
Affiliation(s)
- Panos Zanos
- Department of Psychiatry, University of Maryland School of Medicine, Rm. 934F MSTF, 685 W. Baltimore St., Baltimore, MD, 21201, USA.
| | - Scott M Thompson
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Physiology, University of Maryland School of Medicine, St. BRB 5-007, 655 W. Baltimore St., Baltimore, MD, 21201, USA, Baltimore, MD, 21201, USA
| | - Ronald S Duman
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Todd D Gould
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Psychiatry, University of Maryland School of Medicine, Rm. 936 MSTF, 685 W. Baltimore St., Baltimore, MD, 21201, USA
| |
Collapse
|
25
|
A Homer 1 gene variant influences brain structure and function, lithium effects on white matter, and antidepressant response in bipolar disorder: A multimodal genetic imaging study. Prog Neuropsychopharmacol Biol Psychiatry 2018; 81:88-95. [PMID: 29079138 DOI: 10.1016/j.pnpbp.2017.10.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/28/2017] [Accepted: 10/21/2017] [Indexed: 11/24/2022]
Abstract
BACKGROUND The Homer family of postsynaptic scaffolding proteins plays a crucial role in glutamate-mediated synaptic plasticity, a phenotype associated with Bipolar Disorder (BD). Homer is a target for antidepressants and mood stabilizers. The AA risk genotype of the Homer rs7713917 A>G SNP has been associated with mood disorders and suicide, and in healthy humans with brain function. Despite the evidence linking Homer 1 gene and function to mood disorder, as well as its involvement in animal models of depression, no study has yet investigated the role of Homer in bipolar depression and treatment response. METHODS We studied 199 inpatients, affected by a major depressive episode in course of BD. 147 patients were studied with structural MRI of grey and white matter, and 50 with BOLD functional MRI of emotional processing. 158 patients were treated with combined total sleep deprivation and light therapy. RESULTS At neuroimaging, patients with the AA genotype showed lower grey matter volumes in medial prefrontal cortex, higher BOLD fMRI neural responses to emotional stimuli in anterior cingulate cortex, and lower fractional anisotropy in bilateral frontal WM tracts. Lithium treatment increased axial diffusivity more in AA patients than in G*carriers. At clinical evaluation, the same AA homozygotes showed a worse antidepressant response to combined SD and LT. CONCLUSIONS rs7713917 influenced brain grey and white matter structure and function in BD, long term effects of lithium on white matter structure, and antidepressant response to chronotherapeutics, thus suggesting that glutamatergic neuroplasticity and Homer 1 function might play a role in BD psychopathology and response to treatment.
Collapse
|
26
|
Hashimoto K. Metabolomics of Major Depressive Disorder and Bipolar Disorder: Overview and Future Perspective. Adv Clin Chem 2018; 84:81-99. [PMID: 29478517 DOI: 10.1016/bs.acc.2017.12.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Major depressive disorder (MDD) and bipolar disorder (BD) are the most common mood disorders. They are etiologically related, but clinically distinct psychiatric illnesses. Their shared clinical features result in high rates of misdiagnosis due to a lack of biomarkers that allow their differentiation. BD is more frequently misdiagnosed as MDD because of overlapping symptomology, often later onset of mania, and frequent occurrence of depressive episodes in patients with BD. Misdiagnosis is also increased when patients with BD present symptoms indicative of a clinically significant depressive episode, but are premorbid for manic symptoms, or previous manic states not recognized. Therefore, the development of specific biomarkers for these disorders would be invaluable for establishing the correct diagnosis and treatment of MDD and BD. This chapter presents an overview and future perspective of the identification of biomarkers for mood disorders using metabolomics.
Collapse
Affiliation(s)
- Kenji Hashimoto
- Chiba University Center for Forensic Mental Health, Chiba, Japan.
| |
Collapse
|
27
|
Javitt DC, Carter CS, Krystal JH, Kantrowitz JT, Girgis RR, Kegeles LS, Ragland JD, Maddock RJ, Lesh TA, Tanase C, Corlett PR, Rothman DL, Mason G, Qiu M, Robinson J, Potter WZ, Carlson M, Wall MM, Choo TH, Grinband J, Lieberman JA. Utility of Imaging-Based Biomarkers for Glutamate-Targeted Drug Development in Psychotic Disorders: A Randomized Clinical Trial. JAMA Psychiatry 2018; 75:11-19. [PMID: 29167877 PMCID: PMC5833531 DOI: 10.1001/jamapsychiatry.2017.3572] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
IMPORTANCE Despite strong theoretical rationale and preclinical evidence, several glutamate-targeted treatments for schizophrenia have failed in recent pivotal trials, prompting questions as to target validity, compound inadequacy, or lack of target engagement. A key limitation for glutamate-based treatment development is the lack of functional target-engagement biomarkers for translation between preclinical and early-stage clinical studies. We evaluated the utility of 3 potential biomarkers-ketamine-evoked changes in the functional magnetic imaging (fMRI) blood oxygen level-dependent response (pharmacoBOLD), glutamate proton magnetic resonance spectroscopy (1H MRS), and task-based fMRI-for detecting ketamine-related alterations in brain glutamate. OBJECTIVE To identify measures with sufficient effect size and cross-site reliability to serve as glutamatergic target engagement biomarkers within early-phase clinical studies. DESIGN, SETTING, AND PARTICIPANTS This randomized clinical trial was conducted at an academic research institution between May 2014 and October 2015 as part of the National Institute of Mental Health-funded Fast-Fail Trial for Psychotic Spectrum Disorders project. All raters were blinded to study group. Healthy volunteers aged 18 to 55 years of either sex and free of significant medical or psychiatric history were recruited from 3 sites. Data were analyzed between November 2015 and December 2016. INTERVENTIONS Volunteers received either sequential ketamine (0.23 mg/kg infusion over 1 minute followed by 0.58 mg/kg/h infusion over 30 minutes and then 0.29 mg/kg/h infusion over 29 minutes) or placebo infusions. MAIN OUTCOMES AND MEASURES Ketamine-induced changes in pharmacoBOLD, 1H MRS, and task-based fMRI measures, along with symptom ratings. Measures were prespecified prior to data collection. RESULTS Of the 65 volunteers, 41 (63%) were male, and the mean (SD) age was 31.1 (9.6) years; 59 (91%) had at least 1 valid scan. A total of 53 volunteers (82%) completed both ketamine infusions. In pharmacoBOLD, a highly robust increase (Cohen d = 5.4; P < .001) in fMRI response was observed, with a consistent response across sites. A smaller but significant signal (Cohen d = 0.64; P = .04) was also observed in 1H MRS-determined levels of glutamate+glutamine immediately following ketamine infusion. By contrast, no significant differences in task-activated fMRI responses were found between groups. CONCLUSIONS AND RELEVANCE These findings demonstrate robust effects of ketamine on pharmacoBOLD across sites, supporting its utility for definitive assessment of functional target engagement. Other measures, while sensitive to ketamine effects, were not sufficiently robust for use as cross-site target engagement measures. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT02134951.
Collapse
Affiliation(s)
- Daniel C. Javitt
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York,Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, New York
| | | | - John H. Krystal
- Department of Psychiatry, Yale University, New Haven, Connecticut
| | - Joshua T. Kantrowitz
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York,Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, New York
| | - Ragy R. Girgis
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York
| | - Lawrence S. Kegeles
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York
| | | | | | - Tyler A. Lesh
- Department of Psychiatry, University of California, Davis
| | - Costin Tanase
- Department of Psychiatry, University of California, Davis
| | | | | | - Graeme Mason
- Department of Psychiatry, Yale University, New Haven, Connecticut
| | - Maolin Qiu
- Department of Psychiatry, Yale University, New Haven, Connecticut
| | - James Robinson
- Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, New York
| | | | - Marlene Carlson
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York
| | - Melanie M. Wall
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York,National Institute of Mental Health, Rockville, Maryland
| | - Tse-Hwei Choo
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York
| | - Jack Grinband
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York
| | - Jeffrey A. Lieberman
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York
| |
Collapse
|
28
|
Niciu MJ, Iadarola ND, Banerjee D, Luckenbaugh DA, Park M, Lener M, Park L, Ionescu DF, Ballard ED, Brutsche NE, Akula N, McMahon FJ, Machado-Vieira R, Nugent AC, Zarate CA. The antidepressant efficacy of subanesthetic-dose ketamine does not correlate with baseline subcortical volumes in a replication sample with major depressive disorder. J Psychopharmacol 2017; 31:1570-1577. [PMID: 29039254 PMCID: PMC5863225 DOI: 10.1177/0269881117732514] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND This study sought to reproduce, in a larger sample, previous findings of a correlation between smaller raw 3-Tesla (3T) hippocampal volumes and improved antidepressant efficacy of ketamine in individuals with major depressive disorder (MDD). A secondary analysis stratified subjects according to functional BDNF rs6265 (val66met) genotype. METHODS Unmedicated subjects with treatment-resistant MDD ( n=55) underwent baseline structural 3T MRI. Data processing was conducted with FSL/FIRST and Freesurfer software. The amygdala, hippocampus, and thalamus were selected a priori for analysis. All subjects received a single 0.5mg/kg × 40-minute ketamine infusion. Pearson correlations were performed with subcortical volumes and percent change in MADRS score (from baseline to 230 minutes, 1 day, and 1 week post-infusion). RESULTS Raw and corrected subcortical volumes did not correlate with antidepressant response at any timepoint. In val/val subjects ( n=23), corrected left and right thalamic volume positively correlated with antidepressant response to ketamine at 230 minutes post-infusion but did not reach statistical significance. In met carriers ( n=14), corrected left and right thalamic volume negatively correlated with antidepressant response to ketamine. CONCLUSION Baseline subcortical volumes implicated in MDD did not correlate with ketamine's antidepressant efficacy. Baseline thalamic volume and BDNF genotype may be a combinatorial rapid antidepressant response biomarker.
Collapse
Affiliation(s)
- Mark J Niciu
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Nicolas D Iadarola
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Dipavo Banerjee
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - David A Luckenbaugh
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Minkyung Park
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Marc Lener
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Lawrence Park
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Dawn F Ionescu
- Depression Clinical and Research Program, Massachusetts General Hospital, Boston, USA
| | - Elizabeth D Ballard
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Nancy E Brutsche
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Nirmala Akula
- Human Genetics Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Francis J McMahon
- Human Genetics Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Rodrigo Machado-Vieira
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Allison C Nugent
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| |
Collapse
|
29
|
Buji RI, Abdul Murad NA, Chan LF, Maniam T, Mohd Shahrir MS, Rozita M, Shamsul AS, Mohamad Hussain R, Abdullah N, Jamal R, Nik Jaafar NR. Suicidal ideation in systemic lupus erythematosus: NR2A gene polymorphism, clinical and psychosocial factors. Lupus 2017; 27:744-752. [PMID: 29161964 DOI: 10.1177/0961203317742711] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Background Systemic lupus erythematosus (SLE) patients are a high-risk population for suicide. Glutamatergic neurosystem genes have been implicated in the neurobiology of depression in SLE and suicidal behaviour in general. However, the role of glutamate receptor gene polymorphisms in suicidal behaviour among SLE patients remains unclear in the context of established clinical and psychosocial factors. We aimed to investigate the association of NR2A gene polymorphism with suicidal ideation in SLE while accounting for the interaction between clinical and psychosocial factors. Methods A total of 130 SLE patients were assessed for mood disorders (MINI International Neuropsychiatric Interview), severity of depression (Patient Health Questionnaire-9), suicidal behaviour (Columbia-Suicide Severity Rating Scale), socio-occupational functioning (Work and Social Adjustment Scale), recent life events (Social Readjustment Rating Scale) and lupus disease activity (SELENA-SLE Disease Activity Index). Eighty-six out of the 130 study participants consented for NR2A genotyping. Results Multivariable logistic regression showed nominal significance for the interaction effect between the NR2A rs2072450 AC genotype and higher severity of socio-occupational impairment with lifetime suicidal ideation in SLE patients ( p = 0.038, odds ratio = 1.364, 95% confidence interval = 1.018-1.827). However, only the association between lifetime mood disorder and lifetime suicidal ideation remained significant after Bonferroni correction ( p < 0.001, odds ratio = 33.834, 95% confidence interval = 7.624-150.138). Conclusions Lifetime mood disorder emerged as a more significant factor for suicidal ideation in SLE compared with NR2A gene polymorphism main and interaction effects. Clinical implications include identification and treatment of mood disorders as an early intervention for suicidal behaviour in SLE. More adequately-powered gene-environment interaction studies are required in the future to clarify the role of glutamate receptor gene polymorphisms in the risk stratification of suicidal behaviour among SLE patients.
Collapse
Affiliation(s)
- R I Buji
- 1 Hospital Mesra Bukit Padang, Kota Kinabalu, Sabah, Malaysia
| | - N A Abdul Murad
- 2 UKM Medical Molecular Biology Institute (UMBI), Kuala Lumpur, Malaysia
| | - L F Chan
- 3 Department of Psychiatry, National University of Malaysia Medical Centre (UKMMC), Kuala Lumpur, Malaysia
| | - T Maniam
- 3 Department of Psychiatry, National University of Malaysia Medical Centre (UKMMC), Kuala Lumpur, Malaysia.,7 Mind Faculty, Mont Kiara, Kuala Lumpur, Malaysia
| | - M S Mohd Shahrir
- 4 Department of Medicine, National University of Malaysia Medical Centre (UKMMC), Kuala Lumpur, Malaysia
| | - M Rozita
- 4 Department of Medicine, National University of Malaysia Medical Centre (UKMMC), Kuala Lumpur, Malaysia
| | - A S Shamsul
- 2 UKM Medical Molecular Biology Institute (UMBI), Kuala Lumpur, Malaysia.,5 Department of Community Health, National University of Malaysia Medical Centre (UKMMC), Kuala Lumpur, Malaysia
| | - R Mohamad Hussain
- 2 UKM Medical Molecular Biology Institute (UMBI), Kuala Lumpur, Malaysia
| | - N Abdullah
- 2 UKM Medical Molecular Biology Institute (UMBI), Kuala Lumpur, Malaysia
| | - R Jamal
- 2 UKM Medical Molecular Biology Institute (UMBI), Kuala Lumpur, Malaysia.,6 Department of Pediatrics, National University of Malaysia Medical Centre (UKMMC), Kuala Lumpur, Malaysia
| | - N R Nik Jaafar
- 3 Department of Psychiatry, National University of Malaysia Medical Centre (UKMMC), Kuala Lumpur, Malaysia
| |
Collapse
|
30
|
Nugent AC, Zarate CA. Using Neuroimaging to Decipher the Mechanism of Action of Ketamine: A Pathway to Novel Therapeutics? BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2017; 2:549-551. [PMID: 29560906 PMCID: PMC6251409 DOI: 10.1016/j.bpsc.2017.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 08/22/2017] [Indexed: 11/30/2022]
Affiliation(s)
- Allison C Nugent
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland.
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
31
|
Machado-Vieira R, Henter ID, Zarate CA. New targets for rapid antidepressant action. Prog Neurobiol 2017; 152:21-37. [PMID: 26724279 PMCID: PMC4919246 DOI: 10.1016/j.pneurobio.2015.12.001] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 11/30/2015] [Accepted: 12/07/2015] [Indexed: 02/08/2023]
Abstract
Current therapeutic options for major depressive disorder (MDD) and bipolar disorder (BD) are associated with a lag of onset that can prolong distress and impairment for patients, and their antidepressant efficacy is often limited. All currently approved antidepressant medications for MDD act primarily through monoaminergic mechanisms. Glutamate is the major excitatory neurotransmitter in the central nervous system, and glutamate and its cognate receptors are implicated in the pathophysiology of MDD, and in the development of novel therapeutics for this disorder. The rapid and robust antidepressant effects of the N-methyl-d-aspartate (NMDA) antagonist ketamine were first observed in 2000. Since then, other NMDA receptor antagonists have been studied in MDD. Most have demonstrated relatively modest antidepressant effects compared to ketamine, but some have shown more favorable characteristics. This article reviews the clinical evidence supporting the use of novel glutamate receptor modulators with direct affinity for cognate receptors: (1) non-competitive NMDA receptor antagonists (ketamine, memantine, dextromethorphan, AZD6765); (2) subunit (GluN2B)-specific NMDA receptor antagonists (CP-101,606/traxoprodil, MK-0657); (3) NMDA receptor glycine-site partial agonists (GLYX-13); and (4) metabotropic glutamate receptor (mGluR) modulators (AZD2066, RO4917523/basimglurant). We also briefly discuss several other theoretical glutamate receptor targets with preclinical antidepressant-like efficacy that have yet to be studied clinically; these include α-amino-3-hydroxyl-5-methyl-4-isoxazoleproprionic acid (AMPA) agonists and mGluR2/3 negative allosteric modulators. The review also discusses other promising, non-glutamatergic targets for potential rapid antidepressant effects, including the cholinergic system (scopolamine), the opioid system (ALKS-5461), corticotropin releasing factor (CRF) receptor antagonists (CP-316,311), and others.
Collapse
Affiliation(s)
- Rodrigo Machado-Vieira
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Ioline D Henter
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
32
|
Gould TD, Georgiou P, Brenner LA, Brundin L, Can A, Courtet P, Donaldson ZR, Dwivedi Y, Guillaume S, Gottesman II, Kanekar S, Lowry CA, Renshaw PF, Rujescu D, Smith EG, Turecki G, Zanos P, Zarate CA, Zunszain PA, Postolache TT. Animal models to improve our understanding and treatment of suicidal behavior. Transl Psychiatry 2017; 7:e1092. [PMID: 28398339 PMCID: PMC5416692 DOI: 10.1038/tp.2017.50] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 01/16/2017] [Accepted: 02/01/2017] [Indexed: 02/08/2023] Open
Abstract
Worldwide, suicide is a leading cause of death. Although a sizable proportion of deaths by suicide may be preventable, it is well documented that despite major governmental and international investments in research, education and clinical practice suicide rates have not diminished and are even increasing among several at-risk populations. Although nonhuman animals do not engage in suicidal behavior amenable to translational studies, we argue that animal model systems are necessary to investigate candidate endophenotypes of suicidal behavior and the neurobiology underlying these endophenotypes. Animal models are similarly a critical resource to help delineate treatment targets and pharmacological means to improve our ability to manage the risk of suicide. In particular, certain pathophysiological pathways to suicidal behavior, including stress and hypothalamic-pituitary-adrenal axis dysfunction, neurotransmitter system abnormalities, endocrine and neuroimmune changes, aggression, impulsivity and decision-making deficits, as well as the role of critical interactions between genetic and epigenetic factors, development and environmental risk factors can be modeled in laboratory animals. We broadly describe human biological findings, as well as protective effects of medications such as lithium, clozapine, and ketamine associated with modifying risk of engaging in suicidal behavior that are readily translatable to animal models. Endophenotypes of suicidal behavior, studied in animal models, are further useful for moving observed associations with harmful environmental factors (for example, childhood adversity, mechanical trauma aeroallergens, pathogens, inflammation triggers) from association to causation, and developing preventative strategies. Further study in animals will contribute to a more informed, comprehensive, accelerated and ultimately impactful suicide research portfolio.
Collapse
Affiliation(s)
- T D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - P Georgiou
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - L A Brenner
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Mental Illness Research Education and Clinical Center, Denver, CO, USA
- Military and Veteran Microbiome Consortium for Research and Education, U.S. Department of Veterans Affairs, Washington, DC, USA
- Department of Physical Medicine and Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - L Brundin
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - A Can
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Psychology, Notre Dame of Maryland University, Baltimore, MD, USA
| | - P Courtet
- Department of Emergency Psychiatry and Post Acute Care, CHU Montpellier, Montpellier, France
- Université Montpellier, Inserm U1061, Montpellier, France
| | - Z R Donaldson
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- Department of Psychology, University of Colorado, Boulder, Boulder, CO, USA
- Department of Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Y Dwivedi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - S Guillaume
- Department of Emergency Psychiatry and Post Acute Care, CHU Montpellier, Montpellier, France
- Université Montpellier, Inserm U1061, Montpellier, France
| | - I I Gottesman
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, USA
| | - S Kanekar
- Department of Psychiatry, University of Utah, Salt Lake City, UT, USA
| | - C A Lowry
- Rocky Mountain Mental Illness Research Education and Clinical Center, Denver, CO, USA
- Military and Veteran Microbiome Consortium for Research and Education, U.S. Department of Veterans Affairs, Washington, DC, USA
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
- Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - P F Renshaw
- Rocky Mountain Mental Illness Research Education and Clinical Center, Denver, CO, USA
- Department of Psychiatry, University of Utah, Salt Lake City, UT, USA
| | - D Rujescu
- Department of Psychiatry, University of Halle-Wittenberg, Halle, Germany
| | - E G Smith
- Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, USA
| | - G Turecki
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - P Zanos
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - C A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - P A Zunszain
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - T T Postolache
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
- Rocky Mountain Mental Illness Research Education and Clinical Center, Denver, CO, USA
- Military and Veteran Microbiome Consortium for Research and Education, U.S. Department of Veterans Affairs, Washington, DC, USA
- VISN 5 Mental Illness Research Education and Clinical Center, Baltimore MD, USA
| |
Collapse
|
33
|
Zhang JC, Yao W, Hashimoto K. Brain-derived Neurotrophic Factor (BDNF)-TrkB Signaling in Inflammation-related Depression and Potential Therapeutic Targets. Curr Neuropharmacol 2017; 14:721-31. [PMID: 26786147 PMCID: PMC5050398 DOI: 10.2174/1570159x14666160119094646] [Citation(s) in RCA: 345] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 10/08/2015] [Accepted: 11/10/2015] [Indexed: 12/20/2022] Open
Abstract
Depression is the most prevalent and among the most debilitating of psychiatric disorders. The precise neurobiology of this illness is unknown. Several lines of evidence suggest that peripheral and central inflammation plays a role in depressive symptoms, and that anti-inflammatory drugs can improve depressive symptoms in patients with inflammation-related depression. Signaling via brain-derived neurotrophic factor (BDNF) and its receptor, tropomycin receptor kinase B (TrkB) plays a key role in the pathophysiology of depression and in the therapeutic mechanisms of antidepressants. A recent paper showed that lipopolysaccharide (LPS)-induced inflammation gave rise to depression-like phenotype by altering BDNF-TrkB signaling in the prefrontal cortex, hippocampus, and nucleus accumbens, areas thought to be involved in the antidepressant effects of TrkB agonist, 7,8-dihydroxyflavone (7,8-DHF) and TrkB antagonist, ANA-12. Here we provide an overview of the tryptophan-kynurenine pathway and BDNF-TrkB signaling in the pathophysiology of inflammation-induced depression, and propose mechanistic actions for potential therapeutic agents. Additionally, the authors discuss the putative role of TrkB agonists and antagonists as novel therapeutic drugs for inflammation-related depression.
Collapse
Affiliation(s)
| | | | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, 1-8-1 Inohana, Chiba 260-8670, Japan
| |
Collapse
|
34
|
Saland SK, Duclot F, Kabbaj M. Integrative analysis of sex differences in the rapid antidepressant effects of ketamine in preclinical models for individualized clinical outcomes. Curr Opin Behav Sci 2016; 14:19-26. [PMID: 28584860 DOI: 10.1016/j.cobeha.2016.11.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In major depressive disorder, women exhibit higher lifetime prevalence and different antidepressant response rates than men, which illustrates the importance of examining individual differences in the pathophysiology of depression and therapeutic response. In recent years, the consideration of sex in related preclinical research has thus gained interest-particularly in light of novel evidence for rapid-acting antidepressants. Notably, the literature recently revealed a higher sensitivity of females to the antidepressant effects of the N-methyl-D-aspartate receptor antagonist ketamine, in both baseline and preclinical conditions. Combined with its fast-acting and relatively sustained properties, this evidence highlights ketamine as a particularly interesting therapeutic alternative for this sensitive population, and supports the value in considering sex as a critical factor for improved individualized therapeutic strategies.
Collapse
Affiliation(s)
- Samantha K Saland
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL-32306.,Program in Neuroscience, Florida State University, Tallahassee, FL-32306
| | - Florian Duclot
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL-32306.,Program in Neuroscience, Florida State University, Tallahassee, FL-32306
| | - Mohamed Kabbaj
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL-32306.,Program in Neuroscience, Florida State University, Tallahassee, FL-32306
| |
Collapse
|
35
|
Taylor O, Audenaert K, Baeken C, Saunders J, Peremans K. Nuclear medicine for the investigation of canine behavioral disorders. J Vet Behav 2016. [DOI: 10.1016/j.jveb.2016.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
36
|
Muzio L, Brambilla V, Calcaterra L, D’Adamo P, Martino G, Benedetti F. Increased neuroplasticity and hippocampal microglia activation in a mice model of rapid antidepressant treatment. Behav Brain Res 2016; 311:392-402. [DOI: 10.1016/j.bbr.2016.05.063] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 05/27/2016] [Accepted: 05/29/2016] [Indexed: 10/21/2022]
|
37
|
Szczepanik J, Nugent AC, Drevets WC, Khanna A, Zarate CA, Furey ML. Amygdala response to explicit sad face stimuli at baseline predicts antidepressant treatment response to scopolamine in major depressive disorder. Psychiatry Res Neuroimaging 2016; 254:67-73. [PMID: 27366831 PMCID: PMC6711385 DOI: 10.1016/j.pscychresns.2016.06.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 06/08/2016] [Accepted: 06/15/2016] [Indexed: 12/20/2022]
Abstract
The muscarinic antagonist scopolamine produces rapid antidepressant effects in individuals with major depressive disorder (MDD). In healthy subjects, manipulation of acetyl-cholinergic transmission modulates attention in a stimulus-dependent manner. This study tested the hypothesis that baseline amygdalar activity in response to emotional stimuli correlates with antidepressant treatment response to scopolamine and could thus potentially predict treatment outcome. MDD patients and healthy controls performed an attention shifting task involving emotional faces while undergoing functional magnetic resonance imaging (fMRI). We found that blood oxygenation level dependent (BOLD) signal in the amygdala acquired while MDD patients processed sad face stimuli correlated positively with antidepressant response to scopolamine. Amygdalar response to sad faces in MDD patients who did not respond to scopolamine did not differ from that of healthy controls. This suggests that the pre-treatment task elicited amygdalar activity that may constitute a biomarker of antidepressant treatment response to scopolamine. Furthermore, in MDD patients who responded to scopolamine, we observed a post-scopolamine stimulus processing shift towards a pattern demonstrated by healthy controls, indicating a change in stimulus-dependent neural response potentially driven by attenuated cholinergic activity in the amygdala.
Collapse
Affiliation(s)
- Joanna Szczepanik
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Allison C Nugent
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Wayne C Drevets
- Janssen Pharmaceuticals, LLC of Johnson and Johnson, Inc., Titusville, NJ, USA
| | - Ashish Khanna
- Physical Medicine and Rehabilitation, Jewish Medical Center, Brooklyn Hospital Center, Brooklyn, NY, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Maura L Furey
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA; Neuroscience Biomarkers Division, Janssen Research and Development, San Diego, CA, USA
| |
Collapse
|
38
|
Pešić V, Petrović J, M Jukić M. Molecular Mechanism and Clinical Relevance of Ketamine as Rapid-Acting Antidepressant. Drug Dev Res 2016; 77:414-422. [DOI: 10.1002/ddr.21335] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Vesna Pešić
- Department of Physiology; Faculty of Pharmacy, University of Belgrade; Belgrade Serbia
| | - Jelena Petrović
- Department of Physiology; Faculty of Pharmacy, University of Belgrade; Belgrade Serbia
| | - Marin M Jukić
- Department of Physiology and Pharmacology; Karolinska Institute; Stockholm Sweden
| |
Collapse
|
39
|
Dallaspezia S, Locatelli C, Lorenzi C, Pirovano A, Colombo C, Benedetti F. Sleep homeostatic pressure and PER3 VNTR gene polymorphism influence antidepressant response to sleep deprivation in bipolar depression. J Affect Disord 2016; 192:64-9. [PMID: 26707349 DOI: 10.1016/j.jad.2015.11.039] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 10/20/2015] [Accepted: 11/22/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Combined Total sleep deprivation (TSD) and light therapy (LT) cause a rapid improvement in bipolar depression which has been hypothesized to be paralleled by changes in sleep homeostasis. Recent studies showed that bipolar patients had lower changes of EEG theta power after sleep and responders to antidepressant TSD+LT slept less and showed a lower increase of EEG theta power then non-responders. A polymorphism in PER3 gene has been associated with diurnal preference, sleep structure and homeostatic response to sleep deprivation in healthy subjects. We hypothesized that the individual variability in the homeostatic response to TSD could be a correlate of antidepressant response and be influenced by genetic factors. METHODS We administered three TSD+LT cycles to bipolar depressed patients. Severity of depression was rated on Hamilton Depression Rating Scale. Actigraphic recordings were performed in a group of patients. RESULTS PER3 polymorphism influenced changes in total sleep time (F=2.24; p=0.024): while PER3(4/4) and PER3(4/5) patients showed a reduction in it after treatment, PER3(5/5) subjects showed an increase of about 40min, suggesting a higher homeostatic pressure. The same polymorphism influenced the change of depressive symptomatology during treatment (F=3.72; p=0.028). LIMITATIONS Sleep information was recorded till the day after the end of treatment: a longer period of observation could give more information about the possible maintenance of allostatic adaptation. CONCLUSIONS A higher sleep homeostatic pressure reduced the antidepressant response to TSD+LT, while an allostatic adaptation to sleep loss was associated with better response. This process seems to be under genetic control.
Collapse
Affiliation(s)
- Sara Dallaspezia
- Department of Clinical Neurosciences, Scientific Institute and University Vita-Salute, San Raffaele, Milan, Italy.
| | - Clara Locatelli
- Department of Clinical Neurosciences, Scientific Institute and University Vita-Salute, San Raffaele, Milan, Italy
| | - Cristina Lorenzi
- Department of Clinical Neurosciences, Scientific Institute and University Vita-Salute, San Raffaele, Milan, Italy
| | - Adele Pirovano
- Department of Clinical Neurosciences, Scientific Institute and University Vita-Salute, San Raffaele, Milan, Italy
| | - Cristina Colombo
- Department of Clinical Neurosciences, Scientific Institute and University Vita-Salute, San Raffaele, Milan, Italy
| | - Francesco Benedetti
- Department of Clinical Neurosciences, Scientific Institute and University Vita-Salute, San Raffaele, Milan, Italy
| |
Collapse
|
40
|
Abstract
The article provides an overview of common and differentiating self-reported and objective sleep disturbances seen in mood-disordered populations. The importance of considering sleep disturbances in the context of mood disorders is emphasized, because a large body of evidence supports the notion that sleep disturbances are a risk factor for onset, exacerbation, and relapse of mood disorders. In addition, potential mechanisms for sleep disturbance in depression, other primary sleep disorders that often occur with mood disorders, effects of antidepressant and mood-stabilizing drugs on sleep, and the adjunctive effect of treating sleep in patients with mood disorders are discussed.
Collapse
Affiliation(s)
- Meredith E Rumble
- Department of Psychiatry, University of Wisconsin, 6001 Research Park Boulevard, Madison, WI 53719, USA.
| | - Kaitlin Hanley White
- Department of Psychiatry, University of Wisconsin, 6001 Research Park Boulevard, Madison, WI 53719, USA
| | - Ruth M Benca
- Department of Psychiatry, University of Wisconsin, 6001 Research Park Boulevard, Madison, WI 53719, USA
| |
Collapse
|
41
|
Grimm O, Gass N, Weber-Fahr W, Sartorius A, Schenker E, Spedding M, Risterucci C, Schweiger JI, Böhringer A, Zang Z, Tost H, Schwarz AJ, Meyer-Lindenberg A. Acute ketamine challenge increases resting state prefrontal-hippocampal connectivity in both humans and rats. Psychopharmacology (Berl) 2015; 232:4231-41. [PMID: 26184011 DOI: 10.1007/s00213-015-4022-y] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 07/06/2015] [Indexed: 12/27/2022]
Abstract
RATIONALE Aberrant prefrontal-hippocampal (PFC-HC) connectivity is disrupted in several psychiatric and at-risk conditions. Advances in rodent functional imaging have opened the possibility that this phenotype could serve as a translational imaging marker for psychiatric research. Recent evidence from functional magnetic resonance imaging (fMRI) studies has indicated an increase in PFC-HC coupling during working-memory tasks in both schizophrenic patients and at-risk populations, in contrast to a decrease in resting-state PFC-HC connectivity. Acute ketamine challenge is widely used in both humans and rats as a pharmacological model to study the mechanisms of N-methyl-D-aspartate (NMDA) receptor hypofunction in the context of psychiatric disorders. OBJECTIVES We aimed to establish whether acute ketamine challenge has consistent effects in rats and humans by investigating resting-state fMRI PFC-HC connectivity and thus to corroborate its potential utility as a translational probe. METHODS Twenty-four healthy human subjects (12 females, mean age 25 years) received intravenous doses of either saline (placebo) or ketamine (0.5 mg/kg body weight). Eighteen Sprague-Dawley male rats received either saline or ketamine (25 mg/kg). Resting-state fMRI measurements took place after injections, and the data were analyzed for PFC-HC functional connectivity. RESULTS In both species, ketamine induced a robust increase in PFC-HC coupling, in contrast to findings in chronic schizophrenia. CONCLUSIONS This translational comparison demonstrates a cross-species consistency in pharmacological effect and elucidates ketamine-induced alterations in PFC-HC coupling, a phenotype often disrupted in pathological conditions, which may give clue to understanding of psychiatric disorders and their onset, and help in the development of new treatments.
Collapse
Affiliation(s)
- Oliver Grimm
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany
| | - Natalia Gass
- Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany.
| | - Wolfgang Weber-Fahr
- Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany
| | - Alexander Sartorius
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany.,Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany
| | - Esther Schenker
- Neuroscience Drug Discovery Unit, Institut de Recherches Servier, Croissy s/Seine, France
| | | | - Celine Risterucci
- CNS Biomarker, Pharmaceuticals Division, F. Hoffmann-La Roche, Basel, Switzerland
| | - Janina Isabel Schweiger
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany
| | - Andreas Böhringer
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany
| | - Zhenxiang Zang
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany
| | - Heike Tost
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany
| | - Adam James Schwarz
- Tailored Therapeutics, Eli Lilly and Company, Indianapolis, IN, USA.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA.,Department of Radiology and Imaging Sciences, Indiana University, Indianapolis, IN, USA
| | - Andreas Meyer-Lindenberg
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany
| |
Collapse
|
42
|
Affiliation(s)
- Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mark J Niciu
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
43
|
Vai B, Poletti S, Radaelli D, Dallaspezia S, Bulgarelli C, Locatelli C, Bollettini I, Falini A, Colombo C, Smeraldi E, Benedetti F. Successful antidepressant chronotherapeutics enhance fronto-limbic neural responses and connectivity in bipolar depression. Psychiatry Res 2015. [PMID: 26195295 DOI: 10.1016/j.pscychresns.2015.07.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The identification of antidepressant response predictors in bipolar disorder (BD) may provide new potential enhancements in treatment selection. Repeated total sleep deprivation combined with light therapy (TSD+LT) can acutely reverse depressive symptoms and has been proposed as a model antidepressant treatment. This study aims at investigating the effect of TSD+LT on effective connectivity and neural response in cortico-limbic circuitries during implicit processing of fearful and angry faces in patients with BD. fMRI and Dynamic Causal Modeling (DCM) were combined to study the effect of chronotherapeutics on neural responses in healthy controls (HC, n = 35) and BD patients either responder (RBD, n = 26) or non responder (nRBD, n = 11) to 3 consecutive TSD+LT sessions. Twenty-four DCMs exploring connectivity between anterior cingulate cortex (ACC), dorsolateral prefrontal cortex (DLPFC), Amygdala (Amy), fusiform gyrus and visual cortex were constructed. After treatment, patients significantly increased their neural responses in DLPFC, ACC and insula. nRBD showed lower baseline and endpoint neural responses than RBD. The increased activity in ACC and in medial prefrontal cortex, associated with antidepressant treatment, was positively associated with the improvement of depressive symptomatology. Only RBD patients increased intrinsic connectivity from DLPFC to ACC and reduced the modulatory effect of the task on Amy-DLPFC connection. A successful antidepressant treatment was associated with an increased functional activity and connectivity within cortico-limbic networks, suggesting the possible role of these measures in providing possible biomarkers for treatment efficacy.
Collapse
Affiliation(s)
- Benedetta Vai
- Department of Clinical Neurosciences, Scientific Institute Ospedale San Raffaele, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), Università Vita-Salute San Raffaele, Italy; Department of Human Studies, Libera Università Maria Ss. Assunta, Roma, Italy.
| | - Sara Poletti
- Department of Clinical Neurosciences, Scientific Institute Ospedale San Raffaele, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), Università Vita-Salute San Raffaele, Italy
| | - Daniele Radaelli
- Department of Clinical Neurosciences, Scientific Institute Ospedale San Raffaele, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), Università Vita-Salute San Raffaele, Italy
| | - Sara Dallaspezia
- Department of Clinical Neurosciences, Scientific Institute Ospedale San Raffaele, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), Università Vita-Salute San Raffaele, Italy
| | - Chiara Bulgarelli
- Department of Clinical Neurosciences, Scientific Institute Ospedale San Raffaele, Milan, Italy
| | - Clara Locatelli
- Department of Clinical Neurosciences, Scientific Institute Ospedale San Raffaele, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), Università Vita-Salute San Raffaele, Italy
| | - Irene Bollettini
- Department of Clinical Neurosciences, Scientific Institute Ospedale San Raffaele, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), Università Vita-Salute San Raffaele, Italy; PhD in Philosophy and Sciences of Mind, Università Vita-Saluta San Raffaele, Milan, Italy
| | - Andrea Falini
- Department of Neuroradiology, Scientific Institute Ospedale San Raffaele, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), Università Vita-Salute San Raffaele, Italy
| | - Cristina Colombo
- Department of Clinical Neurosciences, Scientific Institute Ospedale San Raffaele, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), Università Vita-Salute San Raffaele, Italy
| | - Enrico Smeraldi
- Department of Clinical Neurosciences, Scientific Institute Ospedale San Raffaele, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), Università Vita-Salute San Raffaele, Italy
| | - Francesco Benedetti
- Department of Clinical Neurosciences, Scientific Institute Ospedale San Raffaele, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), Università Vita-Salute San Raffaele, Italy
| |
Collapse
|
44
|
Pekary AE, Sattin A, Lloyd RL. Ketamine modulates TRH and TRH-like peptide turnover in brain and peripheral tissues of male rats. Peptides 2015; 69:66-76. [PMID: 25882008 DOI: 10.1016/j.peptides.2015.04.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 04/01/2015] [Accepted: 04/02/2015] [Indexed: 01/10/2023]
Abstract
Major depression is the largest single healthcare burden with treatments of slow onset and often limited efficacy. Ketamine, a NMDA antagonist used extensively as a pediatric and veterinary anesthetic, has recently been shown to be a rapid acting antidepressant, making it a potential lifesaver for suicidal patients. Side effects and risk of abuse limit the chronic use of ketamine. More complete understanding of the neurobiochemical mechanisms of ketamine should lead to safer alternatives. Some of the physiological and pharmacological actions of ketamine are consistent with increased synthesis and release of TRH (pGlu-His-Pro-NH2), and TRH-like peptides (pGlu-X-Pro-NH2) where "X" can be any amino acid residue. Moreover, TRH-like peptides are themselves potential therapeutic agents for the treatment of major depression, anxiety, bipolar disorder, epilepsy, Alzheimer's and Parkinson's diseases. For these reasons, male Sprague-Dawley rats were anesthetized with 162 mg/kg ip ketamine and then infused intranasally with 20 μl of sterile saline containing either 0 or 5 mg/ml Glu-TRH. One, 2 or 4h later, the brain levels of TRH and TRH-like peptides were measured in various brain regions and peripheral tissues. At 1h in brain following ketamine only, the levels of TRH and TRH-like peptides were significantly increased in 52 instances (due to increased biosynthesis and/or decreased release) or decreased in five instances. These changes, listed by brain region in order of decreasing number of significant increases (↑) and/or decreases (↓), were: hypothalamus (9↑); piriform cortex (8↑); entorhinal cortex (7↑); nucleus accumbens (7↑); posterior cingulate (5↑); striatum (4↑); frontal cortex (2↑,3↓); amygdala (3↑); medulla oblongata (1↑,2↓); cerebellum (2↑); hippocampus (2↑); anterior cingulate (2↑). The corresponding changes in peripheral tissues were: adrenals (8↑); epididymis (4↑); testis (1↑,3↓); pancreas (1↑); prostate (1↑). We conclude that TRH and TRH-like peptides may be downstream mediators of the rapid antidepressant actions of ketamine.
Collapse
Affiliation(s)
- A Eugene Pekary
- Research Services, VA Greater Los Angeles Healthcare System, University of California, Los Angeles, CA 90073, United States; Center for Ulcer Research and Education, VA Greater Los Angeles Healthcare System, University of California, Los Angeles, CA 90073, United States; Department of Medicine, University of California, Los Angeles, CA 90073, United States.
| | - Albert Sattin
- Research Services, VA Greater Los Angeles Healthcare System, University of California, Los Angeles, CA 90073, United States; Psychiatry Services, VA Greater Los Angeles Healthcare System, University of California, Los Angeles, CA 90073, United States; Departments of Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, CA 90073, United States; Brain Research Institute, University of California, Los Angeles, CA 90073, United States
| | - Robert L Lloyd
- Department of Psychology, University of Minnesota, 332 Bohannon Hall, 10 University Drive, Duluth, MN 55812-2494, United States
| |
Collapse
|
45
|
Moaddel R, Sanghvi M, Dossou KSS, Ramamoorthy A, Green C, Bupp J, Swezey R, O'Loughlin K, Wainer IW. The distribution and clearance of (2S,6S)-hydroxynorketamine, an active ketamine metabolite, in Wistar rats. Pharmacol Res Perspect 2015; 3:e00157. [PMID: 26171236 PMCID: PMC4492732 DOI: 10.1002/prp2.157] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 05/15/2015] [Accepted: 05/21/2015] [Indexed: 12/22/2022] Open
Abstract
The distribution, clearance, and bioavailability of (2S,6S)-hydroxynorketamine has been studied in the Wistar rat. The plasma and brain tissue concentrations over time of (2S,6S)-hydroxynorketamine were determined after intravenous (20 mg/kg) and oral (20 mg/kg) administration of (2S,6S)-hydroxynorketamine (n = 3). After intravenous administration, the pharmacokinetic parameters were estimated using noncompartmental analysis and the half-life of drug elimination during the terminal phase (t1/2) was 8.0 ± 4.0 h and the apparent volume of distribution (Vd) was 7352 ± 736 mL/kg, clearance (Cl) was 704 ± 139 mL/h per kg, and the bioavailability was 46.3%. Significant concentrations of (2S,6S)-hydroxynorketamine were measured in brain tissues at 10 min after intravenous administration, ∼30 μg/mL per g tissue which decreased to 6 μg/mL per g tissue at 60 min. The plasma and brain concentrations of (2S,6S)-hydroxynorketamine were also determined after the intravenous administration of (S)-ketamine, where significant plasma and brain tissue concentrations of (2S,6S)-hydroxynorketamine were observed 10 min after administration. The (S)-ketamine metabolites (S)-norketamine, (S)-dehydronorketamine, (2S,6R)-hydroxynorketamine, (2S,5S)-hydroxynorketamine and (2S,4S)-hydroxynorketamine were also detected in both plasma and brain tissue. The enantioselectivity of the conversion of (S)-ketamine and (R)-ketamine to the respective (2,6)-hydroxynorketamine metabolites was also investigated over the first 60 min after intravenous administration. (S)-Ketamine produced significantly greater plasma and brain tissue concentrations of (2S,6S)-hydroxynorketamine relative to the (2R,6R)-hydroxynorketamine observed after the administration of (R)-ketamine. However, the relative brain tissue: plasma concentrations of the enantiomeric (2,6)-hydroxynorketamine metabolites were not significantly different indicating that the penetration of the metabolite is not enantioselective.
Collapse
Affiliation(s)
- Ruin Moaddel
- Laboratory of Clinical Investigation, Division of Intramural Research Programs, National Institute on Aging, National Institutes of Health Baltimore, Maryland, 21224
| | - Mitesh Sanghvi
- Laboratory of Clinical Investigation, Division of Intramural Research Programs, National Institute on Aging, National Institutes of Health Baltimore, Maryland, 21224
| | - Katina Sourou Sylvestre Dossou
- Laboratory of Clinical Investigation, Division of Intramural Research Programs, National Institute on Aging, National Institutes of Health Baltimore, Maryland, 21224
| | - Anuradha Ramamoorthy
- Laboratory of Clinical Investigation, Division of Intramural Research Programs, National Institute on Aging, National Institutes of Health Baltimore, Maryland, 21224
| | - Carol Green
- Biosciences, SRI International Menlo Park, California
| | - James Bupp
- Biosciences, SRI International Menlo Park, California
| | - Robert Swezey
- Biosciences, SRI International Menlo Park, California
| | | | - Irving W Wainer
- Laboratory of Clinical Investigation, Division of Intramural Research Programs, National Institute on Aging, National Institutes of Health Baltimore, Maryland, 21224
| |
Collapse
|
46
|
Abstract
The wide spectrum of disruptions that characterizes major depressive disorder (MDD) and bipolar disorder (BD) highlights the difficulties researchers are posed with as they try to mimic these disorders in the laboratory. Nonetheless, numerous attempts have been made to create rodent models of mood disorders or at least models of the symptoms of MDD and BD. Present antidepressants are all descendants of the serendipitous findings in the 1950s that the monoamine oxidase inhibitor iproniazid and the tricyclic antidepressant imipramine were effective antidepressants. Thus, the need for improved animal models to provide insights into the neuropathology underlying the disease is critical. Such information is in turn crucial for identifying new antidepressants and mood stabilisers. Currently, there is a shift away from traditional animal models to more focused research dealing with an endophenotype-style approach, genetic models, and incorporation of new findings from human neuroimaging and genetic studies. Such approaches are opening up more tractable avenues for understanding the neurobiological and genetic bases of these disorders. Further, such models promise to yield better translational animal models and hence more fruitful therapeutic targets. This overview focuses on such animal models and tests and how they can be used to assess MDD and BD in rodents.
Collapse
|
47
|
Furey ML, Drevets WC, Szczepanik J, Khanna A, Nugent A, Zarate CA. Pretreatment Differences in BOLD Response to Emotional Faces Correlate with Antidepressant Response to Scopolamine. Int J Neuropsychopharmacol 2015; 18:pyv028. [PMID: 25820840 PMCID: PMC4571629 DOI: 10.1093/ijnp/pyv028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 03/01/2015] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Faster acting antidepressants and biomarkers that predict treatment response are needed to facilitate the development of more effective treatments for patients with major depressive disorders. Here, we evaluate implicitly and explicitly processed emotional faces using neuroimaging to identify potential biomarkers of treatment response to the antimuscarinic, scopolamine. METHODS Healthy participants (n=15) and unmedicated-depressed major depressive disorder patients (n=16) participated in a double-blind, placebo-controlled crossover infusion study using scopolamine (4 μg/kg). Before and following scopolamine, blood oxygen-level dependent signal was measured using functional MRI during a selective attention task. Two stimuli comprised of superimposed pictures of faces and houses were presented. Participants attended to one stimulus component and performed a matching task. Face emotion was modulated (happy/sad) creating implicit (attend-houses) and explicit (attend-faces) emotion processing conditions. The pretreatment difference in blood oxygen-level dependent response to happy and sad faces under implicit and explicit conditions (emotion processing biases) within a-priori regions of interest was correlated with subsequent treatment response in major depressive disorder. RESULTS Correlations were observed exclusively during implicit emotion processing in the regions of interest, which included the subgenual anterior cingulate (P<.02) and middle occipital cortices (P<.02). CONCLUSIONS The magnitude and direction of differential blood oxygen-level- dependent response to implicitly processed emotional faces prior to treatment reflect the potential to respond to scopolamine. These findings replicate earlier results, highlighting the potential for pretreatment neural activity in the middle occipital cortices and subgenual anterior cingulate to inform us about the potential to respond clinically to scopolamine.
Collapse
Affiliation(s)
- Maura L Furey
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute on Mental Health, National Institutes of Health, Bethesda, MD (Dr Furey, Ms Szczepanik, Dr Nugent, and Dr Zarate); Janssen Pharmaceuticals, LLC, of Johnson & Johnson, Inc., Titusville, NJ (Dr Drevets); Physical Medicine & Rehabilitation, Jewish Medical Center, Brooklyn Hospital Center, Brooklyn, NY (Dr Khanna).Registry number NCT00055575.
| | - Wayne C Drevets
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute on Mental Health, National Institutes of Health, Bethesda, MD (Dr Furey, Ms Szczepanik, Dr Nugent, and Dr Zarate); Janssen Pharmaceuticals, LLC, of Johnson & Johnson, Inc., Titusville, NJ (Dr Drevets); Physical Medicine & Rehabilitation, Jewish Medical Center, Brooklyn Hospital Center, Brooklyn, NY (Dr Khanna).Registry number NCT00055575
| | - Joanna Szczepanik
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute on Mental Health, National Institutes of Health, Bethesda, MD (Dr Furey, Ms Szczepanik, Dr Nugent, and Dr Zarate); Janssen Pharmaceuticals, LLC, of Johnson & Johnson, Inc., Titusville, NJ (Dr Drevets); Physical Medicine & Rehabilitation, Jewish Medical Center, Brooklyn Hospital Center, Brooklyn, NY (Dr Khanna).Registry number NCT00055575
| | - Ashish Khanna
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute on Mental Health, National Institutes of Health, Bethesda, MD (Dr Furey, Ms Szczepanik, Dr Nugent, and Dr Zarate); Janssen Pharmaceuticals, LLC, of Johnson & Johnson, Inc., Titusville, NJ (Dr Drevets); Physical Medicine & Rehabilitation, Jewish Medical Center, Brooklyn Hospital Center, Brooklyn, NY (Dr Khanna).Registry number NCT00055575
| | - Allison Nugent
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute on Mental Health, National Institutes of Health, Bethesda, MD (Dr Furey, Ms Szczepanik, Dr Nugent, and Dr Zarate); Janssen Pharmaceuticals, LLC, of Johnson & Johnson, Inc., Titusville, NJ (Dr Drevets); Physical Medicine & Rehabilitation, Jewish Medical Center, Brooklyn Hospital Center, Brooklyn, NY (Dr Khanna).Registry number NCT00055575
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute on Mental Health, National Institutes of Health, Bethesda, MD (Dr Furey, Ms Szczepanik, Dr Nugent, and Dr Zarate); Janssen Pharmaceuticals, LLC, of Johnson & Johnson, Inc., Titusville, NJ (Dr Drevets); Physical Medicine & Rehabilitation, Jewish Medical Center, Brooklyn Hospital Center, Brooklyn, NY (Dr Khanna).Registry number NCT00055575
| |
Collapse
|
48
|
Hashimoto K. Inflammatory biomarkers as differential predictors of antidepressant response. Int J Mol Sci 2015; 16:7796-801. [PMID: 25856677 PMCID: PMC4425050 DOI: 10.3390/ijms16047796] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 04/03/2015] [Accepted: 04/06/2015] [Indexed: 11/16/2022] Open
Abstract
Although antidepressants are generally effective in the treatment of major depressive disorder (MDD), it can still take weeks before patients feel the full antidepressant effects. Despite the efficacy of standard treatments, approximately two-thirds of patients with MDD fail to respond to pharmacotherapy. Therefore, the identification of blood biomarkers that can predict the treatment response to antidepressants would be highly useful in order to improve this situation. This article discusses inflammatory molecules as predictive biomarkers for antidepressant responses to several classes of antidepressants, including the N-methyl-d-aspartate (NMDA) receptor antagonist ketamine.
Collapse
Affiliation(s)
- Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, 1-8-1 Inohana, Chiba 260-7680, Japan.
| |
Collapse
|
49
|
Murrough JW, Burdick KE, Levitch CF, Perez AM, Brallier JW, Chang LC, Foulkes A, Charney DS, Mathew SJ, Iosifescu DV. Neurocognitive effects of ketamine and association with antidepressant response in individuals with treatment-resistant depression: a randomized controlled trial. Neuropsychopharmacology 2015; 40:1084-90. [PMID: 25374095 PMCID: PMC4367458 DOI: 10.1038/npp.2014.298] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 10/13/2014] [Accepted: 10/18/2014] [Indexed: 12/31/2022]
Abstract
The glutamate N-methyl-D-aspartate (NMDA) receptor antagonist ketamine displays rapid antidepressant effects in patients with treatment-resistant depression (TRD); however, the potential for adverse neurocognitive effects in this population has not received adequate study. The current study was designed to investigate the delayed neurocognitive impact of ketamine in TRD and examine baseline antidepressant response predictors in the context of a randomized controlled trial. In the current study, 62 patients (mean age = 46.2 ± 12.2) with TRD free of concomitant antidepressant medication underwent neurocognitive assessments using components of the MATRICS Consensus Cognitive Battery (MCCB) before and after a single intravenous infusion of ketamine (0.5 mg/kg) or midazolam (0.045 mg/kg). Participants were randomized to ketamine or midazolam in a 2:1 fashion under double-blind conditions and underwent depression symptom assessments at 24, 48, 72 h, and 7 days post treatment using the Montgomery-Asberg Depression Rating Scale (MADRS). Post-treatment neurocognitive assessment was conducted once at 7 days. Neurocognitive performance improved following the treatment regardless of treatment condition. There was no differential effect of treatment on neurocognitive performance and no association with antidepressant response. Slower processing speed at baseline uniquely predicted greater improvement in depression at 24 h following ketamine (t = 2.3, p = 0.027), while controlling for age, depression severity, and performance on other neurocognitive domains. In the current study, we found that ketamine was devoid of adverse neurocognitive effects at 7 days post treatment and that slower baseline processing speed was associated with greater antidepressant response. Future studies are required to further define the neurocognitive profile of ketamine in clinical samples and to identify clinically useful response moderators.
Collapse
Affiliation(s)
- James W Murrough
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY 10029, USA, Tel: +1 212 241 7574, Fax: +1 212 241 3354, E-mail:
| | - Katherine E Burdick
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cara F Levitch
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrew M Perez
- Department of Anesthesiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jess W Brallier
- Department of Anesthesiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lee C Chang
- Department of Anesthesiology, Baylor College of Medicine, Houston, TX, USA
| | - Alexandra Foulkes
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Dennis S Charney
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sanjay J Mathew
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA,Michael E. Debakey VA Medical Center, Houston, TX, USA
| | - Dan V Iosifescu
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
50
|
Bollettini I, Poletti S, Locatelli C, Vai B, Smeraldi E, Colombo C, Benedetti F. Disruption of white matter integrity marks poor antidepressant response in bipolar disorder. J Affect Disord 2015; 174:233-40. [PMID: 25527993 DOI: 10.1016/j.jad.2014.11.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 11/07/2014] [Accepted: 11/10/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND Changes of white matter (WM) microstructure have been proposed as structural biomarkers of bipolar disorder (BD). The chronotherapeutic combination of repeated total sleep deprivation and morning light therapy (TSD+LT) can acutely reverse depressive symptoms in approximately 60% of patients, and it has been proposed as a model antidepressant treatment to investigate the neurobiological correlates of rapid antidepressant response. METHODS We tested if baseline DTI measures can predict response to treatment in 70 in-patients affected by a major depressive episode in the course of BD, treated with chronotherapeutics for one week. We performed whole-brain tract-based spatial statistics with threshold-free cluster enhancement for the DTI measures of WM microstructure integrity: fractional anisotropy, axial, radial, and mean diffusivity. RESULTS Increased mean and radial water diffusivity correlated with poor antidepressant response to TSD+LT in core WM tracts which are crucial for the functional integrity of the brain, including corpus callosum, corona radiata, cingulum bundle, superior longitudinal fasciculus, inferior fronto-occipital fasciculus, and thalamic radiation. LIMITATIONS Limitations include issues such as generalizability, possible population stratification, medications and their effects on DTI measures, and no placebo control for chronotherapeutics. We could not consider other factors such as gene-environment interactions. CONCLUSIONS The association of increased radial and mean diffusivity with poor response to chronotherapeutic treatment warrants interest for the study of DTI measures of WM microstructure as markers for treatment response in bipolar depression.
Collapse
Affiliation(s)
- Irene Bollettini
- Department of Clinical Neurosciences, Scientific Institute and University Vita-Salute San Raffaele, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), University Vita-Salute San Raffaele, Milan, Italy; PhD Program in Philosophy and Sciences of Mind; University Vita-Salute San Raffaele, Milan, Italy
| | - Sara Poletti
- Department of Clinical Neurosciences, Scientific Institute and University Vita-Salute San Raffaele, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), University Vita-Salute San Raffaele, Milan, Italy
| | - Clara Locatelli
- Department of Clinical Neurosciences, Scientific Institute and University Vita-Salute San Raffaele, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), University Vita-Salute San Raffaele, Milan, Italy
| | - Benedetta Vai
- Department of Clinical Neurosciences, Scientific Institute and University Vita-Salute San Raffaele, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), University Vita-Salute San Raffaele, Milan, Italy; PhD Program in Evolutionary Psychopathology; Libera Università Maria SS. Assunta, Rome, Italy
| | - Enrico Smeraldi
- Department of Clinical Neurosciences, Scientific Institute and University Vita-Salute San Raffaele, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), University Vita-Salute San Raffaele, Milan, Italy
| | - Cristina Colombo
- Department of Clinical Neurosciences, Scientific Institute and University Vita-Salute San Raffaele, Milan, Italy
| | - Francesco Benedetti
- Department of Clinical Neurosciences, Scientific Institute and University Vita-Salute San Raffaele, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), University Vita-Salute San Raffaele, Milan, Italy.
| |
Collapse
|