1
|
Hing B, Mitchell SB, Filali Y, Eberle M, Hultman I, Matkovich M, Kasturirangan M, Johnson M, Wyche W, Jimenez A, Velamuri R, Ghumman M, Wickramasinghe H, Christian O, Srivastava S, Hultman R. Transcriptomic Evaluation of a Stress Vulnerability Network Using Single-Cell RNA Sequencing in Mouse Prefrontal Cortex. Biol Psychiatry 2024; 96:886-899. [PMID: 38866174 PMCID: PMC11524784 DOI: 10.1016/j.biopsych.2024.05.023] [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: 10/20/2023] [Revised: 04/24/2024] [Accepted: 05/27/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Increased vulnerability to stress is a major risk factor for several mood disorders, including major depressive disorder. Although cellular and molecular mechanisms associated with depressive behaviors following stress have been identified, little is known about the mechanisms that confer the vulnerability that predisposes individuals to future damage from chronic stress. METHODS We used multisite in vivo neurophysiology in freely behaving male and female C57BL/6 mice (n = 12) to measure electrical brain network activity previously identified as indicating a latent stress vulnerability brain state. We combined this neurophysiological approach with single-cell RNA sequencing of the prefrontal cortex to identify distinct transcriptomic differences between groups of mice with inherent high and low stress vulnerability. RESULTS We identified hundreds of differentially expressed genes (padjusted < .05) across 5 major cell types in animals with high and low stress vulnerability brain network activity. This unique analysis revealed that GABAergic (gamma-aminobutyric acidergic) neuron gene expression contributed most to the network activity of the stress vulnerability brain state. Upregulation of mitochondrial and metabolic pathways also distinguished high and low vulnerability brain states, especially in inhibitory neurons. Importantly, genes that were differentially regulated with vulnerability network activity significantly overlapped (above chance) with those identified by genome-wide association studies as having single nucleotide polymorphisms significantly associated with depression as well as genes more highly expressed in postmortem prefrontal cortex of patients with major depressive disorder. CONCLUSIONS This is the first study to identify cell types and genes involved in a latent stress vulnerability state in the brain.
Collapse
Affiliation(s)
- Benjamin Hing
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Sara B Mitchell
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa
| | - Yassine Filali
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa
| | - Maureen Eberle
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Ian Hultman
- Department of Statistics and Actuarial Science, University of Iowa, Iowa City, Iowa
| | - Molly Matkovich
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | | | - Micah Johnson
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa
| | - Whitney Wyche
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Alli Jimenez
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Radha Velamuri
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Mahnoor Ghumman
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Himali Wickramasinghe
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Olivia Christian
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Sanvesh Srivastava
- Department of Statistics and Actuarial Science, University of Iowa, Iowa City, Iowa
| | - Rainbo Hultman
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa; Department of Psychiatry, University of Iowa, Iowa City, Iowa.
| |
Collapse
|
2
|
Herzog S, Bartlett EA, Zanderigo F, Galfalvy HC, Burke A, Mintz A, Schmidt M, Hauser E, Huang YY, Melhem N, Sublette ME, Miller JM, Mann JJ. Neuroinflammation, Stress-Related Suicidal Ideation, and Negative Mood in Depression. JAMA Psychiatry 2024:2825422. [PMID: 39504032 PMCID: PMC11541744 DOI: 10.1001/jamapsychiatry.2024.3543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 08/17/2024] [Indexed: 11/09/2024]
Abstract
Importance Brain translocator protein 18k Da (TSPO) binding, a putative marker of neuroinflammatory processes (eg, gliosis), is associated with stress and elevated in depressed and suicidal populations. However, it is unclear whether neuroinflammation moderates the impact of daily life stress on suicidal ideation and negative affect, thereby increasing risk for suicidal behavior. Objective To examine the association of TSPO binding in participants with depression with real-world daily experiences of acute stress-related suicidal ideation and negative affect, as well as history of suicidal behavior and clinician-rated suicidal ideation. Design, Setting, and Participants Data for this cross-sectional study were collected from June 2019 through July 2023. Procedures were conducted at a hospital-based research center in New York, New York. Participants were recruited via clinical referrals, the Columbia University research subject web portal, and from responses to internet advertisements. Of 148 participants who signed informed consent for study protocols, 53 adults aged 18 to 60 years who met DSM-5 diagnostic criteria for current major depressive disorder completed procedures with approved data and were enrolled. Participants were free of schizophrenia spectrum disorders, active physical illness, cognitive impairment, and substance intoxication or withdrawal at the time of scan. Exposures All participants underwent positron emission tomography imaging of TSPO binding with 11C-ER176 and concurrent arterial blood sampling. Main Outcome and Measures A weighted average of 11C-ER176 total distribution volume (VT) was computed across 11 a priori brain regions and made up the primary outcome measure. Clinician-rated suicidal ideation was measured via the Beck Scale for Suicidal Ideation (BSS). A subset of participants (n = 21) completed 7 days of ecological momentary assessment (EMA), reporting daily on suicidal ideation, negative affect, and stressors. Results In the overall sample of 53 participants (mean [SD] age, 29.5 [9.8] years; 37 [69.8%] female and 16 [30.2%] male), 11C-ER176 VT was associated at trend levels with clinician-rated suicidal ideation severity (β, 0.19; 95% CI, -0.03 to 0.39; P = .09) and did not differ by suicide attempt history (n = 15; β, 0.18; 95% CI, -0.04 to 0.37; P = .11). Exploratory analyses indicated that presence of suicidal ideation (on BSS or EMA) was associated with higher 11C-ER176 VT (β, 0.21; 95% CI, 0.01 to 0.98; P = .045). In 21 participants who completed EMA, 11C-ER176 VT was associated with greater suicidal ideation and negative affect during EMA periods with stressors compared with nonstress periods (β, 0.12; SE, 0.06; 95% CI, 0.01 to 0.23; P = .03 and β, 0.19; SE, 0.06; 95% CI, 0.08 to 0.30; P < .001, respectively). Conclusion and Relevance TSPO binding in individuals with depression may be a marker of vulnerability to acute stress-related increases in suicidal ideation and negative affect. Continued study is needed to determine the causal direction of TSPO binding and stress-related suicidal ideation or negative affect and whether targeting neuroinflammation may improve resilience to life stress in patients with depression.
Collapse
Affiliation(s)
- Sarah Herzog
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - Elizabeth A. Bartlett
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - Francesca Zanderigo
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - Hanga C. Galfalvy
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
- Department of Biostatistics, Columbia University, New York, New York
| | - Ainsley Burke
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - Akiva Mintz
- Department of Radiology, Columbia University Irving Medical Center, New York, New York
| | - Mike Schmidt
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - Eric Hauser
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - Yung-yu Huang
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - Nadine Melhem
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - M. Elizabeth Sublette
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - Jeffrey M. Miller
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - J. John Mann
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
- Department of Radiology, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
3
|
Shao X, Wang Y, Geng Z, Liang G, Zhu X, Liu L, Meng M, Duan L, Zhu G. Novel therapeutic targets for major depressive disorder related to oxidative stress identified by integrative multi-omics and multi-trait study. Transl Psychiatry 2024; 14:443. [PMID: 39426956 PMCID: PMC11490649 DOI: 10.1038/s41398-024-03126-0] [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: 06/26/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/21/2024] Open
Abstract
Oxidative stress (OS) is strongly implicated in the pathophysiology of major depressive disorder (MDD) but the molecular mechanisms remain largely unknown. The purpose of this study is to identify genes related to both OS and MDD, and further to evaluate the utility of these genes as diagnostic markers and potential treatment targets. We searched datasets related to MDD from the Gene Expression Omnibus (GEO) database for differentially expressed genes (DEGs) also related to OS according to GeneCards. Bioinformatics analyses and machine learning algorithms were used to identify hub genes mediating OS-MDD interactions. A summary data-based Mendelian randomization (SMR) approach was employed to identify possible causal genes for MDD from blood tissue eQLT data. These investigations identified 32 genes mediating OS-MDD interactions, while SMR analysis identified KCNE1 (OR = 1.057, 95%CI = 1.013-1.102, P value = 0.010), MAPK3 (OR = 1.023, 95%CI = 1.004-1.043, P value = 0.020), and STIP1 (OR = 0.792, 95%CI = 0.641-0.979, P value = 0.031) as OS-related causal genes for MDD. These genes may thus serve as useful diagnostic markers and potential therapeutic targets.
Collapse
Affiliation(s)
- Xiaojun Shao
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Yuan Wang
- Department of Psychiatry, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Zhongli Geng
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
- Shenyang Mental Health Center, Shenyang, 110168, China
| | | | - Xiaotong Zhu
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
- Department of Psychiatry, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110005, China
| | - Lu Liu
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
- Department of Psychiatry, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110005, China
| | - Ming Meng
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
- Shenyang Mental Health Center, Shenyang, 110168, China
| | - Li Duan
- Department of Health Management Research, Chengde Medical University, Chengde, 067000, China
| | - Gang Zhu
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
4
|
Swann P, Mirza-Davies A, O'Brien J. Associations Between Neuropsychiatric Symptoms and Inflammation in Neurodegenerative Dementia: A Systematic Review. J Inflamm Res 2024; 17:6113-6141. [PMID: 39262651 PMCID: PMC11389708 DOI: 10.2147/jir.s385825] [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: 04/15/2024] [Accepted: 08/30/2024] [Indexed: 09/13/2024] Open
Abstract
Background Neuropsychiatric symptoms are common in dementia and linked to adverse outcomes. Inflammation is increasingly recognized as playing a role as a driver of early disease progression in Alzheimer's disease (AD) and related dementias. Inflammation has also been linked to primary psychiatric disorders, however its association with neuropsychiatric symptoms in neurodegenerative dementias remains uncertain. Methods We conducted a systematic literature review investigating associations between inflammation and neuropsychiatric symptoms in neurodegenerative dementias, including AD, Lewy body, Frontotemporal, Parkinson's (PD) and Huntington's disease dementias. Results Ninety-nine studies met our inclusion criteria, and the majority (n = 59) investigated AD and/or mild cognitive impairment (MCI). Thirty-five studies included PD, and only 6 investigated non-AD dementias. Inflammation was measured in blood, CSF, by genotype, brain tissue and PET imaging. Overall, studies exhibited considerable heterogeneity and evidence for specific inflammatory markers was inconsistent, with lack of replication and few longitudinal studies with repeat biomarkers. Depression was the most frequently investigated symptom. In AD, some studies reported increases in peripheral IL-6, TNF-a associated with depressive symptoms. Preliminary investigations using PET measures of microglial activation found an association with agitation. In PD, studies reported positive associations between TNF-a, IL-6, CRP, MCP-1, IL-10 and depression. Conclusion Central and peripheral inflammation may play a role in neuropsychiatric symptoms in neurodegenerative dementias; however, the evidence is inconsistent. There is a need for multi-site longitudinal studies with detailed assessments of neuropsychiatric symptoms combined with replicable peripheral and central markers of inflammation.
Collapse
Affiliation(s)
- Peter Swann
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge, UK
| | - Anastasia Mirza-Davies
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge, UK
| | - John O'Brien
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge, UK
| |
Collapse
|
5
|
Thomas-Odenthal F, Ringwald K, Teutenberg L, Stein F, Alexander N, Bonnekoh LM, Brosch K, Dohm K, Flinkenflügel K, Grotegerd D, Hahn T, Jansen A, Leehr EJ, Meinert S, Pfarr JK, Renz H, Schürmeyer N, Stief T, Straube B, Thiel K, Usemann P, Winter A, Krug A, Nenadić I, Dannlowski U, Kircher T. Neural foundation of the diathesis-stress model: longitudinal gray matter volume changes in response to stressful life events in major depressive disorder and healthy controls. Mol Psychiatry 2024; 29:2724-2732. [PMID: 38553539 PMCID: PMC11420061 DOI: 10.1038/s41380-024-02526-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 03/03/2024] [Accepted: 03/12/2024] [Indexed: 09/25/2024]
Abstract
Recurrences of depressive episodes in major depressive disorder (MDD) can be explained by the diathesis-stress model, suggesting that stressful life events (SLEs) can trigger MDD episodes in individuals with pre-existing vulnerabilities. However, the longitudinal neurobiological impact of SLEs on gray matter volume (GMV) in MDD and its interaction with early-life adversity remains unresolved. In 754 participants aged 18-65 years (362 MDD patients; 392 healthy controls; HCs), we assessed longitudinal associations between SLEs (Life Events Questionnaire) and whole-brain GMV changes (3 Tesla MRI) during a 2-year interval, using voxel-based morphometry in SPM12/CAT12. We also explored the potential moderating role of childhood maltreatment (Childhood Trauma Questionnaire) on these associations. Over the 2-year interval, HCs demonstrated significant GMV reductions in the middle frontal, precentral, and postcentral gyri in response to higher levels of SLEs, while MDD patients showed no such GMV changes. Childhood maltreatment did not moderate these associations in either group. However, MDD patients who had at least one depressive episode during the 2-year interval, compared to those who did not, or HCs, showed GMV increases in the middle frontal, precentral, and postcentral gyri associated with an increase in SLEs and childhood maltreatment. Our findings indicate distinct GMV changes in response to SLEs between MDD patients and HCs. GMV decreases in HCs may represent adaptive responses to stress, whereas GMV increases in MDD patients with both childhood maltreatment and a depressive episode during the 2-year interval may indicate maladaptive changes, suggesting a neural foundation for the diathesis-stress model in MDD recurrences.
Collapse
Affiliation(s)
- Florian Thomas-Odenthal
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany.
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Gießen, Marburg, Germany.
| | - Kai Ringwald
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Gießen, Marburg, Germany
| | - Lea Teutenberg
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Gießen, Marburg, Germany
| | - Frederike Stein
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Gießen, Marburg, Germany
| | - Nina Alexander
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Gießen, Marburg, Germany
| | - Linda M Bonnekoh
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Department of Child and Adolescent Psychiatry, University Hospital Münster, Münster, Germany
| | - Katharina Brosch
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Gießen, Marburg, Germany
| | - Katharina Dohm
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Kira Flinkenflügel
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Dominik Grotegerd
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Tim Hahn
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Andreas Jansen
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Gießen, Marburg, Germany
- Core-Facility BrainImaging, Faculty of Medicine, University of Marburg, Marburg, Germany
| | - Elisabeth J Leehr
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Susanne Meinert
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Institute for Translational Neuroscience, University of Münster, Münster, Germany
| | - Julia-Katharina Pfarr
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Gießen, Marburg, Germany
| | - Harald Renz
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, University of Marburg, Marburg, Germany
| | - Navid Schürmeyer
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Thomas Stief
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, University of Marburg, Marburg, Germany
| | - Benjamin Straube
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Gießen, Marburg, Germany
| | - Katharina Thiel
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Paula Usemann
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Gießen, Marburg, Germany
| | - Alexandra Winter
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Axel Krug
- Department of Psychiatry and Psychotherapy, University Hospital of Bonn, Bonn, Germany
| | - Igor Nenadić
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Gießen, Marburg, Germany
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Gießen, Marburg, Germany
| |
Collapse
|
6
|
Zheng J, Zong X, Tang L, Guo H, Zhao P, Womer FY, Zhang X, Tang Y, Wang F. Characterizing the distinct imaging phenotypes, clinical behavior, and genetic vulnerability of brain maturational subtypes in mood disorders. Psychol Med 2024; 54:2774-2784. [PMID: 38804091 DOI: 10.1017/s0033291724000886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
BACKGROUND Mood disorders are characterized by great heterogeneity in clinical manifestation. Uncovering such heterogeneity using neuroimaging-based individual biomarkers, clinical behaviors, and genetic risks, might contribute to elucidating the etiology of these diseases and support precision medicine. METHODS We recruited 174 drug-naïve and drug-free patients with major depressive disorder and bipolar disorder, as well as 404 healthy controls. T1 MRI imaging data, clinical symptoms, and neurocognitive assessments, and genetics were obtained and analyzed. We applied regional gray matter volumes (GMV) and quantile normative modeling to create maturation curves, and then calculated individual deviations to identify subtypes within the patients using hierarchical clustering. We compared the between-subtype differences in GMV deviations, clinical behaviors, cell-specific transcriptomic associations, and polygenic risk scores. We also validated the GMV deviations based subtyping analysis in a replication cohort. RESULTS Two subtypes emerged: subtype 1, characterized by increased GMV deviations in the frontal cortex, cognitive impairment, a higher genetic risk for Alzheimer's disease, and transcriptionally associated with Alzheimer's disease pathways, oligodendrocytes, and endothelial cells; and subtype 2, displaying globally decreased GMV deviations, more severe depressive symptoms, increased genetic vulnerability to major depressive disorder and transcriptionally related to microglia and inhibitory neurons. The distinct patterns of GMV deviations in the frontal, cingulate, and primary motor cortices between subtypes were shown to be replicable. CONCLUSIONS Our current results provide vital links between MRI-derived phenotypes, spatial transcriptome, genetic vulnerability, and clinical manifestation, and uncover the heterogeneity of mood disorders in biological and behavioral terms.
Collapse
Affiliation(s)
- Junjie Zheng
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Xiaofen Zong
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lili Tang
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Huiling Guo
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Pengfei Zhao
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Fay Y Womer
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xizhe Zhang
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Yanqing Tang
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
- Brain Function Research Section, The First Hospital of China Medical University, Shenyang, China
- Department of Gerontology, The First Hospital of China Medical University, Shenyang, China
- Department of Psychiatry, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fei Wang
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
- Department of Mental Health, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
7
|
Viudez-Martínez A, Torregrosa AB, Navarrete F, García-Gutiérrez MS. Understanding the Biological Relationship between Migraine and Depression. Biomolecules 2024; 14:163. [PMID: 38397400 PMCID: PMC10886628 DOI: 10.3390/biom14020163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/25/2024] Open
Abstract
Migraine is a highly prevalent neurological disorder. Among the risk factors identified, psychiatric comorbidities, such as depression, seem to play an important role in its onset and clinical course. Patients with migraine are 2.5 times more likely to develop a depressive disorder; this risk becomes even higher in patients suffering from chronic migraine or migraine with aura. This relationship is bidirectional, since depression also predicts an earlier/worse onset of migraine, increasing the risk of migraine chronicity and, consequently, requiring a higher healthcare expenditure compared to migraine alone. All these data suggest that migraine and depression may share overlapping biological mechanisms. Herein, this review explores this topic in further detail: firstly, by introducing the common epidemiological and risk factors for this comorbidity; secondly, by focusing on providing the cumulative evidence of common biological aspects, with a particular emphasis on the serotoninergic system, neuropeptides such as calcitonin-gene-related peptide (CGRP), pituitary adenylate cyclase-activating polypeptide (PACAP), substance P, neuropeptide Y and orexins, sexual hormones, and the immune system; lastly, by remarking on the future challenges required to elucidate the etiopathological mechanisms of migraine and depression and providing updated information regarding new key targets for the pharmacological treatment of these clinical entities.
Collapse
Affiliation(s)
- Adrián Viudez-Martínez
- Hospital Pharmacy Service, Hospital General Dr. Balmis de Alicante, 03010 Alicante, Spain;
| | - Abraham B. Torregrosa
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - María Salud García-Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| |
Collapse
|
8
|
Sciacchitano S, Carola V, Nicolais G, Sciacchitano S, Napoli C, Mancini R, Rocco M, Coluzzi F. To Be Frail or Not to Be Frail: This Is the Question-A Critical Narrative Review of Frailty. J Clin Med 2024; 13:721. [PMID: 38337415 PMCID: PMC10856357 DOI: 10.3390/jcm13030721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/07/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Many factors have contributed to rendering frailty an emerging, relevant, and very popular concept. First, many pandemics that have affected humanity in history, including COVID-19, most recently, have had more severe effects on frail people compared to non-frail ones. Second, the increase in human life expectancy observed in many developed countries, including Italy has led to a rise in the percentage of the older population that is more likely to be frail, which is why frailty is much a more common concern among geriatricians compared to other the various health-care professionals. Third, the stratification of people according to the occurrence and the degree of frailty allows healthcare decision makers to adequately plan for the allocation of available human professional and economic resources. Since frailty is considered to be fully preventable, there are relevant consequences in terms of potential benefits both in terms of the clinical outcome and healthcare costs. Frailty is becoming a popular, pervasive, and almost omnipresent concept in many different contexts, including clinical medicine, physical health, lifestyle behavior, mental health, health policy, and socio-economic planning sciences. The emergence of the new "science of frailty" has been recently acknowledged. However, there is still debate on the exact definition of frailty, the pathogenic mechanisms involved, the most appropriate method to assess frailty, and consequently, who should be considered frail. This narrative review aims to analyze frailty from many different aspects and points of view, with a special focus on the proposed pathogenic mechanisms, the various factors that have been considered in the assessment of frailty, and the emerging role of biomarkers in the early recognition of frailty, particularly on the role of mitochondria. According to the extensive literature on this topic, it is clear that frailty is a very complex syndrome, involving many different domains and affecting multiple physiological systems. Therefore, its management should be directed towards a comprehensive and multifaceted holistic approach and a personalized intervention strategy to slow down its progression or even to completely reverse the course of this condition.
Collapse
Affiliation(s)
- Salvatore Sciacchitano
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy;
- Unit of Anaesthesia, Intensive Care and Pain Medicine, Sant’Andrea University Hospital, 00189 Rome, Italy; (M.R.); (F.C.)
- Department of Life Sciences, Health and Health Professions, Link Campus University, 00165 Rome, Italy
| | - Valeria Carola
- Department of Dynamic and Clinical Psychology and Health Studies, Sapienza University of Rome, 00189 Rome, Italy; (V.C.); (G.N.)
| | - Giampaolo Nicolais
- Department of Dynamic and Clinical Psychology and Health Studies, Sapienza University of Rome, 00189 Rome, Italy; (V.C.); (G.N.)
| | - Simona Sciacchitano
- Department of Psychiatry, La Princesa University Hospital, 28006 Madrid, Spain;
| | - Christian Napoli
- Department of Surgical and Medical Science and Translational Medicine, Sapienza University of Rome, 00189 Rome, Italy;
| | - Rita Mancini
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy;
| | - Monica Rocco
- Unit of Anaesthesia, Intensive Care and Pain Medicine, Sant’Andrea University Hospital, 00189 Rome, Italy; (M.R.); (F.C.)
- Department of Surgical and Medical Science and Translational Medicine, Sapienza University of Rome, 00189 Rome, Italy;
| | - Flaminia Coluzzi
- Unit of Anaesthesia, Intensive Care and Pain Medicine, Sant’Andrea University Hospital, 00189 Rome, Italy; (M.R.); (F.C.)
- Department Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy
| |
Collapse
|
9
|
Bahmani M, Mehrtabar S, Jafarizadeh A, Zoghi S, Heravi FS, Abbasi A, Sanaie S, Rahnemayan S, Leylabadlo HE. The Gut Microbiota and Major Depressive Disorder: Current Understanding and Novel Therapeutic Strategies. Curr Pharm Biotechnol 2024; 25:2089-2107. [PMID: 38288791 DOI: 10.2174/0113892010281892240116081031] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 09/10/2024]
Abstract
Major depressive disorder (MDD) is a common neuropsychiatric challenge that primarily targets young females. MDD as a global disorder has a multifactorial etiology related to the environment and genetic background. A balanced gut microbiota is one of the most important environmental factors involved in human physiological health. The interaction of gut microbiota components and metabolic products with the hypothalamic-pituitary-adrenal system and immune mediators can reverse depression phenotypes in vulnerable individuals. Therefore, abnormalities in the quantitative and qualitative structure of the gut microbiota may lead to the progression of MDD. In this review, we have presented an overview of the bidirectional relationship between gut microbiota and MDD, and the effect of pre-treatments and microbiomebased approaches, such as probiotics, prebiotics, synbiotics, fecal microbiota transplantation, and a new generation of microbial alternatives, on the improvement of unstable clinical conditions caused by MDD.
Collapse
Affiliation(s)
- Mohaddeseh Bahmani
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba Mehrtabar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Jafarizadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sevda Zoghi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Amin Abbasi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sarvin Sanaie
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sama Rahnemayan
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
10
|
Guerrin CG, Prasad K, Vazquez-Matias DA, Zheng J, Franquesa-Mullerat M, Barazzuol L, Doorduin J, de Vries EF. Prenatal infection and adolescent social adversity affect microglia, synaptic density, and behavior in male rats. Neurobiol Stress 2023; 27:100580. [PMID: 37920548 PMCID: PMC10618826 DOI: 10.1016/j.ynstr.2023.100580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/27/2023] [Accepted: 10/12/2023] [Indexed: 11/04/2023] Open
Abstract
Maternal infection during pregnancy and childhood social trauma have been associated with neurodevelopmental and affective disorders, such as schizophrenia, autism spectrum disorders, bipolar disorder and depression. These disorders are characterized by changes in microglial cells, which play a notable role in synaptic pruning, and synaptic deficits. Here, we investigated the effect of prenatal infection and social adversity during adolescence - either alone or in combination - on behavior, microglia, and synaptic density. Male offspring of pregnant rats injected with poly I:C, mimicking prenatal infection, were exposed to repeated social defeat during adolescence. We found that maternal infection during pregnancy prevented the reduction in social behavior and increase in anxiety induced by social adversity during adolescence. Furthermore, maternal infection and social adversity, alone or in combination, induced hyperlocomotion in adulthood. Longitudinal in vivo imaging with [11C]PBR28 positron emission tomography revealed that prenatal infection alone and social adversity during adolescence alone induced a transient increase in translocator protein TSPO density, an indicator of glial reactivity, whereas their combination induced a long-lasting increase that remained until adulthood. Furthermore, only the combination of prenatal infection and social adversity during adolescence induced an increase in microglial cell density in the frontal cortex. Prenatal infection increased proinflammatory cytokine IL-1β protein levels in hippocampus and social adversity reduced anti-inflammatory cytokine IL-10 protein levels in hippocampus during adulthood. This reduction in IL-10 was prevented if rats were previously exposed to prenatal infection. Adult offspring exposed to prenatal infection or adolescent social adversity had a higher synaptic density in the frontal cortex, but not hippocampus, as evaluated by synaptophysin density. Interestingly, such an increase in synaptic density was not observed in rats exposed to the combination of prenatal infection and social adversity, perhaps due to the long-lasting increase in microglial density, which may lead to an increase in microglial synaptic pruning. These findings suggest that changes in microglia activity and cytokine release induced by prenatal infection and social adversity during adolescence may be related to a reduced synaptic pruning, resulting in a higher synaptic density and behavioral changes in adulthood.
Collapse
Affiliation(s)
- Cyprien G.J. Guerrin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Kavya Prasad
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Daniel A. Vazquez-Matias
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Jing Zheng
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Maria Franquesa-Mullerat
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Lara Barazzuol
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Erik F.J. de Vries
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| |
Collapse
|
11
|
Hing B, Mitchell SB, Eberle M, Filali Y, Hultman I, Matkovich M, Kasturirangan M, Wyche W, Jimenez A, Velamuri R, Johnson M, Srivastava S, Hultman R. Single Cell Transcriptome of Stress Vulnerability Network in mouse Prefrontal Cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.14.540705. [PMID: 37662266 PMCID: PMC10473598 DOI: 10.1101/2023.05.14.540705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Increased vulnerability to stress is a major risk factor for the manifestation of several mood disorders, including major depressive disorder (MDD). Despite the status of MDD as a significant donor to global disability, the complex integration of genetic and environmental factors that contribute to the behavioral display of such disorders has made a thorough understanding of related etiology elusive. Recent developments suggest that a brain-wide network approach is needed, taking into account the complex interplay of cell types spanning multiple brain regions. Single cell RNA-sequencing technologies can provide transcriptomic profiling at the single-cell level across heterogenous samples. Furthermore, we have previously used local field potential oscillations and machine learning to identify an electrical brain network that is indicative of a predisposed vulnerability state. Thus, this study combined single cell RNA-sequencing (scRNA-Seq) with electrical brain network measures of the stress-vulnerable state, providing a unique opportunity to access the relationship between stress network activity and transcriptomic changes within individual cell types. We found especially high numbers of differentially expressed genes between animals with high and low stress vulnerability brain network activity in astrocytes and glutamatergic neurons but we estimated that vulnerability network activity depends most on GABAergic neurons. High vulnerability network activity included upregulation of microglia and mitochondrial and metabolic pathways, while lower vulnerability involved synaptic regulation. Genes that were differentially regulated with vulnerability network activity significantly overlapped with genes identified as having significant SNPs by human GWAS for depression. Taken together, these data provide the gene expression architecture of a previously uncharacterized stress vulnerability brain state, enabling new understanding and intervention of predisposition to stress susceptibility.
Collapse
|
12
|
Favretti M, Iannuccelli C, Di Franco M. Pain Biomarkers in Fibromyalgia Syndrome: Current Understanding and Future Directions. Int J Mol Sci 2023; 24:10443. [PMID: 37445618 DOI: 10.3390/ijms241310443] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/16/2023] [Accepted: 06/18/2023] [Indexed: 07/15/2023] Open
Abstract
Fibromyalgia is a complex and heterogeneous clinical syndrome, mainly characterized by the presence of widespread pain, possibly associated with a variety of other symptoms. Fibromyalgia can have an extremely negative impact on the psychological, physical and social lives of people affected, sometimes causing patients to experience dramatically impaired quality of life. Nowadays, the diagnosis of fibromyalgia is still clinical, thus favoring diagnostic uncertainties and making its clear identification challenging to establish, especially in primary care centers. These difficulties lead patients to undergo innumerable clinical visits, investigations and specialist consultations, thus increasing their stress, frustration and even dissatisfaction. Unfortunately, research over the last 25 years regarding a specific biomarker for the diagnosis of fibromyalgia has been fruitless. The discovery of a reliable biomarker for fibromyalgia syndrome would be a critical step towards the early identification of this condition, not only reducing patient healthcare utilization and diagnostic test execution but also providing early intervention with guideline-based treatments. This narrative article reviews different metabolite alterations proposed as possible biomarkers for fibromyalgia, focusing on their associations with clinical evidence of pain, and highlights some new, promising areas of research in this context. Nevertheless, none of the analyzed metabolites emerge as sufficiently reliable to be validated as a diagnostic biomarker. Given the complexity of this syndrome, in the future, a panel of biomarkers, including subtype-specific biomarkers, could be considered as an interesting alternative research area.
Collapse
Affiliation(s)
- Martina Favretti
- Rheumatology Unit, Department of Internal Clinical, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Cristina Iannuccelli
- Rheumatology Unit, Department of Internal Clinical, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Manuela Di Franco
- Rheumatology Unit, Department of Internal Clinical, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
13
|
Gędek A, Szular Z, Antosik AZ, Mierzejewski P, Dominiak M. Celecoxib for Mood Disorders: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J Clin Med 2023; 12:jcm12103497. [PMID: 37240605 DOI: 10.3390/jcm12103497] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/30/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
The effects of celecoxib on a broad spectrum of mood disorders and on inflammatory parameters have not yet been comprehensively evaluated. The aim of this study was to systematically summarize the available knowledge on this topic. Data from both preclinical and clinical studies were analyzed, considering the efficacy and safety of celecoxib in the treatment of mood disorders, as well as the correlation of inflammatory parameters with the effect of celecoxib treatment. Forty-four studies were included. We found evidence supporting the antidepressant efficacy of celecoxib in a dose of 400 mg/day used for 6 weeks as an add-on treatment in major depression (SMD = -1.12 [95%Cl: -1.71,-0.52], p = 0.0002) and mania (SMD = -0.82 [95% CI:-1.62,-0.01], p = 0.05). The antidepressant efficacy of celecoxib in the above dosage used as sole treatment was also confirmed in depressed patients with somatic comorbidity (SMD = -1.35 [95% CI:-1.95,-0.75], p < 0.0001). We found no conclusive evidence for the effectiveness of celecoxib in bipolar depression. Celecoxib at a dose of 400 mg/d used for up to 12 weeks appeared to be a safe treatment in patients with mood disorders. Although an association between celecoxib response and inflammatory parameters has been found in preclinical studies, this has not been confirmed in clinical trials. Further studies are needed to evaluate the efficacy of celecoxib in bipolar depression, as well as long-term studies evaluating the safety and efficacy of celecoxib in recurrent mood disorders, studies involving treatment-resistant populations, and assessing the association of celecoxib treatment with inflammatory markers.
Collapse
Affiliation(s)
- Adam Gędek
- Department of Pharmacology, Institute of Psychiatry and Neurology, Sobieskiego 9, 02-957 Warsaw, Poland
- Praski Hospital, Aleja Solidarności 67, 03-401 Warsaw, Poland
| | - Zofia Szular
- Faculty of Medicine, Medical University of Warsaw, Żwirki i Wigury 61, 02-091 Warsaw, Poland
| | - Anna Z Antosik
- Department of Psychiatry, Faculty of Medicine, Collegium Medicum, Cardinal Wyszynski University in Warsaw, Woycickiego 1/3, 01-938 Warsaw, Poland
| | - Paweł Mierzejewski
- Department of Pharmacology, Institute of Psychiatry and Neurology, Sobieskiego 9, 02-957 Warsaw, Poland
| | - Monika Dominiak
- Department of Pharmacology, Institute of Psychiatry and Neurology, Sobieskiego 9, 02-957 Warsaw, Poland
| |
Collapse
|
14
|
Raval NR, Wetherill RR, Wiers CE, Dubroff JG, Hillmer AT. Positron Emission Tomography of Neuroimmune Responses in Humans: Insights and Intricacies. Semin Nucl Med 2023; 53:213-229. [PMID: 36270830 PMCID: PMC11261531 DOI: 10.1053/j.semnuclmed.2022.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 08/30/2022] [Indexed: 11/06/2022]
Abstract
The brain's immune system plays a critical role in responding to immune challenges and maintaining homeostasis. However, dysregulated neuroimmune function contributes to neurodegenerative disease and neuropsychiatric conditions. In vivo positron emission tomography (PET) imaging of the neuroimmune system has facilitated a greater understanding of its physiology and the pathology of some neuropsychiatric conditions. This review presents an in-depth look at PET findings from human neuroimmune function studies, highlighting their importance in current neuropsychiatric research. Although the majority of human PET studies feature radiotracers targeting the translocator protein 18 kDa (TSPO), this review also considers studies with other neuroimmune targets, including monoamine oxidase B, cyclooxygenase-1 and cyclooxygenase-2, nitric oxide synthase, and the purinergic P2X7 receptor. Promising new targets, such as colony-stimulating factor 1, Sphingosine-1-phosphate receptor 1, and the purinergic P2Y12 receptor, are also discussed. The significance of validating neuroimmune targets and understanding their function and expression is emphasized in this review to better identify and interpret PET results.
Collapse
Affiliation(s)
- Nakul R Raval
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT; Yale PET Center, Yale University, New Haven, CT
| | - Reagan R Wetherill
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Corinde E Wiers
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jacob G Dubroff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ansel T Hillmer
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT; Yale PET Center, Yale University, New Haven, CT; Department of Psychiatry, Yale University, New Haven, CT.
| |
Collapse
|
15
|
Liu ST, Lin SC, Chang JPC, Yang KJ, Chu CS, Yang CC, Liang CS, Sun CF, Wang SC, Satyanarayanan SK, Su KP. The Clinical Observation of Inflammation Theory for Depression: The Initiative of the Formosa Long COVID Multicenter Study (FOCuS). CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2023; 21:10-18. [PMID: 36700308 PMCID: PMC9889898 DOI: 10.9758/cpn.2023.21.1.10] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 01/27/2023]
Abstract
There is growing evidence that the coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is associated with increased risks of psychiatric sequelae. Depression, anxiety, cognitive impairments, sleep disturbance, and fatigue during and after the acute phase of COVID-19 are prevalent, long-lasting, and exerting negative consequences on well-being and imposing a huge burden on healthcare systems and society. This current review presented timely updates of clinical research findings, particularly focusing on the pathogenetic mechanisms underlying the neuropsychiatric sequelae, and identified potential key targets for developing effective treatment strategies for long COVID. In addition, we introduced the Formosa Long COVID Multicenter Study (FOCuS), which aims to apply the inflammation theory to the pathogenesis and the psychosocial and nutrition treatments of post-COVID depression and anxiety.
Collapse
Affiliation(s)
- Shu-Tsen Liu
- Division of Child and Adolescent Psychiatry & Division of Developmental and Behavioral Pediatrics, China Medical University Children’s Hospital, Taichung, Taiwan
| | - Sheng-Che Lin
- An-Nan Hospital, China Medical University, Tainan, Taiwan
| | - Jane Pei-Chen Chang
- Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan,Division of Child Psychiatry, Department of Psychiatry, China Medical University Hospital, Taichung, Taiwan,Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan
| | - Kai-Jie Yang
- Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan,Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan
| | - Che-Sheng Chu
- Department of Psychiatry, Kaohsiung Veterans General Hospital, Kaohsiung, Taipei, Taiwan
| | - Chia-Chun Yang
- Department of Psychiatry, Taoyuan Psychiatric Center, Taoyuan City, Taipei, Taiwan
| | - Chih-Sung Liang
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ching-Fang Sun
- Department of Psychiatry and Behavioral Medicine, Virginia Tech Carilion Clinic School of Medicine, Roanoke, VA, USA
| | - Shao-Cheng Wang
- Department of Psychiatry, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan,Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Kuan-Pin Su
- An-Nan Hospital, China Medical University, Tainan, Taiwan,Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan,Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan,Address for correspondence: Kuan-Pin Su China Medical University, No.2 Yuh-Der Road, North District, Taichung 404332, Taiwan , E-mail: , ORCID: https://orcid.org/0000-0002-4501-2502
| |
Collapse
|
16
|
Guerrin CG, Doorduin J, Prasad K, Vazquez-Matias DA, Barazzuol L, de Vries EF. Social adversity during juvenile age but not adulthood increases susceptibility to an immune challenge later in life. Neurobiol Stress 2023; 23:100526. [PMID: 36844420 PMCID: PMC9945751 DOI: 10.1016/j.ynstr.2023.100526] [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: 11/07/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
Adverse experiences in early life can increase mental vulnerability to immune challenges experienced later in life, which may induce the development of stress-related psychopathologies. Here, we investigated whether the combined effect of both events is higher if the first adverse experience occurs when the brain is still in development. Therefore, male Wistar rats were exposed to repeated social defeat (RSD, first hit) during juvenile age or adulthood and to an immune challenge consisting of a single injection of lipopolysaccharide (LPS, second hit) in adulthood. Control animals were not exposed to RSD, but only to the LPS challenge. Translocator protein density, a marker for reactive microglia, microglia cell density and plasma corticosterone levels were measured using in vivo [11C]PBR28 positron emission tomography, iba1 immunostaining, and corticosterone ELISA, respectively. Anhedonia, social behavior and anxiety were measured with the sucrose preference, social interaction, and open field tests, respectively. Rats exposed to RSD during juvenile age exhibited enhanced anhedonia and social interaction dysfunction after an immune challenge in adulthood. This enhanced susceptibility was not observed in rats exposed to RSD during adulthood. In addition, exposure to RSD synergistically increased microglia cell density and glial reactivity to the LPS challenge. This increase in microglia cell density and reactivity to the LPS challenge was more pronounced in rats exposed to RSD during juvenile age than in adulthood. Exposure to RSD alone in juvenile age or adulthood induced similar short-term anhedonia, a long-lasting increase in plasma corticosterone and microglial activity, but no change in anxiety and social behavior. Our findings indicate that exposure to social stress during juvenile age, but not adulthood, primes the immune system and increases the sensitivity to an immune challenge experienced later in life. This suggests that juvenile social stress can have more deleterious effects in the long term than similar stress in adulthood.
Collapse
Affiliation(s)
- Cyprien G.J. Guerrin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
- Corresponding author.
| | - Kavya Prasad
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Daniel A. Vazquez-Matias
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Lara Barazzuol
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Erik F.J. de Vries
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| |
Collapse
|
17
|
Royse SK, Lopresti BJ, Mathis CA, Tollefson S, Narendran R. Beyond monoamines: II. Novel applications for PET imaging in psychiatric disorders. J Neurochem 2023; 164:401-443. [PMID: 35716057 DOI: 10.1111/jnc.15657] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 11/27/2022]
Abstract
Early applications of positron emission tomography (PET) in psychiatry sought to identify derangements of cerebral blood flow and metabolism. The need for more specific neurochemical imaging probes was soon evident, and these probes initially targeted the sites of action of neuroleptic (dopamine D2 receptors) and psychoactive (serotonin receptors) drugs. For nearly 30 years, the centrality of monoamine dysfunction in psychiatric disorders drove the development of an armamentarium of monoaminergic PET radiopharmaceuticals and imaging methodologies. However, continued investments in monoamine-enhancing drug development realized only modest gains in efficacy and tolerability. As patent protection for many widely prescribed and profitable psychiatric drugs lapsed, drug development pipelines shifted away from monoamines in search of novel targets with the promises of improved efficacy, or abandoned altogether. Over this period, PET radiopharmaceutical development activities closely parallelled drug development priorities, resulting in the development of new PET imaging agents for non-monoamine targets. In part two of this review, we survey clinical research studies using the novel targets and radiotracers described in part one across major psychiatric application areas such as substance use disorders, anxiety disorders, eating disorders, personality disorders, mood disorders, and schizophrenia. Important limitations of the studies described are discussed, as well as key methodologic issues, challenges to the field, and the status of clinical trials seeking to exploit these targets for novel therapeutics.
Collapse
Affiliation(s)
- Sarah K Royse
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Brian J Lopresti
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Chester A Mathis
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Savannah Tollefson
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rajesh Narendran
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
18
|
Lopresti BJ, Royse SK, Mathis CA, Tollefson SA, Narendran R. Beyond monoamines: I. Novel targets and radiotracers for Positron emission tomography imaging in psychiatric disorders. J Neurochem 2023; 164:364-400. [PMID: 35536762 DOI: 10.1111/jnc.15615] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 10/18/2022]
Abstract
With the emergence of positron emission tomography (PET) in the late 1970s, psychiatry had access to a tool capable of non-invasive assessment of human brain function. Early applications in psychiatry focused on identifying characteristic brain blood flow and metabolic derangements using radiotracers such as [15 O]H2 O and [18 F]FDG. Despite the success of these techniques, it became apparent that more specific probes were needed to understand the neurochemical bases of psychiatric disorders. The first neurochemical PET imaging probes targeted sites of action of neuroleptic (dopamine D2 receptors) and psychoactive (serotonin receptors) drugs. Based on the centrality of monoamine dysfunction in psychiatric disorders and the measured success of monoamine-enhancing drugs in treating them, the next 30 years witnessed the development of an armamentarium of PET radiopharmaceuticals and imaging methodologies for studying monoamines. Continued development of monoamine-enhancing drugs over this time however was less successful, realizing only modest gains in efficacy and tolerability. As patent protection for many widely prescribed and profitable psychiatric drugs lapsed, drug development pipelines shifted away from monoamines in search of novel targets with the promises of improved efficacy, or abandoned altogether. Over this period, PET radiopharmaceutical development activities closely paralleled drug development priorities resulting in the development of new PET imaging agents for non-monoamine targets. Part one of this review will briefly survey novel PET imaging targets with relevance to the field of psychiatry, which include the metabotropic glutamate receptor type 5 (mGluR5), purinergic P2 X7 receptor, type 1 cannabinoid receptor (CB1 ), phosphodiesterase 10A (PDE10A), and describe radiotracers developed for these and other targets that have matured to human subject investigations. Current limitations of the targets and techniques will also be discussed.
Collapse
Affiliation(s)
- Brian J Lopresti
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sarah K Royse
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Chester A Mathis
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Savannah A Tollefson
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rajesh Narendran
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Departments of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
19
|
Blocking Two-Pore Domain Potassium Channel TREK-1 Inhibits the Activation of A1-Like Reactive Astrocyte Through the NF-κB Signaling Pathway in a Rat Model of Major Depressive Disorder. Neurochem Res 2023; 48:1737-1754. [PMID: 36670238 PMCID: PMC10119044 DOI: 10.1007/s11064-023-03857-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/15/2022] [Accepted: 01/06/2023] [Indexed: 01/22/2023]
Abstract
Major depressive disorder (MDD) refers to a widespread psychiatric disorder. Astrocytes play a pivotal role in regulating inflammation which is a well-acknowledged key component in depression pathogenesis. However, the effects of the neuroinflammation-inducing A1-like astrocytes on MDD are still unknown. TWIK-related K+ channel 1 (TREK-1) has been demonstrated to regulate the action of antidepressants. Nevertheless, its mechanisms and effects on A1-like astrocyte stimulation in MDD are not clear. Therefore, we conducted in vivo and in vitro experiments using TREK-1 specific inhibitor spadin. In vivo, rats were subjected to a 6-week chronic unpredictable mild stress (CUMS) followed by spadin treatment. Behavioral tests were employed to surveil depressive-like behaviors. Hippocampal proteomic analysis was carried out with the purpose of identifying differentially expressed proteins after CUMS and spadin treatments. In vitro, astrocyte-conditioned medium and spadin were used to treat rat astrocyte cell line. The activated microglia, inflammatory factors, A1 astrocyte markers, and activated nuclear factor kappa B (NF-κB) pathway were later analyzed using immunofluorescence, western blot, and RT-qPCR. Our findings indicated that blockage of TREK-1 reduced CUMS-induced depressive-like behavior in rats, inhibited the microglial stimulation, reduced inflammatory factor levels, and suppressed the activation of A1-like reactive astrocytes in the hippocampus. We also verified that the suppression of A1-like astrocytes by spadin necessitated the NF-κB pathway. According to the findings, blocking TREK-1 inhibited the activation of A1-like reactive astrocytes via the NF-κB signaling pathway in MDD. Our study preliminarily identifies a novel antidepressant mechanism of TREK-1 action and provides a therapeutic path for MDD.
Collapse
|
20
|
Guo B, Zhang M, Hao W, Wang Y, Zhang T, Liu C. Neuroinflammation mechanisms of neuromodulation therapies for anxiety and depression. Transl Psychiatry 2023; 13:5. [PMID: 36624089 PMCID: PMC9829236 DOI: 10.1038/s41398-022-02297-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023] Open
Abstract
Mood disorders are associated with elevated inflammation, and the reduction of symptoms after multiple treatments is often accompanied by pro-inflammation restoration. A variety of neuromodulation techniques that regulate regional brain activities have been used to treat refractory mood disorders. However, their efficacy varies from person to person and lack reliable indicator. This review summarizes clinical and animal studies on inflammation in neural circuits related to anxiety and depression and the evidence that neuromodulation therapies regulate neuroinflammation in the treatment of neurological diseases. Neuromodulation therapies, including transcranial magnetic stimulation (TMS), transcranial electrical stimulation (TES), electroconvulsive therapy (ECT), photobiomodulation (PBM), transcranial ultrasound stimulation (TUS), deep brain stimulation (DBS), and vagus nerve stimulation (VNS), all have been reported to attenuate neuroinflammation and reduce the release of pro-inflammatory factors, which may be one of the reasons for mood improvement. This review provides a better understanding of the effective mechanism of neuromodulation therapies and indicates that inflammatory biomarkers may serve as a reference for the assessment of pathological conditions and treatment options in anxiety and depression.
Collapse
Affiliation(s)
- Bingqi Guo
- grid.413259.80000 0004 0632 3337Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China ,grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Neuromodulation, Beijing, 100053 China
| | - Mengyao Zhang
- grid.413259.80000 0004 0632 3337Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China ,grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Neuromodulation, Beijing, 100053 China
| | - Wensi Hao
- grid.413259.80000 0004 0632 3337Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China ,grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Neuromodulation, Beijing, 100053 China
| | - Yuping Wang
- grid.413259.80000 0004 0632 3337Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China ,grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Neuromodulation, Beijing, 100053 China ,grid.24696.3f0000 0004 0369 153XInstitute of sleep and consciousness disorders, Center of Epilepsy, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069 China
| | - Tingting Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China. .,Beijing Key Laboratory of Neuromodulation, Beijing, 100053, China.
| | - Chunyan Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China. .,Beijing Key Laboratory of Neuromodulation, Beijing, 100053, China.
| |
Collapse
|
21
|
Ji N, Lei M, Chen Y, Tian S, Li C, Zhang B. How Oxidative Stress Induces Depression? ASN Neuro 2023; 15:17590914231181037. [PMID: 37331994 DOI: 10.1177/17590914231181037] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023] Open
Abstract
Depression increasingly affects a wide range and a large number of people worldwide, both physically and psychologically, which makes it a social problem requiring prompt attention and management. Accumulating clinical and animal studies have provided us with substantial insights of disease pathogenesis, especially central monoamine deficiency, which considerably promotes antidepressant research and clinical treatment. The first-line antidepressants mainly target the monoamine system, whose drawbacks mainly include slow action and treatment resistant. The novel antidepressant esketamine, targeting on central glutamatergic system, rapidly and robustly alleviates depression (including treatment-resistant depression), whose efficiency is shadowed by potential addictive and psychotomimetic side effects. Thus, exploring novel depression pathogenesis is necessary, for seeking more safe and effective therapeutic methods. Emerging evidence has revealed vital involvement of oxidative stress (OS) in depression, which inspires us to pursue antioxidant pathway for depression prevention and treatment. Fully uncovering the underlying mechanisms of OS-induced depression is the first step towards the avenue, thus we summarize and expound possible downstream pathways of OS, including mitochondrial impairment and related ATP deficiency, neuroinflammation, central glutamate excitotoxicity, brain-derived neurotrophic factor/tyrosine receptor kinase B dysfunction and serotonin deficiency, the microbiota-gut-brain axis disturbance and hypothalamic-pituitary-adrenocortical axis dysregulation. We also elaborate on the intricate interactions between the multiple aspects, and molecular mechanisms mediating the interplay. Through reviewing the related research progress in the field, we hope to depict an integral overview of how OS induces depression, in order to provide fresh ideas and novel targets for the final goal of efficient treatment of the disease.
Collapse
Affiliation(s)
- Na Ji
- The School of Public Health, Faculty of Basic Medical Sciences, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin Guangxi, China
| | - Mengzhu Lei
- The School of Public Health, Faculty of Basic Medical Sciences, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin Guangxi, China
| | - Yating Chen
- The School of Public Health, Faculty of Basic Medical Sciences, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin Guangxi, China
| | - Shaowen Tian
- The School of Public Health, Faculty of Basic Medical Sciences, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin Guangxi, China
| | - Chuanyu Li
- The School of Public Health, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin Guangxi, China
| | - Bo Zhang
- The School of Public Health, Faculty of Basic Medical Sciences, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin Guangxi, China
| |
Collapse
|
22
|
Acetylsalicylic Acid and Mood Disorders: A Systematic Review. Pharmaceuticals (Basel) 2022; 16:ph16010067. [PMID: 36678565 PMCID: PMC9861965 DOI: 10.3390/ph16010067] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/01/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
The effects of acetylsalicylic acid (ASA) on mood disorders (MD) and on inflammatory parameters in preclinical and clinical studies have not yet been comprehensively evaluated. The aim of this study was to systematically summarize the available knowledge on this topic according to PRISMA guidelines. Data from preclinical and clinical studies were analyzed, considering the safety and efficacy of ASA in the treatment of MD and the correlation of inflammatory parameters with the effect of ASA treatment. Twenty-one studies were included. Both preclinical and clinical studies found evidence indicating the safety and efficacy of low-dose ASA in the treatment of all types of affective episodes in MD. Observational studies have indicated a reduced risk of all types of affective episodes in chronic low-dose ASA users (HR 0.92, 95% CI: 0.88, 0.95, p < 0.0001). An association between ASA response and inflammatory parameters was found in preclinical studies, but this was not confirmed in clinical trials. Further long-term clinical trials evaluating the safety and efficacy of ASA in recurrent MD, as well as assessing the linkage of ASA treatment with inflammatory phenotype and cytokines, are required. There is also a need for preclinical studies to understand the exact mechanism of action of ASA in MD.
Collapse
|
23
|
Is depression the missing link between inflammatory mediators and cancer? Pharmacol Ther 2022; 240:108293. [PMID: 36216210 DOI: 10.1016/j.pharmthera.2022.108293] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/14/2022] [Accepted: 09/26/2022] [Indexed: 11/05/2022]
Abstract
Patients with cancer are at greater risk of developing depression in comparison to the general population and this is associated with serious adverse effects, such as poorer quality of life, worse prognosis and higher mortality. Although the relationship between depression and cancer is now well established, a common underlying pathophysiological mechanism between the two conditions is yet to be elucidated. Existing theories of depression, based on monoamine neurotransmitter system dysfunction, are insufficient as explanations of the disorder. Recent advances have implicated neuroinflammatory mechanisms in the etiology of depression and it has been demonstrated that inflammation at a peripheral level may be mirrored centrally in astrocytes and microglia serving to promote chronic levels of inflammation in the brain. Three major routes to depression in cancer in which proinflammatory mediators are implicated, seem likely. Activation of the kynurenine pathway involving cytokines, increases tryptophan catabolism, resulting in diminished levels of serotonin which is widely acknowledged as being the hallmark of depression. It also results in neurotoxic effects on brain regions thought to be involved in the evolution of major depression. Proinflammatory mediators also play a crucial role in impairing regulatory glucocorticoid mediated feedback of the hypothalamic-pituitary-adrenal axis, which is activated by stress and considered to be involved in both depression and cancer. The third route is via the glutamatergic pathway, whereby glutamate excitotoxicity may lead to depression associated with cancer. A better understanding of the mechanisms underlying these dysregulated and other newly emerging pathways may provide a rationale for therapeutic targeting, serving to improve the care of cancer patients.
Collapse
|
24
|
Lee HL, Jung KM, Fotio Y, Squire E, Palese F, Lin L, Torrens A, Ahmed F, Mabou Tagne A, Ramirez J, Su S, Wong CR, Jung DH, Scarfone VM, Nguyen PU, Wood M, Green K, Piomelli D. Frequent Low-Dose Δ 9-Tetrahydrocannabinol in Adolescence Disrupts Microglia Homeostasis and Disables Responses to Microbial Infection and Social Stress in Young Adulthood. Biol Psychiatry 2022; 92:845-860. [PMID: 35750512 PMCID: PMC10629396 DOI: 10.1016/j.biopsych.2022.04.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/16/2022] [Accepted: 04/20/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND During adolescence, microglia are actively involved in neocortical maturation while concomitantly undergoing profound phenotypic changes. Because the teenage years are also a time of experimentation with cannabis, we evaluated whether adolescent exposure to the drug's psychotropic constituent, Δ9-tetrahydrocannabinol (THC), might persistently alter microglia function. METHODS We administered THC (5 mg/kg, intraperitoneal) once daily to male and female mice from postnatal day (PND) 30 to PND44 and examined the transcriptome of purified microglia in adult animals (PND70 and PND120) under baseline conditions or following either of two interventions known to recruit microglia: lipopolysaccharide injection and repeated social defeat. We used high-dimensional mass cytometry by time-of-flight to map brain immune cell populations after lipopolysaccharide challenge. RESULTS Adolescent THC exposure produced in mice of both sexes a state of microglial dyshomeostasis that persisted until young adulthood (PND70) but receded with further aging (PND120). Key features of this state included broad alterations in genes involved in microglia homeostasis and innate immunity along with marked impairments in the responses to lipopolysaccharide- and repeated social defeat-induced psychosocial stress. The endocannabinoid system was also dysfunctional. The effects of THC were prevented by coadministration of either a global CB1 receptor inverse agonist or a peripheral CB1 neutral antagonist and were not replicated when THC was administered in young adulthood (PND70-84). CONCLUSIONS Daily low-intensity CB1 receptor activation by THC during adolescence may disable critical functions served by microglia until young adulthood with potentially wide-ranging consequences for brain and mental health.
Collapse
Affiliation(s)
- Hye-Lim Lee
- Departments of Anatomy and Neurobiology, University of California Irvine, Irvine, California
| | - Kwang-Mook Jung
- Departments of Anatomy and Neurobiology, University of California Irvine, Irvine, California
| | - Yannick Fotio
- Departments of Anatomy and Neurobiology, University of California Irvine, Irvine, California
| | - Erica Squire
- Departments of Anatomy and Neurobiology, University of California Irvine, Irvine, California
| | - Francesca Palese
- Departments of Anatomy and Neurobiology, University of California Irvine, Irvine, California
| | - Lin Lin
- Departments of Anatomy and Neurobiology, University of California Irvine, Irvine, California
| | - Alexa Torrens
- Departments of Anatomy and Neurobiology, University of California Irvine, Irvine, California
| | - Faizy Ahmed
- Departments of Anatomy and Neurobiology, University of California Irvine, Irvine, California
| | - Alex Mabou Tagne
- Departments of Anatomy and Neurobiology, University of California Irvine, Irvine, California
| | - Jade Ramirez
- Departments of Anatomy and Neurobiology, University of California Irvine, Irvine, California
| | - Shiqi Su
- Departments of Anatomy and Neurobiology, University of California Irvine, Irvine, California
| | - Christina Renee Wong
- Departments of Anatomy and Neurobiology, University of California Irvine, Irvine, California
| | - Daniel Hojin Jung
- Departments of Anatomy and Neurobiology, University of California Irvine, Irvine, California
| | - Vanessa M Scarfone
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, California
| | - Pauline U Nguyen
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, California
| | - Marcelo Wood
- Neurobiology and Behavior, University of California Irvine, Irvine, California
| | - Kim Green
- Neurobiology and Behavior, University of California Irvine, Irvine, California
| | - Daniele Piomelli
- Departments of Anatomy and Neurobiology, University of California Irvine, Irvine, California; Biological Chemistry, University of California Irvine, Irvine, California; Pharmaceutical Sciences, University of California Irvine, Irvine, California.
| |
Collapse
|
25
|
Hodges TE, Lieblich SE, Rechlin RK, Galea LAM. Sex differences in inflammation in the hippocampus and amygdala across the lifespan in rats: associations with cognitive bias. Immun Ageing 2022; 19:43. [PMID: 36203171 PMCID: PMC9535862 DOI: 10.1186/s12979-022-00299-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/19/2022] [Indexed: 11/05/2022]
Abstract
Background Cognitive symptoms of major depressive disorder, such as negative cognitive bias, are more prevalent in women than in men. Cognitive bias involves pattern separation which requires hippocampal neurogenesis and is modulated by inflammation in the brain. Previously, we found sex differences in the activation of the amygdala and the hippocampus in response to negative cognitive bias in rats that varied with age. Given the association of cognitive bias to neurogenesis and inflammation, we examined associations between cognitive bias, neurogenesis in the hippocampus, and cytokine and chemokine levels in the ventral hippocampus (HPC) and basolateral amygdala (BLA) of male and female rats across the lifespan. Results After cognitive bias testing, males had more IFN-γ, IL-1β, IL-4, IL-5, and IL-10 in the ventral HPC than females in adolescence. In young adulthood, females had more IFN-γ, IL-1β, IL-6, and IL-10 in the BLA than males. Middle-aged rats had more IL-13, TNF-α, and CXCL1 in both regions than younger groups. Adolescent male rats had higher hippocampal neurogenesis than adolescent females after cognitive bias testing and young rats that underwent cognitive bias testing had higher levels of hippocampal neurogenesis than controls. Neurogenesis in the dorsal hippocampus was negatively associated with negative cognitive bias in young adult males. Conclusions Overall, the association between negative cognitive bias, hippocampal neurogenesis, and inflammation in the brain differs by age and sex. Hippocampal neurogenesis and inflammation may play greater role in the cognitive bias of young males compared to a greater role of BLA inflammation in adult females. These findings lay the groundwork for the discovery of sex-specific novel therapeutics that target region-specific inflammation in the brain and hippocampal neurogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s12979-022-00299-4. • Adolescent male rats had more hippocampal inflammation than females after cognitive bias testing. • Adult female rats had more basolateral amygdalar inflammation than males after cognitive bias testing. • HPC neurogenesis was negatively associated to cognitive bias in young adult male rats.
Collapse
Affiliation(s)
- Travis E. Hodges
- grid.17091.3e0000 0001 2288 9830Department of Psychology, University of British Columbia, Vancouver, Canada
| | - Stephanie E. Lieblich
- grid.17091.3e0000 0001 2288 9830Department of Psychology, University of British Columbia, Vancouver, Canada ,grid.17091.3e0000 0001 2288 9830Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Rebecca K. Rechlin
- grid.17091.3e0000 0001 2288 9830Department of Psychology, University of British Columbia, Vancouver, Canada ,grid.17091.3e0000 0001 2288 9830Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Liisa A. M. Galea
- grid.17091.3e0000 0001 2288 9830Department of Psychology, University of British Columbia, Vancouver, Canada ,grid.17091.3e0000 0001 2288 9830Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada ,grid.17091.3e0000 0001 2288 9830Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
| |
Collapse
|
26
|
Eggerstorfer B, Kim JH, Cumming P, Lanzenberger R, Gryglewski G. Meta-analysis of molecular imaging of translocator protein in major depression. Front Mol Neurosci 2022; 15:981442. [PMID: 36226319 PMCID: PMC9549359 DOI: 10.3389/fnmol.2022.981442] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
Molecular neuroimaging studies provide mounting evidence that neuroinflammation plays a contributory role in the pathogenesis of major depressive disorder (MDD). This has been the focus of a number of positron emission tomography (PET) studies of the 17-kDa translocator protein (TSPO), which is expressed by microglia and serves as a marker of neuroinflammation. In this meta-analysis, we compiled and analyzed all available molecular imaging studies comparing cerebral TSPO binding in MDD patients with healthy controls. Our systematic literature search yielded eight PET studies encompassing 238 MDD patients and 164 healthy subjects. The meta-analysis revealed relatively increased TSPO binding in several cortical regions (anterior cingulate cortex: Hedges' g = 0.6, 95% CI: 0.36, 0.84; hippocampus: g = 0.54, 95% CI: 0.26, 0.81; insula: g = 0.43, 95% CI: 0.17, 0.69; prefrontal cortex: g = 0.36, 95% CI: 0.14, 0.59; temporal cortex: g = 0.39, 95% CI: -0.04, 0.81). While the high range of effect size in the temporal cortex might reflect group-differences in body mass index (BMI), exploratory analyses failed to reveal any relationship between elevated TSPO availability in the other four brain regions and depression severity, age, BMI, radioligand, or the binding endpoint used, or with treatment status at the time of scanning. Taken together, this meta-analysis indicates a widespread ∼18% increase of TSPO availability in the brain of MDD patients, with effect sizes comparable to those in earlier molecular imaging studies of serotonin transporter availability and monoamine oxidase A binding.
Collapse
Affiliation(s)
- Benjamin Eggerstorfer
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Jong-Hoon Kim
- Department of Psychiatry, Gachon University College of Medicine, Gil Medical Center, Neuroscience Research Institute, GAIHST, Gachon University, Incheon, South Korea
| | - Paul Cumming
- Department of Nuclear Medicine, Inselspital, Bern University, Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD, Australia
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Gregor Gryglewski
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| |
Collapse
|
27
|
Hansen N. Immunopsychiatry – Innovative Technology to Characterize Disease Activity in Autoantibody-Associated Psychiatric Diseases. Front Immunol 2022; 13:867229. [PMID: 35711412 PMCID: PMC9197207 DOI: 10.3389/fimmu.2022.867229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/19/2022] [Indexed: 01/04/2023] Open
Abstract
Background Anti-neural autoantibody-associated psychiatric disease is a novel field in immunopsychiatry that has been attracting attention thanks to its potentially positive therapeutic outcome and distinct prognosis compared with non-organic psychiatric disease. This review aims to describe recent novel technological developments for improving diagnostics in the field of autoantibody-related psychiatric disease.MethodsWe screened for relevant articles in PubMed for this narrative article. We focused on research methods such as neuroimaging, immune cells and inflammation markers, and molecular biomarkers in human biofluids like serum and cerebrospinal fluid and plasma proteomics.ResultsWe introduce several novel methods for investigating autoinflammation with the aim of optimizing therapies for autoantibody-associated psychiatric disease. We describe measuring the translocator protein 18kDa in activated microglia via positron emission tomography imaging, brain volumetric assessment, flow cell cytometry of cerebrospinal fluid and blood, and blood biological probes as well as psychopathological cues to help us gain insights into diagnosing inflammation and brain damage better in psychiatric patients presenting a suspected autoimmune etiology.ConclusionOur short methodological review provides an overview of recent developments in the field of autoantibody-related immunopsychiatry. More research is needed to prove their usefulness in diagnosing and treating autoantibody-associated psychiatric disease and its subtypes.
Collapse
Affiliation(s)
- Niels Hansen
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- Translational Psychoneuroscience, University Medical Center Göttingen, Göttingen, Germany
- *Correspondence: Niels Hansen,
| |
Collapse
|
28
|
Kunugi H, Tikhonova M. Recent advances in understanding depressive disorder: Possible relevance to brain stimulation therapies. PROGRESS IN BRAIN RESEARCH 2022; 270:123-147. [PMID: 35396024 DOI: 10.1016/bs.pbr.2022.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Recent research has provided novel insights into the major depressive disorder (MDD) and identified certain biomarkers of this disease. There are four main mechanisms playing a key role in the related pathophysiology, namely (1) monoamine systems dysfunction, (2) stress response, (3) neuroinflammation, and (4) neurotrophic factors alteration. Robust evidence on the decreased homovanillic acid in the cerebrospinal fluid (CSF) of patients with MDD supports a rationale for therapeutic stimulation of the medial forebrain bundle activating the dopamine reward system. Both activation and suppression of the hypothalamic-pituitary-adrenal (HPA) axis in MDD and related conditions indicate usefulness of its evaluation for the disease subtyping. Elevated proinflammatory cytokines (specifically, interleukin-6) in CSF imply the role of neuroinflammation resulting in activation of the tryptophan-kynurenine pathway. Finally, neuroplasticity and trophic effects of the brain-derived neurotrophic factor (BDNF) may be related to both structural abnormalities of the brain in MDD and the underlying mechanisms of various therapies. In addition, the gut-brain interaction is pivotal, since lack of beneficial microbes confer the risk of MDD through negative effects on the dopamine system, HPA axis, and vagal nerve. All these factors may be highly relevant to treatment of MDD with contemporary brain stimulation therapies.
Collapse
Affiliation(s)
- Hiroshi Kunugi
- Department of Psychiatry, Teikyo University School of Medicine, Tokyo, Japan; Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.
| | - Maria Tikhonova
- Laboratory of the Experimental Models of Neurodegenerative Processes, Department of Experimental Neuroscience, Scientific Research Institute of Neurosciences and Medicine (SRINM), Novosibirsk, Russian Federation
| |
Collapse
|
29
|
Fluoxetine inhibited the activation of A1 reactive astrocyte in a mouse model of major depressive disorder through astrocytic 5-HT2BR/β-arrestin2 pathway. J Neuroinflammation 2022; 19:23. [PMID: 35093099 PMCID: PMC8800238 DOI: 10.1186/s12974-022-02389-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 01/17/2022] [Indexed: 02/08/2023] Open
Abstract
Abstract
Background
Fluoxetine, a selective serotonin reuptake inhibitor, has been reported to directly bind with 5-HT2B receptor (5-HT2BR), but the precise mechanisms, whereby fluoxetine confers the anti-depressive actions via 5-HT2BR is not fully understood. Although neuroinflammation-induced A1 astrocytes are involved in neurodegenerative diseases, the role of A1 astrocyte in the pathogenesis and treatment of major depressive disorder (MDD) remains unclear.
Methods
Mice were subjected to chronic mild stress (CMS) for 6 weeks and subsequently treated with fluoxetine for 4 weeks. The depressive-like and anxiety-like behaviors and the activation of A1 reactive astrocyte in hippocampus and cortex of mice were measured. Primary astrocytes were stimulated with A1 cocktail (tumor necrosis factor (TNF)-α, interleukin (IL)-1α and C1q), activated (LPS) microglia-conditioned medium (MCM) or IL-6 for 24 h and the expression of A1-special and A2-special markers were determined using RT-qPCR and western blot. The role of 5-HT2BR in the effects of fluoxetine on A1 reactive astrocyte was measured using 5-HT2BR inhibitor and siRNA in vitro and AAVs in vivo. The functions of downstream signaling Gq protein and β-arrestins in the effects of fluoxetine on the activation of A1 astrocyte were determined using pharmacological inhibitor and genetic knockout, respectively.
Results
In this study, we found that fluoxetine inhibited the activation of A1 reactive astrocyte and reduced the abnormal behaviors in CMS mice, as well as ameliorated A1 astrocyte reactivity under three different stimulators in primary astrocytes. We also showed that astrocytic 5-HT2BR was required in the inhibitory effects of fluoxetine on A1 reactive astrocyte in MDD in vivo and in vitro. We further found that the functions of fluoxetine in the activation of A1 astrocyte were independent of either Gq protein or β-arrestin1 in vitro. β-arrestin2 pathway was the downstream signaling of astrocytic 5-HT2BR mediated the inhibitory effects of fluoxetine on A1 astrocyte reactivity in primary astrocytes and CMS mice, as well as the improved roles of fluoxetine in behavioral impairments of CMS mice.
Conclusions
These data demonstrate that fluoxetine restricts reactive A1 astrocyte via astrocytic 5-HT2BR/β-arrestin2 pathway in a mouse model of MDD and provide a novel therapeutic avenue for MDD.
Collapse
|
30
|
Han KM, Ham BJ. How Inflammation Affects the Brain in Depression: A Review of Functional and Structural MRI Studies. J Clin Neurol 2021; 17:503-515. [PMID: 34595858 PMCID: PMC8490908 DOI: 10.3988/jcn.2021.17.4.503] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/30/2021] [Accepted: 08/30/2021] [Indexed: 12/27/2022] Open
Abstract
This narrative review discusses how peripheral and central inflammation processes affect brain function and structure in depression, and reports on recent peripheral inflammatory marker-based functional and structural magnetic resonance imaging (MRI) studies from the perspective of neural-circuit dysfunction in depression. Chronic stress stimulates the activity of microglial cells, which increases the production of pro-inflammatory cytokines in the brain. In addition, microglial activation promotes a shift from the synthesis of serotonin to the synthesis of neurotoxic metabolites of the kynurenine pathway, which induces glutamate-mediated excitotoxicity in neurons. Furthermore, the region specificity of microglial activation is hypothesized to contribute to the vulnerability of specific brain regions in the depression-related neural circuits to inflammation-mediated brain injury. MRI studies are increasingly investigating how the blood levels of inflammatory markers such as C-reactive protein, interleukin (IL)-1β, IL-6, and tumor necrosis factor-α are associated with functional and structural neuroimaging markers in depression. Functional MRI studies have found that peripheral inflammatory markers are associated with aberrant activation patterns and altered functional connectivity in neural circuits involved in emotion regulation, reward processing, and cognitive control in depression. Structural MRI studies have suggested that peripheral inflammatory markers are related to reduced cortical gray matter and subcortical volumes, cortical thinning, and decreased integrity of white matter tracts within depression-related neural circuits. These neuroimaging findings may improve our understanding of the relationships between neuroinflammatory processes at the molecular level and macroscale in vivo neuralcircuit dysfunction in depression.
Collapse
Affiliation(s)
- Kyu Man Han
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Byung Joo Ham
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea.
| |
Collapse
|