1
|
Steinholtz L, Bodén R, Wall A, Lubberink M, Fällmar D, Persson J. Alterations in gamma-aminobutyric acid and glutamate neurotransmission linked to intermittent theta-burst stimulation in depression: a sham-controlled study. Transl Psychiatry 2025; 15:133. [PMID: 40199850 PMCID: PMC11978943 DOI: 10.1038/s41398-025-03371-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/19/2025] [Accepted: 04/01/2025] [Indexed: 04/10/2025] Open
Abstract
Gamma-aminobutyric acid (GABA) and glutamate are implicated in the antidepressant effects of repetitive transcranial magnetic stimulation (rTMS), though findings from magnetic resonance spectroscopy (MRS) are inconsistent. Furthermore, the relationship between GABAA-receptor availability and rTMS outcomes remains largely unexplored. In this study, GABA and glutamate levels in the dorsal anterior cingulate cortex (dACC) were measured using a 1H-MRS MEGA-PRESS sequence in 42 patients with bipolar or unipolar depression, both before and after a sham-controlled, double-blind clinical trial involving intermittent theta-burst stimulation (iTBS) over the dorsomedial prefrontal cortex. A subset of 28 patients also underwent [11C]flumazenil positron emission tomography (PET) to measure whole-brain GABAA-receptor availability and mean receptor availability in the nucleus accumbens and dACC. Depressive symptoms were assessed using the self-rated Montgomery Åsberg Depression Rating Scale (MADRS-S). The results indicated no significant changes in neurotransmitter levels or GABAA-receptor availability post-iTBS in either the active or sham conditions. However, changes in MADRS-S scores after active iTBS were positively correlated with changes in GABA levels in the dACC (r(13) = 0.54, p = 0.04) and baseline GABAA-receptor availability in the nucleus accumbens (r(11) = 0.66, p = 0.02). These correlations were absent in the sham group. The findings suggest that a reduction in GABA within targeted frontostriatal circuits can be part of the antidepressant mechanism of iTBS, challenging previous research. Additionally, they indicate a potential predictive role for frontostriatal GABAA-receptor availability in the treatment of depression using dorsomedial prefrontal iTBS.
Collapse
Affiliation(s)
- Linda Steinholtz
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| | - Robert Bodén
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Anders Wall
- PET-Centre, Uppsala University Hospital, Uppsala, Sweden
- Department of Surgical Sciences, Molecular Imaging and Medical Physics, Uppsala University, Uppsala, Sweden
| | - Mark Lubberink
- Department of Surgical Sciences, Molecular Imaging and Medical Physics, Uppsala University, Uppsala, Sweden
| | - David Fällmar
- Department of Surgical Sciences, Neuroradiology, Uppsala University, Uppsala, Sweden
| | - Jonas Persson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
2
|
Liu X, Liu S, Su F, Zhang W, Ke Y, Ming D. Neurophysiological Signatures of Major Depressive Disorder and Frontocentral Gamma Auditory Response Deficits. Depress Anxiety 2025; 2025:7390951. [PMID: 40225721 PMCID: PMC11918825 DOI: 10.1155/da/7390951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 12/08/2024] [Accepted: 01/13/2025] [Indexed: 04/15/2025] Open
Abstract
Background: Aberrant gamma oscillations in major depressive disorder (MDD) have attracted extensive attention, but evidence delineating such neural signatures is lacking. The auditory steady-state response (ASSR) elicited by periodic auditory stimuli is a robust probe of gamma oscillations. Here, we sought to characterize early transient auditory evoked responses (AEPs) and sustained gamma ASSRs in MDD, thereby identifying reliable neurophysiological signatures and providing preliminary interpretations of gamma auditory response deficits in MDD. Methods: Electroencephalographic data were obtained from 40 first-episode drug-naïve patients with MDD and 41 demographically matched healthy controls (HCs) during a 40-Hz ASSR paradigm, encompassing two periodic stimuli-chirp and click stimuli. Source analysis of transient AEPs was performed to identify generators involved in early information processing dysfunction. In addition, spectrotemporal and spatial characteristics of 40-Hz ASSRs were analyzed using event-related spectral perturbation, inter-trial phase coherence, and functional connectivity index. Results: Compared to HCs, patients showed a reduced P200 amplitude that was source-localized to the middle temporal gyrus, possibly reflecting an underlying impairment in the processes of early allocation or auditory information perception within the auditory pathways. Meanwhile, attenuated 40-Hz power and phase coherence, in conjunction with suppressed right frontotemporal and frontocentral connectivity, were observed in MDD, highlighting the multidimensional entrained gamma inhibition. Correlation analyses revealed that the decreased right frontocentral connectivity was strongly related to increased anxiety severity. Importantly, these abnormalities correlated with the patient's symptoms were only found with the chirp stimulus, suggesting that the chirp stimulus has tremendous potential to reveal specific neurophysiological signatures of MDD. Conclusions: Our data reveal impaired gamma auditory responses in first-episode drug-naïve patients with MDD and suggest that right frontocentral connectivity elicited by the chirp stimulus may represent a promising signature for predicting clinical symptoms.
Collapse
Affiliation(s)
- Xiaoya Liu
- Medical School of Tianjin University, Tianjin University, Tianjin, China
| | - Shuang Liu
- Medical School of Tianjin University, Tianjin University, Tianjin, China
| | - Fangyue Su
- College of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin, China
| | - Wenquan Zhang
- Medical School of Tianjin University, Tianjin University, Tianjin, China
| | - Yufeng Ke
- Medical School of Tianjin University, Tianjin University, Tianjin, China
| | - Dong Ming
- Medical School of Tianjin University, Tianjin University, Tianjin, China
- College of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin, China
| |
Collapse
|
3
|
Guet-McCreight A, Mazza F, Prevot TD, Sibille E, Hay E. Therapeutic dose prediction of α5-GABA receptor modulation from simulated EEG of depression severity. PLoS Comput Biol 2024; 20:e1012693. [PMID: 39729407 DOI: 10.1371/journal.pcbi.1012693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 12/03/2024] [Indexed: 12/29/2024] Open
Abstract
Treatment for major depressive disorder (depression) often has partial efficacy and a large portion of patients are treatment resistant. Recent studies implicate reduced somatostatin (SST) interneuron inhibition in depression, and new pharmacology boosting this inhibition via positive allosteric modulators of α5-GABAA receptors (α5-PAM) offers a promising effective treatment. However, testing the effect of α5-PAM on human brain activity is limited, meriting the use of detailed simulations. We utilized our previous detailed computational models of human depression microcircuits with reduced SST interneuron inhibition and α5-PAM effects, to simulate EEG of individual microcircuits across depression severity and α5-PAM doses. We developed machine learning models that predicted optimal dose from EEG with high accuracy and recovered microcircuit activity and EEG. This study provides dose prediction models for α5-PAM administration based on EEG biomarkers of depression severity. Given limitations in doing the above in the living human brain, the results and tools we developed will facilitate translation of α5-PAM treatment to clinical use.
Collapse
Affiliation(s)
| | - Frank Mazza
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Thomas D Prevot
- Department of Psychiatry, University of Toronto, Toronto, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
| | - Etienne Sibille
- Department of Psychiatry, University of Toronto, Toronto, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Etay Hay
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
| |
Collapse
|
4
|
Osborne KJ, Walther S, Mittal VA. Motor actions across psychiatric disorders: A research domain criteria (RDoC) perspective. Clin Psychol Rev 2024; 114:102511. [PMID: 39510028 DOI: 10.1016/j.cpr.2024.102511] [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: 03/14/2024] [Revised: 08/19/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024]
Abstract
The motor system is critical for understanding the pathophysiology and treatment of mental illness. Abnormalities in the processes that allow us to plan and execute movement in a goal-directed, context-appropriate manner (i.e., motor actions) are especially central to clinical motor research. Within this context, the NIMH Research Domain Criteria (RDoC) framework now includes a Motor Actions construct within the recently incorporated Sensorimotor Systems Domain, providing a useful framework for conducting research on motor action processes. However, there is limited available resources for understanding or implementing this framework. We address this gap by providing a comprehensive critical review and conceptual integration of the current clinical literature on the subconstructs comprising the Motor Actions construct. This includes a detailed discussion of each Motor Action subconstruct (e.g., action planning/execution) and its measurement across different units of analysis (e.g., molecules to behavior), the temporal and conceptual relationships among the Motor Action subconstructs (and other relevant RDoC domain constructs), and how abnormalities in these Motor Action subconstructs manifest in mental illness. Together, the review illustrates how motor system dysfunction is implicated in the pathophysiology of many psychiatric conditions and demonstrates shared and distinct mechanisms that may account for similar manifestations of motor abnormalities across disorders.
Collapse
Affiliation(s)
- K Juston Osborne
- Washington University in St. Louis, Department of Psychiatry, 4444 Forest Park Ave., St. Louis, MO, USA; Northwestern University, Department of Psychology, 633 Clark St. Evanston, IL, USA.
| | - Sebastian Walther
- University Hospital Würzburg, Department of Psychiatry, Psychosomatics, and Psychotherapy, Center of Mental Health, Margarete-Höppel-Platz 1, 97080 Würzburg, Germany
| | - Vijay A Mittal
- Northwestern University, Department of Psychology, 633 Clark St. Evanston, IL, USA; Northwestern University, Department of Psychiatry, 676 N. St. Claire, Chicago, IL, USA; Northwestern University, Department of Psychiatry, Institute for Policy Research, Department of Medical Social Sciences, Institute for Innovations in Developmental Sciences (DevSci), 633 Clark St., Evanston, Chicago, IL, USA
| |
Collapse
|
5
|
Lei L, Wang YF, Chen CY, Wang YT, Zhang Y. Novel insight into astrocyte-mediated gliotransmission modulates the synaptic plasticity in major depressive disorder. Life Sci 2024; 355:122988. [PMID: 39153595 DOI: 10.1016/j.lfs.2024.122988] [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: 04/25/2024] [Revised: 07/23/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Major depressive disorder (MDD) is a form of glial cell-based synaptic dysfunction disease in which glial cells interact closely with neuronal synapses and perform synaptic information processing. Glial cells, particularly astrocytes, are active components of the brain and are responsible for synaptic activity through the release gliotransmitters. A reduced density of astrocytes and astrocyte dysfunction have both been identified the brains of patients with MDD. Furthermore, gliotransmission, i.e., active information transfer mediated by gliotransmitters between astrocytes and neurons, is thought to be involved in the pathogenesis of MDD. However, the mechanism by which astrocyte-mediated gliotransmission contributes to depression remains unknown. This review therefore summarizes the alterations in astrocytes in MDD, including astrocyte marker, connexin 43 (Cx43) expression, Cx43 gap junctions, and Cx43 hemichannels, and describes the regulatory mechanisms of astrocytes involved in synaptic plasticity. Additionally, we investigate the mechanisms acting of the glutamatergic, gamma-aminobutyric acidergic, and purinergic systems that modulate synaptic function and the antidepressant mechanisms of the related receptor antagonists. Further, we summarize the roles of glutamate, gamma-aminobutyric acid, d-serine, and adenosine triphosphate in depression, providing a basis for the identification of diagnostic and therapeutic targets for MDD.
Collapse
Affiliation(s)
- Lan Lei
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yu-Fei Wang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Cong-Ya Chen
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ya-Ting Wang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
6
|
Zhang J, Li Y, Liu X, Zhong D, Xue C, Fan J, Xie C, Li J, Jin R. Characteristic Changes of Prefrontal and Motor Areas in Patients with Type 2 Diabetes and Major Depressive Disorder During a Motor Task of Tai Chi Chuan: A Functional Near-Infrared Spectroscopy Study. Brain Behav 2024; 14:e70071. [PMID: 39378277 PMCID: PMC11460607 DOI: 10.1002/brb3.70071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 08/27/2024] [Accepted: 09/05/2024] [Indexed: 10/10/2024] Open
Abstract
AIM This cross-sectional study aims to identify the characteristic changes of prefrontal and motor areas during a tai chi chuan task in patients with Type 2 diabetes mellitus (T2DM) and major depressive disorder (MDD) using wearable functional near-infrared spectroscopy (fNIRS). METHODS Three parallel groups (T2DM with DD group, T2DM group, and healthy group) were recruited from December 10, 2022, to May 31, 2023. Participants in three groups conducted a motor task of tai chi chuan designed by Eprime 3.0, and fNIRS was used to monitor the brain activation, functional connectivity (FC), and lateralization of prefrontal and motor areas. Correlation analyses were performed to examine the relationship between depressive symptoms and the function of prefrontal and motor areas. RESULTS Ninety elder adults (aged ≥ 60), including 30 patients with T2DM and MDD, 30 patients with T2DM, and 30 healthy subjects, were enrolled. In contrast with the patients with T2DM and healthy subjects, the patients with T2DM and MDD had decreased activation and abnormal lateralization in prefrontal and motor areas and decreased FC among supplementary motor area, motor area, and dorsolateral prefrontal cortex (DLPFC). Furthermore, the oxyhemoglobin (HbO2) concentration value of DLPFC in patients with T2DM and MDD was negatively associated with scores of Hamilton Depression Scale-24 (HAMD-24). CONCLUSIONS Patients with T2DM and MDD had characteristic functional changes in prefrontal and motor areas. DLPFC may be a potential target of diagnosis and intervention for patients with T2DM and MDD.
Collapse
Affiliation(s)
- Jiaming Zhang
- School of Health Preservation and RehabilitationChengdu University of Traditional Chinese MedicineChengduChina
- Department for Neural Function Detection and RegulationWest China Xiamen Hospital, Sichuan UniversityXiamenChina
| | - Yuxi Li
- School of Health Preservation and RehabilitationChengdu University of Traditional Chinese MedicineChengduChina
| | - Xiaobo Liu
- School of Health Preservation and RehabilitationChengdu University of Traditional Chinese MedicineChengduChina
| | - Dongling Zhong
- School of Health Preservation and RehabilitationChengdu University of Traditional Chinese MedicineChengduChina
| | - Chen Xue
- School of Health Preservation and RehabilitationChengdu University of Traditional Chinese MedicineChengduChina
| | - Jin Fan
- School of Acumox and TuinaChengdu University of Traditional Chinese MedicineChengduChina
| | - Cheng Xie
- School of Health Preservation and RehabilitationChengdu University of Traditional Chinese MedicineChengduChina
| | - Juan Li
- School of Health Preservation and RehabilitationChengdu University of Traditional Chinese MedicineChengduChina
| | - Rongjiang Jin
- School of Health Preservation and RehabilitationChengdu University of Traditional Chinese MedicineChengduChina
| |
Collapse
|
7
|
Aouci R, Fontaine A, Vion A, Belz L, Levi G, Narboux-Nême N. The Antidepressant Action of Fluoxetine Involves the Inhibition of Dlx5/6 in Cortical GABAergic Neurons through a TrkB-Dependent Pathway. Cells 2024; 13:1262. [PMID: 39120293 PMCID: PMC11311550 DOI: 10.3390/cells13151262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Major depressive disorder (MDD) is a complex and devastating illness that affects people of all ages. Despite the large use of antidepressants in current medical practice, neither their mechanisms of action nor the aetiology of MDD are completely understood. Experimental evidence supports the involvement of Parvalbumin-positive GABAergic neurons (PV-neurons) in the pathogenesis of MDD. DLX5 and DLX6 (DLX5/6) encode two homeodomain transcription factors involved in cortical GABAergic differentiation and function. In the mouse, the level of expression of these genes is correlated with the cortical density of PV-neurons and with anxiety-like behaviours. The same genomic region generates the lncRNA DLX6-AS1, which, in humans, participates in the GABAergic regulatory module downregulated in schizophrenia and ASD. Here, we show that the expression levels of Dlx5/6 in the adult mouse brain are correlated with the immobility time in the forced swim test, which is used to measure depressive-like behaviours. We show that the administration of the antidepressant fluoxetine (Flx) to normal mice induces, within 24 h, a rapid and stable reduction in Dlx5, Dlx6 and Dlx6-AS1 expression in the cerebral cortex through the activation of the TrkB-CREB pathway. Experimental Dlx5 overexpression counteracts the antidepressant effects induced by Flx treatment. Our findings show that one of the short-term effects of Flx administration is the reduction in Dlx5/6 expression in GABAergic neurons, which, in turn, has direct consequences on PV expression and on behavioural profiles. Variants in the DLX5/6 regulatory network could be implicated in the predisposition to depression and in the variability of patients' response to antidepressant treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Nicolas Narboux-Nême
- Molecular Physiology and Adaption, UMR7221 CNRS, Museum National d’Histoire Naturelle, 75005 Paris, France; (R.A.); (A.F.); (L.B.); (G.L.)
| |
Collapse
|
8
|
Luscher B, Jiang T, Feng M, Hutsell A. Sex-specific GABAergic microcircuits that switch vulnerability into resilience to stress and reverse the effects of chronic stress exposure. RESEARCH SQUARE 2024:rs.3.rs-4408723. [PMID: 39041032 PMCID: PMC11261964 DOI: 10.21203/rs.3.rs-4408723/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Clinical and preclinical studies have identified somatostatin (SST)-positive interneurons as key elements that regulate the vulnerability to stress-related psychiatric disorders. Conversely, disinhibition of SST neurons in mice results in resilience to the behavioral effects of chronic stress. Here we established a low-dose chronic chemogenetic protocol to map these changes in positively and negatively motivated behaviors to specific brain regions. AAV-hM3Dq mediated chronic activation of SST neurons in the prelimbic cortex (PLC) had antidepressant drug-like effects on anxiety- and anhedonia-related motivated behaviors in male but not female mice. Analogous manipulation of the ventral hippocampus (vHPC) had such effects in female but not male mice. Moreover, activation of SST neurons in the PLC of male and the vHPC of female mice resulted in stress resilience. Activation of SST neurons in the PLC reversed prior chronic stress-induced defects in motivated behavior in males but was ineffective in females. Conversely, activation of SST neurons in the vHPC reversed chronic stress-induced behavioral alterations in females but not males. Quantitation of c-Fos+ and FosB+ neurons in chronic stress-exposed mice revealed that chronic activation of SST neurons leads to a paradoxical increase in pyramidal cell activity. Collectively, these data demonstrate that GABAergic microcircuits driven by dendrite targeting interneurons enable sex- and brain-region-specific neural plasticity that promotes stress resilience and reverses stress-induced anxiety- and anhedonia-like motivated behavior. Our studies provide a mechanistic rationale for antidepressant efficacy of dendrite-targeting, low-potency GABAA receptor agonists, independent of sex and despite striking sex differences in the relevant brain substrates.
Collapse
|
9
|
Nguyen LTH, Nguyen NPK, Tran KN, Shin HM, Yang IJ. Intranasal administration of the essential oil from Perillae Folium ameliorates social defeat stress-induced behavioral impairments in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 324:117775. [PMID: 38224793 DOI: 10.1016/j.jep.2024.117775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/17/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Perillae Folium, the leaves and twigs of Perilla frutescens (L.) Britton, has been included in many traditional Chinese medicine herbal formulas to treat depression. However, the precise antidepressant mechanism of the essential oil from Perillae Folium (PFEO) has not been fully investigated. AIM OF THE STUDY To assess the effects and potential mechanisms of PFEO on depression using animal models and network pharmacology analysis. MATERIALS AND METHODS PFEO was intranasally administered to a mouse model of social defeat stress (SDS). The antidepressant effects of PFEO on SDS-induced mice were evaluated using behavioral tests. Enzyme-linked immunosorbent assay (ELISA) and western blot were performed to measure the levels of depression-related biomarkers in the hippocampus and serum of the mice. The chemical compounds of PFEO were determined using gas chromatography-mass spectrometry (GC-MS). Network pharmacology and molecular docking analyses were conducted to investigate the potential bioactive components of PFEO and the mechanisms underlying the antidepressant effects. To validate the mechanisms of the bioactive compounds, in vitro models using PC12 and BV2 cells were established and the blood-brain barrier (BBB) permeability was evaluated. RESULTS The intranasal administration of PFEO suppressed SDS-induced depression in mice by increasing the time spent in the social zone and the social interactions in the social interaction test and by decreasing the immobility time in the tail suspension and forced swimming tests. Moreover, the PFEO treatment reduced the SDS-induced anxiety-like behavior, as inferred from the increased activity in the central zone observed in the open field test and in the open arms observed in the elevated plus maze test. PFEO administration recovered the SDS-induced decrease in the levels of 5-HT, NE, gamma-aminobutyric acid (GABA), and p-ERK in the hippocampus of mice. Furthermore, the increased serum corticosterone level was also attenuated by the PFEO treatment. A total of 21 volatile compounds were detected in PFEO using GC-MS, among which elemicin (15.52%), apiol (15.16%), and perillaldehyde (12.79%) were the most abundant ones. The PFEO compounds targeted 32 depression-associated genes, which were mainly related to neural cells and neurotransmission pathways. Molecular docking indicated good binding affinities between the bioactive components of PFEO (apiol, β-caryophyllene, elemicin, and myristicin) and the key targets, including ACHE, IL1B, IL6, MAOB, SLC6A2, SLC6A3, SLC6A4, and tumor necrosis factor. Among the four compounds, β-caryophyllene, elemicin, and myristicin were more effective in reducing neurotoxicity and neuroinflammation. Elemicin showed the highest BBB permeability rate. CONCLUSIONS This study shows the antidepressant activities of PFEO in an SDS-induced mouse model and suggests its potential mechanisms of action: regulation of the corticosterone levels, hippocampal neurotransmitters, and ERK signaling. Apiol, β-caryophyllene, elemicin, and myristicin may be the main contributors to the observed effects induced by PFEO. Further studies are needed to fully elucidate the underlying mechanisms and the main PFEO bioactive components.
Collapse
Affiliation(s)
- Ly Thi Huong Nguyen
- Department of Physiology, Dongguk University College of Korean Medicine, Gyeongju, 38066, Republic of Korea; Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Nhi Phuc Khanh Nguyen
- Department of Physiology, Dongguk University College of Korean Medicine, Gyeongju, 38066, Republic of Korea.
| | - Khoa Nguyen Tran
- Department of Physiology, Dongguk University College of Korean Medicine, Gyeongju, 38066, Republic of Korea.
| | - Heung-Mook Shin
- Department of Physiology, Dongguk University College of Korean Medicine, Gyeongju, 38066, Republic of Korea.
| | - In-Jun Yang
- Department of Physiology, Dongguk University College of Korean Medicine, Gyeongju, 38066, Republic of Korea.
| |
Collapse
|
10
|
Guet-McCreight A, Chameh HM, Mazza F, Prevot TD, Valiante TA, Sibille E, Hay E. In-silico testing of new pharmacology for restoring inhibition and human cortical function in depression. Commun Biol 2024; 7:225. [PMID: 38396202 PMCID: PMC10891083 DOI: 10.1038/s42003-024-05907-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Reduced inhibition by somatostatin-expressing interneurons is associated with depression. Administration of positive allosteric modulators of α5 subunit-containing GABAA receptor (α5-PAM) that selectively target this lost inhibition exhibit antidepressant and pro-cognitive effects in rodent models of chronic stress. However, the functional effects of α5-PAM on the human brain in vivo are unknown, and currently cannot be assessed experimentally. We modeled the effects of α5-PAM on tonic inhibition as measured in human neurons, and tested in silico α5-PAM effects on detailed models of human cortical microcircuits in health and depression. We found that α5-PAM effectively recovered impaired cortical processing as quantified by stimulus detection metrics, and also recovered the power spectral density profile of the microcircuit EEG signals. We performed an α5-PAM dose-response and identified simulated EEG biomarker candidates. Our results serve to de-risk and facilitate α5-PAM translation and provide biomarkers in non-invasive brain signals for monitoring target engagement and drug efficacy.
Collapse
Affiliation(s)
- Alexandre Guet-McCreight
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada.
| | | | - Frank Mazza
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Thomas D Prevot
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Taufik A Valiante
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
- Center for Advancing Neurotechnological Innovation to Application, Toronto, ON, Canada
- Max Planck-University of Toronto Center for Neural Science and Technology, Toronto, ON, Canada
| | - Etienne Sibille
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Etay Hay
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
11
|
Leng L, Zhuang K, Lin H, Ding J, Yang S, Yuan Z, Huang C, Chen G, Chen Z, Wang M, Wang H, Sun H, Li H, Chang H, Chen Z, Xu Q, Yuan T, Zhang J. Menin Reduces Parvalbumin Expression and is Required for the Anti-Depressant Function of Ketamine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305659. [PMID: 38044302 PMCID: PMC10837338 DOI: 10.1002/advs.202305659] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 10/23/2023] [Indexed: 12/05/2023]
Abstract
Dysfunction of parvalbumin (PV) neurons is closely involved in depression, however, the detailed mechanism remains unclear. Based on the previous finding that multiple endocrine neoplasia type 1 (Protein: Menin; Gene: Men1) mutation (G503D) is associated with a higher risk of depression, a Menin-G503D mouse model is generated that exhibits heritable depressive-like phenotypes and increases PV expression in brain. This study generates and screens a serial of neuronal specific Men1 deletion mice, and found that PV interneuron Men1 deletion mice (PcKO) exhibit increased cortical PV levels and depressive-like behaviors. Restoration of Menin, knockdown PV expression or inhibition of PV neuronal activity in PV neurons all can ameliorate the depressive-like behaviors of PcKO mice. This study next found that ketamine stabilizes Menin by inhibiting protein kinase A (PKA) activity, which mediates the anti-depressant function of ketamine. These results demonstrate a critical role for Menin in depression, and prove that Menin is key to the antidepressant function of ketamine.
Collapse
Affiliation(s)
- Lige Leng
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Kai Zhuang
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Hui Lin
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Jinjun Ding
- Shanghai Mental Health CenterShanghai Jiaotong University School of MedicineShanghai200030P. R. China
| | - Shangchen Yang
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Ziqi Yuan
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Changquan Huang
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Guimiao Chen
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Zhenlei Chen
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Mengdan Wang
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Han Wang
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Hao Sun
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Huifang Li
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - He Chang
- Department of GeriatricsXiang'an Hospital of Xiamen universityXiamenFujian361102P. R. China
| | - Zhenyi Chen
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Qi Xu
- State Key Laboratory of Medical Molecular BiologyInstitute of Basic Medical Sciences Chinese Academy of Medical Sciences and Peking Union Medical CollegeNeuroscience CenterChinese Academy of Medical SciencesBeijing100730P. R. China
| | - Tifei Yuan
- Shanghai Mental Health CenterShanghai Jiaotong University School of MedicineShanghai200030P. R. China
| | - Jie Zhang
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| |
Collapse
|
12
|
Cai DB, Qin XD, Qin ZJ, Lan XJ, Wang JJ, Ng CH, Zheng W, Xiang YT. Adjunctive continuous theta burst stimulation for major depressive disorder or bipolar depression: A meta-analysis of randomized controlled studies. J Affect Disord 2024; 346:266-272. [PMID: 37924984 DOI: 10.1016/j.jad.2023.10.161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/06/2023]
Abstract
OBJECTIVES As a novel type of theta burst stimulation (TBS), continuous TBS (cTBS) has been shown to have mixed therapeutic effects for major depressive disorder (MDD) or bipolar depression (BD). Thus, we performed a meta-analysis of randomized controlled trials (RCTs) of cTBS for treating major depressive episodes in patients with MDD or BD. METHODS A systematic search of four major bibliographic databases (PubMed, EMBASE, Cochrane Library, and PsycINFO) was conducted from inception dates to February 3, 2023 to identify eligible studies. The data were analyzed using a random-effects model. RESULTS Three RCTs (n = 78, active cTBS = 37 and sham cTBS = 41) were included the meta-analysis. No significant differences were found in terms of change in Hamilton Depression Rating Scale (HAMD) scores (3 RCTs, n = 78, SMD = -0.09, 95 % CI: -0.53 to 0.36; I2 = 0 %; P = 0.71) and study-defined response (2 RCTs, n = 58, 26.7 % versus 21.4 %, RR = 1.20, 95 % CI: 0.48 to 2.96; I2 = 0 %; P = 0.70) between active and sham cTBS groups. Similarly, no group differences were found in the rates of adverse events and discontinuation due to any reason (P > 0.05). LIMITATIONS Meta-analysis had small sample sizes and low number of included studies. CONCLUSIONS Although cTBS appeared to be a safe and well-tolerated option for treating major depressive episodes in MDD or BD patients, no advantage in treatment effects was found in this meta-analysis. Future large-scale studies are warranted to assess the efficacy of cTBS for MDD or BD patients with a major depressive episode.
Collapse
Affiliation(s)
- Dong-Bin Cai
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Xiu-De Qin
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Zhen-Juan Qin
- The Brain Hospital of Guangxi Zhuang Autonomous Region, LiuZhou, China
| | - Xian-Jun Lan
- The Brain Hospital of Guangxi Zhuang Autonomous Region, LiuZhou, China
| | - Jian-Jun Wang
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Chee H Ng
- Department of Psychiatry, The Melbourne Clinic and St Vincent's Hospital, University of Melbourne, Richmond, Victoria, Australia.
| | - Wei Zheng
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Yu-Tao Xiang
- Unit of Psychiatry, Department of Public Health and Medicinal Administration, & Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macao SAR, China; Centre for Cognitive and Brain Sciences, University of Macau, Macao SAR, China..
| |
Collapse
|
13
|
You Z, Wang C, Lan X, Li W, Shang D, Zhang F, Ye Y, Liu H, Zhou Y, Ning Y. The contribution of polyamine pathway to determinations of diagnosis for treatment-resistant depression: A metabolomic analysis. Prog Neuropsychopharmacol Biol Psychiatry 2024; 128:110849. [PMID: 37659714 DOI: 10.1016/j.pnpbp.2023.110849] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/26/2023] [Accepted: 08/28/2023] [Indexed: 09/04/2023]
Abstract
OBJECTIVES Approximately one-third of major depressive disorder (MDD) patients do not respond to standard antidepressants and develop treatment-resistant depression (TRD). We aimed to reveal metabolic differences and discover promising potential biomarkers in TRD. METHODS Our study recruited 108 participants including healthy controls (n = 40) and patients with TRD (n = 35) and first-episode drug-naive MDD (DN-MDD) (n = 33). Plasma samples were presented to ultra performance liquid chromatography-tandem mass spectrometry. Then, a machine-learning algorithm was conducted to facilitate the selection of potential biomarkers. RESULTS TRD appeared to be a distinct metabolic disorder from DN-MDD and healthy controls (HCs). Compared to HCs, 199 and 176 differentially expressed metabolites were identified in TRD and DN-MDD, respectively. Of all the metabolites that were identified, spermine, spermidine, and carnosine were considered the most promising biomarkers for diagnosing TRD and DN-MDD patients, with the resulting area under the ROC curve of 0.99, 0.99, and 0.93, respectively. Metabolic pathway analysis yielded compelling evidence of marked changes or imbalances in both polyamine metabolism and energy metabolism, which could potentially represent the primary altered pathways associated with MDD. Additionally, L-glutamine, Beta-alanine, and spermine were correlated with HAMD score. CONCLUSIONS A more disordered metabolism structure is found in TRD than in DN-MDD and HCs. Future investigations should prioritize the comprehensive analysis of potential roles played by these differential metabolites and disturbances in polyamine pathways in the pathophysiology of TRD and depression.
Collapse
Affiliation(s)
- Zerui You
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Child and Adolescent Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Chengyu Wang
- Department of Child and Adolescent Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Xiaofeng Lan
- Department of Child and Adolescent Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Weicheng Li
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Child and Adolescent Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Dewei Shang
- Department of Child and Adolescent Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Fan Zhang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Child and Adolescent Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yanxiang Ye
- Department of Child and Adolescent Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Haiyan Liu
- Department of Child and Adolescent Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yanling Zhou
- Department of Child and Adolescent Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China.
| | - Yuping Ning
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Child and Adolescent Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China.
| |
Collapse
|
14
|
Thompson SM. Modulators of GABA A receptor-mediated inhibition in the treatment of neuropsychiatric disorders: past, present, and future. Neuropsychopharmacology 2024; 49:83-95. [PMID: 37709943 PMCID: PMC10700661 DOI: 10.1038/s41386-023-01728-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/14/2023] [Accepted: 08/08/2023] [Indexed: 09/16/2023]
Abstract
The predominant inhibitory neurotransmitter in the brain, γ-aminobutyric acid (GABA), acts at ionotropic GABAA receptors to counterbalance excitation and regulate neuronal firing. GABAA receptors are heteropentameric channels comprised from subunits derived from 19 different genes. GABAA receptors have one of the richest and well-developed pharmacologies of any therapeutic drug target, including agonists, antagonists, and positive and negative allosteric modulators (PAMs, NAMs). Currently used PAMs include benzodiazepine sedatives and anxiolytics, barbiturates, endogenous and synthetic neurosteroids, and general anesthetics. In this article, I will review evidence that these drugs act at several distinct binding sites and how they can be used to alter the balance between excitation and inhibition. I will also summarize existing literature regarding (1) evidence that changes in GABAergic inhibition play a causative role in major depression, anxiety, postpartum depression, premenstrual dysphoric disorder, and schizophrenia and (2) whether and how GABAergic drugs exert beneficial effects in these conditions, focusing on human studies where possible. Where these classical therapeutics have failed to exert benefits, I will describe recent advances in clinical and preclinical drug development. I will also highlight opportunities to advance a generation of GABAergic therapeutics, such as development of subunit-selective PAMs and NAMs, that are engendering hope for novel tools to treat these devastating conditions.
Collapse
Affiliation(s)
- Scott M Thompson
- Center for Novel Therapeutics, Department of Psychiatry, University of Colorado School of Medicine, 12700 E. 19th Ave., Aurora, CO, 80045, USA.
| |
Collapse
|
15
|
Hu YT, Hu XW, Han JF, Zhang JF, Wang YY, Wolff A, Tremblay S, Hirjak D, Tan ZL, Northoff G. Motor cortex repetitive transcranial magnetic stimulation in major depressive disorder - A preliminary randomized controlled clinical trial. J Affect Disord 2024; 344:169-175. [PMID: 37827254 DOI: 10.1016/j.jad.2023.10.058] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/27/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND Repetitive transcranial magnetic stimulation (rTMS) at left dorsolateral prefrontal cortex (lDLPFC) is commonly used in major depressive disorder (MDD), even though its therapeutic efficacy is limited. Given that many MDD patients show psychomotor retardation, we aim to examine whether the left motor cortex (lMC) as a novel rTMS target would provide effective and well-tolerated treatment as being comparable to lDLPFC-rTMS. METHODS In this prospective double-blind randomized single-center study, 131 MDD patients were randomly assigned to the lDLPFC or lMC group and were treated with 10 Hz rTMS (90 % motor threshold) applied twice daily for 4000 pulses continuously over five days. The primary endpoint was the Hamilton Depression Scale (HAMD) total score change after treatment. RESULTS After the five-day rTMS treatment, there was no significant difference in both HAMD reduction rate (lDLPFC 59.3 % ± 20.4 %, lMC 51.3 % ± 26.3 %, P = 0.10) and adverse effects (P = 0.79) between 48 (73.8 %) lMC subjects and 51 (77.3 %) lDLPFC subjects. Furthermore, the lMC study group showed stable HAMD scores at follow-up compared to their endpoint scores (P = 0.08). LIMITATIONS Sham-control group was not included and the sample size was small. Therefore, our results should be seen as exploratory and preliminary. CONCLUSIONS The preliminary good therapeutic response, comparability, and tolerability of lMC-rTMS suggest lMC a potential and more easily accessible rTMS target. Together, our findings raise the possibility of symptom-specific rTMS in motor cortex (psychomotor retardation) or lDLPFC (cognitive deficits). This warrants larger clinical trials of rTMS in MDD with symptom-specific stimulation targets.
Collapse
Affiliation(s)
- Yu-Ting Hu
- Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute of Mental Health Research, University of Ottawa, Ottawa, Canada
| | - Xi-Wen Hu
- Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jin-Fang Han
- Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian-Feng Zhang
- Center for Brain Disorders and Cognitive Sciences, Shenzhen University, Shenzhen, China
| | - Ying-Ying Wang
- Institute of Psychological Sciences, College of Education, Hangzhou Normal University, Hangzhou, China
| | - Annemarie Wolff
- Institute of Mental Health Research, University of Ottawa, Ottawa, Canada
| | - Sara Tremblay
- Institute of Mental Health Research, University of Ottawa, Ottawa, Canada
| | - Dusan Hirjak
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Zhong-Lin Tan
- Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Georg Northoff
- Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute of Mental Health Research, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
16
|
Vecera CM, C. Courtes A, Jones G, Soares JC, Machado-Vieira R. Pharmacotherapies Targeting GABA-Glutamate Neurotransmission for Treatment-Resistant Depression. Pharmaceuticals (Basel) 2023; 16:1572. [PMID: 38004437 PMCID: PMC10675154 DOI: 10.3390/ph16111572] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/22/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Treatment-resistant depression (TRD) is a term used to describe a particular type of major depressive disorder (MDD). There is no consensus about what defines TRD, with various studies describing between 1 and 4 failures of antidepressant therapies, with or without electroconvulsive therapy (ECT). That is why TRD is such a growing concern among clinicians and researchers, and it explains the necessity for investigating novel therapeutic targets beyond conventional monoamine pathways. An imbalance between two primary central nervous system (CNS) neurotransmitters, L-glutamate and γ-aminobutyric acid (GABA), has emerged as having a key role in the pathophysiology of TRD. In this review, we provide an evaluation and comprehensive review of investigational antidepressants targeting these two systems, accessing their levels of available evidence, mechanisms of action, and safety profiles. N-methyl-D-aspartate (NMDA) receptor antagonism has shown the most promise amongst the glutamatergic targets, with ketamine and esketamine (Spravato) robustly generating responses across trials. Two specific NMDA-glycine site modulators, D-cycloserine (DCS) and apimostinel, have also generated promising initial safety and efficacy profiles, warranting further investigation. Combination dextromethorphan-bupropion (AXS-05/Auvelity) displays a unique mechanism of action and demonstrated positive results in particular applicability in subpopulations with cognitive dysfunction. Currently, the most promising GABA modulators appear to be synthetic neurosteroid analogs with positive GABAA receptor modulation (such as brexanolone). Overall, advances in the last decade provide exciting perspectives for those who do not improve with conventional therapies. Of the compounds reviewed here, three are approved by the Food and Drug Administration (FDA): esketamine (Spravato) for TRD, Auvelity (dextromethorphan-bupropion) for major depressive disorder (MDD), and brexanolone (Zulresso) for post-partum depression (PPD). Notably, some concerns have arisen with esketamine and brexanolone, which will be detailed in this study.
Collapse
Affiliation(s)
- Courtney M. Vecera
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center, Houston, TX 77054, USA
| | - Alan C. Courtes
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center, Houston, TX 77054, USA
| | - Gregory Jones
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center, Houston, TX 77054, USA
| | - Jair C. Soares
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center, Houston, TX 77054, USA
| | - Rodrigo Machado-Vieira
- John S. Dunn Behavioral Sciences Center at UTHealth Houston, 5615 H.Mark Crosswell Jr St, Houston, TX 77021, USA
| |
Collapse
|
17
|
García-Gutiérrez MS, Navarro D, Torregrosa AB, Viudez-Martínez A, Giner S, Manzanares J. Alterations of BDNF, mGluR5, Homer1a, p11 and excitatory/inhibitory balance in corticolimbic brain regions of suicide decedents. J Affect Disord 2023; 339:366-376. [PMID: 37437733 DOI: 10.1016/j.jad.2023.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 06/02/2023] [Accepted: 07/08/2023] [Indexed: 07/14/2023]
Abstract
BACKGROUND Developing biological based approaches for preventing suicide has become a priority. In recent years, there has been a surge in studies investigating the role of the glutamatergic system in suicide, although it remains unclear. METHODS We evaluated changes in the gene expression of the metabotropic glutamate receptor 5 (mGluR5) and its scaffolding proteins Homer1a and p11 in the dorsolateral prefrontal cortex (DLPFC), amygdala (AMY), and hippocampus (HIP) of 28 suicide decedents (S) (with no clinical psychiatric history or treatment with anxiolytics or antidepressants) and 26 controls (C) by real-time PCR (qPCR). Indeed, we measured BDNF gene expression and VGluT1 and VGAT immunoreactivities in the HIP by qPCR and immunohistochemistry, respectively. Cases and controls matched for age (C: 48.6 ± 11.6 years; S: 46.9 ± 14.5 years) and postmortem interval (PMI; C: 20.1 ± 13h; S: 16.9 ± 5h). RESULTS In DLPFC, S had lower p11 gene expression levels, but no differences were found in mGluR5 or Homer1a. In the AMY and HIP, mGluR5 and Homer1a were increased, p11 and BDNF were reduced. In the HIP, there were less VGAT-ir and more VGluT1-ir. LIMITATIONS Future studies are necessary to evaluate protein levels, and determine the cell types and potential compensatory mechanisms in a larger sample including S diagnosed with psychiatric disorders, females and different ethnicities. CONCLUSIONS This study identified significant alterations in mGluR5, Homer1a, p11, BDNF and excitatory/inhibitory balance in corticolimbic brain areas of S. These results further characterize the biological basis of suicide, contributing to the identification of potential biomarkers for suicide prevention.
Collapse
Affiliation(s)
- María S García-Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante 03550, Alicante, Spain; Red de Investigación en Atención Primaria de Adicciones, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain; Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Daniela Navarro
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante 03550, Alicante, Spain; Red de Investigación en Atención Primaria de Adicciones, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain; Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Abraham B Torregrosa
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante 03550, Alicante, Spain; Red de Investigación en Atención Primaria de Adicciones, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain; Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | | | - Salvador Giner
- Instituto de Medicina Legal, Avenida Aguilera 53, 03007, Alicante, Spain
| | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante 03550, Alicante, Spain; Red de Investigación en Atención Primaria de Adicciones, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain; Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain.
| |
Collapse
|
18
|
Watson M, Chaves AR, Gebara A, Desforges M, Broomfield A, Landry N, Lemoyne A, Shim S, Drodge J, Cuda J, Kiaee N, Nasr Y, Carleton C, Daskalakis ZJ, Taylor R, Tuominen L, Brender R, Antochi R, McMurray L, Tremblay S. A naturalistic study comparing the efficacy of unilateral and bilateral sequential theta burst stimulation in treating major depression - the U-B-D study protocol. BMC Psychiatry 2023; 23:739. [PMID: 37817124 PMCID: PMC10566125 DOI: 10.1186/s12888-023-05243-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/01/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND Major depressive disorder (MDD) is a prevalent mental health condition affecting millions worldwide, leading to disability and reduced quality of life. MDD poses a global health priority due to its early onset and association with other disabling conditions. Available treatments for MDD exhibit varying effectiveness, and a substantial portion of individuals remain resistant to treatment. Repetitive transcranial magnetic stimulation (rTMS), applied to the left and/or right dorsolateral prefrontal cortex (DLPFC), is an alternative treatment strategy for those experiencing treatment-resistant MDD. The objective of this study is to investigate whether this newer form of rTMS, namely theta burst stimulation (TBS), when performed unilaterally or bilaterally, is efficacious in treatment-resistant MDD. METHODS In this naturalistic, randomized double-blinded non-inferiority trial, participants with a major depressive episode will be randomized to receive either unilateral (i.e., continuous TBS [cTBS] to the right and sham TBS to the left DLPFC) or bilateral sequential TBS (i.e., cTBS to the right and intermittent TBS [iTBS] to the left DLPFC) delivered 5 days a week for 4-6 weeks. Responders will move onto a 6-month flexible maintenance phase where TBS treatment will be delivered at a decreasing frequency depending on degree of symptom mitigation. Several clinical assessments and neuroimaging and neurophysiological biomarkers will be collected to investigate treatment response and potential associated biomarkers. A non-inferiority analysis will investigate whether bilateral sequential TBS is non-inferior to unilateral TBS and regression analyses will investigate biomarkers of treatment response. We expect to recruit a maximal of 256 participants. This trial is approved by the Research Ethics Board of The Royal's Institute of Mental Health Research (REB# 2,019,071) and will follow the Declaration of Helsinki. Findings will be published in peer-reviewed journals. DISCUSSION Comprehensive assessment of symptoms and neurophysiological biomarkers will contribute to understanding the differential efficacy of the tested treatment protocols, identifying biomarkers for treatment response, and shedding light into underlying mechanisms of TBS. Our findings will inform future clinical trials and aid in personalizing treatment selection and scheduling for individuals with MDD. TRIAL REGISTRATION The trial is registered on https://clinicaltrials.gov/ct2/home (#NCT04142996).
Collapse
Affiliation(s)
- Molly Watson
- University of Ottawa Institute of Mental Health Research at The Royal, 1145 Carling Ave, Ottawa, ON, K1Z 7K4, Canada
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada
| | - Arthur R Chaves
- University of Ottawa Institute of Mental Health Research at The Royal, 1145 Carling Ave, Ottawa, ON, K1Z 7K4, Canada
- Faculty of Health Sciences, University of Ottawa, 125 University, Ottawa, ON, K1N6N5, Canada
| | - Abir Gebara
- School of Medicine, Stanford University, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Manon Desforges
- University of Ottawa Institute of Mental Health Research at The Royal, 1145 Carling Ave, Ottawa, ON, K1Z 7K4, Canada
- Département de Psychoéducation Et Psychologie, Université du Québec en Outaouais, 283 Alexandre-Taché Boul, Gatineau, QC, J8X 3X7, Canada
| | - Antoinette Broomfield
- University of Ottawa Institute of Mental Health Research at The Royal, 1145 Carling Ave, Ottawa, ON, K1Z 7K4, Canada
| | - Noémie Landry
- University of Ottawa Institute of Mental Health Research at The Royal, 1145 Carling Ave, Ottawa, ON, K1Z 7K4, Canada
- Département de Psychoéducation Et Psychologie, Université du Québec en Outaouais, 283 Alexandre-Taché Boul, Gatineau, QC, J8X 3X7, Canada
| | - Alexandra Lemoyne
- University of Ottawa Institute of Mental Health Research at The Royal, 1145 Carling Ave, Ottawa, ON, K1Z 7K4, Canada
- Département de Psychoéducation Et Psychologie, Université du Québec en Outaouais, 283 Alexandre-Taché Boul, Gatineau, QC, J8X 3X7, Canada
| | - Stacey Shim
- University of Ottawa Institute of Mental Health Research at The Royal, 1145 Carling Ave, Ottawa, ON, K1Z 7K4, Canada
| | - Jessica Drodge
- University of Ottawa Institute of Mental Health Research at The Royal, 1145 Carling Ave, Ottawa, ON, K1Z 7K4, Canada
| | - Jennifer Cuda
- University of Ottawa Institute of Mental Health Research at The Royal, 1145 Carling Ave, Ottawa, ON, K1Z 7K4, Canada
| | - Nasim Kiaee
- University of Ottawa Institute of Mental Health Research at The Royal, 1145 Carling Ave, Ottawa, ON, K1Z 7K4, Canada
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada
| | - Youssef Nasr
- University of Ottawa Institute of Mental Health Research at The Royal, 1145 Carling Ave, Ottawa, ON, K1Z 7K4, Canada
| | - Christophe Carleton
- University of Ottawa Institute of Mental Health Research at The Royal, 1145 Carling Ave, Ottawa, ON, K1Z 7K4, Canada
- Département de Psychoéducation Et Psychologie, Université du Québec en Outaouais, 283 Alexandre-Taché Boul, Gatineau, QC, J8X 3X7, Canada
| | - Zafiris J Daskalakis
- Department of Psychiatry, University California San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA
| | - Reggie Taylor
- University of Ottawa Institute of Mental Health Research at The Royal, 1145 Carling Ave, Ottawa, ON, K1Z 7K4, Canada
- Department of Physics, Carleton University, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada
| | - Lauri Tuominen
- University of Ottawa Institute of Mental Health Research at The Royal, 1145 Carling Ave, Ottawa, ON, K1Z 7K4, Canada
- Department of Psychiatry, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Ram Brender
- Department of Psychiatry, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Royal Ottawa Mental Health Centre, 1145 Carling Ave, Ottawa, ON, K1Z 7K4, Canada
| | - Ruxandra Antochi
- Department of Psychiatry, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Royal Ottawa Mental Health Centre, 1145 Carling Ave, Ottawa, ON, K1Z 7K4, Canada
| | - Lisa McMurray
- Department of Psychiatry, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Royal Ottawa Mental Health Centre, 1145 Carling Ave, Ottawa, ON, K1Z 7K4, Canada
| | - Sara Tremblay
- University of Ottawa Institute of Mental Health Research at The Royal, 1145 Carling Ave, Ottawa, ON, K1Z 7K4, Canada.
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada.
- Département de Psychoéducation Et Psychologie, Université du Québec en Outaouais, 283 Alexandre-Taché Boul, Gatineau, QC, J8X 3X7, Canada.
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
19
|
Strafella R, Momi D, Zomorrodi R, Lissemore J, Noda Y, Chen R, Rajji TK, Griffiths JD, Vila-Rodriguez F, Downar J, Daskalakis ZJ, Blumberger DM, Voineskos D. Identifying Neurophysiological Markers of Intermittent Theta Burst Stimulation in Treatment-Resistant Depression Using Transcranial Magnetic Stimulation-Electroencephalography. Biol Psychiatry 2023; 94:454-465. [PMID: 37084864 DOI: 10.1016/j.biopsych.2023.04.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 03/12/2023] [Accepted: 04/03/2023] [Indexed: 04/23/2023]
Abstract
BACKGROUND Intermittent theta burst stimulation (iTBS) targeting the left dorsolateral prefrontal cortex is effective for treatment-resistant depression, but the effects of iTBS on neurophysiological markers remain unclear. Here, we indexed transcranial magnetic stimulation-electroencephalography (TMS-EEG) markers, specifically, the N45 and N100 amplitudes, at baseline and post-iTBS, comparing separated and contiguous iTBS schedules. TMS-EEG markers were also compared between iTBS responders and nonresponders. METHODS TMS-EEG was analyzed from a triple-blind 1:1 randomized trial for treatment-resistant depression, comparing a separated (54-minute interval) and contiguous (0-minute interval) schedule of 2 × 600-pulse iTBS for 30 treatments. Participants underwent TMS-EEG over the left dorsolateral prefrontal cortex at baseline and posttreatment. One hundred fourteen participants had usable TMS-EEG at baseline, and 98 at posttreatment. TMS-evoked potential components (N45, N100) were examined via global mean field analysis. RESULTS The N100 amplitude decreased from baseline to posttreatment, regardless of the treatment group (F1,106 = 5.20, p = .02). There were no changes in N45 amplitude in either treatment group. In responders, the N100 amplitude decreased after iTBS (F1,102 = 11.30, p = .001, pcorrected = .0004). Responders showed higher posttreatment N45 amplitude than nonresponders (F1,94 = 4.11, p = .045, pcorrected = .016). Higher baseline N100 amplitude predicted lower post-iTBS depression scores (F4,106 = 6.28, p = .00014). CONCLUSIONS These results provide further evidence for an association between the neurophysiological effects of iTBS and treatment efficacy in treatment-resistant depression. Future studies are needed to test the predictive potential for clinical applications of TMS-EEG markers.
Collapse
Affiliation(s)
- Rebecca Strafella
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada; Temerty Centre for Therapeutic Brain Intervention, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Davide Momi
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Krembil Centre for Neuroinformatics, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Reza Zomorrodi
- Temerty Centre for Therapeutic Brain Intervention, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Jennifer Lissemore
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, California
| | - Yoshihiro Noda
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Robert Chen
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada; Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada; Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Tarek K Rajji
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada; Temerty Centre for Therapeutic Brain Intervention, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Toronto Dementia Research Alliance, University of Toronto, Toronto, Ontario, Canada
| | - John D Griffiths
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada; Krembil Centre for Neuroinformatics, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Fidel Vila-Rodriguez
- Non-Invasive Neurostimulation Therapies Laboratory, Vancouver, British Columbia, Canada; Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jonathan Downar
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Zafiris J Daskalakis
- Department of Psychiatry, University of California San Diego, La Jolla, California
| | - Daniel M Blumberger
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada; Temerty Centre for Therapeutic Brain Intervention, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Daphne Voineskos
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada; Temerty Centre for Therapeutic Brain Intervention, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
20
|
Cutler AJ, Mattingly GW, Maletic V. Understanding the mechanism of action and clinical effects of neuroactive steroids and GABAergic compounds in major depressive disorder. Transl Psychiatry 2023; 13:228. [PMID: 37365161 PMCID: PMC10293235 DOI: 10.1038/s41398-023-02514-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 05/12/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023] Open
Abstract
The pathophysiology of major depressive disorder (MDD) is thought to result from impaired connectivity between key brain networks. Gamma-aminobutyric acid (GABA) is the key inhibitory neurotransmitter in the brain, working primarily via GABAA receptors, with an important role in virtually all physiologic functions in the brain. Some neuroactive steroids (NASs) are positive allosteric modulators (PAMs) of GABAA receptors and potentiate phasic and tonic inhibitory responses via activation of synaptic and extrasynaptic GABAA receptors, respectively. This review first discusses preclinical and clinical data that support the association of depression with diverse defects in the GABAergic system of neurotransmission. Decreased levels of GABA and NASs have been observed in adults with depression compared with healthy controls, while treatment with antidepressants normalized the altered levels of GABA and NASs. Second, as there has been intense interest in treatment approaches for depression that target dysregulated GABAergic neurotransmission, we discuss NASs approved or currently in clinical development for the treatment of depression. Brexanolone, an intravenous NAS and a GABAA receptor PAM, is approved by the U.S. Food and Drug Administration for the treatment of postpartum depression (PPD) in patients 15 years and older. Other NASs include zuranolone, an investigational oral GABAA receptor PAM, and PH10, which acts on nasal chemosensory receptors; clinical data to date have shown improvement in depressive symptoms with these investigational NASs in adults with MDD or PPD. Finally, the review discusses how NAS GABAA receptor PAMs may potentially address the unmet need for novel and effective treatments with rapid and sustained antidepressant effects in patients with MDD.
Collapse
|
21
|
Han S, Li XX, Wei S, Zhao D, Ding J, Xu Y, Yu C, Chen Z, Zhou DS, Yuan TF. Orbitofrontal cortex-hippocampus potentiation mediates relief for depression: A randomized double-blind trial and TMS-EEG study. Cell Rep Med 2023:101060. [PMID: 37263267 DOI: 10.1016/j.xcrm.2023.101060] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 02/01/2023] [Accepted: 05/07/2023] [Indexed: 06/03/2023]
Abstract
It has been 15 years since repetitive transcranial magnetic stimulation (rTMS) targeting the dorsolateral prefrontal cortex (DLPFC) was approved by the FDA for clinical depression treatment. Yet, the underlying mechanisms for rTMS-induced depression relief are not fully elucidated. This study analyzes TMS-electroencephalogram (EEG) data from 64 healthy control (HC) subjects and 53 patients with major depressive disorder (MDD) before and after rTMS treatment. Prior to treatment, patients with MDD have lower activity in the DLPFC, the hippocampus (HPC), the orbitofrontal cortex (OFC), and DLPFC-OFC connectivity compared with HCs. Following active rTMS treatment, patients with MDD show a significant increase in the DLPFC, HPC, and OFC. Notably, the increase in HPC activity is specifically associated with amelioration of depressive symptoms but not anxiety or sleep quality. The orbitofrontal-hippocampal pathway plays a crucial role in mediating depression relief following rTMS treatment. These findings suggest potential alternative targets for brain stimulation therapy against depression (chictr.org.cn: ChiCTR2100052007).
Collapse
Affiliation(s)
- Sizhu Han
- Department of Psychiatry, Ningbo Kangning Hospital, Ningbo 315201, China; Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai 200030, China
| | - Xing-Xing Li
- Department of Psychiatry, Ningbo Kangning Hospital, Ningbo 315201, China
| | - Shuochi Wei
- Department of Psychiatry, Ningbo Kangning Hospital, Ningbo 315201, China
| | - Di Zhao
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai 200030, China
| | - Jinjun Ding
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai 200030, China
| | - Yongming Xu
- Department of Psychiatry, Ningbo Kangning Hospital, Ningbo 315201, China
| | - Chang Yu
- Department of Psychiatry, Ningbo Kangning Hospital, Ningbo 315201, China
| | - Zan Chen
- Department of Psychiatry, Ningbo Kangning Hospital, Ningbo 315201, China
| | - Dong-Sheng Zhou
- Department of Psychiatry, Ningbo Kangning Hospital, Ningbo 315201, China.
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai 200030, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China.
| |
Collapse
|
22
|
Yuruk D, Ozger C, Garzon JF, Leffler JM, Shekunov J, Vande Voort JL, Zaccariello MJ, Nakonezny PA, Croarkin PE. Sequential bilateral accelerated theta burst stimulation in adolescents with suicidal ideation associated with major depressive disorder: Protocol for a randomized controlled trial. PLoS One 2023; 18:e0280010. [PMID: 37053246 PMCID: PMC10101506 DOI: 10.1371/journal.pone.0280010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 01/26/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND Suicide is a leading cause of death in adolescents worldwide. Previous research findings suggest that suicidal adolescents with depression have pathophysiological dorsolateral prefrontal cortex (DLPFC) deficits in γ-aminobutyric acid neurotransmission. Interventions with transcranial magnetic stimulation (TMS) directly address these underlying pathophysiological deficits in the prefrontal cortex. Theta burst stimulation (TBS) is newer dosing approach for TMS. Accelerated TBS (aTBS) involves administering multiple sessions of TMS daily as this dosing may be more efficient, tolerable, and rapid acting than standard TMS. MATERIALS AND METHODS This is a randomized, double-blind, sham-controlled trial of sequential bilateral aTBS in adolescents with major depressive disorder (MDD) and suicidal ideation. Three sessions are administered daily for 10 days. During each session, continuous TBS is administered first to the right DPFC, in which 1,800 pulses are delivered continuously over 120 seconds. Then intermittent TBS is applied to the left DPFC, in which 1,800 pulses are delivered in 2-second bursts and repeated every 10 seconds for 570 seconds. The TBS parameters were adopted from prior research, with 3-pulse, 50-Hz bursts given every 200 ms (at 5 Hz) with an intensity of 80% active motor threshold. The comparison group will receive 3 daily sessions of bilateral sham TBS treatment for 10 days. All participants will receive the standard of care for patients with depression and suicidal ideation including daily psychotherapeutic skill sessions. Long-interval intracortical inhibition (LICI) biomarkers will be measured before and after treatment. Exploratory measures will be collected with TMS and electroencephalography for biomarker development. DISCUSSION This is the first known randomized controlled trial to examine the efficacy of sequential bilateral aTBS for treating suicidal ideation in adolescents with MDD. Results from this study will also provide opportunities to further understand the neurophysiological and molecular mechanisms of suicidal ideation in adolescents. TRIAL REGISTRATION Investigational device exemption (IDE) Number: G200220, ClinicalTrials.gov (ID: NCT04701840). Registered August 6, 2020. https://clinicaltrials.gov/ct2/show/NCT04502758?term=NCT04701840&draw=2&rank=1.
Collapse
Affiliation(s)
- Deniz Yuruk
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Can Ozger
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Juan F. Garzon
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Jarrod M. Leffler
- Virginia Treatment Center for Children, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Julia Shekunov
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, United States of America
- Mayo Clinic Depression Center, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Jennifer L. Vande Voort
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, United States of America
- Mayo Clinic Depression Center, Mayo Clinic, Rochester, Minnesota, United States of America
- Mayo Clinic Children’s Research Center, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Michael J. Zaccariello
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, United States of America
- Mayo Clinic Children’s Research Center, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Paul A. Nakonezny
- Department of Population and Data Sciences, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Paul E. Croarkin
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, United States of America
- Mayo Clinic Depression Center, Mayo Clinic, Rochester, Minnesota, United States of America
- Mayo Clinic Children’s Research Center, Mayo Clinic, Rochester, Minnesota, United States of America
| |
Collapse
|
23
|
Chen MH, Su TP, Chen LF, Li CT, Wu HJ, Lin WC, Tsai SJ, Bai YM, Mao WC, Tu PC, Jeng JS, Li WC. Cortical inhibition function is associated with baseline suicidal symptoms and post-ketamine suicidal symptom reduction among patients with treatment-resistant depression and strong suicidal ideation. J Psychiatr Res 2023; 161:48-53. [PMID: 36898326 DOI: 10.1016/j.jpsychires.2023.03.010] [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: 10/28/2022] [Revised: 02/14/2023] [Accepted: 03/02/2023] [Indexed: 03/12/2023]
Abstract
BACKGROUND Whether cortical excitation and inhibition functions differ between patients with treatment-resistant depression (TRD) and strong suicidal ideation (SI) and healthy subjects and whether 0.5 mg/kg ketamine infusion can modulate cortical excitation and inhibition functions among patients with TRD-SI remain unclear. METHODS A total of 29 patients with TRD-SI and 35 age- and sex-matched healthy controls were assessed using paired-pulse transcranial magnetic stimulation. The patients were randomly assigned to receive either a single 0.5-mg/kg ketamine or 0.045-mg/kg midazolam infusion. Depressive and suicidal symptoms were assessed at baseline and 240 min after infusion. Intracortical facilitation (ICF), short-interval intracortical inhibition (SICI), and long-interval intracortical inhibition (LICI), all of which reflect cortical excitability and inhibition functions, were measured at the same time points. RESULTS The patients with TRD-SI had lower ICF (p < 0.001) estimates (worse cortical excitatory function) and higher SICI (p = 0.032) and LICI (p < 0.001) estimates (worse cortical inhibitory function) compared with the control group. Higher SICI estimates at baseline were associated with greater baseline suicidal symptoms. No differences were found in the SICI, ICF, and LICI estimates at 240 min after the infusion between the two groups. Low-dose ketamine did not alter the cortical excitation and inhibition functions of the patients with TRD-SI. However, decreased SICI estimates (greater cortical inhibition function) were related to the reduction of suicidal symptoms. DISCUSSION Dysfunction of cortical excitation and inhibition may play a crucial role in the pathomechanisms of TRD and suicidal symptoms. However, we found a lack of predictive ability of the baseline cortical excitation and inhibition parameters on the antidepressant and antisuicidal effect of low-dose ketamine infusion.
Collapse
Affiliation(s)
- Mu-Hong Chen
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Tung-Ping Su
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Psychiatry, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Li-Fen Chen
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan; Integrated Brain Research Unit, Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Cheng-Ta Li
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hui-Ju Wu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wei-Chen Lin
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ya-Mei Bai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Chung Mao
- Department of Psychiatry, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Pei-Chi Tu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jia-Shyun Jeng
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Chi Li
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan; Integrated Brain Research Unit, Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
24
|
Knyahnytska Y, Zomorrodi R, Kaster T, Voineskos D, Trevizol A, Blumberger D. Neural Correlates of the DEEPP (Anti-suicidal Response to Ketamine in Treatment-Resistant Bipolar Depression) Study: Protocol for a Pilot, Open-Label Clinical Trial. JMIR Res Protoc 2023; 12:e41013. [PMID: 36573651 PMCID: PMC9919457 DOI: 10.2196/41013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 12/17/2022] [Accepted: 12/27/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Suicide is among the top 10 leading causes of death worldwide. Of people who died by suicide, the majority are diagnosed with depression. It is estimated that 25%-60% of people with bipolar depression (BD) will attempt suicide at least once, and 10%-15% will die by suicide. Several treatments, such as lithium, clozapine, electroconvulsive therapy, and cognitive behavioral therapy, have been shown to be effective in treating suicidality. However, these treatments can be difficult to tolerate or may take months to take effect. Ketamine, a glutamate N-methyl-D-aspartate antagonist, has been shown to have rapid antisuicidal effect and antidepressant qualities, and is thus a promising intervention to target acute suicidality in patients with BD. However, the biological mechanism underlying its therapeutic action remains poorly understood. Enhancing our understanding of underlying mechanisms of action for ketamine's effectiveness in reducing suicidality is critical to establishing biological markers of treatment response and developing tailored, personalized interventions for patients with BD. OBJECTIVE This is an open-label clinical trial to test the safety and feasibility of repeated ketamine infusions to treat acute suicidality. The primary objective is to test the safety and feasibility of ketamine intervention. The secondary objective is to examine ketamine's potential neurophysiological mechanisms of action by assessing cortical excitation and inhibition to determine potential biomarkers of clinical response. Other objectives are to evaluate the effect of ketamine on acute suicidality and other clinical outcomes, such as depressive symptoms and quality of life, to inform a future larger trial. METHODS This open-label clinical trial aims to test the safety and feasibility of repeated ketamine infusions in patients with BD for suicidality and to assess ketamine's neurophysiological effects. A sterile form of racemic ketamine hydrochloride will be administered over a 40-minute intravenous infusion 2 times per week on nonconsecutive days for 4 weeks (8 sessions). We will recruit 30 adults (24-65 year olds) over 2 years from an academic psychiatric hospital in Toronto, Canada. RESULTS This study is currently ongoing and actively recruiting participants. So far, 5 participants have completed the trial, 1 is currently in active treatment, and 8 participants are on the waitlist to be screened. We anticipate initial results being available in the fall of 2023. This proposal was presented as a poster presentation at the Research to Reality Global Summit on Psychedelic-Assisted Therapies and Medicine, held in May 2022 in Toronto, Canada. CONCLUSIONS Developing effective interventions for acute suicidality in high-risk populations such as those with BD remains a major therapeutic challenge. Ketamine is a promising treatment due to its rapid antidepressant and antisuicidal effects, but its underlying neurophysiological mechanisms of action remain unknown. TRIAL REGISTRATION ClinicalTrials.gov NCT05177146; https://clinicaltrials.gov/ct2/show/NCT05177146. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/41013.
Collapse
Affiliation(s)
| | - Reza Zomorrodi
- Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Tyler Kaster
- Centre for Addiction and Mental Health, Toronto, ON, Canada
| | | | | | | |
Collapse
|
25
|
Abo Aoun M, Meek BP, Clair L, Wikstrom S, Prasad B, Modirrousta M. Prognostic factors in major depressive disorder: comparing responders and non-responders to Repetitive Transcranial Magnetic Stimulation (rTMS), a naturalistic retrospective chart review. Psychiatry Clin Neurosci 2023; 77:38-47. [PMID: 36207801 DOI: 10.1111/pcn.13488] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 08/18/2022] [Accepted: 10/04/2022] [Indexed: 01/06/2023]
Abstract
AIM Repetitive transcranial magnetic stimulation (rTMS) is widely utilized as an effective treatment for major depressive disorder (MDD) with varying response rates. Factors associated with better treatment outcome remain scarce. This naturalistic retrospective chart review hopes to shed light on easily obtainable and measurable predictive factors for patients referred to rTMS. METHODS Protocol parameters, medication, rated scales, rTMS protocols, and treatment outcomes were reviewed for 196 patients with MDD who received rTMS at Saint Boniface Hospital between 2013 and 2019. Logistic regression and marginal effects were used to assess the different predictor variables for response (50% reduction or more on the Hamilton Depression Rating Scale (Ham-D)) and remission (Ham-D of ≤7 by the last session). RESULTS HamD at 10 sessions was predictive of remission, and Sheehan Disability Scale (SDS) at 10 sessions was predictive of response to rTMS. Ham-D, SDS, and Beck Anxiety Inventory were predictive of remission and response by Beck Anxiety Inventory 20 sessions. High frequency rTMS had a similar response and remission rate to low frequency, but higher response rate to intermittent Theta Burst Stimulation with no difference in remission rate. Positive predictive factors of response were lower age and bupropion use. Negative predictive factors were antipsychotics, anticonvulsants, or benzodiazepine use. For remission, antipsychotics or anticonvulsants use were negative predictors; bupropion use and higher resting motor threshold were positive predictors. Severity of depression as measured by baseline HamD was not associated with different probabilities of treatment success.
Collapse
Affiliation(s)
| | - Benjamin P Meek
- Department of Clinical Health Psychology, University of Manitoba, Winnipeg, Canada
| | - Luc Clair
- Department of Economics, University of Winnipeg, Winnipeg, Canada.,Canadian Centre for Agri-Food Research in Health and Medicine, Saint Boniface Research Hospital, Winnipeg, Canada
| | - Sara Wikstrom
- Saint Boniface Hospital, Psychiatry, Winnipeg, Canada
| | | | - Mandana Modirrousta
- BrainWave Clinic, Winnipeg, Canada.,Department of Psychiatry, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
26
|
Habets PC, Kalafatakis K, Dzyubachyk O, van der Werff SJ, Keo A, Thakrar J, Mahfouz A, Pereira AM, Russell GM, Lightman SL, Meijer OC. Transcriptional and cell type profiles of cortical brain regions showing ultradian cortisol rhythm dependent responses to emotional face stimulation. Neurobiol Stress 2023; 22:100514. [PMID: 36660181 PMCID: PMC9842700 DOI: 10.1016/j.ynstr.2023.100514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 01/02/2023] [Accepted: 01/02/2023] [Indexed: 01/05/2023] Open
Abstract
The characteristic endogenous circadian rhythm of plasma glucocorticoid concentrations is made up from an underlying ultradian pulsatile secretory pattern. Recent evidence has indicated that this ultradian cortisol pulsatility is crucial for normal emotional response in man. In this study, we investigate the anatomical transcriptional and cell type signature of brain regions sensitive to a loss of ultradian rhythmicity in the context of emotional processing. We combine human cell type and transcriptomic atlas data of high spatial resolution with functional magnetic resonance imaging (fMRI) data. We show that the loss of cortisol ultradian rhythm alters emotional processing response in cortical brain areas that are characterized by transcriptional and cellular profiles of GABAergic function. We find that two previously identified key components of rapid non-genomic GC signaling - the ANXA1 gene and retrograde endocannabinoid signaling - show most significant differential expression (q = 3.99e-10) and enrichment (fold enrichment = 5.56, q = 9.09e-4). Our results further indicate that specific cell types, including a specific NPY-expressing GABAergic neuronal cell type, and specific G protein signaling cascades underly the cerebral effects of a loss of ultradian cortisol rhythm. Our results provide a biological mechanistic underpinning of our fMRI findings, indicating specific cell types and cascades as a target for manipulation in future experimental studies.
Collapse
Affiliation(s)
- Philippe C. Habets
- Leiden University Medical Center, Department of Medicine, Division of Endocrinology, 2300 RC Leiden, the Netherlands
- Amsterdam University Medical Centre, Department of Psychiatry, Department of Anatomy and Neurosciences, 1081 HZ, Amsterdam, the Netherlands
| | - Konstantinos Kalafatakis
- Henry Wellcome Laboratories of Integrative Neuroscience and Endocrinology, Bristol Medical School, University of Bristol, BS1 3NY, Bristol, United Kingdom
- Institute of Health Science Education, Barts and the London School of Medicine & Dentistry, Queen Mary University of London Malta Campus, VCT 2520, Victoria Gozo, Malta
| | - Oleh Dzyubachyk
- Department of Radiology, Division of Medical Image Processing, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
- Leiden University Medical Center, Department of Cell and Chemical Biology, Section Electron Microscopy, 2300 RC, Leiden, the Netherlands
| | - Steven J.A. van der Werff
- Department of Psychiatry, Leiden University Medical Center LUMC, Leiden, the Netherlands
- Leiden Institute for Brain and Cognition, Leiden, the Netherlands
| | - Arlin Keo
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, the Netherlands
- Delft Bioinformatics Lab, Delft University of Technology, Delft, the Netherlands
| | - Jamini Thakrar
- Henry Wellcome Laboratories of Integrative Neuroscience and Endocrinology, Bristol Medical School, University of Bristol, BS1 3NY, Bristol, United Kingdom
| | - Ahmed Mahfouz
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, the Netherlands
- Delft Bioinformatics Lab, Delft University of Technology, Delft, the Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Alberto M. Pereira
- Leiden University Medical Center, Department of Medicine, Division of Endocrinology, 2300 RC Leiden, the Netherlands
- Department of Endocrinology & Metabolism, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Georgina M. Russell
- Henry Wellcome Laboratories of Integrative Neuroscience and Endocrinology, Bristol Medical School, University of Bristol, BS1 3NY, Bristol, United Kingdom
| | - Stafford L. Lightman
- Henry Wellcome Laboratories of Integrative Neuroscience and Endocrinology, Bristol Medical School, University of Bristol, BS1 3NY, Bristol, United Kingdom
| | - Onno C. Meijer
- Leiden University Medical Center, Department of Medicine, Division of Endocrinology, 2300 RC Leiden, the Netherlands
- Leiden Institute for Brain and Cognition, Leiden, the Netherlands
| |
Collapse
|
27
|
Tallarico M, Pisano M, Leo A, Russo E, Citraro R, De Sarro G. Antidepressant Drugs for Seizures and Epilepsy: Where do we Stand? Curr Neuropharmacol 2023; 21:1691-1713. [PMID: 35761500 PMCID: PMC10514547 DOI: 10.2174/1570159x20666220627160048] [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: 02/26/2022] [Revised: 06/13/2022] [Accepted: 06/18/2022] [Indexed: 11/22/2022] Open
Abstract
People with epilepsy (PWE) are more likely to develop depression and both these complex chronic diseases greatly affect health-related quality of life (QOL). This comorbidity contributes to the deterioration of the QOL further than increasing the severity of epilepsy worsening prognosis. Strong scientific evidence suggests the presence of shared pathogenic mechanisms. The correct identification and management of these factors are crucial in order to improve patients' QOL. This review article discusses recent original research on the most common pathogenic mechanisms of depression in PWE and highlights the effects of antidepressant drugs (ADs) against seizures in PWE and animal models of seizures and epilepsy. Newer ADs, such as selective serotonin reuptake inhibitors (SRRI) or serotonin-noradrenaline reuptake inhibitors (SNRI), particularly sertraline, citalopram, mirtazapine, reboxetine, paroxetine, fluoxetine, escitalopram, fluvoxamine, venlafaxine, duloxetine may lead to improvements in epilepsy severity whereas the use of older tricyclic antidepressant (TCAs) can increase the occurrence of seizures. Most of the data demonstrate the acute effects of ADs in animal models of epilepsy while there is a limited number of studies about the chronic antidepressant effects in epilepsy and epileptogenesis or on clinical efficacy. Much longer treatments are needed in order to validate the effectiveness of these new alternatives in the treatment and the development of epilepsy, while further clinical studies with appropriate protocols are warranted in order to understand the real potential contribution of these drugs in the management of PWE (besides their effects on mood).
Collapse
Affiliation(s)
- Martina Tallarico
- System and Applied Pharmacology, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Maria Pisano
- System and Applied Pharmacology, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Antonio Leo
- System and Applied Pharmacology, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Emilio Russo
- System and Applied Pharmacology, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Rita Citraro
- System and Applied Pharmacology, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Giovambattista De Sarro
- System and Applied Pharmacology, Science of Health Department, School of Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| |
Collapse
|
28
|
Zheng N, Ou Y, Li H, Liu F, Xie G, Li P, Lang B, Guo W. Shared and differential fractional amplitude of low-frequency fluctuation patterns at rest in major depressive disorders with or without sleep disturbance. Front Psychol 2023; 14:1153335. [PMID: 37034932 PMCID: PMC10075231 DOI: 10.3389/fpsyg.2023.1153335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 03/01/2023] [Indexed: 04/11/2023] Open
Abstract
Objective Sleep disturbances (SD) are commonly found in patients with major depressive disorder (MDD). This study aims to explore the influence of SD symptoms on clinical characteristics in patients with MDD and to investigate the shared and distinct fractional amplitude of low-frequency fluctuation (fALFF) patterns in these patients with or without SD symptoms. Methods Twenty-four MDD patients with SD symptoms (Pa_s), 33 MDD patients without SD symptoms (Pa_ns) and 32 healthy controls (HCs) were included in this study. The fALFF and correlation analyses were applied to analyze the features of imaging and clinical data. Results Pa_s showed more severe anxiety and depression than Pa_ns. Compared with Pa_ns, Pa_s exhibited increased fALFF value in the left precuneus. Patients shared abnormal fALFF in the frontal-occipital brain regions. There was a positive correlation between fALFF values of the left precuneus and sleep disturbance scores (r = 0.607, p = 0.0000056734) in all patients in addition to a negative correlation between fALFF values of the left MOG/cuneus and HAMD-17 total scores (r = -0.595, p = 0.002141) in Pa_s. The receiver operating characteristic (ROC) results of the fALFF could be used to discriminate Pa_s from Pa_ns with a specificity of 72.73% and a sensitivity of 70.83%. Conclusion Pa_s displayed more serious anxiety and depression symptoms. Patients shared abnormal fALFF in the frontal-occipital brain regions, which may be a common characteristic for MDD. And increased fALFF value in the left precuneus might be a specific neuroimaging feature of MDD patients with SD symptoms.
Collapse
Affiliation(s)
- Nanxi Zheng
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yangpan Ou
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Huabing Li
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Feng Liu
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Guojun Xie
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan, Guangdong, China
| | - Ping Li
- Department of Psychiatry, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Bing Lang
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- *Correspondence: Bing Lang,
| | - Wenbin Guo
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Wenbin Guo,
| |
Collapse
|
29
|
Huang J, Xu F, Yang L, Tuolihong L, Wang X, Du Z, Zhang Y, Yin X, Li Y, Lu K, Wang W. Involvement of the GABAergic system in PTSD and its therapeutic significance. Front Mol Neurosci 2023; 16:1052288. [PMID: 36818657 PMCID: PMC9928765 DOI: 10.3389/fnmol.2023.1052288] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
The neurobiological mechanism of post-traumatic stress disorder (PTSD) is poorly understood. The inhibition of GABA neurons, especially in the amygdala, is crucial for the precise regulation of the consolidation, expression, and extinction of fear conditioning. The GABAergic system is involved in the pathophysiological process of PTSD, with several studies demonstrating that the function of the GABAergic system decreases in PTSD patients. This paper reviews the preclinical and clinical studies, neuroimaging techniques, and pharmacological studies of the GABAergic system in PTSD and summarizes the role of the GABAergic system in PTSD. Understanding the role of the GABAergic system in PTSD and searching for new drug targets will be helpful in the treatment of PTSD.
Collapse
Affiliation(s)
| | - Fei Xu
- Department of Psychiatry of School of Public Health, Southern Medical University, Guangzhou, China
| | - Liping Yang
- Department of Applied Psychology of School of Public Health, Southern Medical University, Guangzhou, China
| | - Lina Tuolihong
- Department of Basic Medical of Basic Medical College, Southern Medical University, Guangzhou, China
| | - Xiaoyu Wang
- Eight-Year Master's and Doctoral Program in Clinical Medicine of the First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Zibo Du
- Eight-Year Master's and Doctoral Program in Clinical Medicine of the First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Yiqi Zhang
- Eight-Year Master's and Doctoral Program in Clinical Medicine of the First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Xuanlin Yin
- Department of Basic Medical of Basic Medical College, Southern Medical University, Guangzhou, China
| | - Yingjun Li
- Department of Medical Laboratory Science, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Kangrong Lu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, China
| | - Wanshan Wang
- Department of Laboratory Animal Center, Southern Medical University, Guangzhou, China
| |
Collapse
|
30
|
Jannati A, Oberman LM, Rotenberg A, Pascual-Leone A. Assessing the mechanisms of brain plasticity by transcranial magnetic stimulation. Neuropsychopharmacology 2023; 48:191-208. [PMID: 36198876 PMCID: PMC9700722 DOI: 10.1038/s41386-022-01453-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/10/2022]
Abstract
Transcranial magnetic stimulation (TMS) is a non-invasive technique for focal brain stimulation based on electromagnetic induction where a fluctuating magnetic field induces a small intracranial electric current in the brain. For more than 35 years, TMS has shown promise in the diagnosis and treatment of neurological and psychiatric disorders in adults. In this review, we provide a brief introduction to the TMS technique with a focus on repetitive TMS (rTMS) protocols, particularly theta-burst stimulation (TBS), and relevant rTMS-derived metrics of brain plasticity. We then discuss the TMS-EEG technique, the use of neuronavigation in TMS, the neural substrate of TBS measures of plasticity, the inter- and intraindividual variability of those measures, effects of age and genetic factors on TBS aftereffects, and then summarize alterations of TMS-TBS measures of plasticity in major neurological and psychiatric disorders including autism spectrum disorder, schizophrenia, depression, traumatic brain injury, Alzheimer's disease, and diabetes. Finally, we discuss the translational studies of TMS-TBS measures of plasticity and their therapeutic implications.
Collapse
Affiliation(s)
- Ali Jannati
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Lindsay M Oberman
- Center for Neuroscience and Regenerative Medicine, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Alexander Rotenberg
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Alvaro Pascual-Leone
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- Hinda and Arthur Marcus Institute for Aging Research and Deanna and Sidney Wolk Center for Memory Health, Hebrew SeniorLife, Boston, MA, USA.
- Guttmann Brain Health Institute, Institut Guttmann, Barcelona, Spain.
| |
Collapse
|
31
|
Wang Y, Meng W, Liu Z, An Q, Hu X. Cognitive impairment in psychiatric diseases: Biomarkers of diagnosis, treatment, and prevention. Front Cell Neurosci 2022; 16:1046692. [DOI: 10.3389/fncel.2022.1046692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
Psychiatric diseases, such as schizophrenia, bipolar disorder, autism spectrum disorder, and major depressive disorder, place a huge health burden on society. Cognitive impairment is one of the core characteristics of psychiatric disorders and a vital determinant of social function and disease recurrence in patients. This review thus aims to explore the underlying molecular mechanisms of cognitive impairment in major psychiatric disorders and identify valuable biomarkers for diagnosis, treatment and prevention of patients.
Collapse
|
32
|
Wu C, Yang L, Feng S, Zhu L, Yang L, Liu TCY, Duan R. Therapeutic non-invasive brain treatments in Alzheimer's disease: recent advances and challenges. Inflamm Regen 2022; 42:31. [PMID: 36184623 PMCID: PMC9527145 DOI: 10.1186/s41232-022-00216-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/13/2022] [Indexed: 11/10/2022] Open
Abstract
Alzheimer's disease (AD) is one of the major neurodegenerative diseases and the most common form of dementia. Characterized by the loss of learning, memory, problem-solving, language, and other thinking abilities, AD exerts a detrimental effect on both patients' and families' quality of life. Although there have been significant advances in understanding the mechanism underlying the pathogenesis and progression of AD, there is no cure for AD. The failure of numerous molecular targeted pharmacologic clinical trials leads to an emerging research shift toward non-invasive therapies, especially multiple targeted non-invasive treatments. In this paper, we reviewed the advances of the most widely studied non-invasive therapies, including photobiomodulation (PBM), transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), and exercise therapy. Firstly, we reviewed the pathological changes of AD and the challenges for AD studies. We then introduced these non-invasive therapies and discussed the factors that may affect the effects of these therapies. Additionally, we review the effects of these therapies and the possible mechanisms underlying these effects. Finally, we summarized the challenges of the non-invasive treatments in future AD studies and clinical applications. We concluded that it would be critical to understand the exact underlying mechanisms and find the optimal treatment parameters to improve the translational value of these non-invasive therapies. Moreover, the combined use of non-invasive treatments is also a promising research direction for future studies and sheds light on the future treatment or prevention of AD.
Collapse
Affiliation(s)
- Chongyun Wu
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Luoman Yang
- Department of Anesthesiology, Peking University Third Hospital (PUTH), Beijing, 100083, China
| | - Shu Feng
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Ling Zhu
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Luodan Yang
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA.
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| | - Timon Cheng-Yi Liu
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| | - Rui Duan
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| |
Collapse
|
33
|
Hormonal Agents for the Treatment of Depression Associated with the Menopause. Drugs Aging 2022; 39:607-618. [PMID: 35908135 PMCID: PMC9355926 DOI: 10.1007/s40266-022-00962-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2022] [Indexed: 11/29/2022]
Abstract
Perimenopause marks the transition from a woman’s reproductive stage to menopause. Usually occurring between 42 and 52 years of age, it is determined clinically by the onset of irregular menstrual cycles or variable cycle lengths. Women are at an increased risk of depression and anxiety during perimenopause and the menopausal transition. Depressive symptoms experienced in perimenopause are often more severe compared to pre- and post-menopause. During menopausal transition, the impact of fluctuating estrogen in the central nervous system (CNS) can have negative psychological effects for some women. Traditional first-line management of menopausal depression involves antidepressants, with modest outcomes. The positive effects of estrogen treatment in the CNS are becoming increasingly recognised, and hormonal therapy (HT) with estrogen may have a role in the treatment of menopausal depression. In this review we will outline the prevalence, impact and neurochemical basis of menopausal-associated depression, as well as hormone-based approaches that have increasing promise as effective treatments.
Collapse
|
34
|
Newton DF, Oh H, Shukla R, Misquitta K, Fee C, Banasr M, Sibille E. Chronic Stress Induces Coordinated Cortical Microcircuit Cell-Type Transcriptomic Changes Consistent With Altered Information Processing. Biol Psychiatry 2022; 91:798-809. [PMID: 34861977 DOI: 10.1016/j.biopsych.2021.10.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/29/2021] [Accepted: 10/13/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Information processing in cortical cell microcircuits involves regulation of excitatory pyramidal (PYR) cells by inhibitory somatostatin- (SST), parvalbumin-, and vasoactive intestinal peptide-expressing interneurons. Human postmortem and rodent studies show impaired PYR cell dendritic morphology and decreased SST cell markers in major depressive disorder or after chronic stress. However, knowledge of coordinated changes across microcircuit cell types is virtually absent. METHODS We investigated the transcriptomic effects of unpredictable chronic mild stress (UCMS) on distinct microcircuit cell types in the medial prefrontal cortex (cingulate regions 24a, 24b, and 32) in mice. C57BL/6 mice, exposed to UCMS or control housing for 5 weeks, were assessed for anxiety- and depressive-like behaviors. Microcircuit cell types were laser microdissected and processed for RNA sequencing. RESULTS UCMS induced predicted elevations in behavioral emotionality in mice. DESeq2 analysis revealed unique differentially expressed genes in each cell type after UCMS. Presynaptic functions, oxidative stress response, metabolism, and translational regulation were differentially dysregulated across cell types, whereas nearly all cell types showed downregulated postsynaptic gene signatures. Across the cortical microcircuit, we observed a shift from a distributed transcriptomic coordination across cell types in control mice toward UCMS-induced increased coordination between PYR, SST, and parvalbumin cells and a hub-like role for PYR cells. Finally, we identified a microcircuit-wide coexpression network enriched in synaptic, bioenergetic, and oxidative stress response genes that correlated with UCMS-induced behaviors. CONCLUSIONS These findings suggest cell-specific deficits, microcircuit-wide synaptic reorganization, and a shift in cells regulating the cortical excitation-inhibition balance, suggesting increased coordinated regulation of PYR cells by SST and parvalbumin cells.
Collapse
Affiliation(s)
- Dwight F Newton
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Campbell Family Mental Health Research Institute of the Centre of Addiction and Mental Health, Toronto, Ontario, Canada
| | - Hyunjung Oh
- Campbell Family Mental Health Research Institute of the Centre of Addiction and Mental Health, Toronto, Ontario, Canada
| | - Rammohan Shukla
- Campbell Family Mental Health Research Institute of the Centre of Addiction and Mental Health, Toronto, Ontario, Canada; Department of Neurosciences, University of Toledo, Toledo, Ohio
| | - Keith Misquitta
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Campbell Family Mental Health Research Institute of the Centre of Addiction and Mental Health, Toronto, Ontario, Canada
| | - Corey Fee
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Campbell Family Mental Health Research Institute of the Centre of Addiction and Mental Health, Toronto, Ontario, Canada
| | - Mounira Banasr
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Campbell Family Mental Health Research Institute of the Centre of Addiction and Mental Health, Toronto, Ontario, Canada
| | - Etienne Sibille
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Campbell Family Mental Health Research Institute of the Centre of Addiction and Mental Health, Toronto, Ontario, Canada.
| |
Collapse
|
35
|
Transcranial Magnetic Stimulation Indices of Cortical Excitability Enhance the Prediction of Response to Pharmacotherapy in Late-Life Depression. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:265-275. [PMID: 34311121 PMCID: PMC8783923 DOI: 10.1016/j.bpsc.2021.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/16/2021] [Accepted: 07/14/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND Older adults with late-life depression (LLD) often experience incomplete or lack of response to first-line pharmacotherapy. The treatment of LLD could be improved using objective biological measures to predict response. Transcranial magnetic stimulation (TMS) can be used to measure cortical excitability, inhibition, and plasticity, which have been implicated in LLD pathophysiology and associated with brain stimulation treatment outcomes in younger adults with depression. TMS measures have not yet been investigated as predictors of treatment outcomes in LLD or pharmacotherapy outcomes in adults of any age with depression. METHODS We assessed whether pretreatment single-pulse and paired-pulse TMS measures, combined with clinical and demographic measures, predict venlafaxine treatment response in 76 outpatients with LLD. We compared the predictive performance of machine learning models including or excluding TMS predictors. RESULTS Two single-pulse TMS measures predicted venlafaxine response: cortical excitability (neuronal membrane excitability) and the variability of cortical excitability (dynamic fluctuations in excitability levels). In cross-validation, models using a combination of these TMS predictors, clinical markers of treatment resistance, and age classified patients with 73% ± 11% balanced accuracy (average correct classification rate of responders and nonresponders; permutation testing, p < .005); these models significantly outperformed (corrected t test, p = .025) models using clinical and demographic predictors alone (60% ± 10% balanced accuracy). CONCLUSIONS These preliminary findings suggest that single-pulse TMS measures of cortical excitability may be useful predictors of response to pharmacotherapy in LLD. Future studies are needed to confirm these findings and determine whether combining TMS predictors with other biomarkers further improves the accuracy of predicting LLD treatment outcome.
Collapse
|
36
|
Stoby KS, Rafique SA, Oeltzschner G, Steeves JKE. Continuous and intermittent theta burst stimulation to the visual cortex do not alter GABA and glutamate concentrations measured by magnetic resonance spectroscopy. Brain Behav 2022; 12:e2478. [PMID: 35029058 PMCID: PMC8865152 DOI: 10.1002/brb3.2478] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/07/2021] [Accepted: 12/14/2021] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Theta burst stimulation (TBS), a form of repetitive transcranial magnetic stimulation (rTMS), uses repeated high-frequency bursts to non-invasively modulate neural processes in the brain. An intermittent TBS (iTBS) protocol is generally considered "excitatory," while continuous TBS (cTBS) is considered "inhibitory." However, the majority of work that has led to these effects being associated with the respective protocols has been done in the motor cortex, and it is well established that TMS can have variable effects across the brain. OBJECTIVES AND METHOD We investigated the effects of iTBS and cTBS to the primary visual cortex (V1) on composite levels of gamma-aminobutyric acid + co-edited macromolecules (GABA+) and glutamate + glutamine (Glx) since these are key inhibitory and excitatory neurotransmitters, respectively. Participants received a single session of cTBS, iTBS, or sham TBS to V1. GABA+ and Glx were quantified in vivo at the stimulation site using spectral-edited proton magnetic resonance spectroscopy (1 H-MRS) at 3T. Baseline pre-TBS GABA+ and Glx levels were compared to immediate post-TBS and 1 h post-TBS levels. RESULTS There were no significant changes in GABA+ or Glx following either of the TBS conditions. Visual cortical excitability, measured using phosphene thresholds, remained unchanged following both cTBS and iTBS conditions. There was no relationship between excitability thresholds and GABA+ or Glx levels. However, TBS did alter the relationship between GABA+ and Glx for up to 1 h following stimulation. CONCLUSIONS These findings demonstrate that a single session of TBS to the visual cortex can be used without significant effects on the tonic levels of these key neurotransmitters; and add to our understanding that TBS has differential effects at visual, motor, and frontal cortices.
Collapse
Affiliation(s)
- Karlene S Stoby
- Centre for Vision Research and Department of Psychology, York University, Toronto, ON, Canada
| | - Sara A Rafique
- Centre for Vision Research and Department of Psychology, York University, Toronto, ON, Canada
| | - Georg Oeltzschner
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Jennifer K E Steeves
- Centre for Vision Research and Department of Psychology, York University, Toronto, ON, Canada
| |
Collapse
|
37
|
Knyahnytska Y, Zomorrodi R, Kaster T, Voineskos D, Trevizol A, Blumberger D. The Safety, Clinical, and Neurophysiological Effects of Intranasal Ketamine in Patients Who Do Not Respond to Electroconvulsive Therapy: Protocol for a Pilot, Open-Label Clinical Trial. JMIR Res Protoc 2022; 11:e30163. [PMID: 34882570 PMCID: PMC8804953 DOI: 10.2196/30163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 12/03/2021] [Accepted: 12/08/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Major depressive disorder is among the most disabling illnesses worldwide, with a lifetime prevalence of 16.2%. Research suggests that 20% to 40% of patients with depression do not respond to pharmacotherapy, developing treatment-resistant depression. Electroconvulsive therapy is the gold standard for treating individuals with treatment-resistant depression, with remission rates of approximately 75% to 90%. However, 10% to 25% of patients do not respond to electroconvulsive therapy, and many are unable to tolerate it due to the side effects. Both groups are considered to be patients who do not respond to electroconvulsive therapy, because both groups continue to exhibit symptoms of severe depression, have a limited number of treatment options available, and are in need of rapid treatment. Ketamine, an N-methyl-D-aspartate receptor antagonist, has been shown to exert rapid antidepressant effects in patients with treatment-resistant depression when administered in subanesthetic doses through 40-minute intravenous infusions. Recently, a ketamine compound, esketamine (Spravato), that is administered through the intranasal route received regulatory approval by the US Food and Drug Administration and Health Canada to treat depression. However, esketamine is challenging to access due to high costs and limited availability. Racemic ketamine (rketamine) is cheap and easy to access; however, the effects in patients who have not responded to electroconvulsive therapy have yet to be understood or tested. This study will use transcranial magnetic stimulation to study mechanisms of human brain cortical physiology at the systemic level to identify neurobiomarkers of response. OBJECTIVE The objective of this open-label pilot clinical trial is to test the feasibility and safety of intranasal ketamine in patients who have not responded to electroconvulsive therapy. The primary outcome is to determine the feasibility of a larger randomized controlled trial to test the efficacy of intranasal ketamine for patients who have not responded to electroconvulsive therapy for clinical indicators in unipolar depression. The secondary outcome is to determine the preliminary effects of an intervention on clinical outcomes, such as depressive symptoms, suicidal ideation, and quality of living. The third outcome is to explore neurophysiological changes as measured by transcranial magnetic stimulation electromyography and electroencephalography to measure changes in cortical excitability as potential predictors of clinical response. METHODS A sterile solution of racemic ketamine hydrochloride will be administered twice per week for 4 weeks (8 sessions) intranasally to patients with treatment-resistant depression who did not respond to or could not tolerate an acute course of electroconvulsive therapy. We will recruit 25 adults (24-65 years old) over the course of 2 years from an academic psychiatric hospital in Toronto, Canada. RESULTS This study has received ethics approval, and funding has been secured. The study is currently active. CONCLUSIONS This is the first study to test repeated doses of intranasal rketamine in patients who have not responded to electroconvulsive therapy for depression. Results from this study will (1) inform the development of a larger adequately powered randomized controlled trial to test the efficacy of intranasal ketamine for depression and (2) determine potential neurophysiological markers of clinical response. TRIAL REGISTRATION Clinical Trials.gov NCT05137938; http://clinicaltrials.gov/ct2/show/NCT05137938. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) PRR1-10.2196/30163.
Collapse
Affiliation(s)
| | - Reza Zomorrodi
- Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Tyler Kaster
- Centre for Addiction and Mental Health, Toronto, ON, Canada
| | | | | | | |
Collapse
|
38
|
Yao HK, Guet-McCreight A, Mazza F, Moradi Chameh H, Prevot TD, Griffiths JD, Tripathy SJ, Valiante TA, Sibille E, Hay E. Reduced inhibition in depression impairs stimulus processing in human cortical microcircuits. Cell Rep 2022; 38:110232. [PMID: 35021088 DOI: 10.1016/j.celrep.2021.110232] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 10/07/2021] [Accepted: 12/16/2021] [Indexed: 12/01/2022] Open
Abstract
Cortical processing depends on finely tuned excitatory and inhibitory connections in neuronal microcircuits. Reduced inhibition by somatostatin-expressing interneurons is a key component of altered inhibition associated with treatment-resistant major depressive disorder (depression), which is implicated in cognitive deficits and rumination, but the link remains to be better established mechanistically in humans. Here we test the effect of reduced somatostatin interneuron-mediated inhibition on cortical processing in human neuronal microcircuits using a data-driven computational approach. We integrate human cellular, circuit, and gene expression data to generate detailed models of human cortical microcircuits in health and depression. We simulate microcircuit baseline and response activity and find a reduced signal-to-noise ratio and increased false/failed detection of stimuli due to a higher baseline activity in depression. We thus apply models of human cortical microcircuits to demonstrate mechanistically how reduced inhibition impairs cortical processing in depression, providing quantitative links between altered inhibition and cognitive deficits.
Collapse
Affiliation(s)
- Heng Kang Yao
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Alexandre Guet-McCreight
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R7, Canada
| | - Frank Mazza
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | | | - Thomas D Prevot
- Department of Psychiatry, University of Toronto, Toronto, ON M5S 1A1, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R7, Canada
| | - John D Griffiths
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R7, Canada; Department of Psychiatry, University of Toronto, Toronto, ON M5S 1A1, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Shreejoy J Tripathy
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A1, Canada; Department of Psychiatry, University of Toronto, Toronto, ON M5S 1A1, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Taufik A Valiante
- Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A1, Canada; Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON M5S 1A1; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 1A1, Canada; Department of Surgery, University of Toronto, Toronto, ON M5S 1A1, Canada; Max Planck-University of Toronto Center for Neural Science and Technology, University of Toronto, Toronto, ON M5S 1A1, Canada; Center for Advancing Neurotechnological Innovation to Application, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Etienne Sibille
- Department of Psychiatry, University of Toronto, Toronto, ON M5S 1A1, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R7, Canada; Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Etay Hay
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A1, Canada; Department of Psychiatry, University of Toronto, Toronto, ON M5S 1A1, Canada.
| |
Collapse
|
39
|
Du H, Shen X, Du X, Zhao L, Zhou W. Altered Visual Cortical Excitability Is Associated With Psychopathological Symptoms in Major Depressive Disorder. Front Psychiatry 2022; 13:844434. [PMID: 35321224 PMCID: PMC8936091 DOI: 10.3389/fpsyt.2022.844434] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/14/2022] [Indexed: 12/03/2022] Open
Abstract
Previous studies suggest that in people with major depressive disorder (MDD), there exists a perturbation of the normal balance between the excitatory and inhibitory neurotransmitter systems in the visual cortex, indicating the possibility of altered visual cortical excitability. However, investigations into the neural activities of the visual cortex in MDD patients yielded inconsistent findings. The present study aimed to evaluate the visual cortical excitability utilizing a paired-pulse stimulation paradigm in patients with MDD and to access the paired-pulse behavior of recording visual evoked potentials (VEPs) as a marker of MDD. We analyzed the amplitudes of VEPs and paired-pulse suppression (PPS) at four different stimulus onset asynchronies (SOAs) spanning 93 ms to 133 ms. Further, the relationship between PPS and the symptom severity of depression was investigated using Spearman's correlation. We found that, whereas the first VEP amplitude remained unchanged, the second VEP amplitude was significantly higher in the MDD group compared to the healthy controls. As a result, the amplitude ratio (second VEP amplitude/first VEP amplitude) increased, indicating reduced PPS and thus increased excitability in the visual cortex. Moreover, we found the amplitude ratios had a significantly positive correlation with the symptom severity of depression in MDD, indicating a clinically useful biomarker for MDD. Our findings provide new insights into the changes in the excitation-inhibition balance of visual cortex in MDD, which could pave the way for specific clinical interventions.
Collapse
Affiliation(s)
- Hongheng Du
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China.,Division of Clinical Electrophysiology Center, Chongqing Key Laboratory of Cerebrovascular Disease Research, Chongqing, China
| | - Xue Shen
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China.,Division of Clinical Electrophysiology Center, Chongqing Key Laboratory of Cerebrovascular Disease Research, Chongqing, China
| | - Xiaoyan Du
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China.,Division of Clinical Electrophysiology Center, Chongqing Key Laboratory of Cerebrovascular Disease Research, Chongqing, China
| | - Libo Zhao
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China.,Division of Clinical Electrophysiology Center, Chongqing Key Laboratory of Cerebrovascular Disease Research, Chongqing, China
| | - Wenjun Zhou
- Department of Ophthalmology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
40
|
Castricum J, Birkenhager TK, Kushner SA, Elgersma Y, Tulen JHM. Cortical Inhibition and Plasticity in Major Depressive Disorder. Front Psychiatry 2022; 13:777422. [PMID: 35153873 PMCID: PMC8825489 DOI: 10.3389/fpsyt.2022.777422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/05/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Major depressive disorder (MDD) is a severe psychiatric disorder that is associated with various cognitive impairments, including learning and memory deficits. As synaptic plasticity is considered an important mechanism underlying learning and memory, deficits in cortical plasticity might play a role in the pathophysiology of patients with MDD. We used Transcranial Magnetic Stimulation (TMS) to assess inhibitory neurotransmission and cortical plasticity in the motor cortex of MDD patients and controls. METHODS We measured the cortical silent period (CSP) and short interval cortical inhibition (SICI), as well as intermittent theta-burst stimulation (iTBS), in 9 drug-free MDD inpatients and 18 controls. RESULTS The overall response to the CSP, SICI, and iTBS paradigms was not significantly different between the patient and control groups. iTBS induction resulted in significant potentiation after 20 mins in the control group (t (17) = -2.8, p = 0.01), whereas no potentiation was observed in patients. CONCLUSIONS Potentiation of MEP amplitudes was not observed within the MDD group. No evidence was found for medium-to-large effect size differences in CSP and SICI measures in severely depressed drug-free patients, suggesting that reduced cortical inhibition is unlikely to be a robust correlate of the pathophysiological mechanism in MDD. However, these findings should be interpreted with caution due to the high inter-subject variability and the small sample size. SIGNIFICANCE These findings advance our understanding of neurophysiological functioning in drug-free severely depressed inpatients.
Collapse
Affiliation(s)
- Jesminne Castricum
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Neuroscience, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Psychiatry, Erasmus University Medical Center, Rotterdam, Netherlands.,ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Netherlands
| | - Tom K Birkenhager
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Steven A Kushner
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Ype Elgersma
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Neuroscience, Erasmus University Medical Center, Rotterdam, Netherlands.,ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Netherlands
| | - Joke H M Tulen
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, Netherlands.,ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
41
|
Abstract
Gamma-aminobutyric acid (GABA), the main inhibitory neurotransmitter in the brain, acts at the ionotropic GABAA and GABAC receptors, and the metabotropic GABAB receptor. This chapter summarizes the studies that have investigated the role of the GABAB receptor in stress-related psychiatric disorders including anxiety and mood disorders. Overall, clinical and preclinical evidences strongly suggest that the GABAB receptor is a therapeutic candidate for depression and anxiety disorders. However, the clinical development of GABAB receptor-based drugs to treat these disorders has been hampered by their potential side-effects, particularly those of agonists. Nevertheless, the discovery of novel GABAB receptor allosteric modulators, and increasing understanding of the influence of specific intracellular GABAB receptor-associated proteins on GABAB receptor activity, may now pave the way towards GABAB receptor therapeutics in the treatment of mood and anxiety disorders.
Collapse
Affiliation(s)
- Daniela Felice
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| | - Olivia F O'Leary
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|
42
|
Nayeri T, Sarvi S, Daryani A. Toxoplasmosis: Targeting neurotransmitter systems in psychiatric disorders. Metab Brain Dis 2022; 37:123-146. [PMID: 34476718 DOI: 10.1007/s11011-021-00824-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/14/2021] [Indexed: 12/30/2022]
Abstract
The most common form of the disease caused by Toxoplasma gondii (T. gondii) is latent toxoplasmosis due to the formation of tissue cysts in various organs, such as the brain. Latent toxoplasmosis is probably a risk factor in the development of some neuropsychiatric disorders. Behavioral changes after infection are caused by the host immune response, manipulation by the parasite, central nervous system (CNS) inflammation, as well as changes in hormonal and neuromodulator relationships. The present review focused on the exact mechanisms of T. gondii effect on the alteration of behavior and neurotransmitter levels, their catabolites and metabolites, as well as the interaction between immune responses and this parasite in the etiopathogenesis of psychiatric disorders. The dysfunction of neurotransmitters in the neural transmission is associated with several neuropsychiatric disorders. However, further intensive studies are required to determine the effect of this parasite on altering the level of neurotransmitters and the role of neurotransmitters in the etiology of host behavioral changes.
Collapse
Affiliation(s)
- Tooran Nayeri
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahabeddin Sarvi
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ahmad Daryani
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
43
|
Oh H, Newton D, Lewis D, Sibille E. Lower Levels of GABAergic Function Markers in Corticotropin-Releasing Hormone-Expressing Neurons in the sgACC of Human Subjects With Depression. Front Psychiatry 2022; 13:827972. [PMID: 35280164 PMCID: PMC8913899 DOI: 10.3389/fpsyt.2022.827972] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/21/2022] [Indexed: 11/13/2022] Open
Abstract
RATIONALE A previous transcriptome meta-analysis revealed significantly lower levels of corticotropin-releasing hormone (CRH) mRNA in corticolimbic brain regions in major depressive disorder (MDD) subjects, suggesting that cortical CRH-expressing (CRH+) cells are affected in MDD. Rodent studies show that cortical CRH is mostly expressed in GABAergic interneurons; however, the characteristic features of CRH+ cells in human brain cortex and their association with MDD are largely unknown. METHODS Subgenual anterior cingulate cortex (sgACC) of human subjects without brain disorders were labeled using fluorescent in situ hybridization (FISH) for CRH and markers of excitatory (SLC17A7), inhibitory (GAD1) neurons, as well as markers of other interneuron subpopulations (PVALB, SST, VIP). MDD-associated changes in CRH+ cell density and cellular CRH expression (n = 6/group) were analyzed. RNA-sequencing was performed on sgACC CRH+ interneurons from comparison and MDD subjects (n = 6/group), and analyzed for group differences. The effect of reduced BDNF on CRH expression was tested in mice with blocked TrkB function. RESULTS About 80% of CRH+ cells were GABAergic and 17.5% were glutamatergic. CRH+ GABAergic interneurons co-expressed VIP (52%), SST (7%), or PVALB (7%). MDD subjects displayed lower CRH mRNA levels in GABAergic interneurons relative to comparison subjects without changes in cell density. CRH+ interneurons show transcriptomic profile suggesting lower excitability and less GABA release and reuptake. Further analyses suggested that these molecular changes are not mediated by altered glucocorticoid feedback and potentially occur downstream for a common modulator of neurotrophic function. SUMMARY CRH+ cells in human sgACC are a heterogeneous population of GABAergic interneurons, although largely co-expressing VIP. Our data suggest that MDD is associated with reduced markers of inhibitory function in sgACC CRH+ interneurons, and provide further evidence for impaired GABAergic function in the cortex in MDD.
Collapse
Affiliation(s)
- Hyunjung Oh
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
| | - Dwight Newton
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada.,Departments of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - David Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada.,Departments of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
44
|
Daskalakis ZJ, McClintock SM, Hadas I, Kallioniemi E, Zomorrodi R, Throop A, Palmer L, Farzan F, Thorpe KE, Tamminga C, Blumberger DM. Confirmatory Efficacy and Safety Trial of Magnetic Seizure Therapy for Depression (CREST-MST): protocol for identification of novel biomarkers via neurophysiology. Trials 2021; 22:906. [PMID: 34895296 PMCID: PMC8666076 DOI: 10.1186/s13063-021-05873-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 11/24/2021] [Indexed: 11/18/2022] Open
Abstract
Background Electroconvulsive therapy (ECT) is the most effective treatment for treatment-resistant depression (TRD), especially for acute suicidal ideation, but the associated cognitive adverse effects and negative stigma limit its use. Another seizure therapy under development is magnetic seizure therapy (MST), which could potentially overcome the restrictions associated with ECT with similar efficacy. The neurophysiological targets and mechanisms of seizure therapy, however, remain poorly understood. Methods/design This neurophysiological study protocol is published as a companion to the overall Confirmatory Efficacy and Safety Trial of Magnetic Seizure Therapy for Depression (CREST-MST) protocol that describes our two-site, double-blind, randomized, non-inferiority clinical trial to develop MST as an effective and safe treatment for TRD. Our aim for the neurophysiological component of the study is to evaluate two biomarkers, one to predict remission of suicidal ideation (primary outcome) and the other to predict cognitive impairment (secondary outcome). Suicidal ideation will be assessed through cortical inhibition, which according to our preliminary studies, correlates with remission of suicidal ideation. Cortical inhibition will be measured with simultaneous transcranial magnetic stimulation (TMS) and electroencephalography (EEG), TMS-EEG, which measures TMS-evoked EEG activity. Cognitive adverse effects associated with seizure therapy, on the contrary, will be evaluated via multiscale entropy analysis reflecting the complexity of ongoing resting-state EEG activity. Discussion ECT and MST are known to influence cortical inhibition associated with depression, suicidal ideation severity, and clinical outcome. Therefore, evaluating cortical inhibition and brain temporal dynamics will help understand the pathophysiology of depression and suicidal ideation and define new biological targets that could aid clinicians in diagnosing and selecting treatments. Resting-state EEG complexity was previously associated with the degree of cognitive side effects after a seizure therapy. This neurophysiological metric may help clinicians assess the risk for adverse effects caused by these useful and effective treatments. Trial registration ClinicalTrials.govNCT03191058. Registered on June 19, 2017.
Collapse
Affiliation(s)
- Zafiris J Daskalakis
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA.
| | - Shawn M McClintock
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Itay Hadas
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Elisa Kallioniemi
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Reza Zomorrodi
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Alanah Throop
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Lucy Palmer
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Faranak Farzan
- School of Mechatronic Systems Engineering, Simon Fraser University, Surrey, British Columbia, Canada
| | - Kevin E Thorpe
- Applied Health Research Centre, Li Ka Shing Knowledge Institute of St. Michael's, Toronto, Ontario, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Carol Tamminga
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Daniel M Blumberger
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Institute of Medical Science and Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
45
|
Della Vecchia A, Arone A, Piccinni A, Mucci F, Marazziti D. GABA System in Depression: Impact on Pathophysiology and Psychopharmacology. Curr Med Chem 2021; 29:5710-5730. [PMID: 34781862 DOI: 10.2174/0929867328666211115124149] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/21/2021] [Accepted: 09/30/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The pathophysiology of major depressive disorder (MDD), one of the major causes of worldwide disability, is still largely unclear, despite the increasing data reporting evidence of multiple alterations of different systems. Recently, there was a renewed interest in the signalling of gamma aminobutyric acid (GABA) - the main inhibitory neurotransmitter. OBJECTIVE The aim of this study was to review and comment on the available literature about the involvement of GABA in MDD, as well as on novel GABAergic compounds possibly useful as antidepressants. METHODS We carried out a narrative review through Pubmed, Google Scholar and Scopus, by using specific keywords. RESULTS The results, derived from various research tools, strongly support the presence of a deficiency of the GABA system in MDD, which appears to be restored by common antidepressant treatments. More recent publications would indicate the complex interactions between GABA and all the other processes involved in MDD, such as monoamine neurotransmission, hypothalamus-pituitary adrenal axis functioning, neurotrophism, and immune response. Taken together, all these findings seem to further support the complexity of the pathophysiology of MDD, possibly reflecting the heterogeneity of the clinical pictures. CONCLUSION Although further data are necessary to support the specificity of GABA deficiency in MDD, the available findings would suggest that novel GABAergic compounds might constitute innovative therapeutic strategies in MDD.
Collapse
Affiliation(s)
- Alessandra Della Vecchia
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa. Italy
| | - Alessandro Arone
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa. Italy
| | - Armando Piccinni
- Saint Camillus International University of Health and Medical Sciences, Rome. Italy
| | - Federico Mucci
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena. Italy
| | - Donatella Marazziti
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa. Italy
| |
Collapse
|
46
|
Zhang J, Kaye AP, Wang J, Girgenti MJ. Transcriptomics of the depressed and PTSD brain. Neurobiol Stress 2021; 15:100408. [PMID: 34703849 PMCID: PMC8524242 DOI: 10.1016/j.ynstr.2021.100408] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 10/07/2021] [Accepted: 10/09/2021] [Indexed: 12/13/2022] Open
Abstract
Stress is the response of an organism to demands for change, yet excessive or chronic stress contributes to nearly all psychiatric disorders. The advent of high-throughput transcriptomic methods such as single cell RNA sequencing poses new opportunities to understand the neurobiology of stress, yet substantial barriers to understanding stress remain. Stress adaptation is an organismal survival mechanism conserved across all organisms, yet there is an infinity of potential stressful experiences. Unraveling shared and separate transcriptional programs for adapting to stressful experience remains a challenge, despite methodological and analytic advances. Here we review the state of the field focusing on the technologies used to study the transcriptome for the stress neurobiologist, and also attempt to identify central questions about the heterogeneity of stress for those applying transcriptomic approaches. We further explore how postmortem transcriptome studies aided by preclinical animal models are converging on common molecular pathways for adaptation to aversive experience. Finally, we discuss approaches to integrate large genomic datasets with human neuroimaging and other datasets.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Computer Science, University of California- Irvine, Irvine, CA, USA
| | - Alfred P. Kaye
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Jiawei Wang
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Matthew J. Girgenti
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- National Center for PTSD, U.S. Department of Veterans Affairs, USA
| |
Collapse
|
47
|
Lybech LKM, Calabró M, Briuglia S, Drago A, Crisafulli C. Suicide Related Phenotypes in a Bipolar Sample: Genetic Underpinnings. Genes (Basel) 2021; 12:genes12101482. [PMID: 34680877 PMCID: PMC8535342 DOI: 10.3390/genes12101482] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/31/2022] Open
Abstract
Suicide in Bipolar Disorder (BD) is a relevant clinical concern. Genetics may shape the individual risk for suicide behavior in BD, together with known clinical factors. The lack of consistent replication in BD may be associated with its multigenetic component. In the present contribution we analyzed a sample of BD individuals (from STEP-BD database) to identify the genetic variants potentially associated with three different suicide-related phenotypes: (1) a feeling that the life was not worth living; (2) fantasies about committing a violent suicide; (3) previous attempted suicide. The sample under analysis included 1115 BD individuals. None of the SNPs reached genome-wide significance. However, a trend of association was evidenced for rs2767403, an intron variant of AOPEP gene, in association with phenotype #1 (p = 5.977 × 10−6). The molecular pathway analysis showed a significant enrichment in all the investigated phenotypes on pathways related to post synaptic signaling, neurotransmission and neurodevelopment. Further, NOTCH signaling or the γ-aminobutyric acid (GABA)-ergic signaling were found to be associated with specific suicide-related phenotypes. The present investigation contributes to the hypothesis that the genetic architecture of suicide behaviors in BD is related to alteration of entire pathways rather than single genes. In particular, our molecular pathway analysis points on some specific molecular events that could be the focus of further research in this field.
Collapse
Affiliation(s)
- Line K. M. Lybech
- Unit for Psychiatric Research, Psychiatry, Aalborg University Hospital, DK-9100 Aalborg, Denmark;
| | - Marco Calabró
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (M.C.); (S.B.)
| | - Silvana Briuglia
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (M.C.); (S.B.)
| | - Antonio Drago
- Unit for Psychiatric Research, Psychiatry, Aalborg University Hospital, DK-9100 Aalborg, Denmark;
- Correspondence: (A.D.); (C.C.); Tel.: +45-97-64-30-00 (A.D.); +39-(0)9-0221-3373 (C.C.)
| | - Concetta Crisafulli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (M.C.); (S.B.)
- Correspondence: (A.D.); (C.C.); Tel.: +45-97-64-30-00 (A.D.); +39-(0)9-0221-3373 (C.C.)
| |
Collapse
|
48
|
Banaie Boroujeni K, Tiesinga P, Womelsdorf T. Interneuron-specific gamma synchronization indexes cue uncertainty and prediction errors in lateral prefrontal and anterior cingulate cortex. eLife 2021; 10:69111. [PMID: 34142661 PMCID: PMC8248985 DOI: 10.7554/elife.69111] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/17/2021] [Indexed: 12/27/2022] Open
Abstract
Inhibitory interneurons are believed to realize critical gating functions in cortical circuits, but it has been difficult to ascertain the content of gated information for well-characterized interneurons in primate cortex. Here, we address this question by characterizing putative interneurons in primate prefrontal and anterior cingulate cortex while monkeys engaged in attention demanding reversal learning. We find that subclasses of narrow spiking neurons have a relative suppressive effect on the local circuit indicating they are inhibitory interneurons. One of these interneuron subclasses showed prominent firing rate modulations and (35–45 Hz) gamma synchronous spiking during periods of uncertainty in both, lateral prefrontal cortex (LPFC) and anterior cingulate cortex (ACC). In LPFC, this interneuron subclass activated when the uncertainty of attention cues was resolved during flexible learning, whereas in ACC it fired and gamma-synchronized when outcomes were uncertain and prediction errors were high during learning. Computational modeling of this interneuron-specific gamma band activity in simple circuit motifs suggests it could reflect a soft winner-take-all gating of information having high degree of uncertainty. Together, these findings elucidate an electrophysiologically characterized interneuron subclass in the primate, that forms gamma synchronous networks in two different areas when resolving uncertainty during adaptive goal-directed behavior.
Collapse
Affiliation(s)
| | - Paul Tiesinga
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, Netherlands
| | - Thilo Womelsdorf
- Department of Psychology, Vanderbilt University, Nashville, United States.,Department of Biology, Centre for Vision Research, York University, Toronto, Canada
| |
Collapse
|
49
|
Spatial inhibition of return is impaired in mild cognitive impairment and mild Alzheimer's disease. PLoS One 2021; 16:e0252958. [PMID: 34125847 PMCID: PMC8202934 DOI: 10.1371/journal.pone.0252958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 05/25/2021] [Indexed: 11/26/2022] Open
Abstract
Spatial inhibition of return (IOR) refers to the phenomenon by which individuals are slower to respond to stimuli appearing at a previously cued location compared to un-cued locations. Here with a group of older adults (n = 56, 58–80 (67.9±5.2) year old, 31 females, 18.7±3.6 years of education), we provide evidence supporting the notion that spatial IOR is mildly impaired in individuals with mild cognitive impairment (MCI) or mild Alzheimer’s disease (AD), and the impairment is detectable using a double cue paradigm. Furthermore, reduced spatial IOR in high-risk healthy older individuals is associated with reduced memory and other neurocognitive task performance, suggesting that the double cue spatial IOR paradigm may be useful in detecting MCI and early AD.
Collapse
|
50
|
Yin YY, Wang YH, Liu WG, Yao JQ, Yuan J, Li ZH, Ran YH, Zhang LM, Li YF. The role of the excitation:inhibition functional balance in the mPFC in the onset of antidepressants. Neuropharmacology 2021; 191:108573. [PMID: 33945826 DOI: 10.1016/j.neuropharm.2021.108573] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/30/2021] [Accepted: 04/14/2021] [Indexed: 12/15/2022]
Abstract
Currently available antidepressants, such as selective serotonin reuptake inhibitors (SSRIs) and serotonin and norepinephrine reuptake inhibitors (SNRIs), generally require weeks to months to produce a therapeutic response, but the mechanism of action underlying the delayed onset of antidepressant-like action remains to be elucidated. The balance between excitatory glutamatergic pyramidal neurons and inhibitory γ-aminobutyric acid (GABA) interneurons, i.e., the excitation:inhibition functional (E:I) balance, in the medial prefrontal cortex (mPFC) is critical in regulating several behaviors and might play an important mediating role in the mechanism of rapid antidepressant-like action reported by several studies. In the present study, the multichannel electrophysiological technique was used to record the firing activities of pyramidal neurons and interneurons and investigate the effects of a single dose of fluoxetine and ketamine (both 10 mg/kg, i.p.) on the E:I functional balance in the rat mPFC after 90 min or 24 h, and the forced swimming test (FST) was used to evaluate the antidepressant-like effects of fluoxetine and ketamine. The present study also explored the effects of chronic treatment with fluoxetine (10 mg/kg, i.g.) for 7 d or 21 d on the E:I functional balance in the mPFC. The present results suggested that a single dose of ketamine could both significantly increase the firing activities of pyramidal neurons and significantly decrease the firing activities of interneurons in the mPFC and exerted significant antidepressant-like action on the FST after 90 min and 24 h, but fluoxetine had no such effects under the same conditions. However, chronic treatment with fluoxetine for 21 d (but not 7 d) could significantly affect the firing activities of pyramidal neurons and interneurons in the mPFC. Taken together, the present results indicated that rapid regulation of the E:I functional balance in the mPFC might be an important common mechanism of rapid-acting antidepressants and the delayed onset of SSRIs might be partly attributed to their inability to rapidly regulate the E:I functional balance in the mPFC. The present study provided a new entry point to the development of rapid-acting antidepressants.
Collapse
Affiliation(s)
- Yong-Yu Yin
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Yun-Hui Wang
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | | | - Jun-Qi Yao
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Jin Yuan
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Ze-Han Li
- Capital Normal University High School, Beijing, China
| | - Yu-Hua Ran
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Li-Ming Zhang
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China.
| | - Yun-Feng Li
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China; Beijing Institute of Basic Medical Sciences, Beijing, China.
| |
Collapse
|